1
|
Deda O, Armitage EG, Mouskeftara T, Kachrimanidou M, Zervos I, Malousi A, Loftus NJ, Taitzoglou I, Gika H. Unraveling Cecal Alterations in Clostridioides difficile Colonized Mice through Comprehensive Metabolic Profiling. J Proteome Res 2024; 23:5462-5475. [PMID: 39480487 PMCID: PMC11629376 DOI: 10.1021/acs.jproteome.4c00578] [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: 07/08/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 12/07/2024]
Abstract
The disruption of gut microbiota caused by antibiotics favors the intestinal colonization of Clostridioides difficile - a Gram-positive, spore-forming anaerobic bacterium that causes potentially fatal gastrointestinal infections. In an endeavor to elucidate the complexities of the gut-brain axis in the context of Clostridium difficile infection (CDI), a murine model has been used to investigate the potential effects of antibiotic administration and subsequent colonization by C. difficile, as well as the impact of three different 10-day treatments (metronidazole, probiotics, and fecal microbiota transplantation), on the cecal metabolome for the first time. This follows our previous research which highlighted the metabolic effect of CDI and these treatments in the brain and employs the same four different metabolomics-based methods (targeted GC-MS/MS, targeted HILIC-MS/MS, untargeted RP-LC-HRMS/MS and untargeted GC-MS). A total of 286 unique metabolites have been identified in the mouse cecal profiles and statistical analysis revealed that CDI, as well as the subsequent treatments, significantly alters cecal metabolites and lipids implicated in various biochemical pathways centered around amino acid metabolism, glycerophospholipid metabolism, and central carbon metabolism. To our knowledge, this study represents the first exploration of the effects of C. difficile-induced colitis and potential treatments on the cecal tissue metabolome.
Collapse
Affiliation(s)
- Olga Deda
- Laboratory
of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic
AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi
Road, GR 57001 Thessaloniki, Greece
| | | | - Thomai Mouskeftara
- Laboratory
of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic
AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi
Road, GR 57001 Thessaloniki, Greece
| | - Melina Kachrimanidou
- 1 Laboratory of Microbiology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Zervos
- Laboratory
of Animal Physiology, Faculty of Veterinary Medicine, School of Health
Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Andigoni Malousi
- Laboratory
of Biological Chemistry, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Ioannis Taitzoglou
- Laboratory
of Development-Breeding of Animal Models and Biochemical Research,
School of Health Sciences, Aristotle University
of Thessaloniki, 54124 Thessaloniki, Greece
| | - Helen Gika
- Laboratory
of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic
AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi
Road, GR 57001 Thessaloniki, Greece
| |
Collapse
|
2
|
Han S, Oh E, Shin H, Kumaran S, Ko DH, Choi HJ. Antimicrobial Face Masks and Mask Covers with a Salt-Coated Stacked Spunbond Polypropylene Fabric: Effective Inactivation of Resilient Pathogens and Prevention of Contact Transmission. ACS APPLIED BIO MATERIALS 2024; 7:5171-5187. [PMID: 39008660 DOI: 10.1021/acsabm.4c00232] [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: 07/17/2024]
Abstract
In response to the ongoing threat posed by respiratory diseases, ensuring effective transmission protection is crucial for public health. To address the drawbacks of single-use face masks/respirators, which can be a potential source of contact-based transmission, we have designed an antimicrobial face mask and mask covering utilizing a stack of salt-coated spunbond (SB) fabric. This fabric acts as an outer layer for the face mask and as a covering over a conventional mask, respectively. We evaluated the universal antimicrobial performance of the salt-coated three-stacked SB fabric against enveloped/nonenveloped viruses and spore-forming/nonspore-forming bacteria. The distinctive pathogen inactivation efficiency was confirmed, including resistant pathogens such as human rhinovirus and Clostridium difficile. In addition, we tested other filter attributes, such as filtration efficiency and breathability, to determine the optimal layer for salt coating and its effects on performance. Our findings revealed that the outer layer of a conventional face mask plays a crucial role in contact transmission through contaminated face masks and respirators. Through contact transmission experiments using droplets involving three types of contaminants (fluorescent dyes, bacteria, and viruses), the salt-coated stacked SB fabric demonstrated a superior effect in preventing contact transmission compared to SB or meltblown polypropylene fabrics─an issue challenging to existing masks. Our results demonstrate that the use of salt-coated stacked SB fabrics as (i) the outer layer of a mask and (ii) a mask cover over a mask enhances overall filter performance against infectious droplets, achieving high pathogen inactivation and low contact-based transmission while maintaining breathability.
Collapse
Affiliation(s)
- Sumin Han
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada
| | - Euna Oh
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada
| | - Hyerin Shin
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Surjith Kumaran
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada
| | - Dae-Hong Ko
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada
| |
Collapse
|
3
|
Mendo-Lopez R, Alonso CD, Villafuerte-Gálvez JA. Best Practices in the Management of Clostridioides difficile Infection in Developing Nations. Trop Med Infect Dis 2024; 9:185. [PMID: 39195623 PMCID: PMC11359346 DOI: 10.3390/tropicalmed9080185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Clostridioides difficile infection (CDI) is a well-known cause of hospital-acquired infectious diarrhea in developed countries, though it has not been a top priority in the healthcare policies of developing countries. In the last decade, several studies have reported a wide range of CDI rates between 1.3% and 96% in developing nations, raising the concern that this could represent a healthcare threat for these nations. This review defines developing countries as those with a human development index (HDI) below 0.8. We aim to report the available literature on CDI epidemiology, diagnostics, management, and prevention in developing countries. We identify limitations for CDI diagnosis and management, such as limited access to CDI tests and unavailable oral vancomycin formulation, and identify opportunities to enhance CDI care, such as increased molecular test capabilities and creative solutions for CDI. We also discuss infection prevention strategies, including antimicrobial stewardship programs and opportunities emerging from the COVID-19 pandemic, which could impact CDI care.
Collapse
Affiliation(s)
- Rafael Mendo-Lopez
- Division of Infectious Disease, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Carolyn D. Alonso
- Division of Infectious Disease, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
- Harvard Medical School, Harvard University, Boston, MA 02215, USA;
| | - Javier A. Villafuerte-Gálvez
- Harvard Medical School, Harvard University, Boston, MA 02215, USA;
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| |
Collapse
|
4
|
Osman AH, Darkwah S, Kotey FCN, Odoom A, Hotor P, Dayie NTKD, Donkor ES. Reservoirs of Nosocomial Pathogens in Intensive Care Units: A Systematic Review. ENVIRONMENTAL HEALTH INSIGHTS 2024; 18:11786302241243239. [PMID: 38828046 PMCID: PMC11141231 DOI: 10.1177/11786302241243239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/14/2024] [Indexed: 06/05/2024]
Abstract
Background Nosocomial pathogens are known to exacerbate morbidity and mortality in contemporary critical healthcare. Hospital fomites, which include inanimate surfaces, have been identified as "breeding grounds" for pathogens that cause nosocomial infections. This systematic review aimed to deliver incisive insights on nosocomial pathogens in intensive care units (ICUs) and the role of fomites as potential reservoirs for their transmission. Method An extensive exploration of electronic databases, including PubMed and Scopus, from 1990 to 2023, was carried out between 25th and 29th May 2023, per standard PRISMA guidelines. Information were extracted from articles that reported on fomites in the ICU. Studies that did not quantitatively report the fomite contamination, and those that exclusively took samples from patients in the ICU were excluded from the analysis. Results About 40% of the total samples collected on fomites from all the studies yielded microbial growth, with species of Staphylococcus being the most predominant. Other prevalent microbes were Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Candida spp., Enterococcus sp., and Enterobacter sp. The neonatal intensive care unit (NICU) had the highest proportion of contaminated fomites. Among known fomites, the sphygmomanometer exhibited a 100% detection rate of nosocomial pathogens. This included E. aerogenes, Staphylococcus aureus, coagulase-negative Staphylococci (CoNS), E. coli, and K. pneumoniae. Multidrug-resistant (MDR) bacteria, such as methicillin-resistant S. aureus (MRSA), vancomycin-resistant Enterococci (VRE), extended-spectrum beta-lactamase (ESBL)-producing E. coli, and MDR Pseudomonas aeruginosa were commonly isolated on fomites in the ICUs. Conclusion Many fomites that are readily used in patient care in the ICU harbour nosocomial pathogens. The most common fomite appeared to be mobile phones, sphygmomanometers, and stethoscopes, with Staphylococcus being the most common contaminant. Consequently, the need for rigorous disinfection and sterilization protocols on fomites in the ICU cannot be overemphasized. Additionally, heightened awareness on the subject among health professionals is crucial to mitigating the risk and burden of nosocomial infections caused by drug-resistant bacteria.
Collapse
Affiliation(s)
- Abdul-Halim Osman
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Samuel Darkwah
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Fleischer C N Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Alex Odoom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Prince Hotor
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Nicholas T K D Dayie
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Eric S Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| |
Collapse
|
5
|
Ke S, Villafuerte Gálvez JA, Sun Z, Cao Y, Pollock NR, Chen X, Kelly CP, Liu YY. Rational Design of Live Biotherapeutic Products for the Prevention of Clostridioides difficile Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591969. [PMID: 38746249 PMCID: PMC11092666 DOI: 10.1101/2024.04.30.591969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Clostridioides difficile infection (CDI) is one of the leading causes of healthcare- and antibiotic-associated diarrhea. While fecal microbiota transplantation (FMT) has emerged as a promising therapy for recurrent CDI, its exact mechanisms of action and long-term safety are not fully understood. Defined consortia of clonal bacterial isolates, known as live biotherapeutic products (LBPs), have been proposed as an alternative therapeutic option. However, the rational design of LBPs remains challenging. Here, we employ a computational pipeline and three independent metagenomic datasets to systematically identify microbial strains that have the potential to inhibit CDI. We first constructed the CDI-related microbial genome catalog, comprising 3,741 non-redundant metagenome-assembled genomes (nrMAGs) at the strain level. We then identified multiple potential protective nrMAGs that can be candidates for the design of microbial consortia targeting CDI, including strains from Dorea formicigenerans, Oscillibacter welbionis, and Faecalibacterium prausnitzii. Importantly, some of these potential protective nrMAGs were found to play an important role in the success of FMT, and the majority of the top protective nrMAGs can be validated by various previously reported findings. Our results demonstrate a computational framework for the rational selection of microbial strains targeting CDI, paving the way for the computational design of microbial consortia against other enteric infections.
Collapse
Affiliation(s)
- Shanlin Ke
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Javier A Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Zheng Sun
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yangchun Cao
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, People’s Republic of China
| | - Nira R Pollock
- Division of Infectious Disease, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Ciarán P Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
6
|
Singh KB, Khouri A, Singh D, Prieto J, Dutta P, Nnadozie MC, Clanton C, Morrison E, Sonnier W. Testing and Diagnosis of Clostridioides difficile Infection in Special Scenarios: A Systematic Review. Cureus 2024; 16:e59016. [PMID: 38800338 PMCID: PMC11127751 DOI: 10.7759/cureus.59016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
INTRODUCTION Clostridioides difficile infection (CDI) is a clinical and laboratory diagnosis. Populations at higher risk of developing disease require a high clinical index of suspicion for laboratory testing to avoid incorrect assumptions of colonization. Common risk factors include recent antibiotic use, elderly (>65 years old), and immunocompromised patients. C. difficile assays should be ordered in an algorithm approach to diagnose an infection rather than colonization. Screening tests are widely available in hospital systems, but novel molecular testing may aid in diagnosis in patients with inconclusive or discordant antigen and toxin test results. Methods: Data was extracted from PubMed, Scopus, and Cumulative Index of Nursing and Allied Health Literature (CINAHL) databases based on the keywords "clostridioides difficile", "toxin assay", and "toxic megacolon". The data extracted is based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 guidelines. A total of 27 reports were included in this systematic review. RESULTS Testing patients with a significant gastrointestinal surgical history, hypogammaglobulinemia, inflammatory bowel disease, intensive care unit, and immunocompromised patients for CDI is highly recommended. Diarrhea in these subsets of patients requires correlation of clinical context and an understanding of assay results to avoid over- and under-treating. CONCLUSION CDI should be considered in all patients with traditional risk factors. Heightened clinical suspicion of CDI is required in patients with hypogammaglobulinemia, transplant recipients, patients with gastrointestinal surgical history, and inflammatory bowel disease. Testing should be limited to patients with clinical manifestations of CDI to ensure a high pretest probability for test interpretation. Healthcare workers should adhere to testing algorithms to optimize yield in the appropriate clinical context. Diagnostic assays should follow a sequential, stepwise approach to categorize the toxin expression status of the bacteria accurately.
Collapse
Affiliation(s)
- Karan B Singh
- Internal Medicine, Frederick P. Whiddon College of Medicine at the University of South Alabama, Mobile, USA
| | - Anas Khouri
- Internal Medicine, Frederick P. Whiddon College of Medicine at the University of South Alabama, Mobile, USA
| | - Deepak Singh
- Internal Medicine, Caribbean Medical University, Willemstad, CUW
| | - Jose Prieto
- Internal Medicine, Loyola University MacNeal Hospital, Berwyn, USA
| | - Priyata Dutta
- Internal Medicine, Trinity Health St. Joseph Mercy Ann Arbor, Ann Arbor, USA
| | - Maduka C Nnadozie
- Internal Medicine, AtlantiCare Regional Medical Center, Atlantic City, USA
| | - Clista Clanton
- Biomedical Research, Frederick P. Whiddon College of Medicine at the University of South Alabama, Mobile, USA
| | - Esther Morrison
- Infectious Diseases, Frederick P. Whiddon College of Medicine at the University of South Alabama, Mobile, USA
| | - William Sonnier
- Gastroenterology and Hepatology, Frederick P. Whiddon College of Medicine at the University of South Alabama, Mobile, USA
| |
Collapse
|
7
|
McCoy R, Oldroyd S, Yang W, Wang K, Hoven D, Bulmer D, Zilbauer M, Owens RM. In Vitro Models for Investigating Intestinal Host-Pathogen Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306727. [PMID: 38155358 PMCID: PMC10885678 DOI: 10.1002/advs.202306727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Indexed: 12/30/2023]
Abstract
Infectious diseases are increasingly recognized as a major threat worldwide due to the rise of antimicrobial resistance and the emergence of novel pathogens. In vitro models that can adequately mimic in vivo gastrointestinal physiology are in high demand to elucidate mechanisms behind pathogen infectivity, and to aid the design of effective preventive and therapeutic interventions. There exists a trade-off between simple and high throughput models and those that are more complex and physiologically relevant. The complexity of the model used shall be guided by the biological question to be addressed. This review provides an overview of the structure and function of the intestine and the models that are developed to emulate this. Conventional models are discussed in addition to emerging models which employ engineering principles to equip them with necessary advanced monitoring capabilities for intestinal host-pathogen interrogation. Limitations of current models and future perspectives on the field are presented.
Collapse
Affiliation(s)
- Reece McCoy
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Sophie Oldroyd
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Woojin Yang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Kaixin Wang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Darius Hoven
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - David Bulmer
- Department of PharmacologyUniversity of CambridgeCambridgeCB2 1PDUK
| | - Matthias Zilbauer
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| |
Collapse
|
8
|
Douillard FP, Portinha IM, Derman Y, Woudstra C, Mäklin T, Dorner MB, Korkeala H, Henriques AO, Lindström M. A Novel Prophage-like Insertion Element within yabG Triggers Early Entry into Sporulation in Clostridium botulinum. Viruses 2023; 15:2431. [PMID: 38140671 PMCID: PMC10747680 DOI: 10.3390/v15122431] [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: 05/25/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Sporulation is a finely regulated morphogenetic program important in the ecology and epidemiology of Clostridium botulinum. Exogenous elements disrupting sporulation-associated genes contribute to sporulation regulation and introduce diversity in the generally conserved sporulation programs of endospore formers. We identified a novel prophage-like DNA segment, termed the yin element, inserted within yabG, encoding a sporulation-specific cysteine protease, in an environmental isolate of C. botulinum. Bioinformatic analysis revealed that the genetic structure of the yin element resembles previously reported mobile intervening elements associated with sporulation genes. Within a pure C. botulinum culture, we observed two subpopulations of cells with the yin element either integrated into the yabG locus or excised as a circular DNA molecule. The dynamics between the two observed conformations of the yin element was growth-phase dependent and likely mediated by recombination events. The yin element was not required for sporulation by C. botulinum but triggered an earlier entry into sporulation than in a related isolate lacking this element. So far, the yin element has not been found in any other C. botulinum strains or other endospore-forming species. It remains to be demonstrated what kind of competitive edge it provides for C. botulinum survival and persistence.
Collapse
Affiliation(s)
- François P. Douillard
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00350 Helsinki, Finland; (F.P.D.); (I.M.P.); (Y.D.); (C.W.); (H.K.)
| | - Inês Martins Portinha
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00350 Helsinki, Finland; (F.P.D.); (I.M.P.); (Y.D.); (C.W.); (H.K.)
| | - Yağmur Derman
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00350 Helsinki, Finland; (F.P.D.); (I.M.P.); (Y.D.); (C.W.); (H.K.)
| | - Cédric Woudstra
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00350 Helsinki, Finland; (F.P.D.); (I.M.P.); (Y.D.); (C.W.); (H.K.)
| | - Tommi Mäklin
- Department of Mathematics and Statistics, Faculty of Science, University of Helsinki, 00560 Helsinki, Finland;
| | - Martin B. Dorner
- Centre for Biological Threats and Special Pathogens, ZBS3—Biological Toxins, Robert Koch Institute, 13353 Berlin, Germany;
| | - Hannu Korkeala
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00350 Helsinki, Finland; (F.P.D.); (I.M.P.); (Y.D.); (C.W.); (H.K.)
| | - Adriano O. Henriques
- Institute of Chemical and Biological Technology, NOVA University Lisbon, 2780-157 Oeiras, Portugal;
| | - Miia Lindström
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00350 Helsinki, Finland; (F.P.D.); (I.M.P.); (Y.D.); (C.W.); (H.K.)
| |
Collapse
|
9
|
Moore JH, Salahi A, Honrado C, Warburton C, Tate S, Warren CA, Swami NS. Correlating Antibiotic-Induced Dysbiosis to Clostridioides difficile Spore Germination and Host Susceptibility to Infection Using an Ex Vivo Assay. ACS Infect Dis 2023; 9:1878-1888. [PMID: 37756389 PMCID: PMC10581205 DOI: 10.1021/acsinfecdis.3c00192] [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: 04/26/2023] [Indexed: 09/29/2023]
Abstract
Antibiotic-induced microbiota disruption and its persistence create conditions for dysbiosis and colonization by opportunistic pathogens, such as those causing Clostridioides difficile (C. difficile) infection (CDI), which is the most severe hospital-acquired intestinal infection. Given the wide differences in microbiota across hosts and in their recovery after antibiotic treatments, there is a need for assays to assess the influence of dysbiosis and its recovery dynamics on the susceptibility of the host to CDI. Germination of C. difficile spores is a key virulence trait for the onset of CDI, which is influenced by the level of primary vs secondary bile acids in the intestinal milieu that is regulated by the microbiota composition. Herein, the germination of C. difficile spores in fecal supernatant from mice that are subject to varying degrees of antibiotic treatment is utilized as an ex vivo assay to predict intestinal dysbiosis in the host based on their susceptibility to CDI, as determined by in vivo CDI metrics in the same mouse model. Quantification of spore germination down to lower detection limits than the colony-forming assay is achieved by using impedance cytometry to count single vegetative bacteria that are identified based on their characteristic electrical physiology for distinction vs aggregated spores and cell debris in the media. As a result, germination can be quantified at earlier time points and with fewer spores for correlation to CDI outcomes. This sets the groundwork for a point-of-care tool to gauge the susceptibility of human microbiota to CDI after antibiotic treatments.
Collapse
Affiliation(s)
- John H. Moore
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
| | - Armita Salahi
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
| | - Carlos Honrado
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
| | - Christopher Warburton
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
| | - Steven Tate
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
| | - Cirle A. Warren
- Infectious
Diseases, School of Medicine, University
of Virginia, Charlottesville, Virginia 22903, United States
| | - Nathan S. Swami
- Electrical
and Computer Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
- Chemistry, University
of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
10
|
Biswas R, Dudani H, Lakhera P, Pal AK, Kurbah P, Bhatia D, Dhok A, Kashyap RS. Challenges and future solutions for detection of Clostridioides difficile in adults. Ann Gastroenterol 2023; 36:369-377. [PMID: 37396008 PMCID: PMC10304531 DOI: 10.20524/aog.2023.0802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/17/2023] [Indexed: 07/04/2023] Open
Abstract
There are no established standards for the diagnosis of Clostridioides difficile infection (CDI), even though the importance of this infection in humans is well known. The effectiveness of the commercially available techniques, which are all standardized for use with human feces, is also limited in terms of the accuracy of the tests. Furthermore, the current approach lacks a point-of-care diagnosis with an acceptable range of sensitivity and specificity. This article reviews the challenges and possible future solutions for the detection of CDI in adults. Existing diagnostic methods, such as enzyme-linked immunoassays and microbial culturing for the detection of toxins A and B, appear to work poorly in samples but exhibit great sensitivity for glutamate dehydrogenase. Real-time polymerase chain reaction and nucleic acid amplification tests have been investigated in a few studies on human samples, but so far have shown poor turnaround times. Thus, developing a multiplex point-of-care test assay with high sensitivity and specificity is required as a bedside approach for diagnosing this emerging infection.
Collapse
Affiliation(s)
- Rima Biswas
- Research Laboratory, Dr. G.M. Taori Central India Institute of Medical Sciences, Nagpur (Rima Biswas, Hemanshi Dudani, Rajpal Singh Kashyap)
| | - Hemanshi Dudani
- Research Laboratory, Dr. G.M. Taori Central India Institute of Medical Sciences, Nagpur (Rima Biswas, Hemanshi Dudani, Rajpal Singh Kashyap)
| | - Praveen Lakhera
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh and CSIR-Central Scientific Instruments Organization, Chandigarh (Praveen Lakhera)
| | - Arun Kumar Pal
- Department of Molecular and Cellular Engineering, Sam Higginbottom University of Agriculture, Technology, and Sciences, Prayagraj (Arun Kumar Pal)
| | - Phibalari Kurbah
- Department of Biomedical Engineering, North Eastern Hill University, Shillong, Meghalaya (Phibalari Kurbah, Dinesh Bhatia)
| | - Dinesh Bhatia
- Department of Biomedical Engineering, North Eastern Hill University, Shillong, Meghalaya (Phibalari Kurbah, Dinesh Bhatia)
| | - Archana Dhok
- Department of Biochemistry, Datta Meghe Institute of Medical Sciences, Sawangi (Archana Dhok), India
| | - Rajpal Singh Kashyap
- Research Laboratory, Dr. G.M. Taori Central India Institute of Medical Sciences, Nagpur (Rima Biswas, Hemanshi Dudani, Rajpal Singh Kashyap)
| |
Collapse
|
11
|
Bocchetti M, Ferraro MG, Melisi F, Grisolia P, Scrima M, Cossu AM, Yau TO. Overview of current detection methods and microRNA potential in Clostridioides difficile infection screening. World J Gastroenterol 2023; 29:3385-3399. [PMID: 37389232 PMCID: PMC10303512 DOI: 10.3748/wjg.v29.i22.3385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/23/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Clostridioides difficile (formerly called Clostridium difficile, C. difficile) infection (CDI) is listed as an urgent threat on the 2019 antibiotic resistance threats report in the United States by the Centers for Disease Control and Prevention. Early detection and appropriate disease management appear to be essential. Meanwhile, although the majority of cases are hospital-acquired CDI, community-acquired CDI cases are also on the rise, and this vulnerability is not limited to immunocompromised patients. Gastrointestinal treatments and/or gastrointestinal tract surgeries may be required for patients diagnosed with digestive diseases. Such treatments could suppress or interfere with the patient’s immune system and disrupt gut flora homeostasis, creating a suitable microecosystem for C. difficile overgrowth. Currently, stool-based non-invasive screening is the first-line approach to CDI diagnosis, but the accuracy is varied due to different clinical microbiology detection methods; therefore, improving reliability is clearly required. In this review, we briefly summarised the life cycle and toxicity of C. difficile, and we examined existing diagnostic approaches with an emphasis on novel biomarkers such as microRNAs. These biomarkers can be easily detected through non-invasive liquid biopsy and can yield crucial information about ongoing pathological phenomena, particularly in CDI.
Collapse
Affiliation(s)
- Marco Bocchetti
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples 80138, Italy
- Department of Molecular Oncology, Precision Medicine Laboratory and COVID19 Laboratory, Biogem Scarl, Ariano Irpino 83031, Italy
| | - Maria Grazia Ferraro
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II,” Naples 80131, Italy
| | - Federica Melisi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples 80138, Italy
- Department of Molecular Oncology, Precision Medicine Laboratory and COVID19 Laboratory, Biogem Scarl, Ariano Irpino 83031, Italy
| | - Piera Grisolia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples 80138, Italy
- Department of Molecular Oncology, Precision Medicine Laboratory and COVID19 Laboratory, Biogem Scarl, Ariano Irpino 83031, Italy
| | - Marianna Scrima
- Department of Molecular Oncology, Precision Medicine Laboratory and COVID19 Laboratory, Biogem Scarl, Ariano Irpino 83031, Italy
| | - Alessia Maria Cossu
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples 80138, Italy
- Department of Molecular Oncology, Precision Medicine Laboratory and COVID19 Laboratory, Biogem Scarl, Ariano Irpino 83031, Italy
| | - Tung On Yau
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
- Department of Rural Land Use, Scotland’s Rural College, Aberdeen AB21 9YA, Scotland, United Kingdom
- Department of Health Science, University of the People, Pasadena, CA 9110112, United States
| |
Collapse
|
12
|
Francis D, Bhairaddy A, Joy A, Hari GV, Francis A. Secretory proteins in the orchestration of microbial virulence: The curious case of Staphylococcus aureus. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:271-350. [PMID: 36707204 DOI: 10.1016/bs.apcsb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | | | - Ashik Francis
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| |
Collapse
|
13
|
Enkirch T, Mernelius S, Magnusson C, Kühlmann‐Berenzon S, Bengnér M, Åkerlund T, Rizzardi K. Molecular epidemiology of community- and hospital-associated Clostridioides difficile infections in Jönköping, Sweden, October 2017 - March 2018. APMIS 2022; 130:661-670. [PMID: 35980252 PMCID: PMC9826108 DOI: 10.1111/apm.13270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/17/2022] [Indexed: 01/11/2023]
Abstract
Clostridioides difficile infections (CDIs) in Sweden are mostly hospital-associated (HA) with limited knowledge regarding community-associated (CA) infections. Here, we investigated the molecular epidemiology of clinical isolates of CA-CDI and HA-CDI in a Swedish county. Data and isolates (n = 156) of CDI patients (n = 122) from Jönköping county, October 2017-March 2018, were collected and classified as CA (without previous hospital care or onset ≤2 days after admission or >12 weeks after discharge from hospital) or HA (onset >3 days after hospital admission or within 4 weeks after discharge). Molecular characterization of isolates included PCR ribotyping (n = 156 isolates) and whole genome sequencing with single nucleotide polymorphisms (SNP) analysis (n = 53 isolates). We classified 47 patients (39%) as CA-CDI and 75 (61%) as HA-CDI. Between CA-CDI and HA-CDI patients, we observed no statistically significant differences regarding gender, age, 30-day mortality or recurrence. Ribotype 005 (RR 3.1; 95% CI: 1.79-5.24) and 020 (RR 2.5; 95% CI: 1.31-4.63) were significantly associated with CA-CDI. SNP analysis identified seven clusters (0-2 SNP difference) involving 17/53 isolates of both CA-CDI and HA-CDI. Molecular epidemiology differed between CA-CDI and HA-CDI and WGS analysis suggests transmission of CDI within and between hospitals and communities.
Collapse
Affiliation(s)
- Theresa Enkirch
- Public Health Agency of SwedenSolnaSweden,European Programme for Public Health Microbiology Training (EUPHEM), European Centre for Disease Prevention and Control (ECDC)StockholmSweden
| | - Sara Mernelius
- Laboratory MedicineRegion Jönköping CountyJönköpingSweden,Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| | - Cecilia Magnusson
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden,Department of Infectious DiseasesRegion Jönköping CountyJönköpingSweden
| | | | - Malin Bengnér
- Office for Control of Communicable DiseasesRegion Jönköping CountyJönköpingSweden
| | | | | |
Collapse
|
14
|
Raeisi H, Azimirad M, Nabavi-Rad A, Asadzadeh Aghdaei H, Yadegar A, Zali MR. Application of recombinant antibodies for treatment of Clostridioides difficile infection: Current status and future perspective. Front Immunol 2022; 13:972930. [PMID: 36081500 PMCID: PMC9445313 DOI: 10.3389/fimmu.2022.972930] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Clostridioides difficile (C. difficile), known as the major cause of antibiotic-associated diarrhea, is regarded as one of the most common healthcare-associated bacterial infections worldwide. Due to the emergence of hypervirulent strains, development of new therapeutic methods for C. difficile infection (CDI) has become crucially important. In this context, antibodies have been introduced as valuable tools in the research and clinical environments, as far as the effectiveness of antibody therapy for CDI was reported in several clinical investigations. Hence, production of high-performance antibodies for treatment of CDI would be precious. Traditional approaches of antibody generation are based on hybridoma technology. Today, application of in vitro technologies for generating recombinant antibodies, like phage display, is considered as an appropriate alternative to hybridoma technology. These techniques can circumvent the limitations of the immune system and they can be exploited for production of antibodies against different types of biomolecules in particular active toxins. Additionally, DNA encoding antibodies is directly accessible in in vitro technologies, which enables the application of antibody engineering in order to increase their sensitivity and specificity. Here, we review the application of antibodies for CDI treatment with an emphasis on recombinant fragment antibodies. Also, this review highlights the current and future prospects of the aforementioned approaches for antibody-mediated therapy of CDI.
Collapse
Affiliation(s)
- Hamideh Raeisi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Abbasi BA, Dharan A, Mishra A, Saraf D, Ahamad I, Suravajhala P, Valadi J. In Silico Characterization of Uncharacterized Proteins From Multiple Strains of Clostridium Difficile. Front Genet 2022; 13:878012. [PMID: 36035185 PMCID: PMC9403866 DOI: 10.3389/fgene.2022.878012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Clostridium difficile (C. difficile) is a multi-strain, spore-forming, Gram-positive, opportunistic enteropathogen bacteria, majorly associated with nosocomial infections, resulting in severe diarrhoea and colon inflammation. Several antibiotics including penicillin, tetracycline, and clindamycin have been employed to control C. difficile infection, but studies have suggested that injudicious use of antibiotics has led to the development of resistance in C. difficile strains. However, many proteins from its genome are still considered uncharacterized proteins that might serve crucial functions and assist in the biological understanding of the organism. In this study, we aimed to annotate and characterise the 6 C. difficile strains using in silico approaches. We first analysed the complete genome of 6 C. difficile strains using standardised approaches and analysed hypothetical proteins (HPs) employing various bioinformatics approaches coalescing, including identifying contigs, coding sequences, phage sequences, CRISPR-Cas9 systems, antimicrobial resistance determination, membrane helices, instability index, secretory nature, conserved domain, and vaccine target properties like comparative homology analysis, allergenicity, antigenicity determination along with structure prediction and binding-site analysis. This study provides crucial supporting information about the functional characterization of the HPs involved in the pathophysiology of the disease. Moreover, this information also aims to assist in mechanisms associated with bacterial pathogenesis and further design candidate inhibitors and bona fide pharmaceutical targets.
Collapse
Affiliation(s)
| | | | | | | | | | - Prashanth Suravajhala
- Bioclues.org, Hyderabad, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana, India
- *Correspondence: Prashanth Suravajhala, ; Jayaraman Valadi,
| | - Jayaraman Valadi
- Bioclues.org, Hyderabad, India
- School of Computational and Data Sciences, Vidyashilp University, Bengaluru, India
- Department of Computer Science, FLAME University, Pune, India
- *Correspondence: Prashanth Suravajhala, ; Jayaraman Valadi,
| |
Collapse
|
16
|
Kunishima H, Ohge H, Suzuki H, Nakamura A, Matsumoto K, Mikamo H, Mori N, Morinaga Y, Yanagihara K, Yamagishi Y, Yoshizawa S. Japanese Clinical Practice Guidelines for Management of Clostridioides (Clostridium) difficile infection. J Infect Chemother 2022; 28:1045-1083. [PMID: 35618618 DOI: 10.1016/j.jiac.2021.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/16/2021] [Accepted: 12/13/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Hiroyuki Kunishima
- Department of Infectious Diseases, St. Marianna University School of Medicine, Japan.
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University Hospital, Japan
| | - Hiromichi Suzuki
- Division of Infectious Diseases, Department of Medicine, Tsukuba Medical Center Hospital, Japan
| | - Atsushi Nakamura
- Division of Infection Control and Prevention, Nagoya City University Hospital, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Japan
| | - Hiroshige Mikamo
- Clinical Infectious Diseases, Graduate School of Medicine, Aichi Medical University, Japan
| | - Nobuaki Mori
- Division of General Internal Medicine and Infectious Diseases, National Hospital Organization Tokyo Medical Center, Japan
| | - Yoshitomo Morinaga
- Department of Microbiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Yuka Yamagishi
- Clinical Infectious Diseases, Graduate School of Medicine, Aichi Medical University, Japan
| | - Sadako Yoshizawa
- Department of Clinical Laboratory/Department of Microbiology and Infectious Diseases, Toho University School of Medicine, Japan
| |
Collapse
|
17
|
Guiberson ER, Good CJ, Wexler AG, Skaar EP, Spraggins JM, Caprioli RM. Multimodal Imaging Mass Spectrometry of Murine Gastrointestinal Tract with Retained Luminal Content. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:1073-1076. [PMID: 35545232 PMCID: PMC9264265 DOI: 10.1021/jasms.1c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The gastrointestinal tract, including luminal content, harbors a complex mixture of microorganisms, host dietary content, and immune factors. Existing imaging approaches remove luminal content and only visualize small regions of the GI tract. Here, we demonstrate a workflow for multimodal imaging using matrix-assisted laser desorption/ionization imaging mass spectrometry, autofluorescence, and bright field microscopy for mapping intestinal tissue and luminal content. Results comparing tissue and luminal content in control murine tissue show both unique molecular and elemental distributions and abundances using multimodal protein, lipid, and elemental imaging. For instance, lipid PC(42:1) is 2× higher intensity in luminal content than tissue, while PC(32:0) is 80× higher intensity in tissue. Additionally, some ions such as the protein at m/z 3443 and the element manganese are only detected in luminal content, while the protein at m/z 8564 was only detected in tissue and phosphorus had 2× higher abundance in tissue. These data highlight the robust molecular information that can be gained from the gastrointestinal tract with the inclusion of luminal content.
Collapse
Affiliation(s)
- Emma R Guiberson
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37203, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37203, United States
| | - Christopher J Good
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37203, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37203, United States
| | - Aaron G Wexler
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37203, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37203, United States
| | - Eric P Skaar
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37203, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37203, United States
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37203, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37203, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37203, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee 37203, United States
| | - Richard M Caprioli
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37203, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37203, United States
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37203g, United States
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37203, United States
| |
Collapse
|
18
|
Role of Antimicrobial Drug in the Development of Potential Therapeutics. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2500613. [PMID: 35571735 PMCID: PMC9098294 DOI: 10.1155/2022/2500613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 12/17/2022]
Abstract
Population of the world run into several health-related emergencies among mankind and humans as it creates a challenge for the evolution of novel drug discoveries. One such can be the emergence of multidrug-resistant (MDR) strains in both hospital and community settings, which have been due to an inappropriate use and inadequate control of antibiotics that has led to the foremost human health concerns with a high impact on the global economy. So far, there has been application of two strategies for the development of anti-infective agents either by classical antibiotics that have been derived for their synthetic analogs with increased efficacy or screening natural compounds along with the synthetic compound libraries for the antimicrobial activities. However, need for newer treatment options for infectious diseases has led research to develop new generation of antimicrobial activity to further lessen the spread of antibiotic resistance. Currently, the principles aim to find novel mode of actions or products to target the specific sites and virulence factors in pathogens by a series of better understanding of physiology and molecular aspects of the microbial resistance, mechanism of infection process, and gene-pathogenicity relationship. The design various novel strategies tends to provide us a path for the development of various antimicrobial therapies that intends to have a broader and wider antimicrobial spectrum that helps to combat MDR strains worldwide. The development of antimicrobial peptides, metabolites derived from plants, microbes, phage-based antimicrobial agents, use of metal nanoparticles, and role of CRISPR have led to an exceptional strategies in designing and developing the next-generation antimicrobials. These novel strategies might help to combat the seriousness of the infection rates and control the health crisis system.
Collapse
|
19
|
Abay S, Ahmed EF, Aydin F, Karakaya E, Müştak HK. Presence of Clostridioides difficile in cattle feces, carcasses, and slaughterhouses: Molecular characterization and antibacterial susceptibility of the recovered isolates. Anaerobe 2022; 75:102575. [PMID: 35477095 DOI: 10.1016/j.anaerobe.2022.102575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 04/13/2022] [Accepted: 04/17/2022] [Indexed: 11/28/2022]
Abstract
The aims of this study were to isolate and identify Clostridioides difficile from cattle feces and carcasses, and slaughterhouse samples, and to determine the molecular characteristics and antibacterial susceptibility of the recovered isolates. A total of 220 samples, including 100 cattle fecal samples, 100 cattle carcass surface samples, and 20 slaughterhouse samples were used as the study material. In total, 12 (5.45%) samples, including 11 (11%) cattle fecal samples and 1 (5%) slaughterhouse sample, were found to be positive for C. difficile. On the other hand, all of the carcass samples were negative for C. difficile. A total of 11 (91.66%) isolates, including 10 fecal isolates and 1 slaughterhouse wastewater isolate, were found to be positive for the presence of the toxin genes tcdA and tcdB, whilst 1 fecal isolate was found to be negative for both genes. In addition, 3 different ERIC-PCR profiles were identified in the 11 fecal isolates. The ERIC-PCR profile of the slaughterhouse wastewater isolate was found to be similar to one of the ERIC-PCR profiles obtained from the fecal isolates. All of the isolates were resistant to ciprofloxacin and levofloxacin. Considering that the agent is a spore-forming bacterium shed in feces, the detection of C. difficile isolates of different genotypes, some carrying toxin genes, suggests that feces and slaughterhouse wastewater carrying this bacterium may pose a risk for the contamination of carcasses. The current study revealed that hygiene conditions should be performed to the maximum extent in slaughterhouses.
Collapse
Affiliation(s)
- Seçil Abay
- Erciyes University, Faculty of Veterinary Medicine, Department of Microbiology Kayseri, Turkey.
| | | | - Fuat Aydin
- Erciyes University, Faculty of Veterinary Medicine, Department of Microbiology Kayseri, Turkey
| | - Emre Karakaya
- Erciyes University, Faculty of Veterinary Medicine, Department of Microbiology Kayseri, Turkey
| | - Hamit Kaan Müştak
- Ankara University, Faculty of Veterinary Medicine, Department of Microbiology, Ankara, Turkey
| |
Collapse
|
20
|
Di Domenico M, Ballini A, Boccellino M, Scacco S, Lovero R, Charitos IA, Santacroce L. The Intestinal Microbiota May Be a Potential Theranostic Tool for Personalized Medicine. J Pers Med 2022; 12:523. [PMID: 35455639 PMCID: PMC9024566 DOI: 10.3390/jpm12040523] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
The human intestine is colonized by a huge number of microorganisms from the moment of birth. This set of microorganisms found throughout the human body, is called the microbiota; the microbiome indicates the totality of genes that the microbiota can express, i.e., its genetic heritage. Thus, microbiota participates in and influences the proper functioning of the organism. The microbiota is unique for each person; it differs in the types of microorganisms it contains, the number of each microorganism, and the ratio between them, but mainly it changes over time and under the influence of many factors. Therefore, the correct functioning of the human body depends not only on the expression of its genes but also on the expression of the genes of the microorganisms it coexists with. This fact makes clear the enormous interest of community science in studying the relationship of the human microbiota with human health and the incidence of disease. The microbiota is like a unique personalized "mold" for each person; it differs quantitatively and qualitatively for the microorganisms it contains together with the relationship between them, and it changes over time and under the influence of many factors. We are attempting to modulate the microbial components in the human intestinal microbiota over time to provide positive feedback on the health of the host, from intestinal diseases to cancer. These interventions to modulate the intestinal microbiota as well as to identify the relative microbiome (genetic analysis) can range from dietary (with adjuvant prebiotics or probiotics) to fecal transplantation. This article researches the recent advances in these strategies by exploring their advantages and limitations. Furthermore, we aim to understand the relationship between intestinal dysbiosis and pathologies, through the research of resident microbiota, that would allow the personalization of the therapeutic antibiotic strategy.
Collapse
Affiliation(s)
- Marina Di Domenico
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (M.B.)
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (M.B.)
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Mariarosaria Boccellino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.D.D.); (M.B.)
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Roberto Lovero
- AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, Clinical Pathology Unit, Policlinico University Hospital of Bari, 70124 Bari, Italy;
| | - Ioannis Alexandros Charitos
- Department of Emergency and Urgency, National Poisoning Centre, Riuniti University Hospital of Foggia, 71122 Foggia, Italy;
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| |
Collapse
|
21
|
Tagliaferri AR, Murray K, Michael P. A Case of Pseudomembranous Colitis of Unknown Etiology. Cureus 2022; 14:e21914. [PMID: 35265432 PMCID: PMC8898612 DOI: 10.7759/cureus.21914] [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] [Accepted: 02/02/2022] [Indexed: 11/12/2022] Open
Abstract
Pseudomembranous colitis (PC) is a nonspecific bowel injury resulting from decreased oxygenation, endothelial damage, and impaired blood flow to the mucosa. Although the most well-known cause of PC is Clostridium difficile (C. difficile), several diseases and medications can cause or predispose individuals to PC, such as microscopic colitis, infectious organisms, inflammatory conditions, nonsteroidal anti-inflammatory drugs, and chemotherapy agents. Here, we present the case of a patient who completed treatment for C. difficile infection but developed worsening PC of unknown etiology.
Collapse
|
22
|
Patangia DV, Anthony Ryan C, Dempsey E, Paul Ross R, Stanton C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022; 11:e1260. [PMID: 35212478 PMCID: PMC8756738 DOI: 10.1002/mbo3.1260] [Citation(s) in RCA: 250] [Impact Index Per Article: 125.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
It is well established that the gut microbiota plays an important role in host health and is perturbed by several factors including antibiotics. Antibiotic-induced changes in microbial composition can have a negative impact on host health including reduced microbial diversity, changes in functional attributes of the microbiota, formation, and selection of antibiotic-resistant strains making hosts more susceptible to infection with pathogens such as Clostridioides difficile. Antibiotic resistance is a global crisis and the increased use of antibiotics over time warrants investigation into its effects on microbiota and health. In this review, we discuss the adverse effects of antibiotics on the gut microbiota and thus host health, and suggest alternative approaches to antibiotic use.
Collapse
Affiliation(s)
- Dhrati V. Patangia
- School of MicrobiologyUniversity College CorkCorkIreland
- Teagasc Food Research Centre, MooreparkFermoy Co.CorkIreland
- APC MicrobiomeCorkIreland
| | | | - Eugene Dempsey
- School of MicrobiologyUniversity College CorkCorkIreland
| | - Reynolds Paul Ross
- School of MicrobiologyUniversity College CorkCorkIreland
- APC MicrobiomeCorkIreland
| | - Catherine Stanton
- Teagasc Food Research Centre, MooreparkFermoy Co.CorkIreland
- APC MicrobiomeCorkIreland
| |
Collapse
|
23
|
Dassanayake MK, Khoo TJ, An J. Antibiotic resistance modifying ability of phytoextracts in anthrax biological agent Bacillus anthracis and emerging superbugs: a review of synergistic mechanisms. Ann Clin Microbiol Antimicrob 2021; 20:79. [PMID: 34856999 PMCID: PMC8641154 DOI: 10.1186/s12941-021-00485-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 11/22/2021] [Indexed: 01/17/2023] Open
Abstract
Background and objectives The chemotherapeutic management of infections has become challenging due to the global emergence of antibiotic resistant pathogenic bacteria. The recent expansion of studies on plant-derived natural products has lead to the discovery of a plethora of phytochemicals with the potential to combat bacterial drug resistance via various mechanisms of action. This review paper summarizes the primary antibiotic resistance mechanisms of bacteria and also discusses the antibiotic-potentiating ability of phytoextracts and various classes of isolated phytochemicals in reversing antibiotic resistance in anthrax agent Bacillus anthracis and emerging superbug bacteria. Methods Growth inhibitory indices and fractional inhibitory concentration index were applied to evaluate the in vitro synergistic activity of phytoextract-antibiotic combinations in general. Findings A number of studies have indicated that plant-derived natural compounds are capable of significantly reducing the minimum inhibitory concentration of standard antibiotics by altering drug-resistance mechanisms of B. anthracis and other superbug infection causing bacteria. Phytochemical compounds allicin, oleanolic acid, epigallocatechin gallate and curcumin and Jatropha curcas extracts were exceptional synergistic potentiators of various standard antibiotics. Conclusion Considering these facts, phytochemicals represents a valuable and novel source of bioactive compounds with potent antibiotic synergism to modulate bacterial drug-resistance.
Collapse
Affiliation(s)
- Mackingsley Kushan Dassanayake
- School of Pharmacy, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia.
| | - Teng-Jin Khoo
- School of Pharmacy, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia
| | - Jia An
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
24
|
Ke S, Pollock NR, Wang XW, Chen X, Daugherty K, Lin Q, Xu H, Garey KW, Gonzales-Luna AJ, Kelly CP, Liu YY. Integrating gut microbiome and host immune markers to understand the pathogenesis of Clostridioides difficile infection. Gut Microbes 2021; 13:1-18. [PMID: 34132169 PMCID: PMC8210874 DOI: 10.1080/19490976.2021.1935186] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Clostridioides difficile (C.difficile) infection is the most common cause of healthcare-associated infection and an important cause of morbidity and mortality among hospitalized patients. A comprehensive understanding of C.difficile infection (CDI) pathogenesis is crucial for disease diagnosis, treatment, and prevention. Here, we characterized gut microbial compositions and a broad panel of innate and adaptive immunological markers in 243 well-characterized human subjects (including 187 subjects with both microbiota and immune marker data), who were divided into four phenotype groups: CDI, Asymptomatic Carriage, Non-CDI Diarrhea, and Control. We found that the interactions between gut microbiota and host immune markers are very sensitive to the status of C.difficile colonization and infection. We demonstrated that incorporating both gut microbiome and host immune marker data into classification models can better distinguish CDI from other groups than can either type of data alone. Our classification models display robust diagnostic performance to differentiate CDI from Asymptomatic carriage (AUC~0.916), Non-CDI Diarrhea (AUC~0.917), or Non-CDI that combines all other three groups (AUC~0.929). Finally, we performed symbolic classification using selected features to derive simple mathematic formulas that explicitly quantify the interactions between the gut microbiome and host immune markers. These findings support the potential roles of gut microbiota and host immune markers in the pathogenesis of CDI. Our study provides new insights for a microbiome-immune marker-derived signature to diagnose CDI and design therapeutic strategies for CDI.
Collapse
Affiliation(s)
- Shanlin Ke
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MassachusettsUSA,School of Animal Science and Technology, State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University 330045, China
| | - Nira R. Pollock
- Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA,Department of Laboratory Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Xu-Wen Wang
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MassachusettsUSA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Kaitlyn Daugherty
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Qianyun Lin
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Hua Xu
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Kevin W. Garey
- Department of Pharmacy Practice and Translation Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Anne J. Gonzales-Luna
- Department of Pharmacy Practice and Translation Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Ciarán P. Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA,Ciarán P. Kelly Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MassachusettsUSA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MassachusettsUSA,CONTACT Yang-Yu Liu Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MassachusettsUSA
| |
Collapse
|
25
|
Music of metagenomics-a review of its applications, analysis pipeline, and associated tools. Funct Integr Genomics 2021; 22:3-26. [PMID: 34657989 DOI: 10.1007/s10142-021-00810-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/25/2021] [Accepted: 10/03/2021] [Indexed: 10/20/2022]
Abstract
This humble effort highlights the intricate details of metagenomics in a simple, poetic, and rhythmic way. The paper enforces the significance of the research area, provides details about major analytical methods, examines the taxonomy and assembly of genomes, emphasizes some tools, and concludes by celebrating the richness of the ecosystem populated by the "metagenome."
Collapse
|
26
|
Arthithanyaroj S, Chankhamhaengdecha S, Chaisri U, Aunpad R, Aroonnual A. Effective inhibition of Clostridioides difficile by the novel peptide CM-A. PLoS One 2021; 16:e0257431. [PMID: 34516580 PMCID: PMC8437281 DOI: 10.1371/journal.pone.0257431] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
Clostridioides difficile infection is the most common cause of nosocomial and antibiotic-associated diarrhea. C. difficile treatment is increasingly likely to fail, and the recurrence rate is high. Antimicrobial peptides are considered an alternative treatment for many infectious diseases, including those caused by antibiotic resistant bacteria. In the present study, we identified a CM peptide, a hybrid of cecropin A and melittin, and its derivative which possesses potent antimicrobial activity against C. difficile strain 630. CM peptide exhibited antibacterial activity with minimum inhibitory concentration of 3.906 μg/ml (2.21 μM). A modified derivative of CM, CM-A, exhibited even greater activity with a minimum inhibitory concentration of 1.953 μg/ml (1.06 μM) and a minimum bactericidal concentration of 7.8125 μg/ml (4.24 μM), which indicates that CM-A peptide is more efficient than its parent peptide. A fluorescence-activated cell sorter analysis revealed that the membrane of C. difficile 630 could be an important target for CM-A. This peptide induced high levels of cell depolarization and cell permeability on C. difficile cell membrane. Moreover, electron microscopy imaging showed that CM-A interferes with the C. difficile cell membrane. Hence, the antimicrobial peptide CM-A may represent a promising novel approach for the treatment of C. difficile infections.
Collapse
Affiliation(s)
- Sirirak Arthithanyaroj
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Ratchaneewan Aunpad
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| | - Amornrat Aroonnual
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
27
|
Păduraru DN, Ion D, Dumitrașcu MC, Petca R, Petca A, Șandru F, Andronic O, Radu G, Bolocan A. Clostridium difficile infection characteristics in a general surgery clinic. Exp Ther Med 2021; 22:1112. [PMID: 34504566 DOI: 10.3892/etm.2021.10546] [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: 05/25/2021] [Accepted: 06/23/2021] [Indexed: 11/05/2022] Open
Abstract
Clostridium difficile (CD) is an anaerobic, gram-positive bacterium that can produce a spectrum of gastrointestinal diseases ranging from pseudomembranous colitis to diarrhea to toxic megacolon. The infection is even more difficult to manage as CD produces high-end spores, suggesting that this may be the cause of the dangerous recurrent disease as well as dissemination among healthy members in the community. Spores can be hosted in the digestive tract of both symptomatic and asymptomatic patients. The most relevant risk factor in the development of Clostridium difficile infection (CDI) seems to be the overuse of antimicrobials. Comorbidities are another risk factor that may predispose towards more serious CDI. Treatment options vary from oral antibiotics to extensive surgical interventions. The present study aimed to analyze the prevalence, severity, and management of CDIs in a general surgery department in an effort to determine the correlative elements between the infection and surgical pathology.
Collapse
Affiliation(s)
- Dan Nicolae Păduraru
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Third Clinic of General Surgery and Emergency, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Daniel Ion
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Third Clinic of General Surgery and Emergency, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Mihai Cristian Dumitrașcu
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Gynecology, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Răzvan Petca
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Urology, 'Prof. Dr. Th. Burghele' Hospital, 061344 Bucharest, Romania
| | - Aida Petca
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Gynecology, 'Elias' University Emergency Hospital, 011461 Bucharest, Romania
| | - Florica Șandru
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Dermatology, 'Elias' University Emergency Hospital, 011461 Bucharest, Romania
| | - Octavian Andronic
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Third Clinic of General Surgery and Emergency, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Georgiana Radu
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Third Clinic of General Surgery and Emergency, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Alexandra Bolocan
- Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Third Clinic of General Surgery and Emergency, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| |
Collapse
|
28
|
Bile Salt Hydrolases: At the Crossroads of Microbiota and Human Health. Microorganisms 2021; 9:microorganisms9061122. [PMID: 34067328 PMCID: PMC8224655 DOI: 10.3390/microorganisms9061122] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota has been increasingly linked to metabolic health and disease over the last few decades. Several factors have been suggested to be involved in lipid metabolism and metabolic responses. One mediator that has gained great interest as a clinically important enzyme is bile salt hydrolase (BSH). BSH enzymes are widely distributed in human gastrointestinal microbial communities and are believed to play key roles in both microbial and host physiology. In this review, we discuss the current evidence related to the role of BSHs in health and provide useful insights that may pave the way for new therapeutic targets in human diseases.
Collapse
|
29
|
Abstract
Clostridium difficile (C. difficile) is a kind of obligate anaerobic gram-positive Bacillus related with intestinal diseases and antibiotic treatment. In present study, the C. difficile genome was studied employing met genomic technology. Genome sequencing identified C. difficile LCL126 has total size of 4,301,949 bp with a 27.97% of GC content. Specifically, 4119 predicted coding genes, 188 repeat sequences, 13 prophages and 8 gene islands were detected. Additionally, gene function analysis aspect of the function annotation, effector, and virulence were concluded that total of 3367 cluster of orthologous groups of proteins genes and classified into 24 categories, while the most outstanding class was metabolic process (1533) and catalytic activity (1498). The carbohydrate-active enzymes have been detected 127 genes, pathogenicity analysis revealed that 133 reduced and 22 increased virulence (hypervirulence) genes, while 54 unaffected and 10 loss of pathogenicity genes were found. Furthermore, perform the visualization and methylation expression were revealed that the dominant types comprised m4C, m5C, and m6C with the number of 6989, 36,666, and 3534, respectively. Overall, whole genome sequence information of C. difficile LCL126 was obtained and functional prediction was revealed its possible toxicological potential genes existence.
Collapse
Affiliation(s)
- Jianfeng Wang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China.,Laboratory of Clostridium, Lanzhou Institute of Biological Products, Lanzhou, Gansu Province, China
| | - Chu Yang
- Laboratory of Clostridium, Lanzhou Institute of Biological Products, Lanzhou, Gansu Province, China
| | - Chao Zhang
- Laboratory of Clostridium, Lanzhou Institute of Biological Products, Lanzhou, Gansu Province, China
| | - Xiaoyan Mao
- Laboratory of Clostridium, Lanzhou Institute of Biological Products, Lanzhou, Gansu Province, China
| | - An Lizhe
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
30
|
Bhaskara V, Leal MT, Seigner J, Friedrich T, Kreidl E, Gadermaier E, Tesarz M, Rogalli A, Stangl L, Wallwitz J, Hammel K, Rothbauer M, Moll H, Ertl P, Hahn R, Himmler G, Bauer A, Casanova E. Efficient production of recombinant secretory IgA against Clostridium difficile toxins in CHO-K1 cells. J Biotechnol 2021; 331:1-13. [PMID: 33689865 DOI: 10.1016/j.jbiotec.2021.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/25/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022]
Abstract
Despite the essential role secretory IgAs play in the defense against pathogenic invasion and the proposed value of recombinant secretory IgAs as novel therapeutics, currently there are no IgA-based therapies in clinics. Secretory IgAs are complex molecules and the major bottleneck limiting their therapeutic potential is a reliable recombinant production system. In this report, we addressed this issue and established a fast and robust production method for secretory IgAs in CHO-K1 cells using BAC-based expression vectors. As a proof of principle, we produced IgAs against Clostridium difficile toxins TcdA and TcdB. Recombinant secretory IgAs produced using our expression system showed comparable titers to IgGs, widely used as therapeutic biologicals. Importantly, secretory IgAs produced using our method were functional and could efficiently neutralize Clostridium difficile toxins TcdA and TcdB. These results show that recombinant secretory IgAs can be efficiently produced, thus opening the possibility to use them as therapeutic agents in clinics.
Collapse
Affiliation(s)
- Venugopal Bhaskara
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria.
| | - Maria Trinidad Leal
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Jacqueline Seigner
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Theresa Friedrich
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | - Laura Stangl
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | | | - Katharina Hammel
- Department for Biotechnology, University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Mario Rothbauer
- Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, 1060 Vienna, Austria
| | - Herwig Moll
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Ertl
- Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, 1060 Vienna, Austria
| | - Rainer Hahn
- Department for Biotechnology, University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | | | - Anton Bauer
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria; The Antibody Lab GmbH, 1210 Vienna, Austria.
| | - Emilio Casanova
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
31
|
Tawam D, Baladi M, Jungsuwadee P, Earl G, Han J. The Positive Association between Proton Pump Inhibitors and Clostridium Difficile Infection. Innov Pharm 2021; 12:10.24926/iip.v12i1.3439. [PMID: 34007671 PMCID: PMC8102963 DOI: 10.24926/iip.v12i1.3439] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are among the most commonly used medications by patients due to its availability over the counter and frequent prescribing by physicians to treat and alleviate symptoms of gastroesophageal reflux disease. Recently, the FDA issued a warning with respect to the utilization of PPIs and risk of developing Clostridium difficile infections (CDI). The most commonly known medications to cause CDI are antibiotics. However, available studies suggest an association and increase in risk for CDI with PPI use as well. OBJECTIVE The purpose of this research is to review and summarize data currently available on the association between PPIs and CDI. METHODS To search for eligible studies, EBSCO engines were investigated using proton pump inhibitors or PPIs and Clostridium difficile or C. diff. as search terms. Meta analyses and systematic reviews published between 2000 and 2020 on adult patients were considered. RESULTS Eight meta-analyses and systematic reviews met the inclusion criteria. They included studies conducted in the US, Europe, Asia and Canada on inpatient and outpatient adults. The final result for all 8 studies showed a statistically significant association between PPIs and CDI ranging from mild to high risk. CONCLUSION Currently available data suggest a positive association between PPIs and CDI.
Collapse
Affiliation(s)
- Dania Tawam
- Fairleigh Dickinson University School of Pharmacy and Health Sciences
| | - Michael Baladi
- Fairleigh Dickinson University School of Pharmacy and Health Sciences
| | | | - Grace Earl
- Fairleigh Dickinson University School of Pharmacy and Health Sciences
| | - Jayoung Han
- Fairleigh Dickinson University School of Pharmacy and Health Sciences
| |
Collapse
|
32
|
AAV-mediated delivery of actoxumab and bezlotoxumab results in serum and mucosal antibody concentrations that provide protection from C. difficile toxin challenge. Gene Ther 2021; 30:455-462. [PMID: 33608675 DOI: 10.1038/s41434-021-00236-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/16/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022]
Abstract
Clostridium difficile is the leading cause of antibiotic-associated nosocomial diarrhea in the developed world. When the host-associated colon microbiome is disrupted by the ingestion of antibiotics, C. difficile spores can germinate, resulting in infection. C. difficile secretes enterotoxin A (TcdA) and cytotoxin B (TcdB) that are responsible for disease pathology. Treatment options are limited as the bacterium demonstrates resistance to many antibiotics, and even with antibacterial therapies, recurrences of C. difficile are common. Actotoxumab and bezlotoxumab are human monoclonal antibodies that bind and neutralize TcdA and TcdB, respectively. In 2016, the US food and drug administration (FDA) approved bezlotoxumab for use in the prevention of C. difficile infection recurrence. To ensure the long-term expression of antibodies, gene therapy can be used. Here, adeno-associated virus (AAV)6.2FF, a novel triple mutant of AAV6, was engineered to express either actotoxumab or bezlotoxumab in mice and hamsters. Both antibodies expressed at greater than 90 μg/mL in the serum and were detected at mucosal surfaces in both models. Hundred percent of mice given AAV6.2FF-actoxumab survived a lethal dose of TcdA. This proof of concept study demonstrates that AAV-mediated expression of C. difficile toxin antibodies is a viable approach for the prevention of recurrent C. difficile infections.
Collapse
|
33
|
Baghani A, Alimohammadi M, Aliramezani A, Talebi M, Mesdaghinia A, Douraghi M. Isolation and characterization of a multidrug-resistant Clostridioides difficile toxinotype V from municipal wastewater treatment plant. JOURNAL OF ENVIRONMENTAL HEALTH SCIENCE & ENGINEERING 2020; 18:1281-1288. [PMID: 33312642 PMCID: PMC7721768 DOI: 10.1007/s40201-020-00546-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 09/07/2020] [Accepted: 09/21/2020] [Indexed: 06/10/2023]
Abstract
PURPOSE Wastewater treatment plant (WWTP) is regarded as a potential source for transmission of Clostridioides difficile from urban areas into the surface water, through feces of human and animals. The aim of this study was to screen and characterize the C. difficile bacteria in inlet and outlet wastewater of different WWTPs in Tehran, Iran. METHODS Totally, 72 samples were collected from three different WWTPs (inlet site and outlet sites) during a year. C. difficile was isolated and characterized in terms of toxins, toxinotype, resistance profile and genes, and colonization factors using PCR. RESULTS One C. difficile toxinotype V was isolated from the outlet samples. The isolate was susceptible to vancomycin but resistant to metronidazole, tetracycline, ciprofloxacin, and moxifloxacin using MIC Test Strips. The isolated C. difficile was toxigenic (tcdA, tcdB, cdtA, cdtB positive and CPE positive) and had tcdC-A genotype. No mutations were found in fliC and fliD. The slpA sequence type was 078 - 01. The C. difficile was positive for tetM, int, but negative for vanA, nim, and tndX genes. Mutations were not observed in gyrA and gyrB genes. CONCLUSIONS This study provided evidence of presence of a multidrug-resistant C. difficile toxinotype V in one of the municipal WWTP. The transmission of such isolate to the environment and reuse of treated wastewater by human pose a threat to human health and dissemination of antibiotic resistant bacteria which are untreatable.
Collapse
Affiliation(s)
- Akram Baghani
- Division of Microbiology, Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, PO Box: 14155-6446, Tehran, Iran
| | - Mahmood Alimohammadi
- Center for Water Quality Research (CWQR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran
- Department of Environmental Health Engineering, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Aliramezani
- Division of Microbiology, Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, PO Box: 14155-6446, Tehran, Iran
| | - Maliheh Talebi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Mesdaghinia
- Center for Water Quality Research (CWQR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran
- Department of Environmental Health Engineering, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Center for Water Quality Research (CWQR), Department of Environmental Health Engineering, School of Public Health, Institute for Environmental Research (IER), Tehran University of Medical Sciences, PO Box: 14155-6446, Tehran, Iran
| | - Masoumeh Douraghi
- Division of Microbiology, Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, PO Box: 14155-6446, Tehran, Iran
- Center for Water Quality Research (CWQR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Tanaka M, Onizuka S, Mishima R, Nakayama J. Cultural isolation of spore-forming bacteria in human feces using bile acids. Sci Rep 2020; 10:15041. [PMID: 32929101 PMCID: PMC7490687 DOI: 10.1038/s41598-020-71883-1] [Citation(s) in RCA: 13] [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: 03/19/2020] [Accepted: 07/31/2020] [Indexed: 01/01/2023] Open
Abstract
Structurally-diversified bile acids (BAs) are involved in shaping of intestinal microbiota as well as absorption of dietary lipids. Taurocholic acid, a conjugated form of BA, has been reported to be a factor triggering germination of a wide range of spore-forming bacteria in intestine. To test a hypothesis that other BAs also promote germination of intestinal bacteria, we attempted culture of bacteria from ethanol-treated feces by using a series of BAs. It was found that conjugated-BAs, notably three glycine-conjugated BAs, glycodeoxycholic acid and glycochenodeoxycholic acid, significantly increased the number and the species variety of colonies formed on the agar plate. These colonized bacteria mostly belonged to class Clostridia, mainly consisting of families Lachnospiraceae, Clostridiaceae, and Peptostreptococcaceae. There were several types of bacteria associated with different sensitivity to each BA. Eventually, we isolated 72 bacterial species of which 61 are known and 11 novel. These results demonstrate that the culturable range of bacteria in intestine can be widened using the germination-inducing activity of BAs. This approach would advance the research on spore-forming Clostridia that contains important but difficult-to-cultured bacteria associate with host health and diseases.
Collapse
Affiliation(s)
- Masaru Tanaka
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Sakura Onizuka
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Riko Mishima
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Jiro Nakayama
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
35
|
Moore JH, Salahi A, Honrado C, Warburton C, Warren CA, Swami NS. Quantifying bacterial spore germination by single-cell impedance cytometry for assessment of host microbiota susceptibility to Clostridioides difficile infection. Biosens Bioelectron 2020; 166:112440. [PMID: 32745926 DOI: 10.1016/j.bios.2020.112440] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/18/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022]
Abstract
The germination of ingested spores is often a necessary first step required for enabling bacterial outgrowth and host colonization, as in the case of Clostridioides difficile (C. difficile) infection. Spore germination rate in the colon depends on microbiota composition and its level of disruption by antibiotic treatment since secretions by commensal bacteria modulate primary to secondary bile salt levels to control germination. Assessment of C. difficile spore germination typically requires measurement of colony-forming units, which is labor intensive and takes at least 24 h to perform but is regularly required due to the high recurrence rates of nosocomial antibiotic-associated diarrhea. We present a rapid method to assess spore germination by using high throughput single-cell impedance cytometry (>300 events/s) to quantify live bacterial cells, by gating for their characteristic electrophysiology versus spores, so that germination can be assessed after just 4 h of culture at a detection limit of ~100 live cells per 50 μL sample. To detect the phenotype of germinated C. difficile bacteria, we utilize its characteristically higher net conductivity versus that of spore aggregates and non-viable C. difficile forms, which causes a distinctive high-frequency (10 MHz) impedance phase dispersion within moderately conductive media (0.8 S/m). In this manner, we can detect significant differences in spore germination rates within just 4 h, with increasing primary bile salt levels in vitro and using ex vivo microbiota samples from an antibiotic-treated mouse model to assess susceptibility to C. difficile infection. We envision a rapid diagnostic tool for assessing host microbiota susceptibility to bacterial colonization after key antibiotic treatments.
Collapse
Affiliation(s)
- John H Moore
- Electrical & Computer Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - Armita Salahi
- Electrical & Computer Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - Carlos Honrado
- Electrical & Computer Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | | | - Cirle A Warren
- Infectious Diseases, School of Medicine, University of Virginia, VA, 22904, USA
| | - Nathan S Swami
- Electrical & Computer Engineering, University of Virginia, Charlottesville, VA, 22904, USA.
| |
Collapse
|
36
|
Clostridioides difficile Enteritis Induced Anastomotic Rupture: A Case Report and Literature Review. Case Rep Surg 2020; 2020:9794823. [PMID: 32607274 PMCID: PMC7313094 DOI: 10.1155/2020/9794823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/23/2020] [Accepted: 06/11/2020] [Indexed: 12/04/2022] Open
Abstract
Background. A 76-year-old male patient who suffered small bowel anastomotic dehiscence believed to be a complication provoked by Clostridioides difficile enteritis. Case Presentation. The patient was a 76-year-old male who underwent small bowel resection with primary anastomosis for a small bowel obstruction. On postoperative day #7, he rapidly decompensated and upon return to the operating room was found to have complete anastomotic dehiscence with copious enteric spillage. The presentation appeared as if the staple line had burst open. Enteric contents confirmed the diagnosis of Clostridioides difficile enteritis. Subsequent hospital course was complicated by ventilatory-dependent respiratory failure, hemodynamic instability, and persistent anemia secondary to gastric ulcer requiring endoscopic cauterization. After a prolonged hospital course, he eventually progressed and was transferred to a skilled nursing facility on hospital day #42. Discussion. Clostridioides difficile causes inflammation and copious large volume secretions that would theoretically increase intraluminal pressures creating an internal tension. This tension along with other factors from the infection itself would likely be inhibitory of anastomotic healing. Although it is rare, Clostridioides difficile enteritis is being reported with increasing frequency, and in the setting of recent small bowel anastomosis, it should be considered a possible risk factor for anastomotic leak.
Collapse
|
37
|
Parvataneni S, Dasari AR. Zinc Level and Its Role in Recurrent Clostridium difficile Infection: A Case Report and Literature Review. J Investig Med High Impact Case Rep 2020; 8:2324709620941315. [PMID: 32646242 PMCID: PMC7357048 DOI: 10.1177/2324709620941315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Clostridium difficile infection is a common nosocomial infection in US hospitals, accounting for approximately 12 800 deaths annually in the United States. These infections are often associated with the use of antibiotics, which can alter the gut microbiome and thus render patients susceptible to C difficile infection. C difficile is often spread via fecal oral transmission. Multiple medications have been developed, but recurrence rates reach 60% after treatment. Recent data have shown that zinc supplementation decreases the recurrence of C difficile infection. In this article, we present a case of recurrent C difficile infection with zinc deficiency in which zinc supplementation improved the symptoms and reduced the incidence of recurrence.
Collapse
|
38
|
Intestinal Microbiome Changes in Fecal Microbiota Transplant (FMT) vs. FMT Enriched with Lactobacillus in the Treatment of Recurrent Clostridioides difficile Infection. Can J Gastroenterol Hepatol 2019; 2019:4549298. [PMID: 31976311 PMCID: PMC6955117 DOI: 10.1155/2019/4549298] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/19/2019] [Accepted: 11/12/2019] [Indexed: 11/17/2022] Open
Abstract
AIM In this study, we conducted a comparative study to explore the differences in therapeutic efficacy and intestinal microbiome of fecal microbiota transplant (FMT) vs. FMT in addition with Lactobacillus (FMT-L) for treatment of recurrent Clostridioides difficile infection (R-CDI). METHODS We designed a double-blinded randomized comparative two-arm pilot multicenter study to assess the efficacy and impact in the intestinal microbiome of standard capsules of FMT vs. FMT-L enriched with 3 species of Lactobacillus for patients with R-CDI. A 90-day follow-up of 21 patients was performed, starting at the beginning of the study. From the selected patients, fecal samples were obtained at days 0, 3, 7, and 28 after treatment. Fecal samples and FMT were analyzed by 16S rRNA sequencing. RESULTS We included 21 patients (13 in the FMT group and 8 in the FMT-L group). Overall, both groups had a reduction in bowel movements per day, from 8.6 to 3.2 in the first 48 h (62.7% reduction, p=0.001). No severe adverse reactions or recurrences were recorded. Firmicutes were the most abundant phylum in donors. A low relative abundance of Proteobacteria was detected and mostly found in patients even at higher proportions than the donor. The donor's pool also had relatively few Bacteroidetes, and some patients showed a higher abundance of this phylum. Based on the ANOSIM R values, there is a significant difference between the microbial communities of basal samples and samples collected on day 7 (p=0.045) and at day 28 (0.041). CONCLUSION Fecal microbiota transplant by capsules was clinically and genomically similar between traditional FMT and enriched FMT with Lactobacillus spp. Restoration of bacterial diversity and resolution of dysbiosis at days 7 and 28 were observed. Patients with a first episode of recurrence treated with FMT had an excellent response without severe adverse events; FMT should be considered as an early treatment during R-CDI.
Collapse
|
39
|
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: 1.0] [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
|
40
|
Khodaparast S, Mohabati Mobarez A, Saberifiroozi M. A Two-Step Approach for Diagnosing Glutamate Dehydrogenase Genes by Conventional Polymerase Chain Reaction from Clostridium difficile Isolates. Middle East J Dig Dis 2019; 11:135-140. [PMID: 31687111 PMCID: PMC6819966 DOI: 10.15171/mejdd.2019.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 05/10/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Clostridium difficile is the major causative agent of nosocomial antibiotic-associated colitis. The gold standard for C. difficile detection is stool culture followed by cytotoxic assay, although it is laborious and time-consuming. We developed a screening test based on a two-step conventional polymerase chain reaction (PCR) approach to detect gluD, the glutamate dehydrogenase (GDH) enzyme gene, which is a marker for screening of C. difficile. Targeting gluD comparing to the conserved stable genetic element of pathogenicity locus (PaLoc), with an accessory gene of Cdd3, was an effective method for the detection of this pathogen from patients with enterocolitis.
METHODS
Fresh fecal samples of the patients who were clinically suspicious for antibiotic-associated colitis were collected. Stool specimens were cultured on the cycloserine-cefoxitin fructose agar (CCFA) in an anaerobic condition, following alcohol shock treatment and enrichment in Clostridium difficile Brucella broth (CDBB). On confirmed colonies, PCR was carried out for detection of PaLoc subsidiary gene, Cdd3, and toxicogenic genes, tcdA and tcdB. The gluD that is GDH gene detection was performed by conventional PCR on the extracted DNA from 578 fresh stool samples.
RESULTS
57 (9.8%) strains of C. difficile were approved by conventional PCR for gluD and Cdd3 genes, in which 37 (6.4%) colonies had tcdA+/tcdB+ genotype, 2 (0.3%) tcdA+/tcdB-, 4 (0.7%) tcdA-/ tcdB+ and the remaining 14 (2.4%) colonies were tcdA and tcdB negative.
CONCLUSION
These results demonstrate that targeting gluD by PCR is quite promising for rapid detection of C. difficile from fresh fecal samples. Furthermore, the multiple-gene analysis for tcdA and tcdB assay proved a reliable approach for diagnosing of toxigenic strains among clinical samples.
Collapse
Affiliation(s)
- Sepideh Khodaparast
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University. Tehran, Iran
| | - Ashraf Mohabati Mobarez
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University. Tehran, Iran
| | - Mehdi Saberifiroozi
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Muñoz M, Restrepo-Montoya D, Kumar N, Iraola G, Camargo M, Díaz-Arévalo D, Roa-Molina NS, Tellez MA, Herrera G, Ríos-Chaparro DI, Birchenall C, Pinilla D, Pardo-Oviedo JM, Rodríguez-Leguizamón G, Josa DF, Lawley TD, Patarroyo MA, Ramírez JD. Integrated genomic epidemiology and phenotypic profiling of Clostridium difficile across intra-hospital and community populations in Colombia. Sci Rep 2019; 9:11293. [PMID: 31383872 PMCID: PMC6683185 DOI: 10.1038/s41598-019-47688-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/22/2019] [Indexed: 02/06/2023] Open
Abstract
Clostridium difficile, the causal agent of antibiotic-associated diarrhea, has a complex epidemiology poorly studied in Latin America. We performed a robust genomic and phenotypic profiling of 53 C. difficile clinical isolates established from diarrheal samples from either intrahospital (IH) or community (CO) populations in central Colombia. In vitro tests were conducted to evaluate the cytopathic effect, the minimum inhibitory concentration of ten antimicrobial agents, the sporulation efficiency and the colony forming ability. Eleven different sequence types (STs) were found, the majority present individually in each sample, however in three samples two different STs were isolated. Interestingly, CO patients were infected with STs associated with hypervirulent strains (ST-1 in Clade-2). Three coexistence events (two STs simultaneously detected in the same sample) were observed always involving ST-8 from Clade-1. A total of 2,502 genes were present in 99% of the isolates with 95% of identity or more, it represents a core genome of 28.6% of the 8,735 total genes identified in the set of genomes. A high cytopathic effect was observed for the isolates positive for the two main toxins but negative for binary toxin (TcdA+/TcdB+/CDT- toxin production type), found only in Clade-1. Molecular markers conferring resistance to fluoroquinolones (cdeA and gyrA) and to sulfonamides (folP) were the most frequent in the analyzed genomes. In addition, 15 other markers were found mostly in Clade-2 isolates. These results highlight the regional differences that C. difficile isolates display, being in this case the CO isolates the ones having a greater number of accessory genes and virulence-associated factors.
Collapse
Affiliation(s)
- Marina Muñoz
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
- Posgrado Interfacultades Doctorado en Biotecnología, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Daniel Restrepo-Montoya
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
- Genomics and Bioinformatics Department, North Dakota State University, Fargo, North Dakota, USA
| | - Nitin Kumar
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Gregorio Iraola
- Microbial Genomics Laboratory, Institut Pasteur Montevideo, Montevideo, Uruguay
- Center for Integrative Biology, Universidad Mayor, Santiago de Chile, Chile
| | - Milena Camargo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Diana Díaz-Arévalo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- Faculty of Animal Sciences, Universidad de Ciencias Aplicadas y Ambientales (UDCA), Bogotá, Colombia
- Hygea group, Faculty of Health Sciences, Universidad de Boyacá, Tunja, Colombia
| | - Nelly S Roa-Molina
- Centro de Investigaciones Odontológicas, Facultad de Odontología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Mayra A Tellez
- Centro de Investigaciones Odontológicas, Facultad de Odontología, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Giovanny Herrera
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
- PhD Programme in Biomedical and Biological Sciences, Faculty of Natural Sciences and Mathematics/School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Dora I Ríos-Chaparro
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Claudia Birchenall
- Hospital Universitario Mayor - Méderi, Universidad del Rosario, Bogotá, Colombia
| | - Darío Pinilla
- Hospital Universitario Mayor - Méderi, Universidad del Rosario, Bogotá, Colombia
| | - Juan M Pardo-Oviedo
- Hospital Universitario Mayor - Méderi, Universidad del Rosario, Bogotá, Colombia
| | | | | | - Trevor D Lawley
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Manuel A Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
42
|
Nichols RG, Peters JM, Patterson AD. Interplay Between the Host, the Human Microbiome, and Drug Metabolism. Hum Genomics 2019; 13:27. [PMID: 31186074 PMCID: PMC6558703 DOI: 10.1186/s40246-019-0211-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
The human microbiome is composed of four major areas including intestinal, skin, vaginal, and oral microbiomes, with each area containing unique species and unique functionalities. The human microbiome may be modulated with prebiotics, probiotics, and postbiotics to potentially aid in the treatment of diseases like irritable bowel syndrome, bacterial vaginosis, atopic dermatitis, gingivitis, obesity, or cancer. There is also potential for many of the inhabitants of the human microbiome to directly modulate host gene expression and modulate host detoxifying enzyme activity like cytochrome P450s (CYPs), dehydrogenases, and carboxylesterases. Therefore, the microbiome may be important to consider during drug discovery, risk assessment, and dosing regimens for various diseases given that the human microbiome has been shown to impact host detoxification processes.
Collapse
Affiliation(s)
- Robert G. Nichols
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA 16802 USA
| | - Jeffrey M. Peters
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA 16802 USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA 16802 USA
| |
Collapse
|
43
|
Forero AJ, Muñoz M, Camargo M, Soto-De León SC, Ríos-Chaparro DI, Birchenall C, Pinilla D, Pardo JM, Josa DF, Patarroyo MA, Ramírez JD. High frequency of toxigenic Clostridium difficile and Clostridium perfringens coinfection among diarrheic patients at health care facility-onset (HCFO) and community-onset (CO) centers in Bogotá, Colombia. Gut Pathog 2019; 11:27. [PMID: 31171939 PMCID: PMC6545673 DOI: 10.1186/s13099-019-0308-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/29/2019] [Indexed: 12/31/2022] Open
Abstract
Background The aim of this study was to evaluate the frequency of toxigenic C. difficile and C. perfringens infections at health care facility-onset (HCFO) and community-onset (CO), in two health care centers (HCC) in Bogotá, Colombia. A total of 220 stool samples from patients presenting diarrhea acquired at HCFO or CO were analyzed by several PCR tests. Results We found that 65.5% (n = 144) of the population had C. difficile infection, followed by toxigenic C. difficile with 57.3% (n = 126), and finally toxigenic C. perfringens with a frequency of 32.7% (n = 72). Conclusions This study is the first molecular detection and characterization of C. difficile and C. perfringens in HCFO and CO in Latin America and demonstrates a relevant frequency of these two species, including coinfection and strikingly diverse toxigenic profiles, especially in the CO.
Collapse
Affiliation(s)
- Alex J Forero
- 1Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24 # 63C - 69, Bogotá, Colombia.,2Especialización en Microbiología Médica, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Marina Muñoz
- 1Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24 # 63C - 69, Bogotá, Colombia.,3Posgrado Interfacultades Doctorado en Biotecnología, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Milena Camargo
- 4Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Sara C Soto-De León
- 5Laboratorio de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería, Córdoba Colombia
| | - Dora I Ríos-Chaparro
- 1Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24 # 63C - 69, Bogotá, Colombia
| | | | - Darío Pinilla
- Hospital Universitario Mayor-Méderi, Bogotá, Colombia
| | - Juan M Pardo
- Hospital Universitario Mayor-Méderi, Bogotá, Colombia
| | | | - Manuel A Patarroyo
- 4Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,8School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Juan D Ramírez
- 1Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24 # 63C - 69, Bogotá, Colombia
| |
Collapse
|
44
|
Adejumo AC, Akanbi O, Pani L. Among inpatients, ischemic bowel disease predisposes to Clostridium difficile infection with concomitant higher mortality and worse outcomes. Eur J Gastroenterol Hepatol 2019; 31:109-115. [PMID: 30513074 DOI: 10.1097/meg.0000000000001273] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Clinical conditions resulting in hypoxia, hypoperfusion, anaerobic milieu within the gut, and intestinal epithelial breakdown, such as seen in heart failure, precipitates Clostridium difficile infection (CDI). Given that ischemic bowel disease (IB) typically results in similar changes within the gut, we investigated the relationship between CDI and IB, and the impact of CDI on the clinical outcomes of IB. PATIENTS AND METHODS We initially performed a cross-sectional analysis on the 2014 Healthcare Cost and Utilization Project - Nationwide Inpatient Sample (NIS) patient's discharge records of ages 18 years and older, by estimating the crude and adjusted odds ratio (aOR) of CDI and IB as the outcome and predictor respectively. We then pooled data from the 2012-2014 NIS, identified, and compared mortality (and 15 other outcomes) between three groups: IB+CDI, IB-alone, and CDI-alone (Statistical Analysis System 9.4). RESULTS In the 2014 NIS, records with IB (n=27 609), had higher rate and odds of CDI [3.95 vs. 1.17%, aOR: 1.89 (1.77-2.02)] than records without IB (n=5 879 943). The 2012-2014 NIS contained 1105 IB+CDI, 30 960 IB-alone, and 60 758 CDI-alone groups. IB+CDI had higher mortality [aOR: 1.44 (1.11-1.86)], length of stay [9.59 (9.03-10.20) vs. 6.12 (5.99-6.26) days], cost [$93 257 (82 892-104 919) vs. $63 257 (61 029-65 567)], unfavorable discharge disposition [aOR: 2.24 (1.91-2.64)] and poorer results across most of the other outcomes than IB-alone. Comparable results were found for IB+CDI versus CDI-alone. CONCLUSION IB is a risk factor for CDI in hospitals. CDI is associated with higher mortality, longer length of stay, higher cost, unfavorable discharge, and many other poorer health outcomes in patients with IB.
Collapse
Affiliation(s)
- Adeyinka C Adejumo
- Department of Medicine, North Shore Medical Center, Salem
- Department of Medicine, University of Massachusetts Medical School, Worcester
- Department of Medicine, Tufts University Medical School, Boston, Massachusetts
| | - Olalekan Akanbi
- Division of Hospital Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Lydie Pani
- Department of Medicine, North Shore Medical Center, Salem
- Department of Medicine, Tufts University Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Deng H, Yang S, Zhang Y, Qian K, Zhang Z, Liu Y, Wang Y, Bai Y, Fan H, Zhao X, Zhi F. Bacteroides fragilis Prevents Clostridium difficile Infection in a Mouse Model by Restoring Gut Barrier and Microbiome Regulation. Front Microbiol 2018; 9:2976. [PMID: 30619112 PMCID: PMC6308121 DOI: 10.3389/fmicb.2018.02976] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile is currently the leading cause of nosocomial infection. Antibiotics remain the first-line therapy for C. difficile-associated diseases (CDAD), despite the risks of resistance promotion and further gut microbiota perturbation. Notably, the abundance of Bacteroides fragilis was reported to be significantly decreased in CDAD patients. This study aimed to clarify the prophylactic effects of B. fragilis strain ZY-312 in a mouse model of C. difficile infection (CDI). The CDI mouse model was successfully created using C. difficile strain VPI 10463 spores, as confirmed by lethal diarrhea (12.5% survival rate), serious gut barrier disruption, and microbiota disruption. CDI model mice prophylactically treated with B. fragilis exhibited significantly higher survival rates (100% in low dosage group, 87.5% in high dosage group) and improved clinical manifestations. Histopathological analysis of colon and cecum tissue samples revealed an intact gut barrier with strong ZO-1 and Muc-2 expression. The bacterial diversity and relative abundance of gut microbiota were significantly improved. Interestingly, the relative abundance of Akkermansia muciniphila was positively correlated with B. fragilis treatment. In vitro experiments showed that B. fragilis inhibited C. difficile adherence, and attenuated the decrease in CDI-induced transepithelial electrical resistance, ZO-1 and MUC-2 loss, and apoptosis, suggesting that B. fragilis protected against CDI possibly by resisting pathogen colonization and improving gut barrier integrity and functions. In summary, B. fragilis exerted protective effects on a CDI mouse model by modulating gut microbiota and alleviating barrier destruction, thereby relieving epithelial stress and pathogenic colitis triggered by C. difficile. This study provides an alternative preventative measure for CDI and lays the foundations for further investigations of the relationships among opportunistic pathogens, commensal microbiota, and the gut barrier.
Collapse
Affiliation(s)
- Huimin Deng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Siqi Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yucheng Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Qian
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhaohui Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yangyang Liu
- Guangzhou ZhiYi Biotechnology Co., Ltd., Guangzhou, China
| | - Ye Wang
- Guangzhou ZhiYi Biotechnology Co., Ltd., Guangzhou, China
| | - Yang Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongying Fan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xinmei Zhao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
46
|
Kleerebezem M, Binda S, Bron PA, Gross G, Hill C, van Hylckama Vlieg JE, Lebeer S, Satokari R, Ouwehand AC. Understanding mode of action can drive the translational pipeline towards more reliable health benefits for probiotics. Curr Opin Biotechnol 2018; 56:55-60. [PMID: 30296737 DOI: 10.1016/j.copbio.2018.09.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/28/2018] [Accepted: 09/07/2018] [Indexed: 12/18/2022]
Abstract
The different levels of knowledge described in a translational pipeline (the connection of molecular mechanisms with pre-clinical physiological and human health effects) are not complete for many probiotics. At present, we are not in a position to fully understand the mechanistic basis of many well established probiotic health benefits which, in turn, limits our ability to use mechanisms to predict which probiotics are likely to be effective in any given population. Here we suggest that this concept of a translation pipeline connecting mechanistic insights to probiotic efficacy can support the selection and production of improved probiotic products. Such a conceptual pipeline would also provide a framework for the design of clinical trials to convincingly demonstrate the benefit of probiotics to human health in well-defined subpopulations.
Collapse
Affiliation(s)
- Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands.
| | - Sylvie Binda
- Danone Nutricia Research, Centre Daniel Carasso, Palaiseau, France
| | | | - Gabriele Gross
- Innovative Health Sciences, Reckitt Benckiser, Nijmegen, The Netherlands
| | - Colin Hill
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Reetta Satokari
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Arthur C Ouwehand
- Global Health and Nutrition Sciences, DuPont Nutrition and Health, Kantvik, Finland
| |
Collapse
|
47
|
Phothichaisri W, Ounjai P, Phetruen T, Janvilisri T, Khunrae P, Singhakaew S, Wangroongsarb P, Chankhamhaengdecha S. Characterization of Bacteriophages Infecting Clinical Isolates of Clostridium difficile. Front Microbiol 2018; 9:1701. [PMID: 30108562 PMCID: PMC6079236 DOI: 10.3389/fmicb.2018.01701] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022] Open
Abstract
Clostridium difficile is recognized as a problematic pathogen, causing severe enteric diseases including antibiotic-associated diarrhea and pseudomembranous colitis. The emergence of antibiotic resistant C. difficile has driven a search for alternative anti-infection modalities. A promising strategy for controlling bacterial infection includes the use of bacteriophages and their gene products. Currently, knowledge of phages active against C. difficile is still relatively limited by the fact that the isolation of phages for this organism is a technically demanding method since bacterial host themselves are difficult to culture. To isolate and characterize phages specific to C. difficile, a genotoxic agent, mitomycin C, was used to induce temperate phages from 12 clinical isolates of C. difficile. Five temperate phages consisting of ΦHR24, ΦHN10, ΦHN16-1, ΦHN16-2, and ΦHN50 were successfully induced and isolated. Spotting assays were performed against a panel of 92 C. difficile isolates to screen for susceptible bacterial hosts. The results revealed that all the C. difficile phages obtained in this work displayed a relatively narrow host range of 0-6.5% of the tested isolates. Electron microscopic characterization revealed that all isolated phages contained an icosahedral head connected to a long contractile tail, suggesting that they belonged to the Myoviridae family. Restriction enzyme analysis indicated that these phages possess unique double-stranded DNA genome. Further electron microscopic characterization revealed that the ΦHN10 absorbed to the bacterial surface via attachment to cell wall, potentially interacting with S-layer protein. Bacteriophages isolated from this study could lead to development of novel therapeutic agents and detection strategies for C. difficile.
Collapse
Affiliation(s)
- Wichuda Phothichaisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Puey Ounjai
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Tanaporn Phetruen
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Pongsak Khunrae
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Sombat Singhakaew
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Piyada Wangroongsarb
- Department of Medical Sciences, National Institute of Health, Ministry of Public Health, Nonthaburi, Thailand
| | | |
Collapse
|
48
|
Abstract
Germination of Clostridium difficile spores is a crucial early requirement for colonization of the gastrointestinal tract. Likewise, C. difficile cannot cause disease pathologies unless its spores germinate into metabolically active, toxin-producing cells. Recent advances in our understanding of C. difficile spore germination mechanisms indicate that this process is both complex and unique. This review defines unique aspects of the germination pathways of C. difficile and compares them to those of two other well-studied organisms, Bacillus anthracis and Clostridium perfringensC. difficile germination is unique, as C. difficile does not contain any orthologs of the traditional GerA-type germinant receptor complexes and is the only known sporeformer to require bile salts in order to germinate. While recent advances describing C. difficile germination mechanisms have been made on several fronts, major gaps in our understanding of C. difficile germination signaling remain. This review provides an updated, in-depth summary of advances in understanding of C. difficile germination and potential avenues for the development of therapeutics, and discusses the major discrepancies between current models of germination and areas of ongoing investigation.
Collapse
|
49
|
Uzal FA, Navarro MA, Li J, Freedman JC, Shrestha A, McClane BA. Comparative pathogenesis of enteric clostridial infections in humans and animals. Anaerobe 2018; 53:11-20. [PMID: 29883627 DOI: 10.1016/j.anaerobe.2018.06.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/03/2018] [Accepted: 06/04/2018] [Indexed: 02/06/2023]
Abstract
Several enteric clostridial diseases can affect humans and animals. Of these, the enteric infections caused by Clostridium perfringens and Clostridium difficile are amongst the most prevalent and they are reviewed here. C. perfringens type A strains encoding alpha toxin (CPA) are frequently associated with enteric disease of many animal mammalian species, but their role in these diseased mammals remains to be clarified. C. perfringens type B encoding CPA, beta (CPB) and epsilon (ETX) toxins causes necro-hemorrhagic enteritis, mostly in sheep, and these strains have been recently suggested to be involved in multiple sclerosis in humans, although evidence of this involvement is lacking. C. perfringens type C strains encode CPA and CPB and cause necrotizing enteritis in humans and animals, while CPA and ETX producing type D strains of C. perfringens produce enterotoxemia in sheep, goats and cattle, but are not known to cause spontaneous disease in humans. The role of C. perfringens type E in animal or human disease remains poorly defined. The newly revised toxinotype F encodes CPA and enterotoxin (CPE), the latter being responsible for food poisoning in humans, and the less prevalent antibiotic associated and sporadic diarrhea. The role of these strains in animal disease has not been fully described and remains controversial. Another newly created toxinotype, G, encodes CPA and necrotic enteritis toxin B-like (NetB), and is responsible for avian necrotic enteritis, but has not been associated with human disease. C. difficile produces colitis and/or enterocolitis in humans and multiple animal species. The main virulence factors of this microorganism are toxins A, B and an ADP-ribosyltransferase (CDT). Other clostridia causing enteric diseases in humans and/or animals are Clostridium spiroforme, Clostridium piliforme, Clostridium colinum, Clostridium sordellii, Clostridium chauvoei, Clostridium septicum, Clostridium botulinum, Clostridium butyricum and Clostridium neonatale. The zoonotic transmission of some, but not all these clostridsial species, has been demonstrated.
Collapse
Affiliation(s)
- Francisco A Uzal
- California Animal Health and Food Safety Laboratory System, San Bernardino Branch, University of California, Davis, CA, USA.
| | - Mauricio A Navarro
- California Animal Health and Food Safety Laboratory System, San Bernardino Branch, University of California, Davis, CA, USA
| | - Jihong Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John C Freedman
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Archana Shrestha
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bruce A McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
50
|
Arroyo LG, Gomez DE, Martins C. Equine duodenitis-proximal jejunitis: A review. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2018; 59:510-517. [PMID: 29904204 PMCID: PMC5901841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Duodenitis-proximal jejunitis (DPJ) is an inflammatory process of the proximal part of the small intestine and occurs sporadically in horses. It is clinically characterized by an acute onset of ileus and nasogastric reflux leading to systemic signs of toxemia. This review discusses the definition of the disease, potential etiologic agents, clinical findings, epidemiological features, histopathologic and clinico-pathological findings, and medical management of this condition. Salmonella spp., mycotoxins, Clostridium perfringens, and Clostridium difficile have all been associated with the disease but there is limited supporting evidence for any agent other than C. difficile. Particular attention, however, was given to etiological investigations and the data available to support the proposed etiological agents. The potential role of C. difficile as the etiological agent of DPJ, possible pathogenesis, and recent efforts to support this hypothesis are highlighted, but it is recognized that there could be more than one agent that causes the disease.
Collapse
Affiliation(s)
- Luis G Arroyo
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1 (Arroyo, Martins); Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32608, USA (Gomez)
| | - Diego E Gomez
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1 (Arroyo, Martins); Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32608, USA (Gomez)
| | - Candace Martins
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1 (Arroyo, Martins); Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32608, USA (Gomez)
| |
Collapse
|