1
|
Tsai CE, Wang FQ, Yang CW, Yang LL, Nguyen TV, Chen YC, Chen PY, Hwang IS, Ting SY. Surface-mediated bacteriophage defense incurs fitness tradeoffs for interbacterial antagonism. EMBO J 2025; 44:2473-2500. [PMID: 40065098 DOI: 10.1038/s44318-025-00406-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 05/04/2025] Open
Abstract
Bacteria in polymicrobial habitats are constantly exposed to biotic threats from bacteriophages (or "phages"), antagonistic bacteria, and predatory eukaryotes. These antagonistic interactions play crucial roles in shaping the evolution and physiology of bacteria. To survive, bacteria have evolved mechanisms to protect themselves from such attacks, but the fitness costs of resisting one threat and rendering bacteria susceptible to others remain unappreciated. Here, we examined the fitness consequences of phage resistance in Salmonella enterica, revealing that phage-resistant variants exhibited significant fitness loss upon co-culture with competitor bacteria. These phage-resistant strains display varying degrees of lipopolysaccharide (LPS) deficiency and increased susceptibility to contact-dependent interbacterial antagonism, such as the type VI secretion system (T6SS). Utilizing mutational analyses and atomic force microscopy, we show that the long-modal length O-antigen of LPS serves as a protective barrier against T6SS-mediated intoxication. Notably, this competitive disadvantage can also be triggered independently by phages possessing LPS-targeting endoglycosidase in their tail spike proteins, which actively cleave the O-antigen upon infection. Our findings reveal two distinct mechanisms of phage-mediated LPS modifications that modulate interbacterial competition, shedding light on the dynamic microbial interplay within mixed populations.
Collapse
Affiliation(s)
- Chia-En Tsai
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, 11490, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Feng-Qi Wang
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Chih-Wen Yang
- Institute of Physics, Academia Sinica, Taipei, 115201, Taiwan
| | - Ling-Li Yang
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Thao Vp Nguyen
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Yung-Chih Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Po-Yin Chen
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, 11490, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Ing-Shouh Hwang
- Institute of Physics, Academia Sinica, Taipei, 115201, Taiwan
| | - See-Yeun Ting
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, 11490, Taiwan.
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan.
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 106319, Taiwan.
| |
Collapse
|
2
|
Ike KA, Omaliko PC, Fernander MC, Sanders BM, Enikuomehin JM, Alabi JO, Adelusi OO, Kholif AE, Thomas MD, Anele UY. Evaluating the Effect of an Essential Oil Blend on the Growth and Fitness of Gram-Positive and Gram-Negative Bacteria. BIOLOGY 2025; 14:437. [PMID: 40282302 PMCID: PMC12024878 DOI: 10.3390/biology14040437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
The increasing prevalence of antibiotic-resistant bacteria has necessitated the exploration of alternative antimicrobial agents, particularly natural products like essential oils. This study investigated the antibacterial potential of a unique blend of four essential oils (EOB) across a gradient of concentrations (0.1 to 50%) against Gram-positive and Gram-negative bacteria using an adapted broth microdilution method, minimum inhibitory concentrations (MICs), and 24-h growth assays. The Gram-positive bacteria were Staphylococcus epidermidis and Bacillus subtilis, while the Gram-negative bacteria were Escherichia coli and Klebsiella aerogenes. The results demonstrated that the EOB exerted a concentration-dependent inhibitory effect on bacterial growth, with MICs determined at 25% for all the species tested. Growth curve analysis revealed that lower concentrations of the EOB (0.1 to 0.78%) allowed for normal bacterial proliferation, while at intermediate concentrations (1.56 to 3.13%), inconsistent trends in growth were exhibited. At higher concentrations (25 and 50%), the EOB effectively halted bacterial growth, as indicated by flat growth curves. The increase in the lag phase and the decrease in the growth rate at a sub-MIC concentration (12.5%) suggest a significant effect on bacterial adaptation and survival. Relative fitness analyses further highlighted the inhibitory effects of higher essential oil concentrations. S. epidermidis and E. coli had a significant (p < 0.05) reduction in fitness starting from the 6.25% concentration, while the other two species experienced a significant (p < 0.001) reduction in relative fitness from a concentration of 12.5%. These findings underscore the potential of this EOB as an effective antimicrobial agent, particularly in the context of rising antibiotic resistance. Furthermore, the study suggests that the EOB used in the present study could be integrated into therapeutic strategies as a natural alternative or adjunct to traditional antibiotics, offering a promising avenue for combating resistant bacterial strains.
Collapse
Affiliation(s)
- Kelechi A. Ike
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (K.A.I.); (P.C.O.); (J.M.E.); (J.O.A.); (O.O.A.); (A.E.K.)
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (M.C.F.); (B.M.S.); (M.D.T.)
| | - Paul C. Omaliko
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (K.A.I.); (P.C.O.); (J.M.E.); (J.O.A.); (O.O.A.); (A.E.K.)
| | - Mizpha C. Fernander
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (M.C.F.); (B.M.S.); (M.D.T.)
| | - Brittany M. Sanders
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (M.C.F.); (B.M.S.); (M.D.T.)
| | - James M. Enikuomehin
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (K.A.I.); (P.C.O.); (J.M.E.); (J.O.A.); (O.O.A.); (A.E.K.)
| | - Joel O. Alabi
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (K.A.I.); (P.C.O.); (J.M.E.); (J.O.A.); (O.O.A.); (A.E.K.)
| | - Oludotun O. Adelusi
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (K.A.I.); (P.C.O.); (J.M.E.); (J.O.A.); (O.O.A.); (A.E.K.)
| | - Ahmed E. Kholif
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (K.A.I.); (P.C.O.); (J.M.E.); (J.O.A.); (O.O.A.); (A.E.K.)
| | - Misty D. Thomas
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (M.C.F.); (B.M.S.); (M.D.T.)
| | - Uchenna Y. Anele
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA; (K.A.I.); (P.C.O.); (J.M.E.); (J.O.A.); (O.O.A.); (A.E.K.)
| |
Collapse
|
3
|
Hou X, Wang M, Hu T, Wu Z, Liang H, Zhong Y, Ma Z, Zhang H, Xiao L, Zhang W, Zou Y. Evaluation of the safety and probiotic properties of Limosilactobacillus fermentum BGI-AF16, a uric acid-lowering probiotic strain. Microb Pathog 2025; 201:107382. [PMID: 39961375 DOI: 10.1016/j.micpath.2025.107382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 01/08/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
Some beneficial microorganisms in the intestine have the potential to degrade uric acid, offering a novel strategy for the prevention of hyperuricemia. In this study, the safety and probiotic potentials of Limosilactobacillus fermentum BGI-AF16 were evaluated by whole genome sequence analysis and in vitro experiments. Based on the gene analysis of antibiotic resistance and virulence factors, L. fermentum BGI-AF16 has been shown to be safe. We identified probiotic-related genes by genome annotation tools and conducted in vitro experiments to evaluate the ability of L. fermentum BGI-AF16 to inhibit pathogenic bacteria, tolerate a simulated gastrointestinal environment, and degrade uric acid. The results from in vitro experiments showed that L. fermentum BGI-AF16 had inhibitory effects on four clinically relevant pathogens and was highly tolerant to the gastrointestinal environment. In addition, L. fermentum BGI-AF16 was able to rapidly degrade uric acid within the first hour, and the strain could degrade 56.36 ± 2.32 % of uric acid by the third hour. The genome of the strain contains genes encoding flavin adenine dinucleotide (FAD)-dependent urate hydroxylase (EC.1.14.13.113), an enzyme that directly metabolizes uric acid. And the strain has a complete uric acid metabolic pathway. These results suggest that L. fermentum BGI-AF16 is a probiotic candidate with significant potential for reducing uric acid level.
Collapse
Affiliation(s)
- Xiaoxue Hou
- BGI Research, Shenzhen, 518083, China; College of Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Mengmeng Wang
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Zhinan Wu
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Yiyi Zhong
- BGI Precision Nutrition, Shenzhen, 518083, China
| | - Zhihui Ma
- BGI Precision Nutrition, Shenzhen, 518083, China
| | | | - Liang Xiao
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China
| | - Wenjin Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; BGI Genomics, Shenzhen, 518083, China
| | - Yuanqiang Zou
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China.
| |
Collapse
|
4
|
Lacuna ARG, Dato MC, Loterio LMM, Dayrit GB, Villanueva SYAM, Lota MMM. In-Vitro Determination of Minimum Inhibitory Concentration (MIC) and Contact Time of Povidone-Iodine against Staphylococcus aureus and Klebsiella aerogenes Using Micro Suspension Test, Colorimetric Resazurin Microplate Assay, and Dey Engley Neutralizer Assay. ACTA MEDICA PHILIPPINA 2025; 59:113-124. [PMID: 40308795 PMCID: PMC12037336 DOI: 10.47895/amp.v59i4.10222] [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] [Indexed: 05/02/2025]
Abstract
Background and Objective The human nasal passages host major human pathogens. Recent research suggests that the microbial communities inhabiting the epithelial surfaces of the nasal passages play a key factor in maintaining a healthy microenvironment by affecting both resistance to pathogens and immunological responses. Colonization of the nasal cavity by different pathogens such as Staphylococcus aureus and Klebsiella aerogenes, is associated with a higher postoperative infection morbidity. Povidone-iodine (PVP-I) as an antiseptic has been proven to display high antibacterial, antiviral, and antifungal properties even at low concentrations, and was shown to be effective in the control of infections to limit their impact and spread. It can be used as a topical antiseptic for skin decontamination and wound management, as a nasal spray, or as a gargle. There are different methods in testing the efficacy of potential antimicrobial suspensions. This study aimed to determine the concentration of PVP-I that is most effective in nasal decolonization using microsuspension test and colorimetric minimum inhibitory concentration (MIC) determination assays, resazurin microtiter assay (REMA), and Dey-Engley (D/E) neutralizer assay. The findings of this study will contribute to knowledge regarding the intended use of PVP-I in microbial control, particularly in bacterial infections. Methods Several dilutions (2.0%, 1.0%, 0.5%, 0.25%, 0.1% and 0.09%) of commercially bought 10% (10 mg per 100 ml) povidone-iodine were prepared and tested against a standardized inoculum (1x105) of Staphylococcus aureus and Klebsiella aerogenes at different contact times (5 seconds, 10 seconds, 30 seconds, 1 minute, and 5 minutes). Microdilution suspension test was performed to determine the log reduction per variable, while REMA and D/E neutralizer assay were used to determine the MIC. A value of greater than or equal to 5 log reduction was considered effective for microdilution suspension test. Estimates of agreement statistics were used to interpret the results of the assay in which the overall percent agreement (OPA), positive percent agreement (PPA), negative percent agreement (NPA), and Cohen's kappa statistics were calculated. Results Povidone-iodine concentration of 0.25% exhibited ≥5 log reduction against K. aerogenes at the minimum contact time of 5 seconds. On the other hand, a slightly higher PVP-I concentration was required to achieve ≥5 log reduction for S. aureus at 0.5% concentration and a minimum contact time of 1 minute. There was an observed concordance of the results of REMA and D/E neutralizer as MIC colorimetric indicators, which yielded an overall test percent agreement of 90.30% (95% CI: 84.73-94.36), and a strong level of agreement (Κ = 0.8, p<0.0001). A lower overall percent agreement for both REMA and D/E neutralizer versus the microsuspension test was observed at 79.17% (Κ = 0.57, p<0.0001) and 78.18% (Κ = 0.55, p<0.0001), respectively. Conclusion Low povidone-iodine concentrations (i.e., 0.5% against S. aureus and 0.25% against K. aerogenes) were observed to have bactericidal activity of at least 5 log reduction as rapid as the minimum contact time of 5 seconds. Furthermore, D/E and REMA, as colorimetric indicators, had comparable performance (OPA = 90.30%; Κ = 0.8, p<0.0001) suggesting that both REMA and D/E neutralizer assay may detect the same range of minimum inhibitory concentration for the organisms and disinfectant tested in this study.
Collapse
Affiliation(s)
- Azita Racquel G Lacuna
- Department of Medical Microbiology, College of Public Health, University of the Philippines Manila
| | - Micaella C Dato
- Department of Medical Microbiology, College of Public Health, University of the Philippines Manila
| | - Loisse Mikaela M Loterio
- Department of Medical Microbiology, College of Public Health, University of the Philippines Manila
| | - Geraldine B Dayrit
- Department of Medical Microbiology, College of Public Health, University of the Philippines Manila
| | | | - Maria Margarita M Lota
- Department of Medical Microbiology, College of Public Health, University of the Philippines Manila
| |
Collapse
|
5
|
Motiwala T, Nyide B, Khoza T. Molecular dynamic simulations to assess the structural variability of ClpV from Enterobacter cloacae. FRONTIERS IN BIOINFORMATICS 2025; 5:1498916. [PMID: 40201065 PMCID: PMC11975955 DOI: 10.3389/fbinf.2025.1498916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
The Enterobacter cloacae complex (ECC) consists of six Enterobacter species (E. cloacae, hormaechei, kobei, ludwigii, nimipressuralis and asburiae) that have emerged as nosocomial pathogens of interest, with E. cloacae and Enterobacter hormachei being the most frequently isolated ECC species in human clinical specimens and intensive care unit (ICU) patients. Many nosocomial outbreaks of E. cloacae have been related to transmission through contaminated surgical equipment and operative cleaning solutions. As this pathogen evades the action of antibiotics, it is important to find alternative targets to limit the devastating effects of these pathogens. ClpV is a Clp ATPase which dissociates and recycles the contracted sheath of the bacterial type VI secretion system (T6SS), thereby regulating bacterial populations and facilitating environmental colonization. Seventy-one Enterobacter strains were mined for Clp ATPase proteins. All the investigated strains contained ClpA, ClpB, ClpX and ClpV while only 20% contained ClpK. All the investigated strains contained more than one ClpV protein, and the ClpV proteins showed significant variations. Three ClpV proteins from E. cloacae strain E3442 were then investigated to determine the structural difference between each protein. Homology modelling showed the proteins to be structurally similar to each other, however the physicochemical characteristics of the proteins vary. Additionally, physicochemical analysis and molecular dynamic simulations showed that the proteins were highly dynamic and not significantly different from each other. Further investigation of the proteins in silico and in vitro in the presence and absence of various ligands and proteins could be performed to determine whether the proteins all interact with their surroundings in the same manner. This would allow one to determine why multiple homologs of the same protein are expressed by pathogens.
Collapse
Affiliation(s)
| | | | - Thandeka Khoza
- Department of Biochemistry, School of Life Sciences, Pietermaritzburg Campus, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| |
Collapse
|
6
|
Poopedi E, Pierneef R, Singh T, Gomba A. Opportunistic bacterial pathogens in bioaerosols emitted at municipal wastewater treatment plants, South Africa. Sci Rep 2025; 15:10318. [PMID: 40133595 PMCID: PMC11937431 DOI: 10.1038/s41598-025-95484-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/21/2025] [Indexed: 03/27/2025] Open
Abstract
Aeration tanks at wastewater treatment plants (WWTPs) emit significant amounts of bioaerosols containing potentially hazardous infectious material. Occupational exposure to airborne pathogens can pose health risks to WWTP workers. Bioaerosol samples collected at aeration tanks of two typical municipal WWTPs that use different aeration modes were analysed to investigate the composition and diversity of airborne bacteria in wastewater environments, using the Illumina MiSeq platform. Thirty-six potential airborne bacterial pathogens were identified in the air samples, and these were dominated by Bacillus, Enterococcus, Clostridium, Streptococcus, Acinetobacter, Enterobacter, Pseudomonas, Bacteroides fragilis, Acinetobacter baumannii, and Escherichia/Shigella. Bioaerosols from mechanical aeration tanks (72%, 26/36) had a relatively higher richness and diversity of airborne bacterial pathogens than diffused aeration tanks (17%, 6/36). Furthermore, most of the identified airborne bacterial pathogens (78%, 28/36) were classified as Risk Group 2 according to the revised South African Regulation for Hazardous Biological Agents, 2022, and up to 70% of these were gram-negative bacteria. The presence of potentially pathogenic bacteria in the ambient air at WWTPs suggests an elevated risk of bioaerosol exposure for workers. Therefore, further research and site-specific risk assessments are recommended to guide the implementation of effective bioaerosol strategies to protect workers' health, with special attention paid to WWTPs that use mechanical aerators.
Collapse
Affiliation(s)
- Evida Poopedi
- Department of Oral and Maxillofacial Pathology, Oral Microbiology and Oral Biology, Sefako Makgatho Health Sciences University, Pretoria, 0204, South Africa.
- Department of Clinical Microbiology and Infectious Diseases, University of the Witwatersrand, Johannesburg, 2050, South Africa.
| | - Rian Pierneef
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, 0028, South Africa
- Centre for Bioinformatics and Computational Biology, University of Pretoria, Pretoria, 0028, South Africa
| | - Tanusha Singh
- National Health Laboratory Service, National Institute for Occupational Health, Johannesburg, 2000, South Africa
- Department of Environmental Health, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Annancietar Gomba
- National Health Laboratory Service, National Institute for Occupational Health, Johannesburg, 2000, South Africa
| |
Collapse
|
7
|
Chong CSC, Lau YY, Michels PAM, Lim CSY. Insights into biofilm-mediated mechanisms driving last-resort antibiotic resistance in clinical ESKAPE pathogens. Crit Rev Microbiol 2025:1-26. [PMID: 40098357 DOI: 10.1080/1040841x.2025.2473332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025]
Abstract
The rise of antibiotic-resistant bacteria poses a grave threat to global health, with the ESKAPE pathogens, which comprise Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. being among the most notorious. The World Health Organization has reserved a group of last-resort antibiotics for treating multidrug-resistant bacterial infections, including those caused by ESKAPE pathogens. This situation calls for a comprehensive understanding of the resistance mechanisms as it threatens public health and hinder progress toward the Sustainable Development Goal (SDG) 3: Good Health and Well-being. The present article reviews resistance mechanisms, focusing on emerging resistance mutations in multidrug-resistant ESKAPE pathogens, particularly against last-resort antibiotics, and describes the role of biofilm formation in multidrug-resistant ESKAPE pathogens. It discusses the latest therapeutic advances, including the use of antimicrobial peptides and CRISPR-Cas systems, and the modulation of quorum sensing and iron homeostasis, which offer promising strategies for countering resistance. The integration of CRISPR-based tools and biofilm-targeted approaches provides a potential framework for managing ESKAPE infections. By highlighting the spread of current resistance mutations and biofilm-targeted approaches, the review aims to contribute significantly to advancing our understanding and strategies in combatting this pressing global health challenge.
Collapse
Affiliation(s)
- Christina Shook Cheng Chong
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, No 1, Jalan UCSI, UCSI Heights, Taman Connaught, Cheras, Kuala Lumpur, Malaysia
| | - Yin Yin Lau
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, No 1, Jalan UCSI, UCSI Heights, Taman Connaught, Cheras, Kuala Lumpur, Malaysia
| | - Paul A M Michels
- School of Biological Sciences, University of Edinburgh, The King's Buildings, Edinburgh 3FL, UK
| | - Crystale Siew Ying Lim
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, No 1, Jalan UCSI, UCSI Heights, Taman Connaught, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Adeyemi DN, Ajide EO, Adebami GE, Abiala MA. Bacillus Genotypes Exhibit Antagonistic Effects on Lettuce-Based Enterobacter Pathotypes. Curr Microbiol 2025; 82:181. [PMID: 40057914 DOI: 10.1007/s00284-025-04163-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/25/2025] [Indexed: 03/18/2025]
Abstract
Globally, among ready-to-eat (RTE) vegetables, lettuce is well-known as a potential host for Enterobacteriaceae, which theatens public health. However, in addition to E. coli, pathogenic Enterobacter species associated with RTE lettuce is poorly investigated, as well as their genetic relationship with sources of contamination has not been determined. This study investigated the evolutionary relationship between Enterobacter species and their antibiotic resistance attributes in RTE lettuce, irrigation water, and the soil of lettuce farms. Enterobacter species had the highest occurrence in the irrigation water (38%), followed by both the RTE lettuce (31%) and the soil of the lettuce farm (31%). The 16S rRNA-identified the bacteria isolates as Enterobacter species. Phylogenetic analysis revealed that Enterobacter species from RTE lettuce and irrigation water maintained a strong evolutionary relationship in comparison with those from the soil of the lettuce farm. In addition, irrespective of the source of contamination, the isolates demonstrated resistance to 67% of the 12 antibiotics tested. To overcome these challenges, efforts have been made to identify novel antimicrobial agents, especially from eco-friendly soil bacteria. Among the 76 soil bacterial isolates that were assessed via the antagonist‒pathogen agar plug method, only two (IBT42 and VFK47) exhibited outstanding (P = 0.05) antagonism against > 50% of the Enterobacter pathotypes in comparison with the control. The potency of IBT42 and VFK47 were validated via agar well diffusion, the minimum inhibitory concentration, and the minimum bactericidal concentration. The 16S rRNA and phylogenetic analysis revealed that the identities of IBT42 and VFK47 were Bacillus mycoides and Bacillus cereus, respectively.
Collapse
Affiliation(s)
- Damilola N Adeyemi
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria
| | - Esther O Ajide
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria
| | - Gboyega E Adebami
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria
| | - Moses A Abiala
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria.
| |
Collapse
|
9
|
Celebi O, Baser S, Guler MC, Taghizadehghalehjoughi A, Rakici E, Aydin E, Celebi D. Molecular characterization of resistance and biofilm genes of ESKAPE pathogens isolated from clinical samples: examination of the effect of boric acid on biofilm ability by cell culture method. BMC Microbiol 2025; 25:106. [PMID: 40025436 PMCID: PMC11871791 DOI: 10.1186/s12866-025-03830-x] [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: 07/14/2024] [Accepted: 09/18/2024] [Indexed: 03/04/2025] Open
Abstract
Biofilm formation ranks first among the resistance and virulence factors crucial in forming ESKAPE pathogens. Once biofilm is formed, treating the infection with existing drugs is often futile. Therefore, in this study, resistant ESKAPE pathogens were isolated from intensive care units and sent to Atatürk University Yakutiye Research Hospital Microbiology Laboratory. This study investigated the biofilm formation and molecular characterization of resistant ESKAPE pathogens isolated from intensive care units. The bacteria's biofilm formation abilities, genes responsible for biofilm formation, and resistance characteristics were identified. The effect of boric acid (BA) on resistance and bacterial genes was evaluated by a bacterial infection cell culture model. The highest biofilm formation was observed in Escherichia coli, Enterococcus spp., and Pseudomonas aeruginosa Enterococcus spp. isolates showed the vanA gene in 14.6% and the vanC gene in 61% of the samples. Among Staphylococcus spp. isolates, 48.3% were MSSA, 34.5% were MRCNS, and 17.2% were MRSA. The KPC gene was detected in 50%, the OXA-48 gene in 40%, and the NDM gene in 15% of the isolates. In P. aeruginosa, the LasI and LasR quorum sensing system genes were found in 38.5% and 30.8% of the isolates, respectively. In E. coli isolates, OXA-48 was present in 35%, KPC in 31.7%, and TEM in 12.5%. BA demonstrated significant activity against ESKAPE pathogens. The combined antimicrobial activity of boron compounds showed a decrease in the expression level of the resistance gene. It will be promising for preventing hospital-associated infections.
Collapse
Affiliation(s)
- Ozgur Celebi
- Faculty of Medicine, Department of Medical Microbiology, Atatürk University, 25240, Erzurum, Turkey
| | - Sumeyye Baser
- Faculty of Medicine, Department of Medical Microbiology, Atatürk University, 25240, Erzurum, Turkey
| | - Mustafa Can Guler
- Faculty of Medicine, Department of Physiology, Atatürk University, 25240, Erzurum, Turkey
| | - Ali Taghizadehghalehjoughi
- Faculty of Medicine, Department of Medical Pharmacology, Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey
| | - Erva Rakici
- Faculty of Medicine, Department of Medical Microbiology, Recep Tayyip Erdogan University, 53020, Rize, Turkey
| | - Elif Aydin
- Faculty of Medicine, Department of Medical Microbiology, Agri Ibrahim Cecen University, Agri, 04010, Turkey
| | - Demet Celebi
- Faculty of Veterinary Medicine, Department of Microbiology, Atatürk University, 25240, Erzurum, Turkey.
- Ataturk University Vaccine Application and Development Center, Atatürk University, 25240, Erzurum, Turkey.
| |
Collapse
|
10
|
Perault AI, John AS, DuMont AL, Shopsin B, Pironti A, Torres VJ. Enterobacter hormaechei replaces virulence with carbapenem resistance via porin loss. Proc Natl Acad Sci U S A 2025; 122:e2414315122. [PMID: 39977318 PMCID: PMC11874173 DOI: 10.1073/pnas.2414315122] [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: 07/26/2024] [Accepted: 01/08/2025] [Indexed: 02/22/2025] Open
Abstract
Pathogenic Enterobacter species are of increasing clinical concern due to the multidrug-resistant nature of these bacteria, including resistance to carbapenem antibiotics. Our understanding of Enterobacter virulence is limited, hindering the development of new prophylactics and therapeutics targeting infections caused by Enterobacter species. In this study, we assessed the virulence of contemporary clinical Enterobacter hormaechei isolates in a mouse model of intraperitoneal infection and used comparative genomics to identify genes promoting virulence. Through mutagenesis and complementation studies, we found two porin-encoding genes, ompC and ompD, to be required for E. hormaechei virulence. These porins imported clinically relevant carbapenems into the bacteria, and thus loss of OmpC and OmpD desensitized E. hormaechei to the antibiotics. Our genomic analyses suggest porin-related genes are frequently mutated in E. hormaechei, perhaps due to the selective pressure of antibiotic therapy during infection. Despite the importance of OmpC and OmpD during infection of immunocompetent hosts, we found the two porins to be dispensable for virulence in a neutropenic mouse model. Moreover, porin loss provided a fitness advantage during carbapenem treatment in an ex vivo human whole blood model of bacteremia. Our data provide experimental evidence of pathogenic Enterobacter species gaining antibiotic resistance via loss of porins and argue antibiotic therapy during infection of immunocompromised patients is a conducive environment for the selection of porin mutations enhancing the multidrug-resistant profile of these pathogens.
Collapse
Affiliation(s)
- Andrew I. Perault
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY10016
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY10016
- Antimicrobial Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY10016
| | - Amelia St. John
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY10016
- Antimicrobial Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY10016
| | - Ashley L. DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY10016
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY10016
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY10016
- Antimicrobial Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY10016
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY10016
- Antimicrobial Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY10016
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY10016
- Antimicrobial Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY10016
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN38105
| |
Collapse
|
11
|
Vaikkathillam P, Mini M, Mohan A, Jayakumar D, Rajan PP, Asha S, Kumar P. Anti-biofilm effect of ferulic acid against Enterobacter hormaechei and Klebsiella pneumoniae: in vitro and in silico investigation. BIOFOULING 2025; 41:157-170. [PMID: 39757571 DOI: 10.1080/08927014.2024.2446927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 11/18/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Enterobacter hormaechei and Klebsiella pneumoniae, key members of the ESKAPE group of hospital-acquired pathogens, are driving forces behind numerous infections due to their potent biofilm formation and the growing threat of antimicrobial resistance. Ferulic acid (FA) is known for its strong antioxidant properties and is recognized for its numerous physiological benefits, including anti-inflammatory, antimicrobial, anticancer, and antidiabetic effects. The current investigation delves into the antimicrobial and antibiofilm ability of FA against E. hormaechei and K. pneumoniae. Using different assays, we confirmed that FA inhibits the biofilm formation of these pathogens. Through computational studies involving molecular docking and molecular dynamics simulations, it was found that FA exhibits a strong affinity for binding with MrkB in E. hormaechei and MrkH in K. pneumoniae, crucial proteins involved in biofilm formation. We hypothesise that FA might interfere with adhesion-associated molecules and inhibit biofilms through the c-di-GMP pathway and proves as an effective antibiofilm compound.
Collapse
Affiliation(s)
| | - Minsa Mini
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Aparna Mohan
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Devi Jayakumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Pooja P Rajan
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Sneha Asha
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Praveen Kumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| |
Collapse
|
12
|
Beshr G, Sikandar A, Gläser J, Fares M, Sommer R, Wagner S, Köhnke J, Titz A. A fucose-binding superlectin from Enterobacter cloacae with high Lewis and ABO blood group antigen specificity. J Biol Chem 2025; 301:108151. [PMID: 39743000 PMCID: PMC11875179 DOI: 10.1016/j.jbc.2024.108151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025] Open
Abstract
Bacteria frequently employ carbohydrate-binding proteins, so-called lectins, to colonize and persist in a host. Thus, bacterial lectins are attractive targets for the development of new anti-infectives. To find new potential targets for anti-infectives against pathogenic bacteria, we searched for homologs of Pseudomonas aeruginosa lectins and identified homologs of LecA in Enterobacter species. Here, we recombinantly produced and biophysically characterized a homolog that comprises one LecA domain and one additional, novel protein domain. This protein was termed Enterobacter cloacae lectin A (EclA) and found to bind l-fucose. Glycan array analysis revealed a high specificity for the LewisA antigen and the type II H-antigen (blood group O) for EclA, while related antigens LewisX, Y, and B, as well as blood group A or B were not bound. We developed a competitive binding assay to quantify blood group antigen-binding specificity in solution. Finally, the crystal structure of EclA could be solved in complex with methyl α-l-selenofucoside. It revealed the unexpected binding of the carbohydrate ligand to the second domain, which comprises a novel fold that dimerizes via strand-swapping resulting in an intertwined beta sheet.
Collapse
Affiliation(s)
- Ghamdan Beshr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, Saarbrücken, Germany
| | - Asfandyar Sikandar
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarbrücken, Germany
| | - Julia Gläser
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig
| | - Mario Fares
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, Saarbrücken, Germany
| | - Roman Sommer
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, Saarbrücken, Germany
| | - Stefanie Wagner
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig
| | - Jesko Köhnke
- Institut für Lebensmittelchemie, Leibniz Universität Hannover, Hannover, Germany.
| | - Alexander Titz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, Saarbrücken, Germany.
| |
Collapse
|
13
|
Bošnjak Z, Hasman H, Hansen F, Hammerum AM, Roer L, Jurić I, Budimir A. Co-occurrence of triple carbapenemase genes, blaVIM-2, blaNDM-1, and blaOXA-48 in Enterobacter hormaechei clinical isolates -first report from Croatia. J Chemother 2025; 37:10-14. [PMID: 38741515 DOI: 10.1080/1120009x.2024.2354107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
Two Enterobacter hormaechei isolates harbouring three carbapenemase genes each, were isolated from two patients from different ICUs at University Hospital Centre Zagreb, Croatia, which is to our knowledge, the first report of triple carbapenemase (blaVIM-2, blaNDM-1, and blaOXA-48) co-existence in E. hormachei strains and also among Enterobacterales members in Croatia. Antimicrobial susceptibility testing showed susceptibility only to colistin and amikacin. The production of carbapenemases was phenotypically tested by immunochromatographic assay and confirmed by PCR. Detailed analysis by Whole Genome Sequencing (WGS) of short reads by Illumina and long reads by Oxford Nanopore Technologies (ONT) was additionally performed and showed that both isolates belonged to ST200. They were separated by 98 Single Nucleotide Polymorphisms (SNPs) having variations in the number of blaVIM-2 genes on the chromosome, the number of blaNDM-1 genes on the plasmid, non-identical blaNDM-1 plasmids, different plasmid content in general, and only one isolate carried a 94 kb prophage.
Collapse
Affiliation(s)
- Zrinka Bošnjak
- Department of Clinical Microbiology, Infection Prevention and Control, University Hospital Center, Zagreb, Croatia
- Department of Microbiology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Henrik Hasman
- Department for Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Hansen
- Department for Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Anette M Hammerum
- Department for Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Louise Roer
- Department for Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Ivana Jurić
- Department of Clinical Microbiology, Infection Prevention and Control, University Hospital Center, Zagreb, Croatia
| | - Ana Budimir
- Department of Clinical Microbiology, Infection Prevention and Control, University Hospital Center, Zagreb, Croatia
- Department of Microbiology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
14
|
Han M, Hua M, Xie H, Li J, Wang Y, Shen H, Cao X. Clinical Characteristics and Risk Factors for Multidrug-Resistant Enterobacter cloacae Complex Bacteremia in a Chinese Tertiary Hospital: A Decade Review (2013-2022). Infect Drug Resist 2025; 18:427-440. [PMID: 39867289 PMCID: PMC11766150 DOI: 10.2147/idr.s502509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/01/2025] [Indexed: 01/28/2025] Open
Abstract
Objective This study aimed to analyze the antimicrobial resistance profiles, clinical characteristics and risk factors of bacteremia caused by Enterobacter cloacae complex (ECC) strains. Methods We retrospectively collected clinical data from patients diagnosed with ECC bacteremia between 2013 and 2022 in a tertiary hospital in Jiangsu. Subgroup analyses were performed based on multidrug resistance (MDR), nosocomial acquisition, polymicrobial bacteremia, and mortality. Results Among 188 ECC strains, the highest resistance was to ceftriaxone (39.9%), followed by ceftazidime (36.7%) and aztreonam (31.2%), with low resistance to carbapenems (<8.6%) and amikacin (1.6%). MDR ECC accounted for 30.9% (58/188). Previous antibiotic therapy was an independent risk factor for MDR ECC (OR = 3.193, P < 0.020), while appropriate antibiotic therapy significantly reduced the risk (OR = 0.279, P < 0.001). ICU admission was an independent risk factor for polymicrobial bacteremia, both endoscopy and blood transfusion were associated with mortality. Conclusion Carbapenems and amikacin are the most effective treatments for ECC bacteremia. Previous antibiotic therapy increases the risk of MDR ECC, while appropriate antibiotic therapy reduces it. ICU admission is an independent risk factor for polymicrobial bacteremia, both endoscopy and blood transfusion are linked to higher mortality. Effective control of MDR ECC bacteremia requires comprehensive strategies, including resistance detection, risk factor identification, and infection prevention.
Collapse
Affiliation(s)
- Mei Han
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
- Nanjing Field Epidemiology Training Program, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, People’s Republic of China
| | - Miaomiao Hua
- Department of Laboratory Medicine, The Affiliated Fuyang Hospital of Anhui Medical University, Fuyang, Anhui, People’s Republic of China
| | - Hui Xie
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Jia Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Yijun Wang
- Nanjing Field Epidemiology Training Program, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, People’s Republic of China
- Nanjing Jiangning District Center for Disease Control and Prevention, Nanjing, Jiangsu, People’s Republic of China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Xiaoli Cao
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
15
|
Sakalauskienė GV, Malcienė L, Stankevičius E, Radzevičienė A. Unseen Enemy: Mechanisms of Multidrug Antimicrobial Resistance in Gram-Negative ESKAPE Pathogens. Antibiotics (Basel) 2025; 14:63. [PMID: 39858349 PMCID: PMC11762671 DOI: 10.3390/antibiotics14010063] [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: 11/19/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Multidrug antimicrobial resistance (AMR) represents a formidable challenge in the therapy of infectious diseases, triggered by the particularly concerning gram-negative Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp. (ESKAPE) pathogens. Designated as a "priority" in 2017, these bacteria continue to pose a significant threat in 2024, particularly during the worldwide SARS-CoV-2 pandemic, where coinfections with ESKAPE members contributed to worsened patient outcomes. The declining effectiveness of current treatments against these pathogens has led to an increased disease burden and an increase in mortality rates globally. This review explores the sophisticated mechanisms driving AMR in gram-negative ESKAPE bacteria, focusing on Acinetobacter baumannii, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Enterobacter spp. Key bacterial mechanisms contributing to resistance include limitations in drug uptake, production of antibiotic-degrading enzymes, alterations in drug target sites, and enhanced drug efflux systems. Comprehending these pathways is vital for formulating innovative therapeutic strategies and tackling the ongoing threat posed by these resistant pathogens.
Collapse
Affiliation(s)
- Giedrė Valdonė Sakalauskienė
- Institute of Physiology and Pharmacology, Faculty of Medicine, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.M.); (E.S.); (A.R.)
| | | | | | | |
Collapse
|
16
|
Winters AD, Francescutti DM, Kracht DJ, Chaudhari DS, Zagorac B, Angoa-Perez M. The Gut Microbiome Regulates the Psychomotor Effects and Context-Dependent Rewarding Responses to Cocaine in Germ-Free and Antibiotic-Treated Animal Models. Microorganisms 2025; 13:77. [PMID: 39858845 PMCID: PMC11767876 DOI: 10.3390/microorganisms13010077] [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: 12/18/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Cocaine use disorder remains a major global health concern, with growing evidence that the gut microbiome modulates drug-related behaviors. This study examines the microbiome's role in cocaine-induced psychomotor activation and context-dependent reward responses using germ-free (GF) and antibiotic-treated (ABX) models. In GF mice, the absence of a microbiome blunted cocaine-induced psychomotor activation (p = 0.013), which was restored after conventionalization. GF mice also showed reduced cocaine-conditioned place preference (CPP) (p = 0.002), which normalized after conventionalization. Dopaminergic function, critical for psychomotor responses and reward, was microbiome-dependent, with increased dopamine levels (p = 0.009) and normalized turnover ratios after conventionalization. In the ABX model, microbiome depletion reduced both cocaine-induced locomotion and CPP responses (p ≤ 0.009), further supporting the role of gut microbes in modulating psychomotor and reward behaviors. ABX-treated mice also showed significant declines in microbial diversity, shifts in bacterial structure, and dysregulation in metabolic, immune, and neurotransmitter pathways (p ≤ 0.0001), including alterations in short-chain fatty acids and gamma-aminobutyric acid metabolism. These findings highlight the gut microbiome's critical role in regulating cocaine's psychomotor and rewarding effects, offering insights into potential therapeutic strategies for cocaine use disorder.
Collapse
Affiliation(s)
- Andrew D. Winters
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Dina M. Francescutti
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - David J. Kracht
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Diptaraj S. Chaudhari
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Branislava Zagorac
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Mariana Angoa-Perez
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| |
Collapse
|
17
|
Peng Q, Zheng H, Xue J, Xu Y, Hou Q, Yang K, Xia H, Xie G. Mechanism of Polygonum hydropiper reducing ethyl carbamate in Chinese rice wine (Huangjiu) brewing. Food Microbiol 2025; 125:104628. [PMID: 39448146 DOI: 10.1016/j.fm.2024.104628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 10/26/2024]
Abstract
Polygonum hydropiper (PH) is a rich source of active compounds and serves as a pivotal ingredient in Chinese rice wine (Huangjiu) production. This study investigates the impact of PH and Polygonum hydropiper extract (PHE) on ethyl carbamate (EC) production during Huangjiu fermentation. Our findings reveal that PH enhances the relative abundance of Bacillus subtilis in Huangjiu fermentation, thereby facilitating its interaction with Saccharomyces cerevisiae. Furthermore, PH modulates the urea metabolism of S. cerevisiae. In the PH-B. subtilis-S. cerevisiae fermentation system, the expression of DUR1,2 and DUR3 genes in S. cerevisiae is upregulated. This augmentation leads to increased urea uptake and metabolism by S. cerevisiae in the fermentation broth, subsequently reducing the urea concentration in the fermentation medium (The EC content in the CK group was approximately 355.55 % and 356.05 % higher than those in the PH and PHE groups, respectively). Consequently, PH demonstrates promise in reducing the EC concentration of Huangjiu, offering a novel approach to enhance the safety of Huangjiu consumption.
Collapse
Affiliation(s)
- Qi Peng
- National Engineering Research Center for Chinese CRW (Branch Center), School of Life and Environmental Sciences, Shaoxing University, 900 Chengnan Road, Shaoxing, 312000, China
| | - Huajun Zheng
- National Engineering Research Center for Chinese CRW (Branch Center), School of Life and Environmental Sciences, Shaoxing University, 900 Chengnan Road, Shaoxing, 312000, China
| | - Jingrun Xue
- National Engineering Research Center for Chinese CRW (Branch Center), School of Life and Environmental Sciences, Shaoxing University, 900 Chengnan Road, Shaoxing, 312000, China
| | - Yuezheng Xu
- Zhejiang Guyuelongshan Shaoxing Wine Co., Ltd., Shaoxing, 312000, China
| | - Qifan Hou
- National Engineering Research Center for Chinese CRW (Branch Center), School of Life and Environmental Sciences, Shaoxing University, 900 Chengnan Road, Shaoxing, 312000, China
| | - Kaiming Yang
- National Engineering Research Center for Chinese CRW (Branch Center), School of Life and Environmental Sciences, Shaoxing University, 900 Chengnan Road, Shaoxing, 312000, China
| | - Huangjia Xia
- National Engineering Research Center for Chinese CRW (Branch Center), School of Life and Environmental Sciences, Shaoxing University, 900 Chengnan Road, Shaoxing, 312000, China
| | - Guangfa Xie
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, 310015, China.
| |
Collapse
|
18
|
Bérot V, Monsel G, Dauendorffer JN, Aubry A, Nebbad B, Schneider P, Caumes E, Chosidow O. Klebsiella aerogenes-related facial folliculitis in men having sex with men: A hypothetical new STI? J Eur Acad Dermatol Venereol 2025; 39:e10-e12. [PMID: 38619375 DOI: 10.1111/jdv.20008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Affiliation(s)
- V Bérot
- Service des Maladies Infectieuses et Tropicales, Groupe Hospitalier Pitié-Salpêtrière Charles-Foix, AP-HP, Paris, France
- IPLESP, Equipe 3, Inserm UMR-S 1136, Paris, France
| | - G Monsel
- Service des Maladies Infectieuses et Tropicales, Groupe Hospitalier Pitié-Salpêtrière Charles-Foix, AP-HP, Paris, France
| | - J-N Dauendorffer
- Service de Dermatologie, Hôpital Saint-Louis, AP-HP, Paris, France
| | - A Aubry
- Centre National de Référence des Mycobactéries, Groupe Hospitalier Pitié-Salpêtrière Charles-Foix, AP-HP, Paris, France
- Centre d'immunologie et de Maladies Infectieuses (Cimi-Paris), INSERM U1135, Sorbonne-Université, Paris, France
| | - B Nebbad
- Service de Bactériologie, Hôpital Henri Mondor, AP-HP, Créteil, France
- Groupe Dynamic, EA7380, Faculté de Santé de Créteil, Ecole Nationale Vétérinaire d'Alfort, USC ANSES, Université Paris-Est Créteil, Créteil, France
| | - P Schneider
- Service de Dermatologie, Hôpital Saint-Louis, AP-HP, Paris, France
| | - E Caumes
- Service des Maladies Infectieuses et Tropicales, Groupe Hospitalier Pitié-Salpêtrière Charles-Foix, AP-HP, Paris, France
- IPLESP, Equipe 3, Inserm UMR-S 1136, Paris, France
| | - O Chosidow
- Groupe Dynamic, EA7380, Faculté de Santé de Créteil, Ecole Nationale Vétérinaire d'Alfort, USC ANSES, Université Paris-Est Créteil, Créteil, France
- Service de Dermatologie, Hôpital Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
19
|
Joos M, Vackier T, Mees MA, Coppola G, Alexandris S, Geunes R, Thielemans W, Steenackers HPL. Antimicrobial Activity of Glycyrrhizinic Acid Is pH-Dependent. ACS APPLIED BIO MATERIALS 2024; 7:8223-8235. [PMID: 39592134 PMCID: PMC11655076 DOI: 10.1021/acsabm.4c00942] [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: 07/10/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024]
Abstract
In recent years, antimicrobial hydrogels have attracted much attention in biomedical applications due to their biocompatibility and high water content. Glycyrrhizin (GA) is an antimicrobial that can form pH-dependent hydrogels due to the three carboxyl groups of GA that differ in pKa value. The influence of GA protonation on the antimicrobial activity, however, has never been studied before. Therefore, we investigated the effect of the pH on the antimicrobial activity of GA against Pseudomonas aeruginosa, Staphylococcus aureus, MRSA, Staphylococcus epidermidis, Acinetobacter baumannii, Klebsiella pneumoniae, Klebsiella aerogenes, and two strains of Escherichia coli. In general, the antimicrobial activity of GA increases as a function of decreasing pH (and thus increasing protonation of GA). More specifically, fully protonated GA hydrogels (pH = 3) are required for growth inhibition and killing of E. coli UTI89 and Klebsiella in the suspension above the hydrogel, while the staphylococci strains and A. baumannii are already inhibited by fully deprotonated GA (pH = 6.8). P. aeruginosa and E. coli DH5α showed moderate susceptibility, as they are completely inhibited by a hydrogel at pH 3.8, containing partly protonated GA, but not by fully deprotonated GA (pH = 6.8). The antimicrobial activity of the hydrogel cannot solely be attributed to the resulting pH decrease of the suspension, as the presence of GA significantly increases the activity. Instead, this increased activity is due to the release of GA from the hydrogel into the suspension, where it directly interacts with the bacteria. Moreover, we provide evidence indicating that the pH dependency of the antimicrobial activity is due to differences in GA protonation state by treating the pathogens with GA solutions differing in their GA protonation distribution. Finally, we show by LC-MS that there is no chemical or enzymatic breakdown of GA. Overall, our results demonstrate that the pH influences not only the physical but also the antimicrobial properties of the GA hydrogels.
Collapse
Affiliation(s)
- Mathieu Joos
- Department
of Microbial and Molecular Systems, Centre of Microbial and Plant
Genetics (CMPG), KU Leuven, Leuven 3001, Belgium
| | - Thijs Vackier
- Department
of Microbial and Molecular Systems, Centre of Microbial and Plant
Genetics (CMPG), KU Leuven, Leuven 3001, Belgium
| | - Maarten A. Mees
- Department
of Chemical Engineering, KU Leuven, Sustainable
Materials Lab (SusMat), Kortrijk 8500, Belgium
| | - Guglielmo Coppola
- Department
of Microbial and Molecular Systems, Centre of Microbial and Plant
Genetics (CMPG), KU Leuven, Leuven 3001, Belgium
- Department
of Chemistry, KU Leuven - Laboratory for
Organic & Microwave-Assisted Chemistry (LOMAC), Leuven 3001, Belgium
| | - Stelios Alexandris
- Department
of Chemical Engineering, KU Leuven - Laboratory
for Soft Matter, Rheology and Technology (SMaRT), Leuven 3001, Belgium
| | - Robbe Geunes
- Department
of Chemical Engineering, KU Leuven, Sustainable
Materials Lab (SusMat), Kortrijk 8500, Belgium
| | - Wim Thielemans
- Department
of Chemical Engineering, KU Leuven, Sustainable
Materials Lab (SusMat), Kortrijk 8500, Belgium
| | - Hans P. L. Steenackers
- Department
of Microbial and Molecular Systems, Centre of Microbial and Plant
Genetics (CMPG), KU Leuven, Leuven 3001, Belgium
| |
Collapse
|
20
|
da Rosa EEB, Kremer FS. The mobilome landscape of biocide-resistance in Brazilian ESKAPE isolates. Braz J Microbiol 2024; 55:3603-3616. [PMID: 39028534 PMCID: PMC11712051 DOI: 10.1007/s42770-024-01450-7] [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: 05/13/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
The increasing frequency of antibiotic-resistant bacteria is a constant threat to global human health. Therefore, the pathogens of the ESKAPE group (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, and Enterobacter spp.) are among the most relevant causes of hospital infections responsible for millions of deaths every year. However, little has been explored about the danger of microorganisms resistant to biocides such as antiseptics and disinfectants. Widely used in domestic, industrial, and hospital environments, these substances reach the environment and can cause selective pressure for resistance genes and induce cross-resistance to antibiotics, further aggravating the problem. Therefore, it is necessary to use innovative and efficient strategies to monitor the spread of genes related to resistance to biocides. Whole genome sequencing and bioinformatics analysis aiming to search for sequences encoding resistance mechanisms are essential to help monitor and combat these pathogens. Thus, this work describes the construction of a bioinformatics tool that integrates different databases to identify gene sequences that may confer some resistance advantage about biocides. Furthermore, the tool analyzed all the genomes of Brazilian ESKAPE isolates deposited at NCBI and found a series of different genes related to resistance to benzalkonium chloride, chlorhexidine, and triclosan, which were the focus of this work. As a result, the presence of resistance genes was identified in different types of biological samples, environments, and hosts. Regarding mobile genetic elements (MGEs), around 52% of isolates containing genes related to resistance to these compounds had their genes identified in plasmids, and 48.7% in prophages. These data show that resistance to biocides can be a silent, underestimated danger spreading across different environments and, therefore, requires greater attention.
Collapse
Affiliation(s)
- Elias Eduardo Barbosa da Rosa
- Laboratório de Bioinformática (Omixlab), Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Rio Grande Do Sul, Brazil
| | - Frederico Schmitt Kremer
- Laboratório de Bioinformática (Omixlab), Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Rio Grande Do Sul, Brazil.
| |
Collapse
|
21
|
Ullah A, Rehman B, Khan S, Almanaa TN, Waheed Y, Hassan M, Naz T, Ul Haq M, Muhammad R, Sanami S, Irfan M, Ahmad S. An In Silico Multi-epitopes Vaccine Ensemble and Characterization Against Nosocomial Proteus penneri. Mol Biotechnol 2024; 66:3498-3513. [PMID: 37934390 DOI: 10.1007/s12033-023-00949-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/12/2023] [Indexed: 11/08/2023]
Abstract
Proteus penneri (P. penneri) is a bacillus-shaped, gram-negative, facultative anaerobe bacterium that is primarily an invasive pathogen and the etiological agent of several hospital-associated infections. P. penneri strains are naturally resistant to macrolides, amoxicillin, oxacillin, penicillin G, and cephalosporins; in addition, no vaccines are available against these strains. This warrants efforts to propose a theoretical based multi-epitope vaccine construct to prevent pathogen infections. In this research, reverse vaccinology bioinformatics and immunoinformatics approaches were adopted for vaccine target identification and construction of a multi-epitope vaccine. In the first phase, a core proteome dataset of the targeted pathogen was obtained using the NCBI database and subjected to bacterial pan-genome analysis using bacterial pan-genome analysis (BPGA) to predict core protein sequences which were then used to find good vaccine target candidates. This identified two proteins, Hcp family type VI secretion system effector and superoxide dismutase family protein, as promising vaccine targets. Afterward using the IEDB database, different B-cell and T-cell epitopes were predicted. A set of four epitopes "KGSVNVQDRE, NTGKLTGTR, IIHSDSWNER, and KDGKPVPALK" were chosen for the development of a multi-epitope vaccine construct. A 183 amino acid long vaccine design was built along with "EAAAK" and "GPGPG" linkers and a cholera toxin B-subunit adjuvant. The designed vaccine model comprised immunodominant, non-toxic, non-allergenic, and physicochemical stable epitopes. The model vaccine was docked with MHC-I, MHC-II, and TLR-4 immune cell receptors using the Cluspro2.0 web server. The binding energy score of the vaccine was - 654.7 kcal/mol for MHC-I, - 738.4 kcal/mol for MHC-II, and - 695.0 kcal/mol for TLR-4. A molecular dynamic simulation was done using AMBER v20 package for dynamic behavior in nanoseconds. Additionally, MM-PBSA binding free energy analysis was done to test intermolecular binding interactions between docked molecules. The MM-GBSA net binding energy score was - 148.00 kcal/mol, - 118.00 kcal/mol, and - 127.00 kcal/mol for vaccine with TLR-4, MHC-I, and MHC-II, respectively. Overall, these in silico-based predictions indicated that the vaccine is highly promising in terms of developing protective immunity against P. penneri. However, additional experimental validation is required to unveil the real immune response to the designed vaccine.
Collapse
Affiliation(s)
- Asad Ullah
- Department of Health and Biological Sciences, Abasyn University, Peshawar, 2500, Pakistan
- Centre of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Bushra Rehman
- Institute of Biotechnology and Microbiology, Bacha Khan University, Charsadda, Pakistan
| | - Saifullah Khan
- Institute of Biotechnology and Microbiology, Bacha Khan University, Charsadda, Pakistan
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Yasir Waheed
- Office of Research, Innovation and Commercialization, Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), Islamabad, 44000, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, 1401, Lebanon
| | - Muhammad Hassan
- Department of Pharmacy, Bacha Khan University, Charsadda, 24461, Pakistan
| | - Tahira Naz
- Department of Chemical and Life Sciences, Qurtuba University of Science and Technology, Peshawar, Pakistan
| | - Mehboob Ul Haq
- Department of Pharmacy, Abasyn University, Peshawar, 25000, Pakistan
| | - Riaz Muhammad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, 2500, Pakistan
| | - Samira Sanami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Muhammad Irfan
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, 32611, USA
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, 2500, Pakistan.
- Department of Natural Sciences, Lebanese American University, P.O. Box 36, Beirut, Lebanon.
| |
Collapse
|
22
|
Kidenya BR, Mboowa G, Sserwadda I, Kanyerezi S, Nakafu E, Akaro IL, Mkinze B, Joloba ML, Seni J. Whole genome-based characterization of extended-spectrum β-lactamase-producing Enterobacter cloacae from orthopedic patients and environment of a tertiary referral hospital in Tanzania. New Microbes New Infect 2024; 62:101486. [PMID: 39386352 PMCID: PMC11462357 DOI: 10.1016/j.nmni.2024.101486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
Objectives We investigated the genomic epidemiology of extended-spectrum β-lactamase-producing Enterobacter cloacae (ESBL-Ec) isolates from patients and hospital environment to better understand their distribution to help devising effective strategies for infection prevention and control. Methods We screened ESBL-Ec at Bugando Medical Center (BMC) in Mwanza, Tanzania. Rectal swabs from orthopedic patients on admission and swabs from the neighboring inanimate environment were collected. Following microbial culture, DNA was extracted from pure ESBL-Ec, and whole-genome sequencing was done. Sequence typing (ST), plasmid replicons, drug resistance, and virulence genes were deciphered using the Rapid Microbial Analysis Pipeline (rMAP). Results We obtained 209 ESBL isolates, of which 15 (7.2 %) were ESBL-Ec [8 (53.3 %) from patients and 7 (46.7 %) from the environment]. Seven isolates were novel and eight were diverse, each with a unique ST. All isolates harbored two to five β-lactamase genes, with the predominance of bla CTX-M-15 (15/15), bla OXA-1 (14/15), bla TEM (14/15) and bla ACT (12/15). The most common non β-lactam drug resistance genes were aac(3)-IIa (14/15), aac(6')-Ib-cr (14/15), fosA (14/15), and qnrB1 (12/15), aph(3″)-Ib (10/15) and aph(6)-Id (10/15). Eleven different types of plasmid replicons were identified in 14/15 of the isolates, harboring one to five plasmids, with the most common plasmids being IncFII (11/15) and IncFIB (10/15). All isolates harbored the outer membrane protein (ompA), and curli protein (csg) was in 14/15 isolates. Conclusion Admitted orthopedic patients and the hospital environment act as a reservoir of ESBL-Ec with diverse STs and endowed with drug resistance and arsenals of virulence genes, calling for their routine screening on admission for mitigation of potential subsequent infections.
Collapse
Affiliation(s)
- Benson R. Kidenya
- Department of Biochemistry and Molecular Biology, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| | - Gerald Mboowa
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
- African Center of Excellence in Bioinformatics and Data-Intensive Sciences, The Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418, Kampala, Uganda
- Africa Centres for Disease Control and Prevention, African Union Commission, Roosevelt Street, P.O. Box 3243, W21 K19, Addis Ababa, Ethiopia
| | - Ivan Sserwadda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
- African Center of Excellence in Bioinformatics and Data-Intensive Sciences, The Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418, Kampala, Uganda
- Department of Biochemistry and Bioinformatics, School of Pure and Applied Sciences, Pwani University, P.O Box 195-80108, Kilifi, Kenya
| | - Stephen Kanyerezi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
- Africa Centres for Disease Control and Prevention, African Union Commission, Roosevelt Street, P.O. Box 3243, W21 K19, Addis Ababa, Ethiopia
| | - Esther Nakafu
- College of Veterinary Medicine, Animal Resources and Bio-security (COVAB), Makerere University, P. O Box 7062, Kampala, Uganda
| | - Inyasi Lawrence Akaro
- Department of Orthopedic Surgery, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| | - Baraka Mkinze
- Department of Orthopedic Surgery, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| | - Moses L. Joloba
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P. O Box 7072, Kampala, Uganda
| | - Jeremiah Seni
- Department of Microbiology and Immunology, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, P.O. Box 1464, Mwanza, Tanzania
| |
Collapse
|
23
|
Santos da Costa B, Peixoto RS, da Conceição Neto OC, da Silva Pontes L, Tavares E Oliveira TR, Tavares Teixeira CB, de Oliveira Santos IC, Silveira MC, Silva Rodrigues DC, Pribul BR, Rocha-de-Souza CM, D 'Alincourt Carvalho-Assef AP. Polymyxin resistance in Enterobacter cloacae complex in Brazil: phenotypic and molecular characterization. Braz J Microbiol 2024; 55:3541-3550. [PMID: 39210190 PMCID: PMC11712032 DOI: 10.1007/s42770-024-01464-1] [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: 04/20/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
Enterobacter cloacae complex isolates have been reported as an important nosocomial multidrug resistance pathogen. In the present study, we investigated antimicrobial susceptibility and the colistin-resistance rates, their genetic determinants and clonality among clinical E. cloacae complex isolates from different Brazilian states. For this, an initial screening was carried out on 94 clinical isolates of E. clocacae complex received between 2016 and 2018 by LAPIH-FIOCRUZ, using EMB plates containing 4 μg/mL of colistin, followed MIC determination, resulting in the selection of 26 colistin-resistant isolates from the complex. The presence of carbapenemases encoding genes (blaKPC, blaNDM and blaOXA-48), plasmidial genes for resistance to polymyxins (mcr1-9) and mutations in chromosomal genes (pmrA, pmrB, phoP and phoQ) described as associated with resistance to polymyxin were screened by PCR and DNA sequencing. Finally, the hsp60 gene was sequenced to identify species of the E. cloacae complex and genetic diversity was evaluated by PFGE and MLST. The results have shown that among 94 E. cloacae complex isolates, 19 (20.2%) were colistin-resistant. The resistant strains exhibited MIC ranging from 4 to 128 µg / mL and E. hormaechei subsp. steigerwaltii was the prevalent species in the complex (31,6%), followed by E. cloacae subsp. cloacae (26,3%). The antimicrobials with the highest susceptibility rate were gentamicin (21%) and tigecycline (26%). Carbapenemases encoding genes (blaKPC n = 5, blaNDM n = 1) were detected in 6 isolates and mcr-9 in one. Among the modifications found in PmrA, PmrB, PhoP e PhoQ (two-component regulatory system), only the S175I substitution in PmrB found in E. cloacae subsp cloacae isolates were considered deleterious (according to the prediction of PROVEAN). By PFGE, 13 profiles were found among E. cloacae complex isolates, with EcD the most frequent. Furthermore, by MLST 10 ST's, and 1 new ST, were identified in E. cloacae. In conclusion, no prevalence of clones or association among carbapenemase production and polymyxin resistance was found between the E. cloacae. Thereby, the results suggest that the increased polymyxin-resistance is related to the selective pressure exerted by the indiscriminate use in hospitals. Lastly, this study highlights the urgent need to elucidate the mechanism involved in the resistance to polymyxin in the E. cloacae complex and the development of measures to control and prevent infections caused by these multiresistant bacteria.
Collapse
Affiliation(s)
- Bianca Santos da Costa
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Renata Stavracakis Peixoto
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Orlando Carlos da Conceição Neto
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Leilane da Silva Pontes
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Thamirys Rachel Tavares E Oliveira
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Camila Bastos Tavares Teixeira
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Ivson Cassiano de Oliveira Santos
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Melise Chaves Silveira
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Daiana Cristina Silva Rodrigues
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
| | - Bruno Rocha Pribul
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
- Coleção de Culturas de Bactérias de Origem Hospitalar (CCBH), Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Av. Brasil, Rio de Janeiro,RJ, 436521045900, Brazil
| | - Cláudio Marcos Rocha-de-Souza
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil
- Coleção de Culturas de Bactérias de Origem Hospitalar (CCBH), Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Av. Brasil, Rio de Janeiro,RJ, 436521045900, Brazil
| | - Ana Paula D 'Alincourt Carvalho-Assef
- Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Laboratório de Bacteriologia Aplicada a Saúde Única E Resistência Antimicrobiana, Av. Brasil, Rio de Janeiro, RJ, 436521045900, Brazil.
| |
Collapse
|
24
|
González-Montalvo MA, Sorescu JM, Baltes G, Juárez O, Tuz K. The respiratory chain of Klebsiella aerogenes in urine-like conditions: critical roles of NDH-2 and bd-terminal oxidases. Front Microbiol 2024; 15:1479714. [PMID: 39568993 PMCID: PMC11576283 DOI: 10.3389/fmicb.2024.1479714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
Klebsiella aerogenes is an opportunistic nosocomial bacterial pathogen that commonly causes urinary tract infections. Over the past decades, K. aerogenes strains have acquired resistance to common antibiotics that has led to the rise of multidrug-resistant and even pandrug-resistant strains. Infections produced by these strains are nearly impossible to treat, which makes K. aerogenes a global priority to develop new antibiotics and there is an urgent need to identify targets to treat infections against this pathogen. However, very little is known about the metabolism and metabolic adaptations of this bacterium in infection sites. In this work, we investigated the respiratory metabolism of K. aerogenes in conditions that resemble human urine, allowing us to identify novel targets for antibiotic development. Here we describe that, unlike other gram-negative pathogens, K. aerogenes utilizes the type-2 NADH dehydrogenase (NDH-2) as the main entry point for electrons in the respiratory chain in all growth conditions evaluated. Additionally, in urine-like media, the aerobic metabolism as a whole is upregulated, with significant increases in succinate and lactate dehydrogenase activity. Moreover, our data show that the bd-I type oxidoreductases are the main terminal oxidases of this microorganism. Our findings support an initial identification of NDH-2 and bd-I oxidase as attractive targets for the development of new drugs against K. aerogenes as they are not found in human hosts.
Collapse
Affiliation(s)
| | - Jennifer M Sorescu
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, United States
| | - Gabriella Baltes
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, United States
| | - Oscar Juárez
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, United States
| | - Karina Tuz
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, United States
| |
Collapse
|
25
|
Siderius NL, Sapula SA, Hart BJ, Hutchings JL, Venter H. Enterobacter adelaidei sp. nov. Isolation of an extensively drug resistant strain from hospital wastewater in Australia and the global distribution of the species. Microbiol Res 2024; 288:127867. [PMID: 39163716 DOI: 10.1016/j.micres.2024.127867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Enterobacter species are included among the normal human gut microflora and persist in a diverse range of other environmental niches. They have become important opportunistic nosocomial pathogens known to harbour plasmid-mediated multi-class antimicrobial resistance (AMR) determinants. Global AMR surveillance of Enterobacterales isolates shows the genus is second to Klebsiella in terms of frequency of carbapenem resistance. Enterobacter taxonomy is confusing and standard species identification methods are largely inaccurate or insufficient. There are currently 27 named species and a total of 46 taxa in the genus distinguishable via average nucleotide identity (ANI) calculation between pairs of genomic sequences. Here we describe an Enterobacter strain, ECC3473, isolated from the wastewater of an Australian hospital whose species could not be determined by standard methods nor by ribosomal RNA gene multi-locus typing. AIM To characterise ECC3473 in terms of phenotypic and genotypic antimicrobial resistance, biochemical characteristics and taxonomy as well as to determine the global distribution of the novel species to which it belongs. METHODS Standard broth dilution and disk diffusion were used to determine phenotypic AMR. The strain's complete genome, including plasmids, was obtained following long- and short read sequencing and a novel long/short read hybrid assembly and polishing, and the genomic basis of AMR was determined. Phylogenomic analysis and quantitative measures of relatedness (ANI, digital DNA-DNA hybridisation, and difference in G+C content) were used to study the taxonomic relationship between ECC3473 and Enterobacter type-strains. NCBI and PubMLST databases and the literature were searched for additional members of the novel species to determine its global distribution. RESULTS ECC3473 is one of 21 strains isolated globally belonging to a novel Enterobacter species for which the name, Enterobacter adelaidei sp. nov. is proposed. The novel species was found to be resilient in its capacity to persist in contaminated water and adaptable in its ability to accumulate multiple transmissible AMR determinants. CONCLUSION E. adelaidei sp. nov. may become increasingly important to the dissemination of AMR.
Collapse
Affiliation(s)
- Naomi L Siderius
- UniSA Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| | - Sylvia A Sapula
- UniSA Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| | - Bradley J Hart
- UniSA Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| | - Joshua L Hutchings
- UniSA Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| | - Henrietta Venter
- UniSA Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
26
|
Angammana H, Omadevuae K, Bengualid V, Khader R. Silent Invader: A Rare Case of Enterobacter aerogenes Empyema in a Hospice Patient With Complex Comorbidities. Cureus 2024; 16:e73397. [PMID: 39659331 PMCID: PMC11631160 DOI: 10.7759/cureus.73397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/09/2024] [Indexed: 12/12/2024] Open
Abstract
Enterobacter aerogenes (recently renamed Klebsiella aerogenes) is an uncommon pathogen in pleural infections and empyema, typically associated with nosocomial urinary and gastrointestinal infections. This case report describes a 69-year-old male patient with chronic kidney disease, diabetes mellitus, and other comorbidities, who developed empyema despite broad-spectrum antibiotics. Pleural fluid cultures revealed E. aerogenes, known for its ability to develop resistance through beta-lactamase production and efflux pumps, which complicates treatment. Despite initial improvement with cefepime and metronidazole, the patient's respiratory status deteriorated, and due to his do not resuscitate/do not intubate (DNR/DNI) status and extensive comorbidities, no further aggressive interventions were pursued, leading to his passing. This case highlights the diagnostic and therapeutic challenges posed by E. aerogenes in pleural infections, emphasizing its rarity in pulmonary involvement and its potential for antibiotic resistance. It also underscores the importance of considering atypical pathogens in complex infections and the need for multidisciplinary management while balancing aggressive treatments with patient-centered care, particularly in end-of-life scenarios.
Collapse
Affiliation(s)
- Hansani Angammana
- Internal Medicine, Saint Barnabas Hospital Health System, New York, USA
| | - Kafayat Omadevuae
- Internal Medicine, Saint Barnabas Hospital Health System, New York, USA
| | - Victoria Bengualid
- Infectious Diseases, Saint Barnabas Hospital Health System, New York, USA
| | - Rawand Khader
- Geriatrics, Saint Barnabas Hospital Health System, New York, USA
| |
Collapse
|
27
|
Harding-Crooks R, Jones AL, Smith DL, Fanning S, Fox EM. Profiling the Enterobacterales Community Isolated from Retail Foods in England. J Food Prot 2024; 87:100369. [PMID: 39366658 DOI: 10.1016/j.jfp.2024.100369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/21/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024]
Abstract
Enterobacterales include foodborne pathogens of importance to public health and are often targeted in food surveillance programs as both safety and hygiene indicators. Furthermore, Enterobacterales are important in the context of antimicrobial resistance dissemination, also impacting infection treatment efficacy. In this study, the prevalence and characteristics of Enterobacterales in UK retail foods were examined. From 110 retail food samples, 253 Enterobacterales were recovered, with 16S rRNA sequencing revealing a diverse species community, including enteropathogens; the most common were Proteus mirabilis and Escherichia coli (18% each). Antimicrobial resistance was common, with 160/253 (63%) isolates being resistant to at least 1 antimicrobial. Resistance to all tested antimicrobials was observed. Thirteen percent of isolates were multidrug resistant, including 2 isolates each resistant to 8 or 9 of 9 antimicrobials tested. Klebsiella isolates possessed relatively higher levels of antimicrobial resistance to other species. Hafnia, Kluyvera, and Proteus isolates produced significantly higher biofilm biomass than Klebsiella (p = 0.038, 0.028, and 0.042, respectively) or Escherichia (p = 0.001, 0.008, and 0.001, respectively). Simultaneous curli fimbriae and cellulose production was noted in 7% of isolates at 37 °C, but not at 15 °C. This research demonstrates a high diversity of Enterobacterales within UK retail foods, alongside notable antimicrobial resistance phenotypes in enteropathogenic species, highlighting the need for effective surveillance and interventions.
Collapse
Affiliation(s)
- Richard Harding-Crooks
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, United Kingdom
| | - Amanda L Jones
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, United Kingdom
| | - Darren L Smith
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, United Kingdom
| | - Séamus Fanning
- UCD-Centre for Food Safety, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin D04 N2E5, Ireland; Institute for Global Food Security, Queen's University Belfast, Chlorine Gardens, Belfast BT5 6AG, United Kingdom
| | - Edward M Fox
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, United Kingdom.
| |
Collapse
|
28
|
Singh A, Tanwar M, Singh TP, Sharma S, Sharma P. An escape from ESKAPE pathogens: A comprehensive review on current and emerging therapeutics against antibiotic resistance. Int J Biol Macromol 2024; 279:135253. [PMID: 39244118 DOI: 10.1016/j.ijbiomac.2024.135253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
The rise of antimicrobial resistance has positioned ESKAPE pathogens as a serious global health threat, primarily due to the limitations and frequent failures of current treatment options. This growing risk has spurred the scientific community to seek innovative antibiotic therapies and improved oversight strategies. This review aims to provide a comprehensive overview of the origins and resistance mechanisms of ESKAPE pathogens, while also exploring next-generation treatment strategies for these infections. In addition, it will address both traditional and novel approaches to combating antibiotic resistance, offering insights into potential new therapeutic avenues. Emerging research underscores the urgency of developing new antimicrobial agents and strategies to overcome resistance, highlighting the need for novel drug classes and combination therapies. Advances in genomic technologies and a deeper understanding of microbial pathogenesis are crucial in identifying effective treatments. Integrating precision medicine and personalized approaches could enhance therapeutic efficacy. The review also emphasizes the importance of global collaboration in surveillance and stewardship, as well as policy reforms, enhanced diagnostic tools, and public awareness initiatives, to address resistance on a worldwide scale.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - T P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
29
|
Cai S, Quan J, Wang Z, Hu H, Han X, Jiang Y, Yang Q, Yu Y, Zhou Z. High prevalence of carbapenem-resistant Enterobacter cloacae complex in a tertiary hospital over a decade. Microbiol Spectr 2024; 12:e0078024. [PMID: 39475294 PMCID: PMC11619405 DOI: 10.1128/spectrum.00780-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/07/2024] [Indexed: 12/08/2024] Open
Abstract
The aim of this study was to explore the mechanisms and molecular epidemiology of carbapenem resistance in the carbapenem-resistant Enterobacter cloacae complex (CRECC) over a decade in a tertiary hospital in Zhejiang, China. From January 2011 to December 2021, we collected a total of 931 Enterobacter cloacae complex (ECC) isolates from a tertiary hospital in Zhejiang, China. Antimicrobial susceptibility tests were performed. Whole-genome sequencing was used to analyze the molecular characteristics of the CRECC isolates. For carbapenem-resistant strains, efflux inhibitor assay and quantitative real-time PCR (qRT-PCR) were performed to evaluate the function of efflux pumps. A total of 82 CRECC isolates were detected, and the rate of resistance for carbapenems was 8.8%, increasing from 5.5% in 2011 to 18.3% in 2019, with an overall increasing trend, with Enterobacter hormaechei subsp. hoffmannii being the predominant species. Among the CRECC, 24 (24/931) isolates were found to produce carbapenemases, including NDM-1, NDM-5, IMP-4, and KPC-2. Among all carbapenemases, NDM-1 was the most prevalent, accounting for 62.5% (15/24) of carbapenemases, followed by NDM-5 (5/24). Genes encoding extended-spectrum beta-lactamases (47/82) and AmpC (76/82) were also identified, with blaSHV-12 and blaACT being the predominant ones, respectively. Multilocus sequence typing revealed 28 different sequence types, among which ST78 was the predominant, followed by ST93 and ST177. IncFIB was the most common type of plasmid replicon. Efflux inhibitor assay and qRT-PCR indicated that the overexpression of efflux pumps was involved in carbapenem resistance mechanisms. Additionally, disrupted outer membrane proteins also contribute to carbapenem resistance. The detection rate of CRECC was rising in the tertiary hospital. BlaNDM-1 and blaNDM-5 were the main carbapenem resistance genes. Our study revealed the presence of carbapenem-resistant ECC strains, emphasizing the need for effective infection prevention approaches to reduce the prevalence of CRECC. IMPORTANCE The emergence and spread of the carbapenem-resistant Enterobacter cloacae complex (CRECC) have become a significant public health problem. CRECC strains frequently harbor multiple drug resistance genes and can be epidemic within healthcare facilities. The study explored the characteristics and prevalence of CRECC strains in the same hospital over a decade, which provides a theoretical basis for epidemiologic surveillance and clinical treatment.
Collapse
Affiliation(s)
- Shiqi Cai
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Quan
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengan Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huangdu Hu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinhong Han
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Jiang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing Yang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihui Zhou
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
- Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
30
|
Lawrence J, O'Hare D, van Batenburg-Sherwood J, Sutton M, Holmes A, Rawson TM. Innovative approaches in phenotypic beta-lactamase detection for personalised infection management. Nat Commun 2024; 15:9070. [PMID: 39433753 PMCID: PMC11494114 DOI: 10.1038/s41467-024-53192-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Beta-lactamase-producing Enterobacteriaceae present a significant therapeutic challenge. Current developments in phenotypic diagnostics focus primarily on rapid minimum inhibitory concentration (MIC) determination. There is a requirement for rapid phenotypic diagnostics to improve antimicrobial susceptibility tests (AST) and aid prescribing decisions. Phenotypic AST are limited in their ability to characterise beta-lactamase-producing Enterobacteriaceae in detail. Despite advances in rapid AST, gaps and opportunities remain for developing additional diagnostic approaches that facilitate personalised antimicrobial prescribing. In this perspective, we highlight the state-of-the-art in beta-lactamase detection, identify gaps in current practice, and discuss barriers for innovation within this field.
Collapse
Affiliation(s)
- Jennifer Lawrence
- The NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, Hammersmith Campus, London, United Kingdom.
- Centre for Antimicrobial Optimisation, Imperial College London, London, United Kingdom.
| | - Danny O'Hare
- Centre for Antimicrobial Optimisation, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Joseph van Batenburg-Sherwood
- Department of Bioengineering, Sir Michael Uren Hub, Imperial College London, White City Campus, London, United Kingdom
| | - Mark Sutton
- Antimicrobial Discovery, Development and Diagnostics (AD3) UK Health Security Agency, Porton Down, Salisbury, Wiltshire, United Kingdom
- Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Alison Holmes
- The NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, Hammersmith Campus, London, United Kingdom
- Centre for Antimicrobial Optimisation, Imperial College London, London, United Kingdom
- David Price Evans Infectious Diseases and Global Health Group, University of Liverpool, Liverpool, United Kingdom
| | - Timothy Miles Rawson
- The NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, Hammersmith Campus, London, United Kingdom
- Centre for Antimicrobial Optimisation, Imperial College London, London, United Kingdom
| |
Collapse
|
31
|
Roque-Borda CA, Carnero Canales CS, Primo LMDG, Colturato VMM, Polinário G, Di Filippo LD, Duarte JL, Chorilli M, da Silva Barud H, Pavan FR. Cellulose from bacteria as a delivery system for improved treatment of infectious diseases: A review of updates and prospects. Int J Biol Macromol 2024; 277:133831. [PMID: 39084978 DOI: 10.1016/j.ijbiomac.2024.133831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
Cellulose from bacteria is a high-purity biomaterial naturally produced by bacteria as part of their metabolic process. Although it inherently lacks antimicrobial activity, its modification with bioactive substances can significantly enhance its efficacy beyond that of the original compounds. This biomaterial features a unique ability to retain substantial quantities of liquids within its three-dimensional network, making it a prime candidate for biomedical applications. Versatile in its properties, it can be utilized across various industries. Previous research has highlighted its capacity to exhibit antimicrobial properties and to encapsulate nanostructured materials, thereby augmenting its antibacterial effectiveness. This review focuses on the use of cellulose from bacteria as a carrier for active compounds, specifically targeting antibacterial activity against drug-resistant strains. We explore its role in innovative bacterial cellulose-based systems, which present a promising solution for tackling bacterial resistance. This review aims to showcase the potential of bacterial cellulose in developing new devices and treatment strategies that address critical concerns in global health.
Collapse
Affiliation(s)
- Cesar Augusto Roque-Borda
- Universidad Católica de Santa María, Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Arequipa, Peru.
| | | | | | | | - Giulia Polinário
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil
| | | | - Jonatas L Duarte
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil
| | - Marlus Chorilli
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil
| | - Hernane da Silva Barud
- University of Araraquara (UNIARA), Biopolymers and Biomaterials Laboratory (BIOPOLMAT), Araraquara, São Paulo, Brazil
| | - Fernando R Pavan
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil.
| |
Collapse
|
32
|
Chan KBT, Rosete RCR, Yap Silva C. Recurrent Vegetative Pyoderma Gangrenosum of the Face: A Case Report. ACTA MEDICA PHILIPPINA 2024; 58:74-78. [PMID: 39431251 PMCID: PMC11484566 DOI: 10.47895/amp.v58i17.7480] [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] [Indexed: 10/22/2024]
Abstract
A 46-year-old male presented with an erythematous papule progressing into a vegetative plaque on the right cheek that resolved with cribriform scarring. Eight months after, a similar looking erythematous papule appeared on his left cheek. This papule rapidly progressed into a vegetative plaque within a week, and was associated with a pain score of 7 out of 10. Histopathology of the second lesion revealed suppurative dermatitis with diffuse dense infiltrates composed mostly of neutrophils. Cultures revealed few colonies of Enterobacter cloacae which was inconclusive. Pathergy test was negative. High dose systemic corticosteroids were started, with an observed rapid reduction of pain, inflammation, and ultimately resolution of the lesion with formation of cribriform scarring, confirming a case of vegetative pyoderma gangrenosum. It is important to note that not all inflamed and purulent lesions are infectious-neutrophilic dermatoses should always be considered.
Collapse
Affiliation(s)
- Koreen Blossom T Chan
- Department of Dermatology, Philippine General Hospital, University of the Philippines Manila
| | - Raisa Celine R Rosete
- Department of Dermatology, Philippine General Hospital, University of the Philippines Manila
| | - Claudine Yap Silva
- Department of Dermatology, Philippine General Hospital, University of the Philippines Manila
| |
Collapse
|
33
|
Li Y, Du Y, Wang M, Ai D. CSER: a gene regulatory network construction method based on causal strength and ensemble regression. Front Genet 2024; 15:1481787. [PMID: 39371416 PMCID: PMC11449711 DOI: 10.3389/fgene.2024.1481787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction Gene regulatory networks (GRNs) reveal the intricate interactions between and among genes, and understanding these interactions is essential for revealing the molecular mechanisms of cancer. However, existing algorithms for constructing GRNs may confuse regulatory relationships and complicate the determination of network directionality. Methods We propose a new method to construct GRNs based on causal strength and ensemble regression (CSER) to overcome these issues. CSER uses conditional mutual inclusive information to quantify the causal associations between genes, eliminating indirect regulation and marginal genes. It considers linear and nonlinear features and uses ensemble regression to infer the direction and interaction (activation or regression) from regulatory to target genes. Results Compared to traditional algorithms, CSER can construct directed networks and infer the type of regulation, thus demonstrating higher accuracy on simulated datasets. Here, using real gene expression data, we applied CSER to construct a colorectal cancer GRN and successfully identified several key regulatory genes closely related to colorectal cancer (CRC), including ADAMDEC1, CLDN8, and GNA11. Discussion Importantly, by integrating immune cell and microbial data, we revealed the complex interactions between the CRC gene regulatory network and the tumor microenvironment, providing additional new biomarkers and therapeutic targets for the early diagnosis and prognosis of CRC.
Collapse
Affiliation(s)
| | | | | | - Dongmei Ai
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| |
Collapse
|
34
|
Al-Shaarani AAQA, Pecoraro L. A review of pathogenic airborne fungi and bacteria: unveiling occurrence, sources, and profound human health implication. Front Microbiol 2024; 15:1428415. [PMID: 39364169 PMCID: PMC11446796 DOI: 10.3389/fmicb.2024.1428415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Airborne fungi and bacteria have been extensively studied by researchers due to their significant effects on human health. We provided an overview of the distribution and sources of airborne pathogenic microbes, and a detailed description of the detrimental effects that these microorganisms cause to human health in both outdoor and indoor environments. By analyzing the large body of literature published in this field, we offered valuable insights into how airborne microbes influence our well-being. The findings highlight the harmful consequences associated with the exposure to airborne fungi and bacteria in a variety of natural and human-mediated environments. Certain demographic groups, including children and the elderly, immunocompromised individuals, and various categories of workers are particularly exposed and vulnerable to the detrimental effect on health of air microbial pollution. A number of studies performed up to date consistently identified Alternaria, Cladosporium, Penicillium, Aspergillus, and Fusarium as the predominant fungal genera in various indoor and outdoor environments. Among bacteria, Bacillus, Streptococcus, Micrococcus, Enterococcus, and Pseudomonas emerged as the dominant genera in air samples collected from numerous environments. All these findings contributed to expanding our knowledge on airborne microbe distribution, emphasizing the crucial need for further research and increased public awareness. Collectively, these efforts may play a vital role in safeguarding human health in the face of risks posed by airborne microbial contaminants.
Collapse
Affiliation(s)
- Amran A. Q. A. Al-Shaarani
- College of Pharmaceutical Science & Moganshan Research Institute at Deqing County, Zhejiang University of Technology, Hangzhou, China
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lorenzo Pecoraro
- College of Pharmaceutical Science & Moganshan Research Institute at Deqing County, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
35
|
Khan M, Rahman MM, Paul SI, Lively JA. Detection of pathogenic bacteria in retailed shrimp from Bangladesh. Food Sci Nutr 2024; 12:6379-6388. [PMID: 39554329 PMCID: PMC11561818 DOI: 10.1002/fsn3.4260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 11/19/2024] Open
Abstract
The presence of pathogenic bacteria is a problem that might be present in farmed shrimp due to exposure in the environment or post-harvest handling. Retail farmed shrimp in Bangladesh (Penaeus monodon and Macrobrachium rosenbergii) were tested for common pathogenic bacteria namely Salmonella, L. monocytogenes, Vibrio spp., and E. coli. None of these bacteria were found and instead Enterobacter cloacae, Escherichia fergusonii, Proteus penneri, Klebsiella aerogenes, Enterococcus faecalis, Serratia marcescens, Citrobacter freundii, and Aeromonas dhakensis were detected. Pathogenic bacteria found in Bangladeshi shrimp may be due to the farm environment, poor handling during harvest or post-harvest, or unhygienic market conditions. The results indicate that retail shrimp from Bangladesh have food safety concerns. Proper laws and policies need to be enforced and implemented to ensure food safety related to fish and shrimp.
Collapse
Affiliation(s)
- Murshida Khan
- Department of Fisheries TechnologyBangabandhu Sheikh Mujibur Rahman Agricultural UniversityGazipurBangladesh
| | - Md. Mahbubur Rahman
- Institute of Biotechnology & Genetics Engineering (IBGE)Bangabandhu Sheikh Mujibur Rahman Agricultural UniversityGazipurBangladesh
| | - Sulav Indra Paul
- Institute of Biotechnology & Genetics Engineering (IBGE)Bangabandhu Sheikh Mujibur Rahman Agricultural UniversityGazipurBangladesh
| | - Julie Anderson Lively
- School of Renewable Natural ResourcesLouisiana State University Agricultural CenterBaton RougeLouisianaUSA
| |
Collapse
|
36
|
Sun L, Meng N, Wang H, Wang Z, Jiao X, Wang J. Occurrence and characteristics of bla OXA-181-carrying Klebsiella aerogenes from swine in China. J Glob Antimicrob Resist 2024; 38:35-41. [PMID: 38763331 DOI: 10.1016/j.jgar.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/09/2023] [Accepted: 04/19/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVES Klebsiella aerogenes is a largely understudied opportunistic pathogen that can cause sepsis and lead to high mortality rates. In this study, we reported the occurrence of carbapenem-resistant blaOXA-181-carrying Klebsiella aerogenes from swine in China and elucidate their genomic characteristics. METHODS A total of 126 samples, including 109 swine fecal swabs, 14 environmental samples, and three feed samples were collected from a pig farm in China. The samples were enriched with LB broth culture and then inoculated into MacConkey agar plates for bacterial isolation. After PCR detection of carbapenemases genes, the blaOXA-181-carrying isolates were subjected to antimicrobial susceptibility testing, and whole-genome sequence analysis. RESULTS Four Klebsiella aerogenes isolates carrying the blaOXA-181 gene were obtained from swine faecal samples. All the 4 strains were belonged to ST438. The blaOXA-181 genes were located in IncX3-ColKP3 hybrid plasmids with the core genetic structure of IS26-ΔIS3000-ΔISEcp1-blaOXA-181-ΔlysR-ΔereA-ΔrepA-ISKpn19-tinR-qnrS1-ΔIS2-IS26, which suggests the potential for horizontal transfer and further dissemination of this resistance gene among Enterobacteriaceae and other sources. CONCLUSIONS This study represents the first instance of OXA-181-producing K. aerogenes being identified from swine faeces in China. It is crucial to maintain continuous monitoring and ongoing attention to the detection of K. aerogenes carrying blaOXA-181 and other resistance genes in pigs.
Collapse
Affiliation(s)
- Lin Sun
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
| | - Nan Meng
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Hanyun Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Zhenyu Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China.
| | - Jing Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China.
| |
Collapse
|
37
|
Si Z, Zhao S, Hao Y, Wang Q, Zhong Y, Liu Y, Chen R, Jin Y, Lu Z. Colistin Resistance Mechanisms and Molecular Epidemiology of Enterobacter c loacae Complex Isolated from a Tertiary Hospital in Shandong, China. Infect Drug Resist 2024; 17:3723-3735. [PMID: 39221183 PMCID: PMC11365514 DOI: 10.2147/idr.s473580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Background Enterobacter cloacae complex (ECC), which includes major nosocomial pathogens, causes urinary, respiratory, and bloodstream infections in humans, for which colistin is one of the last-line drugs. Objective This study aimed to analyse the epidemiology and resistance mechanisms of colistin-resistant Enterobacter cloacae complex (ECC) strains isolated from Shandong, China. Methods Two hundred non-repetitive ECC strains were collected from a tertiary hospital in Shandong Province, China, from June 2020 to June 2022. Whole-genome sequencing and bioinformatics analyses were performed to understand the molecular epidemiology of the colistin-resistant ECC strains. The nucleotide sequences of heat shock protein (hsp60) were analyzed by using BLAST search to classify ECC. The gene expression levels of ramA, soxS, acrA, acrB, phoP, and phoQ were assessed using RT-qPCR. MALDI-TOF MS was used to analyse the modification of lipid A. Results Twenty-three colistin-resistant strains were detected among the 200 ECC clinical strains (11.5%). The hsp60 cluster analysis revealed that 20 of the 23 ECC strains belonged to heterogeneous resistance clusters. Variants of mgrB, phoPQ, and pmrAB, particularly phoQ and pmrB, were detected in the 23 ECC strains. The soxS and acrA genes were significantly overexpressed in all 23 colistin-resistant ECC strains (P < 0.05). Additionally, all 23 ECC strains contained modified lipid A related to colistin resistance, which showed five ion peaks at m/z 1876, 1920, 1955, 2114, and 2158. Among the 23 ECC strains, 6 strains possessed a phosphoethanolamine (pETN) moiety, 16 strains possessed a 4-amino-4-deoxy-L-arabinose (-L-Ara4N) moiety, and one strain had both pETN and -L-Ara4N moieties. Conclusion This study suggests that diverse colistin resistance existed in ECC, including unknown resistance mechanisms, exist in ECC. Mechanistic investigations of colistin resistance are warranted to optimise colistin use in clinical settings and minimise the emergence of resistance.
Collapse
Affiliation(s)
- Zaifeng Si
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Clinical Laboratory, Dezhou Municipal Hospital of Traditional Chinese Medicine, Dezhou, Shandong, People’s Republic of China
| | - Shengmei Zhao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Yingying Hao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Qian Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Yanfa Zhong
- Department of Clinical Laboratory, Dezhou Municipal Hospital of Traditional Chinese Medicine, Dezhou, Shandong, People’s Republic of China
| | - Yue Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Ran Chen
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Yan Jin
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
38
|
Feng Y, Yang Y, Hu Y, Xiao Y, Xie Y, Wei L, Wen H, Zhang L, McNally A, Zong Z. Population genomics uncovers global distribution, antimicrobial resistance, and virulence genes of the opportunistic pathogen Klebsiella aerogenes. Cell Rep 2024; 43:114602. [PMID: 39137112 PMCID: PMC11372444 DOI: 10.1016/j.celrep.2024.114602] [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: 02/13/2024] [Revised: 06/13/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
Klebsiella aerogenes is an understudied and clinically important pathogen. We therefore investigate its population structure by genome analysis aligned with metadata. We sequence 130 non-duplicated K. aerogenes clinical isolates and identify two inter-patient transmission events. We then retrieve all publicly available K. aerogenes genomes (n = 1,026, accessed by January 1, 2023) and analyze them with our 130 genomes. We develop a core-genome multi-locus sequence-typing scheme. We find that K. aerogenes is a species complex comprising four phylogroups undergoing evolutionary divergence, likely forming three species. We delineate remarkable clonal diversity and identify three worldwide-distributed carbapenemase-encoding clonal clusters, representing high-risk lineages. We uncover that K. aerogenes has an open genome equipped by a large arsenal of antimicrobial resistance genes. We identify two genetic regions specific for K. aerogenes, encoding a type VI secretion system and flagella/chemotaxis for motility, respectively, both contributing to the virulence. These results provide much-needed insights into the population structure and pan-genomes of K. aerogenes.
Collapse
Affiliation(s)
- Yu Feng
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China; Center for Pathogen Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yongqiang Yang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China; Center for Pathogen Research, West China Hospital, Sichuan University, Chengdu, China
| | - Ya Hu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China; Center for Pathogen Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yuling Xiao
- Laboratory of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Xie
- Laboratory of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Li Wei
- Department of Infection Control, West China Hospital, Sichuan University, Chengdu, China
| | - Hongxia Wen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China; Center for Pathogen Research, West China Hospital, Sichuan University, Chengdu, China
| | - Linwan Zhang
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Alan McNally
- Institute of Microbiology and Infection, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Zhiyong Zong
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China; Center for Pathogen Research, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
39
|
Burcham ZM. Comparative genomic analysis of an emerging Pseudomonadaceae member, Thiopseudomonas alkaliphila. Microbiol Spectr 2024; 12:e0415723. [PMID: 38934605 PMCID: PMC11302033 DOI: 10.1128/spectrum.04157-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Thiopseudomonas alkaliphila, an organism recently classified within the Pseudomonadaceae family, has been detected in diverse sources such as human tissues, animal guts, industrial fermenters, and decomposition environments, suggesting a diverse ecological role. However, a large knowledge gap exists in how T. alkaliphila functions. In this comparative genomic analysis, adaptations indicative of habitat specificity among strains and genomic similarity to known opportunistic pathogens are revealed. Genomic investigation reveals a core metabolic utilization of multiple oxidative and non-oxidative catabolic pathways, suggesting adaptability to varied environments and carbon sources. The genomic repertoire of T. alkaliphila includes secondary metabolites, such as antimicrobials and siderophores, indicative of its involvement in microbial competition and resource acquisition. Additionally, the presence of transposases, prophages, plasmids, and Clustered Regularly Interspaced Short Palindromic Repeats-Cas systems in T. alkaliphila genomes suggests mechanisms for horizontal gene transfer and defense against viral predation. This comprehensive genomic analysis expands our understanding on the ecological functions, community interactions, and potential virulence of T. alkaliphila, while emphasizing its adaptability and diverse capabilities across environmental and host-associated ecosystems.IMPORTANCEAs the microbial world continues to be explored, new organisms will emerge with beneficial and/or pathogenetic impact. Thiopseudomonas alkaliphila is a species originally isolated from clinical human tissue and fluid samples but has not been attributed to disease. Since its classification, T. alkaliphila has been found in animal guts, animal waste, decomposing remains, and biogas fermentation reactors. This is the first study to provide an in-depth view of the metabolic potential of publicly available genomes belonging to this species through a comparative genomics and draft pangenome calculation approach. It was found that T. alkaliphila is metabolically versatile and likely adapts to diverse energy sources and environments, which may make it useful for bioremediation and in industrial settings. A range of virulence factors and antibiotic resistances were also detected, suggesting T. alkaliphila may operate as an undescribed opportunistic pathogen.
Collapse
Affiliation(s)
- Zachary M. Burcham
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
40
|
Kumari K, Aggarwal Y, Singh RP. Molecular characterization and in-depth genomic analysis to unravel the pathogenic features of an environmental isolate Enterobacter sp. S-33. Int Microbiol 2024; 27:1095-1110. [PMID: 38044418 DOI: 10.1007/s10123-023-00461-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Enterobacter species represent widely distributed opportunistic pathogens, commonly associated with plants and humans. In the present study, we performed a detailed molecular characterization as well as genomic study of a type VI secretion system (T6SS) bacterium belonging to member of the family Enterobacteriaceae and named Enterobacter sp. S-33. The comparative sequence analysis of the 16S rRNA gene showed that the strain was closely related to other Enterobacter species. The complete genome of the strain with a genome size of 4.6 Mbp and GC-content of 55.63% was obtained through high-quality sequencing. The genomic analysis with online tools unravelled the various genes belonging to the bacterial secretion system, antibiotic resistance, virulence, efflux pumps, etc. The isolate showed the motility behavior that contributes to Enterobacter persistence in a stressed environment and further supports infections. PCR amplification and further sequencing confirmed the presence of drug-efflux genes acrA, acrB, and outer membrane genes, viz. OmpA, OmpC, and OmpF. The cell surface hydrophobicity and co-aggregation assay against different bacterial strains illustrated its putative pathogenic nature. Genome mining identified various biosynthetic gene clusters (BGCs) corresponding to non-ribosomal proteins (NRPS), siderophore, and arylpolyene production. Briefly, genome sequencing and detailed characterization of environmental Enterobacter isolate will assist in understanding the epidemiology of Enterobacter species, and the further prevention and treatment of infectious diseases caused by these broad-host range species.
Collapse
Affiliation(s)
- Kiran Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Yogender Aggarwal
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Rajnish Prakash Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India.
| |
Collapse
|
41
|
Mohamed MAF, Benjamin I, Okon GA, Ahmad I, Khan SAPM, Patel H, Agwamba EC, Louis H. Insights into in-vitro studies and molecular modelling of the antimicrobial efficiency of 4-chlorobenzaldehyde and 4-methoxybenzaldehyde derivatives. J Biomol Struct Dyn 2024; 42:6042-6064. [PMID: 37504959 DOI: 10.1080/07391102.2023.2239917] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023]
Abstract
Owing to the significant gap in the knowledge and understanding of the mechanisms of antimicrobial action and the development of resistance, the optimization of antimicrobial therapies therefore becomes a necessity. It is on this note, that this study seeks to both experimentally and theoretically investigate the antimicrobial efficiency of two synthesized compounds namely; 1-((4-methoxyphenyl) (morpholino)methyl)thiourea (MR1) and diethyl 4-(4-chlorophenyl)-2,6-diphenyl-1,4-dihydropyridine-3,5-dicarboxylate (HRC). Utilizing the density functional theory (DFT), the compounds were optimized at ωB97XD/6-31++G(2d, 2p) level of theory. This provided a clear explanation for their distinct reactivity and stability potentials. More so, the natural bond orbital (NBO) analysis confirmed strong intra and intermolecular interactions, which agreed with the calculated reactivity parameters and density of states (DOS). Upon assessing the antimicrobial efficacy of the synthesized compounds, it was found that they exhibited lower activity against Enterobacter and A. niger, but considerable activity against Moraxella. In contrast, they showed higher activity against B. subtilis and Trichophyton, indicating that the compounds are more effective against gram-positive bacteria than gram-negative ones. Hence, it can be asserted that the synthesized compounds have superior antifungal action than antibacterial activity. A fascinating aspect of the data is that they show interactions that are incredibly insightful, totally correlating with the simulations of both molecular docking and molecular dynamics. Therefore, the alignment between experimental findings and computational simulations strengthens the validity of the study's conclusions, emphasizing the significance of the synthesized compounds in the context of optimizing antimicrobial therapies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mashood A F Mohamed
- PG and Research Department of Chemistry, Jamal Mohamed College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - Innocent Benjamin
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Microbiology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Gideon A Okon
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Dhule, Maharashtra, India
| | - Syed A P M Khan
- PG and Research Department of Chemistry, Jamal Mohamed College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamilnadu, India
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Ernest C Agwamba
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
| | - Hitler Louis
- Computational and Bio-Simulation Research Group, University of Calabar, Calabar, Nigeria
- Department of Pure and Applied Chemistry, University of Calabar, Calabar, Nigeria
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
42
|
Habibi N, Uddin S, Behbehani M, Mustafa AS, Al-Fouzan W, Al-Sarawi HA, Safar H, Alatar F, Al Sawan RMZ. Aerosol-Mediated Spread of Antibiotic Resistance Genes: Biomonitoring Indoor and Outdoor Environments. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:983. [PMID: 39200594 PMCID: PMC11353316 DOI: 10.3390/ijerph21080983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/02/2024]
Abstract
Antimicrobial resistance (AMR) has emerged as a conspicuous global public health threat. The World Health Organization (WHO) has launched the "One-Health" approach, which encourages the assessment of antibiotic resistance genes (ARGs) within an environment to constrain and alleviate the development of AMR. The prolonged use and overuse of antibiotics in treating human and veterinary illnesses, and the inability of wastewater treatment plants to remove them have resulted in elevated concentrations of these metabolites in the surroundings. Microbes residing within these settings acquire resistance under selective pressure and circulate between the air-land interface. Initial evidence on the indoor environments of wastewater treatment plants, hospitals, and livestock-rearing facilities as channels of AMR has been documented. Long- and short-range transport in a downwind direction disseminate aerosols within urban communities. Inhalation of such aerosols poses a considerable occupational and public health risk. The horizontal gene transfer (HGT) is another plausible route of AMR spread. The characterization of ARGs in the atmosphere therefore calls for cutting-edge research. In the present review, we provide a succinct summary of the studies that demonstrated aerosols as a media of AMR transport in the atmosphere, strengthening the need to biomonitor these pernicious pollutants. This review will be a useful resource for environmental researchers, healthcare practitioners, and policymakers to issue related health advisories.
Collapse
Affiliation(s)
- Nazima Habibi
- Environment and Life Science Research Centre, Kuwait Institute for Scientific Research, Shuwaikh 13109, Kuwait
| | - Saif Uddin
- Environment and Life Science Research Centre, Kuwait Institute for Scientific Research, Shuwaikh 13109, Kuwait
| | - Montaha Behbehani
- Environment and Life Science Research Centre, Kuwait Institute for Scientific Research, Shuwaikh 13109, Kuwait
| | - Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Jabriya 13060, Kuwait
| | - Wadha Al-Fouzan
- Department of Microbiology, Faculty of Medicine, Kuwait University, Jabriya 13060, Kuwait
| | | | - Hussain Safar
- OMICS-RU, Health Science Centre, Kuwait University, Jabriya 13060, Kuwait
| | - Fatemah Alatar
- Serology and Molecular Microbiology Reference Laboratory, Mubarak Al-Kabeer Hospital, Ministry of Health, Kuwait City 13110, Kuwait
| | - Rima M. Z. Al Sawan
- Neonatology Department, Farwaniya Hospital, Ministry of Health, Sabah Al-Nasser 92426, Kuwait
| |
Collapse
|
43
|
Mosaffa F, Saffari F, Veisi M, Tadjrobehkar O. Some virulence genes are associated with antibiotic susceptibility in Enterobacter cloacae complex. BMC Infect Dis 2024; 24:711. [PMID: 39030479 PMCID: PMC11264964 DOI: 10.1186/s12879-024-09608-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Enterobacter cloacae complex (ECC) including different species are isolated from different human clinical samples. ECC is armed by many different virulence genes (VGs) and they were also classified among ESKAPE group by WHO recently. The present study was designed to find probable association between VGs and antibiotic susceptibility in different ECC species. METHODS Forty-five Enterobacter isolates that were harvested from different clinical samples were classified in four different species. Seven VGs were screened by PCR technique and antibiotic susceptibility assessment was performed by disk-diffusion assay. RESULT Four Enterobacter species; Enterobacter cloacae (33.3%), Enterobacter hormaechei (55.6%), Enterobacter kobei (6.7%) and Enterobacter roggenkampii (4.4%) were detected. Minimum antibiotic resistance was against carbapenem agents and amikacin even in MDR isolates. 33.3% and 13.3% of isolates were MDR and XDR respectively. The rpoS (97.8%) and csgD (11.1%) showed maximum and minimum frequency respectively. Blood sample isolated were highly virulent but less resistant in comparison to the other sample isolates. The csgA, csgD and iutA genes were associated with cefepime sensitivity. CONCLUSION The fepA showed a predictory role for differentiating of E. hormaechei from other species. More evolved iron acquisition system in E. hormaechei was hypothesized. The fepA gene introduced as a suitable target for designing novel anti-virulence/antibiotic agents against E. hormaechei. Complementary studies on other VGs and ARGs and with bigger study population is recommended.
Collapse
Affiliation(s)
- Fatemeh Mosaffa
- Departement of Medical Microbiology (Bacteriology & Virology), Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fereshteh Saffari
- Departement of Medical Microbiology (Bacteriology & Virology), Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahin Veisi
- Departement of Medical Microbiology (Bacteriology & Virology), Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Omid Tadjrobehkar
- Departement of Medical Microbiology (Bacteriology & Virology), Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
44
|
Jin H, Yan Z, Ge X, Wang Q, Wang H, Du X, Liu H, Yang C, Xiang Y, Tian S, Qiu S, Zhou Y. Genomic and Phenotypic Analysis of bla KPC-2 Associated Carbapenem Resistance in Klebsiella aerogenes: Insights into Clonal Spread and Resistance Mechanisms Across Hospital Departments in Beijing. Infect Drug Resist 2024; 17:2735-2749. [PMID: 38974314 PMCID: PMC11227833 DOI: 10.2147/idr.s458182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/29/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose This study conducted an phenotypic and whole-genome sequencing analysis with Klebsiella aerogenes to elucidate its clinical epidemiological characteristics, antimicrobial resistance (AMR) phenotype, biofilm formation ability and hemolytic activity testing, AMR genes and phylogenetic relationships, so as to provide a further understanding of the intra-hospital strain transmission. Methods Samples were collected from a hospital in Beijing between 2020 and 2022. All strains underwent bacterial identification, antimicrobial susceptibility testing (AST) using the VITEK-2 compact system. Biofilm formation ability and hemolytic activity were tested. Second-generation sequencing was applied to all strains, with those carrying the bla KPC gene were selected for third-generation sequencing. Whole-genome analysis identified resistance genes, plasmid types, MLST typing, and phylogenetic relationships. Plasmids were assembled to detect plasmid structures and AMR gene location. Results Among the 42 K. aerogenes isolates, 21 were carbapenem-resistant K. aerogenes (CRKA). All strains exhibited strong biofilm formation and no hemolytic activity. Most were sourced from sputum (83.3%). CRKA demonstrated extensive resistance to antibiotics, particularly β-lactamase inhibitors and Cefotetan. This resistance pattern was closely associated with the presence of an IncFII(pHN7A8) plasmid, which carried multiple resistance genes, including bla KPC-2, bla CTX-M-65, bla TEM-1, rmtB and a large number of mobile elements. The majority of CRKA strains clustered within the same branch of the phylogenetic tree, exhibiting minimal single nucleotide polymorphism (0-13 SNPs) differences, and they shared the same sequence type (ST292), resistance genes, and plasmids, originating from different departments, suggesting clonal transmission among the hospital. Conclusion Our research reveals that the clonal transmission of CRKA occurs across various departments within the hospital. The widespread resistance observed in CRKA, attributed to the presence of bla KPC and ESBLs genes, underscores the need for heightened vigilance to prevent the further dissemination of CRKA within the hospital and, potentially, throughout the wider community.
Collapse
Affiliation(s)
- Hang Jin
- School of Public Health, Zhengzhou University, Zhengzhou, People’s Republic of China
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Zhongqiang Yan
- Department of Disease Prevention and Control, The Second Medical Center of PLA General Hospital, Beijing, People’s Republic of China
| | - Xin Ge
- School of Public Health, Anhui Medical University, Hefei, People’s Republic of China
| | - Qi Wang
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Hui Wang
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Xinying Du
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Hongbo Liu
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Chaojie Yang
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Ying Xiang
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Sai Tian
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Shaofu Qiu
- School of Public Health, Zhengzhou University, Zhengzhou, People’s Republic of China
- Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention of Chinese PLA, Beijing, People’s Republic of China
| | - Yu Zhou
- Department of Clinical Laboratory, National Clinical Research Center for Geriatric Diseases, The Second Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
45
|
González-Gómez JP, Rodríguez-Arellano SN, Gomez-Gil B, Vergara-Jiménez MDJ, Chaidez C. Genomic and biological characterization of bacteriophages against Enterobacter cloacae, a high-priority pathogen. Virology 2024; 595:110100. [PMID: 38714025 DOI: 10.1016/j.virol.2024.110100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/20/2024] [Accepted: 05/02/2024] [Indexed: 05/09/2024]
Abstract
Enterobacter cloacae is a clinically significant pathogen due to its multi-resistance to antibiotics, presenting a challenge in the treatment of infections. As concerns over antibiotic resistance escalate, novel therapeutic approaches have been explored. Bacteriophages, characterized by their remarkable specificity and ability to self-replicate within target bacteria, are emerging as a promising alternative therapy. In this study, we isolated and partially characterized nine lytic bacteriophages targeting E. cloacae, with two selected for comprehensive genomic analysis based on their host range and bacteriolytic activity. All identified phages exhibited a narrow host range, demonstrated stability within a temperature range of 30-60 °C, displayed pH tolerance from 3 to 10, and showed an excellent bacteriolytic capacity for up to 18 h. Notably, the fully characterized phage genomes revealed an absence of lysogenic, virulence, or antibiotic-resistance genes, positioning them as promising candidates for therapeutic intervention against E. cloacae-related diseases. Nonetheless, translating this knowledge into practical therapeutic applications mandates a deeper understanding of bacteriophage interactions within complex biological environments.
Collapse
Affiliation(s)
- Jean Pierre González-Gómez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA), Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Carretera a Eldorado km 5.5, Campo El Diez, 80110, Culiacan, Sinaloa, Mexico
| | | | - Bruno Gomez-Gil
- Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Unidad Mazatlán en Acuicultura y Manejo Ambiental, AP 711, Mazatlan, Sinaloa, Mexico
| | | | - Cristobal Chaidez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA), Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Carretera a Eldorado km 5.5, Campo El Diez, 80110, Culiacan, Sinaloa, Mexico.
| |
Collapse
|
46
|
Ghazawi A, Anes F, Mouftah S, Elbediwi M, Baig A, Alketbi M, Almazrouei F, Alhashmi M, Alzarooni N, Manzoor A, Habib I, Strepis N, Nabi A, Khan M. Genomic Study of High-Risk Clones of Enterobacter hormaechei Collected from Tertiary Hospitals in the United Arab Emirates. Antibiotics (Basel) 2024; 13:592. [PMID: 39061274 PMCID: PMC11274081 DOI: 10.3390/antibiotics13070592] [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: 05/04/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Enterobacter hormaechei has emerged as a significant pathogen within healthcare settings due to its ability to develop multidrug resistance (MDR) and survive in hospital environments. This study presents a genome-based analysis of carbapenem-resistant Enterobacter hormaechei isolates from two major hospitals in the United Arab Emirates. Eight isolates were subjected to whole-genome sequencing (WGS), revealing extensive resistance profiles including the blaNDM-1, blaOXA-48, and blaVIM-4 genes. Notably, one isolate belonging to ST171 harbored dual carbapenemase genes, while five isolates exhibited colistin resistance without mcr genes. The presence of the type VI secretion system (T6SS), various adhesins, and virulence genes contributes to the virulence and competitive advantage of the pathogen. Additionally, our isolates (87.5%) possessed ampC β-lactamase genes, predominantly blaACT genes. The genomic context of blaNDM-1, surrounded by other resistance genes and mobile genetic elements, highlights the role of horizontal gene transfer (HGT) in the spread of resistance. Our findings highlight the need for rigorous surveillance, strategic antibiotic stewardship, and hospital-based WGS to manage and mitigate the spread of these highly resistant and virulent pathogens. Accurate identification and monitoring of Enterobacter cloacae complex (ECC) species and their resistance mechanisms are crucial for effective infection control and treatment strategies.
Collapse
Affiliation(s)
- Akela Ghazawi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| | - Febin Anes
- Veterinary Public Health Research Laboratory, Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (F.A.); (I.H.)
| | - Shaimaa Mouftah
- Department of Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza 12578, Egypt;
| | - Mohammed Elbediwi
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, 14163 Berlin, Germany;
- Animal Health Research Institute, Agriculture Research Centre, Cairo 12618, Egypt
| | - Awase Baig
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| | - Muna Alketbi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| | - Fatema Almazrouei
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| | - Mariam Alhashmi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| | - Norah Alzarooni
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| | - Ashrat Manzoor
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| | - Ihab Habib
- Veterinary Public Health Research Laboratory, Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (F.A.); (I.H.)
| | - Nikolaos Strepis
- Department of Medical Microbiology & Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), 3015 GD Rotterdam, The Netherlands;
| | - Anju Nabi
- Microbiology and Immunology Department, Dubai Hospital, Dubai P.O. Box 53735, United Arab Emirates;
| | - Mushtaq Khan
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.G.); (A.B.); (M.A.); (F.A.); (M.A.); (N.A.); (A.M.)
| |
Collapse
|
47
|
Fanucci V, Chauhan D, Mascio CE, Arora S, Gupta S, Weisse ME, Kohli U. ENTEROBACTER CLOACAE CARDIAC IMPLANTABLE ELECTRONIC DEVICE INFECTION. Pediatr Infect Dis J 2024:00006454-990000000-00913. [PMID: 38916912 DOI: 10.1097/inf.0000000000004433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Enterobacter cloacae cardiac implantable electronic device infections are rare but can be associated with significant morbidity and mortality. We report an 11-year-old female with Enterobacter cloacae infection of a dual-chamber transvenous pacemaker pocket. The report is supplemented by a comprehensive review of the literature on Enterobacter cloacae cardiac implantable electronic device infections.
Collapse
Affiliation(s)
- Victoria Fanucci
- From the West Virginia University School of Medicine, Morgantown, West Virginia
| | - Dhaval Chauhan
- Department of Cardiovascular and Thoracic Surgery, West Virginia University School of Medicine and West Virginia University Children's Heart Center, Morgantown, West Virginia
| | - Christopher E Mascio
- Department of Cardiovascular and Thoracic Surgery, West Virginia University School of Medicine and West Virginia University Children's Heart Center, Morgantown, West Virginia
| | - Sandeep Arora
- Section of Electrophysiology, Heart and Vascular Institute, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Shipra Gupta
- Section of Pediatric Infectious Diseases, Department of Pediatrics, Westchester Medical Center, Valhalla, New York
| | - Martin E Weisse
- Division of Pediatric Infectious Diseases, Department of Pediatrics, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Utkarsh Kohli
- Division of Pediatric Cardiology, Department of Pediatrics, West Virginia University School of Medicine and West Virginia University Children's Heart Center, Morgantown, West Virginia
| |
Collapse
|
48
|
Shenbagavalli K, Suganya K, Sundaram E, Murugan M, Sivasamy Vasantha V. First organic fluorescence immunoassay for the detection of Enterobacter cloacae in food matrixes. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3927-3937. [PMID: 38832637 DOI: 10.1039/d4ay00289j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
For the first time, a novel fluorescent moiety, 2-amino-4-(7-formyl-1,8-dihydropyren-2-yl)-7-hydroxy-4H-chromene-3-carbonitrile (ACC), was synthesized by an ultrasonication method. The synthesis of this moiety was confirmed via structural elucidation using FTIR and NMR spectroscopy techniques. Further, photophysical properties of the fluorescent moiety were tested using UV-visible and emission spectroscopy techniques. In this case, the moiety was tagged with an antibody of Enterobacter cloacae via 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide and N-hydroxysuccinimide (EDC/NHS) coupling and applied as a sensing element for the detection of Enterobacter cloacae (E. cloacae) by UV-visible and emission spectroscopy techniques. The developed fluorescent sensor detected E. cloacae via a FRET mechanism. Under optimized conditions, ACC-anti-E. cloacae detected E. cloacae in the linear range from 101 to 1010 CFU mL-1 with a limit of detection (LOD) of 10.55 CFU mL-1. The developed sensor was applied for the detection of E. cloacae in food samples such as orange, pomegranate, milk, rice, tomato, potato and onion.
Collapse
Affiliation(s)
- Kathiravan Shenbagavalli
- Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai- 625021, TamilNadu, India.
| | - Kannan Suganya
- Central Research Laboratory, Vinayaka Mission's,Medical College and Hospital, Vinayaka Mission's Research Foundation, Karaikal- 609609, India
| | - Ellairaja Sundaram
- Depatment of Chemistry, Vivekanada College, Tiruvedakam, West, Madurai- 625234, Tamilnadu, India
| | - Marudhamuthu Murugan
- Department of Microbial Technology, School of Biological Science, Madurai Kamaraj University, Madurai-625021, Tamil Nadu, India
| | - Vairathevar Sivasamy Vasantha
- Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai- 625021, TamilNadu, India.
| |
Collapse
|
49
|
Panyod S, Wu WK, Chang CT, Wada N, Ho HC, Lo YL, Tsai SP, Chen RA, Huang HS, Liu PY, Chen YH, Chuang HL, Shen TCD, Tang SL, Ho CT, Wu MS, Sheen LY. Common dietary emulsifiers promote metabolic disorders and intestinal microbiota dysbiosis in mice. Commun Biol 2024; 7:749. [PMID: 38902371 PMCID: PMC11190199 DOI: 10.1038/s42003-024-06224-3] [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: 03/24/2023] [Accepted: 04/22/2024] [Indexed: 06/22/2024] Open
Abstract
Dietary emulsifiers are linked to various diseases. The recent discovery of the role of gut microbiota-host interactions on health and disease warrants the safety reassessment of dietary emulsifiers through the lens of gut microbiota. Lecithin, sucrose fatty acid esters, carboxymethylcellulose (CMC), and mono- and diglycerides (MDG) emulsifiers are common dietary emulsifiers with high exposure levels in the population. This study demonstrates that sucrose fatty acid esters and carboxymethylcellulose induce hyperglycemia and hyperinsulinemia in a mouse model. Lecithin, sucrose fatty acid esters, and CMC disrupt glucose homeostasis in the in vitro insulin-resistance model. MDG impairs circulating lipid and glucose metabolism. All emulsifiers change the intestinal microbiota diversity and induce gut microbiota dysbiosis. Lecithin, sucrose fatty acid esters, and CMC do not impact mucus-bacterial interactions, whereas MDG tends to cause bacterial encroachment into the inner mucus layer and enhance inflammation potential by raising circulating lipopolysaccharide. Our findings demonstrate the safety concerns associated with using dietary emulsifiers, suggesting that they could lead to metabolic syndromes.
Collapse
Affiliation(s)
- Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Kai Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Bachelor Program of Biotechnology and Food Nutrition, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Ting Chang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Naohisa Wada
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Han-Chen Ho
- Department of Anatomy, Tzu Chi University, Hualien, Taiwan, ROC
| | - Yi-Ling Lo
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Sing-Ping Tsai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Huai-Syuan Huang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Po-Yu Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
| | - Yi-Hsun Chen
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Ting-Chin David Shen
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sen-Lin Tang
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC.
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC.
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC.
- National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
50
|
Khalil MAM, Sadagah NM, Alqurashi MM, Basha AA, Sakran HIM, Assiri IMN, Aboalsamh GA, Al-Qurashi SH. A Rare Case of Gas Forming Enterobacter cloacae Leading to Bleeding Mycotic Pseudoaneurysm of Transplant Renal Artery Culminating in Graft Nephrectomy. J Med Cases 2024; 15:110-114. [PMID: 38855292 PMCID: PMC11161181 DOI: 10.14740/jmc4231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024] Open
Abstract
Enterobacter cloacae belongs to Enterobacter genus. It is a common gram-negative, facultative anaerobic, rod-shaped organism. It causes a variety of nosocomial infections including urinary tract infection, pneumonia, wound infection, osteomyelitis and endocarditis. Over time Enterobacter cloacae complex (ECC) has developed to be resistant to antibiotics including carbapenem. It has been rarely reported to cause gas gangrene and never been reported to cause pseudoaneurysm (PA) of transplant renal artery. We report and share our experience with this rare case of gas forming and muti-drug resistant ECC which led to mycotic PA of transplant renal artery, complicated by bleeding and infected hematoma and which resulted in graft nephrectomy.
Collapse
Affiliation(s)
| | - Nihal Mohammed Sadagah
- Centre of Renal Diseases and Transplantation, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Moayad Majed Alqurashi
- Division of Adult Infectious Diseases, Department of Medicine, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Ahmed Abdelahad Basha
- Centre of Renal Diseases and Transplantation, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | | | | | - Ghaleb Anas Aboalsamh
- Centre of Renal Diseases and Transplantation, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Salem H. Al-Qurashi
- Centre of Renal Diseases and Transplantation, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| |
Collapse
|