1
|
Tsakiroglou M, Evans A, Doce-Carracedo A, Little M, Hornby R, Roberts P, Zhang E, Miyajima F, Pirmohamed M. Gene Expression Dysregulation in Whole Blood of Patients with Clostridioides difficile Infection. Int J Mol Sci 2024; 25:12653. [PMID: 39684365 DOI: 10.3390/ijms252312653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Clostridioides difficile (C. difficile) is a global threat and has significant implications for individuals and health care systems. Little is known about host molecular mechanisms and transcriptional changes in peripheral immune cells. This is the first gene expression study in whole blood from patients with C. difficile infection. We took blood and stool samples from patients with toxigenic C. difficile infection (CDI), non-toxigenic C. difficile infection (GDH), inflammatory bowel disease (IBD), diarrhea from other causes (DC), and healthy controls (HC). We performed transcriptome-wide RNA profiling on peripheral blood to identify diarrhea common and CDI unique gene sets. Diarrhea groups upregulated innate immune responses with neutrophils at the epicenter. The common signature associated with diarrhea was non-specific and shared by various other inflammatory conditions. CDI had a unique 45 gene set reflecting the downregulation of humoral and T cell memory functions. Dysregulation of immunometabolic genes was also abundant and linked to immune cell fate during differentiation. Whole transcriptome analysis of white cells in blood from patients with toxigenic C. difficile infection showed that there is an impairment of adaptive immunity and immunometabolism.
Collapse
Affiliation(s)
- Maria Tsakiroglou
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Anthony Evans
- Computational Biology Facility, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Alejandra Doce-Carracedo
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Clinical Directorate, GCP Laboratories, University of Liverpool, Liverpool L7 8TX, UK
| | - Margaret Little
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Rachel Hornby
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Paul Roberts
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Faculty of Science and Engineering, School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LZ, UK
| | - Eunice Zhang
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Fabio Miyajima
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Oswaldo Cruz Foundation (Fiocruz), Branch Ceara, Eusebio 61773-270, Brazil
| | - Munir Pirmohamed
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| |
Collapse
|
2
|
Hu J, Zou H, Qiao X, Wang Y, Lv M, Zhang K, Wang F. The relationship between oxidative balance scores and chronic diarrhea and constipation: a population-based study. BMC Public Health 2024; 24:1366. [PMID: 38773415 PMCID: PMC11106991 DOI: 10.1186/s12889-024-18683-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 04/22/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Oxidative stress is closely related to gut health. Exposures to oxidative stress in one's diet and lifestyle can be evaluated by the oxidative balance score (OBS). However, the relationship between OBS and intestinal habits is unknown. This study aimed to investigate the relationships between OBS and intestinal habits (chronic diarrhea and chronic constipation) and the underlying mechanisms involved. METHODS Using data from the National Health and Nutrition Examination Survey (NHANES) database from 2005 to 2010, we included a total of 8065 participants. Twenty dietary and lifestyle factors were selected for the OBS calculates. Chronic constipation and chronic diarrhea were defined using the Bristol stool form scale (BSFS) types 1 and 2 and the BSFS 6 and 7, respectively. Multivariate logistic regression, subgroup analysis, and restricted cubic splines (RCS) analysis were used to evaluate the relationship between OBS and defecation habits. Finally, we used mediation analysis to explore the indirect effects of oxidative stress and inflammatory markers on these associations. RESULTS After adjusting for all the covariates, multivariate logistic regression analysis revealed that OBS was negatively correlated with diarrhea (OR = 0.57; 95%CI = 0.39-0.83; P = 0.008)and positively correlated with constipation (OR = 1.75; 95%CI = 1.19-2.25; P = 0.008). The RCS showed a nonlinear relationship between OBS and diarrhea (P for nonlinearity = 0.02) and a linear relationship between OBS and constipation (P for nonlinearity = 0.19). Mediation analysis showed that the C-reactive protein (CRP) concentration and white blood cell (WBC) count mediated the correlation between OBS and diarrhea by 6.28% and 6.53%, respectively (P < 0.05). CONCLUSIONS OBS is closely related to changes in patients' defecation habits. Oxidative stress and inflammation may play a role in the relationship between the two. This result emphasizes the importance of the public adjusting their lifestyle and dietary habits according to their own situation. However, further prospective studies are needed to analyze the relationship between oxidative stress and changes in defecation habits.
Collapse
Affiliation(s)
- Jiayan Hu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hede Zou
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Xiyun Qiao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxi Wang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Mi Lv
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Kunli Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengyun Wang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
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
|
4
|
Yadav M, Chauhan NS. Role of gut-microbiota in disease severity and clinical outcomes. Brief Funct Genomics 2024; 23:24-37. [PMID: 36281758 DOI: 10.1093/bfgp/elac037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/05/2022] [Accepted: 09/28/2022] [Indexed: 01/21/2024] Open
Abstract
A delicate balance of nutrients, antigens, metabolites and xenobiotics in body fluids, primarily managed by diet and host metabolism, governs human health. Human gut microbiota is a gatekeeper to nutrient bioavailability, pathogens exposure and xenobiotic metabolism. Human gut microbiota starts establishing during birth and evolves into a resilient structure by adolescence. It supplements the host's metabolic machinery and assists in many physiological processes to ensure health. Biotic and abiotic stressors could induce dysbiosis in gut microbiota composition leading to disease manifestations. Despite tremendous scientific advancements, a clear understanding of the involvement of gut microbiota dysbiosis during disease onset and clinical outcomes is still awaited. This would be important for developing an effective and sustainable therapeutic intervention. This review synthesizes the present scientific knowledge to present a comprehensive picture of the role of gut microbiota in the onset and severity of a disease.
Collapse
Affiliation(s)
- Monika Yadav
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
5
|
Fettucciari K, Dini F, Marconi P, Bassotti G. Role of the Alteration in Calcium Homeostasis in Cell Death Induced by Clostridioides difficile Toxin A and Toxin B. BIOLOGY 2023; 12:1117. [PMID: 37627001 PMCID: PMC10452684 DOI: 10.3390/biology12081117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023]
Abstract
Clostridioides difficile (C. difficile), responsible for 15-25% of gastrointestinal infections, causes health problems mainly due to the toxic activity of toxins A and B (Tcds). These are responsible for its clinical manifestations, including diarrhea, pseudomembranous colitis, toxic megacolon and death, with a mortality of 5-30% in primary infection, that increase following relapses. Studies on Tcd-induced cell death have highlighted a key role of caspases, calpains, and cathepsins, with involvement of mitochondria and reactive oxygen species (ROS) in a complex signaling pathway network. The complex response in the execution of various types of cell death (apoptosis, necrosis, pyroptosis and pyknosis) depends on the amount of Tcd, cell types, and Tcd receptors involved, and could have as initial/precocious event the alterations in calcium homeostasis. The entities, peculiarities and cell types involved in these alterations will decide the signaling pathways activated and cell death type. Calcium homeostasis alterations can be caused by calcium influx through calcium channel activation, transient intracellular calcium oscillations, and leakage of calcium from intracellular stores. These increases in cytoplasmic calcium have important effects on all calcium-regulated molecules, which may play a direct role in several cell death types and/or activate other cell death effectors, such as caspases, calpains, ROS and proapoptotic Bcl-2 family members. Furthermore, some support for the possible role of the calcium homeostasis alteration in Tcd-induced cell death originates from the similarity with cytotoxic effects that cause pore-forming toxins, based mainly on calcium influx through plasma membrane pores.
Collapse
Affiliation(s)
- Katia Fettucciari
- Biosciences & Medical Embryology Section, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy;
| | - Fabrizio Dini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy;
| | - Pierfrancesco Marconi
- Biosciences & Medical Embryology Section, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy;
| | - Gabrio Bassotti
- Gastroenterology, Hepatology & Digestive Endoscopy Section, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy;
- Gastroenterology & Hepatology Unit, Santa Maria Della Misericordia Hospital, 06129 Perugia, Italy
| |
Collapse
|
6
|
Clostridium novyi’s Alpha-Toxin Changes Proteome and Phosphoproteome of HEp-2 Cells. Int J Mol Sci 2022; 23:ijms23179939. [PMID: 36077344 PMCID: PMC9456407 DOI: 10.3390/ijms23179939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
C. novyi type A produces the alpha-toxin (TcnA) that belongs to the large clostridial glucosylating toxins (LCGTs) and is able to modify small GTPases by N-acetylglucosamination on conserved threonine residues. In contrast, other LCGTs including Clostridioides difficile toxin A and toxin B (TcdA; TcdB) modify small GTPases by mono-o-glucosylation. Both modifications inactivate the GTPases and cause strong effects on GTPase-dependent signal transduction pathways and the consequent reorganization of the actin cytoskeleton leading to cell rounding and finally cell death. However, the effect of TcnA on target cells is largely unexplored. Therefore, we performed a comprehensive screening approach of TcnA treated HEp-2 cells and analyzed their proteome and their phosphoproteome using LC-MS-based methods. With this data-dependent acquisition (DDA) approach, 5086 proteins and 9427 phosphosites could be identified and quantified. Of these, 35 proteins were found to be significantly altered after toxin treatment, and 1832 phosphosites were responsive to TcnA treatment. By analyzing the TcnA-induced proteomic effects of HEp-2 cells, 23 common signaling pathways were identified to be altered, including Actin Cytoskeleton Signaling, Epithelial Adherens Junction Signaling, and Signaling by Rho Family GTPases. All these pathways are also regulated after application of TcdA or TcdB of C. difficile. After TcnA treatment the regulation on phosphorylation level was much stronger compared to the proteome level, in terms of both strength of regulation and the number of regulated phosphosites. Interestingly, various signaling pathways such as Signaling by Rho Family GTPases or Integrin Signaling were activated on proteome level while being inhibited on phosphorylation level or vice versa as observed for the Role of BRCA1 in DNA Damage Response. ZIP kinase, as well as Calmodulin-dependent protein kinases IV & II, were observed as activated while Aurora-A kinase and CDK kinases tended to be inhibited in cells treated with TcnA based on their substrate regulation pattern.
Collapse
|
7
|
Tweedie A, Nissan T. Hiding in Plain Sight: Formation and Function of Stress Granules During Microbial Infection of Mammalian Cells. Front Mol Biosci 2021; 8:647884. [PMID: 33996904 PMCID: PMC8116797 DOI: 10.3389/fmolb.2021.647884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/01/2021] [Indexed: 01/21/2023] Open
Abstract
Stress granule (SG) formation is a host cell response to stress-induced translational repression. SGs assemble with RNA-binding proteins and translationally silent mRNA. SGs have been demonstrated to be both inhibitory to viruses, as well as being subverted for viral roles. In contrast, the function of SGs during non-viral microbial infections remains largely unexplored. A handful of microbial infections have been shown to result in host SG assembly. Nevertheless, a large body of evidence suggests SG formation in hosts is a widespread response to microbial infection. Diverse stresses caused by microbes and their products can activate the integrated stress response in order to inhibit translation initiation through phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α). This translational response in other contexts results in SG assembly, suggesting that SG assembly can be a general phenomenon during microbial infection. This review explores evidence for host SG formation in response to bacterial, fungal, and protozoan infection and potential functions of SGs in the host and for adaptations of the pathogen.
Collapse
Affiliation(s)
- Alistair Tweedie
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Tracy Nissan
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
8
|
The role of the globular heads of the C1q receptor in TcdA-induced human colonic epithelial cell apoptosis via a mitochondria-dependent pathway. BMC Microbiol 2020; 20:274. [PMID: 32878596 PMCID: PMC7465811 DOI: 10.1186/s12866-020-01958-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 08/25/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Clostridioides (formerly Clostridium) difficile infection is the leading cause of antibiotic-associated colitis. Studies have demonstrated that C. difficile toxin A (TcdA) can cause apoptosis of many human cell types. The purpose of this study was to investigate the relationships among exposure to TcdA, the role of the receptor for the globular heads of C1q (gC1qR) gene and the underlying intracellular apoptotic mechanism in human colonic epithelial cells (NCM 460). In this study, gC1qR expression was examined using real-time polymerase chain reaction (PCR), western blotting and immunohistochemical staining. Cell viability was assessed by the water-soluble tetrazolium salt (WST-1) assay, and cell apoptosis was assessed by flow cytometry and the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assay. Mitochondrial function was assessed based on reactive oxygen species (ROS) generation, changes in the mitochondrial membrane potential (ΔΨm) and the content of ATP. RESULTS Our study demonstrated that increasing the concentration of TcdA from 10 ng/ml to 20 ng/ml inhibited cell viability and induced cell apoptosis (p < 0.01). Moreover, the TcdA-induced gC1qR expression and enhanced expression of gC1qR caused mitochondrial dysfunction (including production of ROS and decreases in the ΔΨm and the content of ATP) and cell apoptosis. However, silencing of the gC1qR gene reversed TcdA-induced cell apoptosis and mitochondrial dysfunction. CONCLUSION These data support a mechanism by which gC1qR plays a crucial role in TcdA-induced apoptosis of human colonic epithelial cells in a mitochondria-dependent manner.
Collapse
|
9
|
Probiotic Supplementation in a Clostridium difficile-Infected Gastrointestinal Model Is Associated with Restoring Metabolic Function of Microbiota. Microorganisms 2019; 8:microorganisms8010060. [PMID: 31905795 PMCID: PMC7023328 DOI: 10.3390/microorganisms8010060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/20/2019] [Accepted: 12/27/2019] [Indexed: 01/19/2023] Open
Abstract
Clostridium (C.) difficile-infection (CDI), a nosocomial gastrointestinal disorder, is of growing concern due to its rapid rise in recent years. Antibiotic therapy of CDI is associated with disrupted metabolic function and altered gut microbiota. The use of probiotics as an adjunct is being studied extensively due to their potential to modulate metabolic functions and the gut microbiota. In the present study, we assessed the ability of several single strain probiotics and a probiotic mixture to change the metabolic functions of normal and C. difficile-infected fecal samples. The production of short-chain fatty acids (SCFAs), hydrogen sulfide (H2S), and ammonia was measured, and changes in microbial composition were assessed by 16S rRNA gene amplicon sequencing. The C. difficile-infection in fecal samples resulted in a significant decrease (p < 0.05) in SCFA and H2S production, with a lower microbial alpha diversity. All probiotic treatments were associated with significantly increased (p < 0.05) levels of SCFAs and restored H2S levels. Probiotics showed no effect on microbial composition of either normal or C. difficile-infected fecal samples. These findings indicate that probiotics may be useful to improve the metabolic dysregulation associated with C. difficile infection.
Collapse
|
10
|
Oludiran A, Courson DS, Stuart MD, Radwan AR, Poutsma JC, Cotten ML, Purcell EB. How Oxygen Availability Affects the Antimicrobial Efficacy of Host Defense Peptides: Lessons Learned from Studying the Copper-Binding Peptides Piscidins 1 and 3. Int J Mol Sci 2019; 20:E5289. [PMID: 31653020 PMCID: PMC6862162 DOI: 10.3390/ijms20215289] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
The development of new therapeutic options against Clostridioides difficile (C. difficile) infection is a critical public health concern, as the causative bacterium is highly resistant to multiple classes of antibiotics. Antimicrobial host-defense peptides (HDPs) are highly effective at simultaneously modulating the immune system function and directly killing bacteria through membrane disruption and oxidative damage. The copper-binding HDPs piscidin 1 and piscidin 3 have previously shown potent antimicrobial activity against a number of Gram-negative and Gram-positive bacterial species but have never been investigated in an anaerobic environment. Synergy between piscidins and metal ions increases bacterial killing aerobically. Here, we performed growth inhibition and time-kill assays against C. difficile showing that both piscidins suppress proliferation of C. difficile by killing bacterial cells. Microscopy experiments show that the peptides accumulate at sites of membrane curvature. We find that both piscidins are effective against epidemic C. difficile strains that are highly resistant to other stresses. Notably, copper does not enhance piscidin activity against C. difficile. Thus, while antimicrobial activity of piscidin peptides is conserved in aerobic and anaerobic settings, the peptide-copper interaction depends on environmental oxygen to achieve its maximum potency. The development of pharmaceuticals from HDPs such as piscidin will necessitate consideration of oxygen levels in the targeted tissue.
Collapse
Affiliation(s)
- Adenrele Oludiran
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| | - David S Courson
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| | - Malia D Stuart
- Biology Department, Palomar College, San Marcos, CA 92069, USA.
| | - Anwar R Radwan
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23185, USA.
| | - John C Poutsma
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Myriam L Cotten
- Department of Applied Science, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Erin B Purcell
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
11
|
Dey A, Manna S, Adhikary J, Chattopadhyay S, De S, Chattopadhyay D, Roy S. Biodistribution and toxickinetic variances of chemical and green Copper oxide nanoparticles in vitro and in vivo. J Trace Elem Med Biol 2019; 55:154-169. [PMID: 31345354 DOI: 10.1016/j.jtemb.2019.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022]
Abstract
In this study, chemical (S1) and green (S2) Copper Oxide nanoparticles (NPs) were synthesized to determine their biodistribution and toxicokinetic variances in vitro and in vivo. Both NPs significantly released Copper ions (Cu) in lymphocytes and were primarily deposited in the mononuclear phagocyte system (MPS) such as the liver and spleen in mice. In particular, S2NPs seemed to be prominently stored in the spleen, whereas the S1NPs were widely stored in more organs including the liver, heart, lungs, kidney and intestine. The circulation in the blood and fecal excretions both showed higher S2NPs contents respectively. Measurements of cell viability, Hemolysis assay, Reactive Oxygen Species (ROS) generation, biochemical estimation and apoptotic or necrotic study in lymphocytes after 24 h and measurements of body and organ weight, serum chemistry evaluation, cytokines level, protein expressions and histopathology of Balb/C mice after 15 days indicated significant toxicity difference between the S1NPs and S2NPs. Our observations proved that the NPs physiochemical properties influence toxicity and Biodistribution profiles in vitro and in vivo.
Collapse
Affiliation(s)
- Aditi Dey
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community health, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Subhankar Manna
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community health, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Jaydeep Adhikary
- Department of Chemical Sciences, Ariel University, Ariel, 40700, Israel
| | - Sourav Chattopadhyay
- Molecular Genetics and Therapeutics Lab, Indian Institute of Technology, Kanpur, India
| | - Sriparna De
- Department of Polymer Science and Technology, USCTA, University of Calcutta, 92 A.P.C road, Kolkata, 700009, West Bengal, India
| | - Dipankar Chattopadhyay
- Department of Polymer Science and Technology, USCTA, University of Calcutta, 92 A.P.C road, Kolkata, 700009, West Bengal, India.
| | - Somenath Roy
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community health, Vidyasagar University, Midnapore, 721102, West Bengal, India.
| |
Collapse
|
12
|
Probiotic Supplementation is Associated with Increased Antioxidant Capacity and Copper Chelation in C. difficile-Infected Fecal Water. Nutrients 2019; 11:nu11092007. [PMID: 31454897 PMCID: PMC6769851 DOI: 10.3390/nu11092007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/19/2019] [Accepted: 08/23/2019] [Indexed: 12/23/2022] Open
Abstract
Probiotic supplementation plays a key role in maintaining intestinal homeostasis due to its ability to modulate gut microbiota. Although their potential as potent antioxidants have previously been explored, their ability to affect the redox status in the gut lumen of healthy subjects or those with gastrointestinal (GI) disorders remains unclear. In our study, we assessed the ability of single strain and multispecies probiotic supplementation to cause a change in the redox status of normal fecal water and in Clostridium (C.) difficile-infected fecal water using a simulated gastrointestinal model. Changes in redox status were assessed by ferric-reducing antioxidant power (FRAP), 2’,2’-diphenyl-1-picrylhydrazyl (DPPH), and iron and copper chelation assays. The findings from our study showed that in normal fecal water, probiotic supplements, apart from Lactobacillus (L.) rhamnosus R0011, showed a significant increase in iron chelation (p < 0.05), which was associated with lower FRAP and copper chelation. In C. difficile-infected fecal water, all probiotic supplements showed a significant increase in FRAP (p < 0.05) and were associated with increased copper chelation. The DPPH assay showed no treatment effect in either fecal water. These findings suggest that C. difficile mediates dysregulation of redox status, which is counteracted by probiotics through ferric-reducing ability and copper chelation.
Collapse
|
13
|
Junemann J, Just I, Gerhard R, Pich A. Quantitative Phosphoproteome Analysis of Clostridioides difficile Toxin B Treated Human Epithelial Cells. Front Microbiol 2018; 9:3083. [PMID: 30619164 PMCID: PMC6304397 DOI: 10.3389/fmicb.2018.03083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/29/2018] [Indexed: 01/02/2023] Open
Abstract
The large clostridial glucosylating toxin B (TcdB) is a major virulence factor of the nosocomial pathogen Clostridioides difficile. TcdB inhibits small GTPases by glucosylation leading to impaired downstream signaling. TcdB also possesses a glucosyltransferase independent effect described as pyknosis. To elucidate the impact of TcdB and its glucosylation-inactive mutant TcdBNXN on the kinome of human cells, SILAC labeled HEp-2 cells were treated with 2 nM TcdB for 8 h. Phosphopeptides were enriched using SCX chromatography, IMAC and TiO2 followed shotgun mass spectrometry analysis. Overall 4,197 phosphopeptides were identified; more than 1,200 phosphosites responded to treatment with TcdB or TcdBNXN. The data suggested that predominantly stress-activated MAPK-dependent signaling pathways were triggered by toxin B treatment.
Collapse
Affiliation(s)
| | - Ingo Just
- Hannover Medical School, Institute for Toxicology, Hanover, Germany
| | - Ralf Gerhard
- Hannover Medical School, Institute for Toxicology, Hanover, Germany
| | - Andreas Pich
- Hannover Medical School, Institute for Toxicology, Hanover, Germany
| |
Collapse
|
14
|
Mechanisms of Bacterial Tolerance and Persistence in the Gastrointestinal and Respiratory Environments. Clin Microbiol Rev 2018; 31:31/4/e00023-18. [PMID: 30068737 DOI: 10.1128/cmr.00023-18] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Pathogens that infect the gastrointestinal and respiratory tracts are subjected to intense pressure due to the environmental conditions of the surroundings. This pressure has led to the development of mechanisms of bacterial tolerance or persistence which enable microorganisms to survive in these locations. In this review, we analyze the general stress response (RpoS mediated), reactive oxygen species (ROS) tolerance, energy metabolism, drug efflux pumps, SOS response, quorum sensing (QS) bacterial communication, (p)ppGpp signaling, and toxin-antitoxin (TA) systems of pathogens, such as Escherichia coli, Salmonella spp., Vibrio spp., Helicobacter spp., Campylobacter jejuni, Enterococcus spp., Shigella spp., Yersinia spp., and Clostridium difficile, all of which inhabit the gastrointestinal tract. The following respiratory tract pathogens are also considered: Staphylococcus aureus, Pseudomonas aeruginosa, Acinetobacter baumannii, Burkholderia cenocepacia, and Mycobacterium tuberculosis Knowledge of the molecular mechanisms regulating the bacterial tolerance and persistence phenotypes is essential in the fight against multiresistant pathogens, as it will enable the identification of new targets for developing innovative anti-infective treatments.
Collapse
|
15
|
Beer LA, Tatge H, Reich N, Tenspolde M, Olling A, Goy S, Rottner K, Alekov AK, Gerhard R. Early cell death induced by Clostridium difficile TcdB: Uptake and Rac1-glucosylation kinetics are decisive for cell fate. Cell Microbiol 2018; 20:e12865. [PMID: 29904993 DOI: 10.1111/cmi.12865] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/23/2018] [Accepted: 05/26/2018] [Indexed: 12/29/2022]
Abstract
Toxin A and Toxin B (TcdA/TcdB) are large glucosyltransferases produced by Clostridium difficile. TcdB but not TcdA induces reactive oxygen species-mediated early cell death (ECD) when applied at high concentrations. We found that nonglucosylated Rac1 is essential for induction of ECD since inhibition of Rac1 impedes this effect. ECD only occurs when TcdB is rapidly endocytosed. This was shown by generation of chimeras using the trunk of TcdB from a hypervirulent strain. TcdB from hypervirulent strain has been described to translocate from endosomes at higher pH values and thus, meaning faster than reference type TcdB. Accordingly, intracellular delivery of the glucosyltransferase domain of reference TcdB by the trunk of TcdB from hypervirulent strain increased ECD. Furthermore, proton transporters such as sodium/proton exchanger (NHE) or the ClC-5 anion/proton exchanger, both of which contribute to endosomal acidification, also affected cytotoxic potency of TcdB: Specific inhibition of NHE reduced cytotoxicity, whereas transfection of cells with the endosomal anion/proton exchanger ClC-5 increased cytotoxicity of TcdB. Our data suggest that both the uptake rate of TcdB into the cytosol and the status of nonglucosylated Rac1 are key determinants that are decisive for whether ECD or delayed apoptosis is triggered.
Collapse
Affiliation(s)
| | - Helma Tatge
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Nicole Reich
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Michel Tenspolde
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Alexandra Olling
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Sebastian Goy
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Erdmann J, Junemann J, Schröder A, Just I, Gerhard R, Pich A. Glucosyltransferase-dependent and -independent effects of TcdB on the proteome of HEp-2 cells. Proteomics 2017; 17. [PMID: 28612519 DOI: 10.1002/pmic.201600435] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 12/16/2022]
Abstract
Toxin B (TcdB) of the nosocomial pathogen C. difficile has been reported to exhibit a glucosyltransferase-dependent and -independent effect on treated HEp-2 cells at toxin concentration above 0.3 nM. In order to investigate and further characterize both effects epithelial cells were treated with wild type TcdB and glucosyltransferase-deficient TcdBNXN and their proteomes were analyzed by LC-MS. Triplex SILAC labeling was used for quantification. Identification of 5212 and quantification of 4712 protein groups was achieved. Out of these 257 were affected by TcdB treatment, 92 by TcdBNXN treatment and 49 by both. TcdB mainly led to changes in proteins that are related to "GTPase mediated signaling" and the "cytoskeleton" while "chromatin" and "cell cycle" related proteins were altered by both, TcdB and TcdBNXN . The obtained dataset of HEp-2 cell proteome helps us to better understand glucosyltransferase-dependent and -independent mechanisms of TcdB and TcdBNXN , particularly those involved in pyknotic cell death. All proteomics data have been deposited in the ProteomeXchange with the dataset identifier PXD006658 (https://proteomecentral.proteomexchange.org/dataset/PXD006658).
Collapse
Affiliation(s)
- Jelena Erdmann
- Hannover Medical School, Institute of Toxicology, Hannover, Germany
| | | | - Anke Schröder
- Hannover Medical School, Institute of Toxicology, Hannover, Germany
| | - Ingo Just
- Hannover Medical School, Institute of Toxicology, Hannover, Germany
| | - Ralf Gerhard
- Hannover Medical School, Institute of Toxicology, Hannover, Germany
| | - Andreas Pich
- Hannover Medical School, Institute of Toxicology, Hannover, Germany
| |
Collapse
|
17
|
Enteric glial cells counteract Clostridium difficile Toxin B through a NADPH oxidase/ROS/JNK/caspase-3 axis, without involving mitochondrial pathways. Sci Rep 2017; 7:45569. [PMID: 28349972 PMCID: PMC5368562 DOI: 10.1038/srep45569] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023] Open
Abstract
Enteric glial cells (EGCs) are components of the intestinal epithelial barrier essential for regulating the enteric nervous system. Clostridium difficile is the most common cause of antibiotic-associated colitis, toxin B (TcdB) being the major virulence factor, due to its ability to breach the intestinal epithelial barrier and to act on other cell types. Here we investigated TcdB effects on EGCs and the activated molecular mechanisms. Already at 2 hours, TcdB triggered ROS formation originating from NADPH-oxidase, as demonstrated by their reduction in the presence of the NADPH-oxidase inhibitor ML171. Although EGCs mitochondria support almost completely the cellular ATP need, TcdB exerted weak effects on EGCs in terms of ATP and mitochondrial functionality, mitochondrial ROS production occurring as a late event. ROS activated the JNK signalling and overexpression of the proapoptotic Bim not followed by cytochrome c or AIF release to activate the downstream apoptotic cascade. EGCs underwent DNA fragmentation through activation of the ROS/JNK/caspase-3 axis, evidenced by the ability of ML171, N-acetylcysteine, and the JNK inhibitor SP600125 to inhibit caspase-3 or to contrast apoptosis. Therefore, TcdB aggressiveness towards EGCs is mainly restricted to the cytosolic compartment, which represents a peculiar feature, since TcdB primarily influences mitochondria in other cellular types.
Collapse
|
18
|
Gigli S, Seguella L, Pesce M, Bruzzese E, D'Alessandro A, Cuomo R, Steardo L, Sarnelli G, Esposito G. Cannabidiol restores intestinal barrier dysfunction and inhibits the apoptotic process induced by Clostridium difficile toxin A in Caco-2 cells. United European Gastroenterol J 2017; 5:1108-1115. [PMID: 29238589 DOI: 10.1177/2050640617698622] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/15/2017] [Indexed: 12/17/2022] Open
Abstract
Background Clostridium difficile toxin A is responsible for colonic damage observed in infected patients. Drugs able to restore Clostridium difficile toxin A-induced toxicity have the potential to improve the recovery of infected patients. Cannabidiol is a non-psychotropic component of Cannabis sativa, which has been demonstrated to protect enterocytes against chemical and/or inflammatory damage and to restore intestinal mucosa integrity. Objective The purpose of this study was to evaluate (a) the anti-apoptotic effect and (b) the mechanisms by which cannabidiol protects mucosal integrity in Caco-2 cells exposed to Clostridium difficile toxin A. Methods Caco-2 cells were exposed to Clostridium difficile toxin A (30 ng/ml), with or without cannabidiol (10-7-10-9 M), in the presence of the specific antagonist AM251 (10-7 M). Cytotoxicity assay, transepithelial electrical resistence measurements, immunofluorescence analysis and immunoblot analysis were performed in the different experimental conditions. Results Clostridium difficile toxin A significantly decreased Caco-2 cells' viability and reduced transepithelial electrical resistence values and RhoA guanosine triphosphate (GTP), bax, zonula occludens-1 and occludin protein expression, respectively. All these effects were significantly and concentration-dependently inhibited by cannabidiol, whose effects were completely abolished in the presence of the cannabinoid receptor type 1 (CB1) antagonist, AM251. Conclusions Cannabidiol improved Clostridium difficile toxin A-induced damage in Caco-2 cells, by inhibiting the apoptotic process and restoring the intestinal barrier integrity, through the involvement of the CB1 receptor.
Collapse
Affiliation(s)
- Stefano Gigli
- Department of Physiology and Pharmacology, La Sapienza University of Rome, Rome, Italy
| | - Luisa Seguella
- Department of Physiology and Pharmacology, La Sapienza University of Rome, Rome, Italy
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - Eugenia Bruzzese
- Department of Translational Medical Science, University of Naples 'Federico II', Naples, Italy
| | - Alessandra D'Alessandro
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - Rosario Cuomo
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - Luca Steardo
- Department of Physiology and Pharmacology, La Sapienza University of Rome, Rome, Italy
| | - Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - Giuseppe Esposito
- Department of Physiology and Pharmacology, La Sapienza University of Rome, Rome, Italy
| |
Collapse
|
19
|
Péchiné S, Collignon A. Immune responses induced by Clostridium difficile. Anaerobe 2016; 41:68-78. [PMID: 27108093 DOI: 10.1016/j.anaerobe.2016.04.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 02/06/2023]
Abstract
The spectrum of Clostridium difficile infections is highly variable, ranging from asymptomatic carriage to fatal colitis depending on the strain virulence and on the host, its gut microbiota and its immune response. After disruption of the gut microbiota, C. difficile pathogenesis can be divided into three steps: 1) contamination by spores and their germination; 2) multiplication of vegetative cells and intestinal colonization using colonization factors; 3) production of the toxins TcdA and TcdB, and for some strains, the binary toxin, which are responsible for the clinical signs. Three lines of defense counteract C. difficile. The first line is the epithelial barrier, which is breached by the toxins. Then, a rapid innate immune response follows, which forms the second line of defense. It provides very quick defense reactions against C. difficile but is non-specific and does not confer memory. C. difficile and its virulence factors, the toxins and colonization factors, induce a highly pro-inflammatory response, which can be either beneficial or harmful, but triggers the adaptive immunity as the third line of defense required to control the infectious process. Adaptive immunity provides a highly specific immune response against C. difficile with memory and long lasting immunity. The innate and adaptive immune responses against the toxins and surface components are analyzed as well as their role in disease susceptibility, severity and recurrences.
Collapse
Affiliation(s)
- Séverine Péchiné
- Faculté de pharmacie, EA 4043 "Unité Bactéries Pathogènes et Santé", Univ. Paris-Sud, Université Paris-Saclay, 5 Rue Jean Baptiste Clément, 92296 Châtenay-Malabry Cedex, France
| | - Anne Collignon
- Faculté de pharmacie, EA 4043 "Unité Bactéries Pathogènes et Santé", Univ. Paris-Sud, Université Paris-Saclay, 5 Rue Jean Baptiste Clément, 92296 Châtenay-Malabry Cedex, France.
| |
Collapse
|
20
|
Abt MC, McKenney PT, Pamer EG. Clostridium difficile colitis: pathogenesis and host defence. Nat Rev Microbiol 2016; 14:609-20. [PMID: 27573580 DOI: 10.1038/nrmicro.2016.108] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Clostridium difficile is a major cause of intestinal infection and diarrhoea in individuals following antibiotic treatment. Recent studies have begun to elucidate the mechanisms that induce spore formation and germination and have determined the roles of C. difficile toxins in disease pathogenesis. Exciting progress has also been made in defining the role of the microbiome, specific commensal bacterial species and host immunity in defence against infection with C. difficile. This Review will summarize the recent discoveries and developments in our understanding of C. difficile infection and pathogenesis.
Collapse
Affiliation(s)
- Michael C Abt
- Immunology Program, Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Peter T McKenney
- Immunology Program, Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Eric G Pamer
- Immunology Program, Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
21
|
Costa CL, López-Ureña D, de Oliveira Assis T, Ribeiro RA, Silva ROS, Rupnik M, Wilcox MH, de Carvalho AF, do Carmo AO, Dias AAM, de Carvalho CBM, Chaves-Olarte E, Rodríguez C, Quesada-Gómez C, de Castro Brito GA. A MLST Clade 2 Clostridium difficile strain with a variant TcdB induces severe inflammatory and oxidative response associated with mucosal disruption. Anaerobe 2016; 40:76-84. [PMID: 27311833 DOI: 10.1016/j.anaerobe.2016.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 06/10/2016] [Accepted: 06/12/2016] [Indexed: 01/13/2023]
Abstract
The epidemiology of Clostridium difficile infections is highly dynamic as new strains continue to emerge worldwide. Here we present a detailed analysis of a new C. difficile strain (ICC-45) recovered from a cancer patient in Brazil that died from severe diarrhea. A polyphasic approach assigned a new PCR-ribotype and PFGE macrorestriction pattern to strain ICC-45, which is toxigenic (tcdA(+), tcdB(+) and ctdB(+)) and classified as ST41 from MLST Clade 2 and toxinotype IXb. Strain ICC-45 encodes for a variant TcdB that induces a distinct CPE in agreement with its toxinotype. Unlike epidemic NAP1/027 strains, which are also classified to MLST Clade 2, strain ICC-45 is susceptible to fluoroquinolones and does not overproduce toxins TcdA and TcdB. However, supernatants from strain ICC-45 and a NAP1/027 strain produced similar expression of pro-inflammatory cytokines, epithelial damage, and oxidative stress response in the mouse ileal loop model. These results highlight inflammation and oxidative stress as common features in the pathogenesis of C. difficile Clade 2 strains. Finally, this work contributes to the description of differences in virulence among various C. difficile strains.
Collapse
Affiliation(s)
- Cecília Leite Costa
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Laboratory of Bacteriology, Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Diana López-Ureña
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica
| | - Thiago de Oliveira Assis
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Department of Morphology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Ronaldo A Ribeiro
- Haroldo Juaçaba Hospital, Cancer Institute of Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Maja Rupnik
- University of Maribor, Faculty of Medicine and National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Mark H Wilcox
- Department of Microbiology, Leeds Teaching Hospitals NHS Trust, The General Infirmary, Leeds, UK
| | - Alex Fiorini de Carvalho
- Experimental Genetics and Laboratory Animal Science, Department of General Biology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anderson Oliveira do Carmo
- Experimental Genetics and Laboratory Animal Science, Department of General Biology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Adriana Abalen Martins Dias
- Experimental Genetics and Laboratory Animal Science, Department of General Biology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cibele Barreto Mano de Carvalho
- Laboratory of Bacteriology, Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Esteban Chaves-Olarte
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica
| | - César Rodríguez
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica
| | - Carlos Quesada-Gómez
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica.
| | | |
Collapse
|