1
|
Pearson T, Furstenau T, Wood C, Rigas V, Drake K, Sahl J, Maltinsky S, Currie BJ, Mayo M, Hall C, Keim P, Fofanov V. Population sequencing for phylogenetic diversity and transmission analyses. Proc Natl Acad Sci U S A 2025; 122:e2424797122. [PMID: 40460116 DOI: 10.1073/pnas.2424797122] [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: 11/26/2024] [Accepted: 04/29/2025] [Indexed: 06/11/2025] Open
Abstract
Genomic diversity in pathogen populations is foundational for evolution and adaptation. Understanding population-level diversity is also essential for tracking sources and revealing detailed pathways of transmission and spread. For bacteria, culturing, isolating, and sequencing the large number of individual colonies required to adequately sample diversity can be prohibitively time-consuming and expensive. While sequencing directly from a mixed population will show variants among reads, they cannot be linked to reveal allele combinations associated with phylogenetic inheritance patterns. Here, we describe the theory and method for using population sequencing directly from a mixed sample, along with a minimal number of individually sequenced colonies, to describe the phylogenetic diversity of a population without haplotype reconstruction. To demonstrate the utility of population sequencing in capturing phylogenetic diversity, we compared isogenic clones to population sequences of Burkholderia pseudomallei from sputum of a single patient. Our results point to the pathogen population being highly structured, suggesting that for some pathogens, sputum sampling may preserve structuring in the lungs and thus present a noninvasive alternative to understanding colonization, movement, and pathogen/host interactions. We also analyzed population sequences of Staphylococcus aureus derived from different people and different body sites to reveal directionality of transmission between hosts and across body sites, demonstrating the power and utility for characterizing the spread of disease and identification of reservoirs at the finest levels. We anticipate that population sequencing and analysis can be broadly applied to accelerate research in a wide range of fields reliant on a foundational understanding of population phylogenetic diversity.
Collapse
Affiliation(s)
- Talima Pearson
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
| | - Tara Furstenau
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
- Health and Bioinformatics, School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011
| | - Colin Wood
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
| | - Vanessa Rigas
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT 0811, Australia
| | - Kylie Drake
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
| | - Jason Sahl
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
| | - Sara Maltinsky
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
| | - Bart J Currie
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT 0811, Australia
- Infectious Diseases Department and Northern Territory Medical Program, Royal Darwin Hospital, Darwin, NT 0811, Australia
| | - Mark Mayo
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT 0811, Australia
| | - Carina Hall
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
| | - Paul Keim
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
| | - Viacheslav Fofanov
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011
- Health and Bioinformatics, School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011
| |
Collapse
|
2
|
Graham CT, Gordon S, Kubes P. A historical perspective of Kupffer cells in the context of infection. Cell Tissue Res 2025; 400:121-136. [PMID: 39392500 DOI: 10.1007/s00441-024-03924-4] [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: 06/28/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
The Kupffer cell was first discovered by Karl Wilhelm von Kupffer in 1876, labeling them as "Sternzellen." Since their discovery as the primary macrophages of the liver, researchers have gradually gained an in-depth understanding of the identity, functions, and influential role of Kupffer cells, particularly in infection. It is becoming clear that Kupffer cells perform important tissue-specific functions in homeostasis and disease. Stationary in the sinusoids of the liver, Kupffer cells have a high phagocytic capacity and are adept in clearing the bloodstream of foreign material, toxins, and pathogens. Thus, they are indispensable to host defense and prevent the dissemination of bacteria during infections. To highlight the importance of this cell, this review will explore the history of the Kupffer cell in the context of infection beginning with its discovery to the present day.
Collapse
Affiliation(s)
- Carolyn T Graham
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wenhua 1st Road Guishan Dist., Taoyuan, Taiwan
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
3
|
Douglas EJ, Palk N, Rudolph ER, Laabei M. Anti-staphylococcal fatty acids: mode of action, bacterial resistance and implications for therapeutic application. MICROBIOLOGY (READING, ENGLAND) 2025; 171:001563. [PMID: 40402078 PMCID: PMC12098983 DOI: 10.1099/mic.0.001563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025]
Abstract
Novel strategies to counter multidrug-resistant pathogens such as methicillin-resistant Staphylococcus aureus are urgently required. The antimicrobial properties of fatty acids (FAs) have long been recognized and offer significant promise as viable alternatives to, or potentiators of, conventional antibiotics. In this review, we examine the interplay between FAs and S. aureus, specifically detailing the underlying molecular mechanisms responsible for FA-mediated inhibition and the counteracting staphylococcal systems evolved to withstand FA onslaught. Finally, we present an update on the recent therapeutic FA applications to combat S. aureus infection, either as a monotherapy or in combination with antibiotics or host-derived antimicrobial peptides. Given the frequency of interaction between FAs and S. aureus during host colonization and infection, understanding FA mode of action and deciphering S. aureus FA resistance strategies are central in rationally designing future anti-staphylococcal FAs and FA-combination therapies.
Collapse
Affiliation(s)
- Edward J.A. Douglas
- Centre for Bacterial Resistance Biology, Imperial College London, London W2 1NY, UK
| | - Nathanael Palk
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Emily R. Rudolph
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK
| | - Maisem Laabei
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
4
|
Basta DW, Campbell IW, Sullivan EJ, Hotinger JA, Hullahalli K, Garg M, Waldor MK. Inducible transposon mutagenesis identifies bacterial fitness determinants during infection in mice. Nat Microbiol 2025; 10:1171-1183. [PMID: 40148565 PMCID: PMC12055562 DOI: 10.1038/s41564-025-01975-z] [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: 06/05/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
Transposon insertion sequencing (Tn-seq) is a powerful method for genome-scale forward genetics in bacteria. However, inefficient transposon delivery or stochastic loss of mutants due to population bottlenecks can limit its effectiveness. Here we have developed 'InducTn-seq', where an arabinose-inducible Tn5 transposase enables temporal control of mini-Tn5 transposition. InducTn-seq generated up to 1.2 million transposon mutants from a single colony of enterotoxigenic Escherichia coli, Salmonella typhimurium, Shigella flexneri and Citrobacter rodentium. This mutant diversity enabled more sensitive detection of subtle fitness defects and measurement of quantitative fitness effects for essential and non-essential genes. Applying InducTn-seq to C. rodentium in a mouse model of infectious colitis bypassed a highly restrictive host bottleneck, generating a diverse population of >5 × 105 unique transposon mutants compared to 10-102 recovered by traditional Tn-seq. This in vivo screen revealed that the C. rodentium type I-E CRISPR system is required to suppress a toxin otherwise activated during gut colonization. Our findings highlight the potential of InducTn-seq for genome-scale forward genetic screens in bacteria.
Collapse
Affiliation(s)
- David W Basta
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ian W Campbell
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Emily J Sullivan
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Julia A Hotinger
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Karthik Hullahalli
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Mehek Garg
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
5
|
Zhang Z, Wang L, Yu Q, Li J, Li P, Luan S, Shi H. Bacterial Specific Recognition of Sulfonium Poly(Amino Acid) Adsorbents for Ultrafast MRSA Capture Against Bloodstream Infection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501298. [PMID: 40223366 DOI: 10.1002/smll.202501298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) bloodstream infections pose significant health risks, potentially leading to severe conditions such as bacteremia. Developing effective treatments to eliminate resistant bacteria from the bloodstream, simultaneously mitigate infection-related complications, and reduce mortality remains challenging. Herein, microspheres are synthesized with bacterial elimination and inflammation prevention by crosslinked sulfonium poly(amino acids). As-synthesized microsphere, PM1 0.6B MS, exhibits an ultrafast adsorption efficiency of 0.41 × 108 CFU mg-1 min-1 for MRSA, which positions the highest index among the reported resin and inorganic adsorptions. This bacterial-specific and efficient capture of PM1 0.6B MS is attributed to its strong interactions with teichoic acids in MRSA (Ka: 1.8 × 105 M-1) rather than acting with phospholipids of mammalian cells. Unlike the present resin-based adsorbent, for example, heparin-modified polyethylene in the only commercial Seraph® 100, PM1 0.6B MS kills adsorbed bacteria within 1 h and can be reused by simple treatment. Meanwhile, PM1 0.6B MS also shows good hemocompatibility and longer thrombin activation time to reduce the risk of thrombosis and hemolysis. In vivo experiments further confirm the abilities of PM1 0.6B MS to prevent inflammation by removing bacteria. This adsorbent is a promising candidate for early treating life-threatening bloodstream infections, potentially preventing bacteremia and subsequent organ damage.
Collapse
Affiliation(s)
- Zhenyan Zhang
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics and MIIT Key Laboratory of Flexible Electronics (KLoFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, P. R. China
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Lei Wang
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Qing Yu
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Jing Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun, Changchun, 130022, P. R. China
| | - Peng Li
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics and MIIT Key Laboratory of Flexible Electronics (KLoFE), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, P. R. China
| | - Shifang Luan
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Hengchong Shi
- State Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
6
|
Xu J, Chen J, Xia H, Gong Y, Xiong F. Integrated Approaches for Discovery of Staphylococcus aureus Antimicrobial Agents: Virtual Screening, Molecular Docking, Molecular Dynamics Simulations, and Density Functional Theory. Chem Biodivers 2025:e202403449. [PMID: 40192570 DOI: 10.1002/cbdv.202403449] [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: 12/26/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Due to the excessive use of antibiotics, Staphylococcus aureus has developed resistance to conventional antibiotics. This study primarily employs virtual screening methods to explore the binding mode, biological stability, electronic properties, and antimicrobial activity of the drugs. Lactate dehydrogenase (LDH) was chosen as the primary target, and virtual screening of approximately 3180 FDA-approved drugs was performed. On the basis of binding affinity scores, the top 12 molecules were shortlisted for further analysis through precise docking and MMGBSA calculations. Molecular docking simulations revealed that these compounds exhibit a strong affinity for the target protein 6BAZ, with Gliquidone demonstrating the highest binding affinity at -76.25 kcal/mol. The top three hit molecules were subjected to 100 ns molecular dynamics simulations, which confirmed the stability of the ligand-protein complexes through hydrophobic and hydrogen bonding interactions, corroborating the docking and MMGBSA findings. Density functional theory (B3LYP level, 6-31 + G (d, p) basis set) was applied to evaluate molecular geometry optimization and vibrational frequencies, offering valuable insights into the structure and stability of the drug molecules, which further supports their potential as lead compounds for LDH inhibition and establishes a strong basis for future drug development and optimization.
Collapse
Affiliation(s)
- Jie Xu
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, China
| | - Jiawei Chen
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, China
| | - Heping Xia
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, China
| | - Yi Gong
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, China
| | - Fei Xiong
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
7
|
Liu S, Gaisford S, Williams GR. Ciprofloxacin-Loaded Spray-Dried Lactose Particles: Formulation Optimization and Antibacterial Efficacy. Pharmaceutics 2025; 17:392. [PMID: 40143055 PMCID: PMC11945521 DOI: 10.3390/pharmaceutics17030392] [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: 02/06/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Bacterial infections in the oral cavity and outer ear require effective and targeted drug delivery systems. This study details the production of drug-loaded lactose microparticles, with the aim of creating antibiotic formulations for ultimate use in combatting oral and outer ear bacterial infections. Methods: Lactose particles were prepared via spray drying and optimized with varying ciprofloxacin (cipro) loadings to maximize the drug content. The particles were characterized to evaluate their performance in terms of physicochemical properties, drug-loading efficiency, drug-release kinetics, and antibacterial activity. Results: The resulting particles exhibited spherical morphology, efficient cipro loading (in the range of 1.1-52.9% w/w) and rapid cipro release within 5 h (achieving 70-81% release). In addition, they demonstrated effective concentration-dependent antibacterial activity against gram-positive Staphylococcus aureus and gram-negative Pseudomonas aeruginosa, with bacterial growth effectively inhibited for more than 24 h when particle concentrations reached the minimum inhibitory concentration. Conclusions: These findings highlight the potential of spray-dried cipro loaded lactose particles as an efficient approach for localized antibacterial treatment, offering a promising solution for managing bacterial infections in the oral cavity and outer ear.
Collapse
Affiliation(s)
| | | | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29–39 Brunswick Square, London WC1N 1AX, UK; (S.L.); (S.G.)
| |
Collapse
|
8
|
Zheng Q, Fu R, Du X, Wu J, Luo A, Ren Y. WITHDRAWN: Intense and vibrant color construction and functionalization for protein macromolecule/polyamide two-component fabric by sustainable microbial nano prodigiosins based on adjustable pigment allocation. Int J Biol Macromol 2025; 293:139326. [PMID: 39743115 DOI: 10.1016/j.ijbiomac.2024.139326] [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: 10/15/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
This article has been withdrawn: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/policies/article-withdrawal). This article has been withdrawn at the request of the editor and publisher. The publisher regrets that an error occurred which led to the premature publication of this paper. This error bears no reflection on the article or its authors. The publisher apologizes to the authors and the readers for this unfortunate error.
Collapse
Affiliation(s)
- Qiumeng Zheng
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao, 266071, China
| | - Ranran Fu
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao, 266071, China.
| | - Xinyu Du
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao, 266071, China
| | - Jing Wu
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao, 266071, China
| | - Anqiao Luo
- Sunvim Group Co., Ltd., Gaomi, 261500, China
| | - Yanfei Ren
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao, 266071, China; Key Laboratory of Clean Dyeing and Finishing Technology of Zhejiang Province, Shaoxing University, Shaoxing, 312000, China.
| |
Collapse
|
9
|
Kim SJ, Ali MS, Kang HS, Moon BY, Hwang YJ, Yoon SS, Park SC, Lim SK. Characterization of fusidic acid-resistant Staphylococcus aureus isolated from food animals during 2010-2021 in South Korea. Int J Food Microbiol 2025; 430:111026. [PMID: 39731987 DOI: 10.1016/j.ijfoodmicro.2024.111026] [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: 08/09/2024] [Revised: 11/15/2024] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
Livestock-associated fusidic acid-resistant Staphylococcus aureus (FRSA) is frequently linked to global public health hazards. This study aimed to ascertain the prevalence and molecular characteristics of FRSA isolated from food animal products in South Korea from 2010 to 2021. We obtained a total of 3980 S. aureus isolates from cattle carcasses (n = 482), pig carcasses (n = 1531), and chicken carcasses (n = 1967). The isolates were evaluated for antimicrobial susceptibility using the broth microdilution method. Antimicrobial resistance genes, spa types, sequence types (STs), and Staphylococcus cassette chromosome mec (SCCmec) types were determined by polymerase chain reaction (PCR) and sequencing analysis. In total, 187 isolates (4.7 %) demonstrated resistance to fusidic acid, with the maximum recovered from cattle (16.2 %), followed by pigs (6.5 %) and chickens (0.5 %). In addition, the majority of the isolates showed resistance to penicillin (86.6 %), while comparatively low resistance rates (7-13.9 %) were observed for erythromycin, gentamicin, kanamycin, and tetracycline. Moreover, multidrug resistance (MDR) comprised 8.6 % (16/187) of the isolates. Among the fusidic acid resistance determinants, the fusA mutation was the highest, containing 54 % (101/187), followed by fusC (29.4 %, 55/187) and fusB (15.5 %, 29/187). A high level of resistance regarding the substitution of L461K in the fusA gene was identified in 97 % of isolates. In addition, the most commonly detected resistance patterns include penicillin (87.1 %, 88/101) among the FRSA. The nucleotide sequencing analysis showed that all 29 fusB-carrying isolates possess the structural gene blaZ of the bla operon and the insertion sequences orf152, orf170, IS257, and orf152. In total, 21 spa types were found, where t126 was detected the most (81.2 %, 82/101) in fusA, followed by t127 (81.8 %, 45/55) in fusC, and t189 (27.6 %, 8/29) in fusB. Furthermore, all t002 harboring fusC were detected as ST5-MRSA-SCCmecII clones. This is the first report of fusA and fusB carrying S. aureus and linkage fusB and blaZ genes in FRSA isolated from food animal products. Taken together, the FRSA in food animals with different resistance determinants and spa types could pose a threat to public health.
Collapse
Affiliation(s)
- Su-Jeong Kim
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Md Sekendar Ali
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Hee-Seung Kang
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Bo-Youn Moon
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Yu-Jeong Hwang
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Soon-Seek Yoon
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, Institute for Veterinary Biomedical Science, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Suk-Kyung Lim
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea.
| |
Collapse
|
10
|
Zrihan S, Itay P, Kroin Y, Davidovich N, Wosnick N, Tchernov D, Koh XP, Lau SCK, Morick D. Monitoring Fish Bacterial Pathogens of Wild Fish Species From the South China Sea by Applying Next-Generation Sequencing on Gill Tissue. JOURNAL OF FISH DISEASES 2025; 48:e14050. [PMID: 39575841 PMCID: PMC11706320 DOI: 10.1111/jfd.14050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 01/11/2025]
Abstract
The classic epidemiological triangle model of host-environment-pathogen is recently being reshaped into a tetrahedron, with the growing understanding of the importance of the microbiome in this array. The gills, being a gateway into the fish body, bearing an important role in fish homeostasis, host a complex microbiome that reflects the ambient water, while also showing resemblance to gut microbiome. Next-generation sequencing (NGS) and improvements in data analysis tools enable researchers to gather and analyse a lot more data than ever before, take a closer, more detailed look at microbiota, and gain a much better understanding of the biological processes at work in these complex relations. Here, 16S rRNA amplicons of bacterial DNA extracted from the gills of 36 asymptomatic specimens of three wild fish species from the South China Sea (Nemipterus japonicus, Alepes djebaba, and Saurida tumbil) were sequenced using NGS. Data analyses revealed the presence of 20 potentially pathogenic species, including several zoonotic agents. Gill microbiota exhibited host species-specificity, and expressed a significant difference between demersal and pelagic-amphidromous fish. It is suggested that this method be more widely implemented, in order to gain more insight on ocean ecosystems' health status, as well as fish stocks of commercial importance.
Collapse
Affiliation(s)
- Shlomi Zrihan
- Morris Kahn Marine Research StationUniversity of HaifaSdot YamIsrael
- Department of Marine Biology, Leon H. Charney School of Marine SciencesUniversity of HaifaSdot YamIsrael
| | - Peleg Itay
- Morris Kahn Marine Research StationUniversity of HaifaSdot YamIsrael
- Department of Blue Biotechnologies and Sustainable Mariculture, The Leon H. Charney School of Marine SciencesUniversity of HaifaSdot YamIsrael
| | - Yael Kroin
- Morris Kahn Marine Research StationUniversity of HaifaSdot YamIsrael
| | - Nadav Davidovich
- Morris Kahn Marine Research StationUniversity of HaifaSdot YamIsrael
- Israeli Veterinary ServicesBet DaganIsrael
| | - Natascha Wosnick
- Programa de Pós‐Graduação Em ZoologiaUniversidade Federal Do ParanáCuritibaBrazil
| | - Dan Tchernov
- Morris Kahn Marine Research StationUniversity of HaifaSdot YamIsrael
- Department of Marine Biology, Leon H. Charney School of Marine SciencesUniversity of HaifaSdot YamIsrael
| | - Xiu Pei Koh
- Department of Ocean ScienceThe Hong Kong University of Science and TechnologyHong KongSARChina
| | - Stanley C. K. Lau
- Department of Ocean ScienceThe Hong Kong University of Science and TechnologyHong KongSARChina
| | - Danny Morick
- Morris Kahn Marine Research StationUniversity of HaifaSdot YamIsrael
- Department of Blue Biotechnologies and Sustainable Mariculture, The Leon H. Charney School of Marine SciencesUniversity of HaifaSdot YamIsrael
| |
Collapse
|
11
|
Herlina T, Rizaldi Akili AW, Nishinarizki V, Hardianto A, Latip JB. Review on antibacterial flavonoids from genus Erythrina: Structure-activity relationship and mode of action. Heliyon 2025; 11:e41395. [PMID: 39811340 PMCID: PMC11729662 DOI: 10.1016/j.heliyon.2024.e41395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
The Fabaceae family, particularly genus Erythrina, is renowned for significant medicinal properties. These plants have been used as natural remedies to address various health issues and are rich in flavonoids. Therefore, this review aimed to provide a comprehensive overview of antibacterial activity, structure-activity relationship, especially against drug-resistance Staphylococcus aureus, and mode of action for flavonoids isolated from Erythrina. Data were collected from reputable electronic scholarly resources focusing on publications from 2000 to 2022. The results showed that the evaluated flavonoids include 31 % pterocarpans, 22 % flavanones, 20 % isoflavanones, 18 % isoflavones, 4 % isoflavans, 3 % isoflav-3-enes, 1 % 3-arylcoumarins, and 1 % coumestans. Most of these compounds in Erythrina plants were extracted from the roots and stem bark. Among these group of flavonoids, pterocarpan stands out as the most active class against S. aureus. Structure-activity relationship study emphasized pivotal contribution of the prenyl functional group to enhance antibacterial activity of flavonoids. Increasing the number of prenyl groups enhanced antibacterial effectiveness while modifying the group reduced this activity. The proposed antibacterial mechanisms of these flavonoids include the suppression of nucleic acid synthesis, disruption of cytoplasmic membrane function, and modulation of energy metabolism. Among the potent antibacterial flavonoids from genus Erythrina, compound 3,9-dihyroxy-10-γ,γ-dimethylallyl-6a,11a-dehydropterocarpan was found as the most potent against Methicillin-Resistant Staphylococcus aureus (MRSA) through the inhibition of nucleic acid synthesis. Other common flavonoids such as genistein, daidzein, apigenin, and luteolin exert antibacterial activity through the inhibition of ATP synthase.
Collapse
Affiliation(s)
- Tati Herlina
- Department of Chemistry, Faculty of Mathematics and Natural Science, Padjadjaran University, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Abd Wahid Rizaldi Akili
- Department of Chemistry, Faculty of Mathematics and Natural Science, Padjadjaran University, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Vicki Nishinarizki
- Department of Chemistry, Faculty of Mathematics and Natural Science, Padjadjaran University, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Ari Hardianto
- Department of Chemistry, Faculty of Mathematics and Natural Science, Padjadjaran University, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Jalifah Binti Latip
- Department of Chemical Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), 46300, Bangi, Selangor, Malaysia
| |
Collapse
|
12
|
Chen Z, Jin Q, Zhong J, Xie Z, Chen Q, Li L, Li J, Zhao C, Wang J, Tang X, Han M, Li R, Li Z, Tong Z, Wang M, Du H, Zhang H. Staphylococcus aureus blocks host autophagy through circSyk/miR-5106/Sik3 axis to promote progression of bone infection. PLoS Pathog 2025; 21:e1012896. [PMID: 39869636 PMCID: PMC11781720 DOI: 10.1371/journal.ppat.1012896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/30/2025] [Accepted: 01/09/2025] [Indexed: 01/29/2025] Open
Abstract
With the rapid increase in the number of implant operations, the incidence of bone infections has increased. Methicillin-resistant Staphylococcus aureus (S. aureus) and other emerging fully drug-resistant strains make the management of bone infections even more challenging. Bone infections are mainly caused by S. aureus and require extensive surgical intervention and long-term antibiotic therapy. The host autophagy response is critical to the elimination of S. aureus infections. In this study, we demonstrate that a circular RNA (circRNA), circSyk, is a potential biological target for the treatment of S. aureus-induced bone infection. Most importantly, S. aureus regulates circSyk to block autophagy and promote bone destruction via the circSyk/miR-5106/Sik3 axis in a nonclassical pathway, which is involved in the S. aureus infection process through a competitive endogenous RNA network. In summary, this study proposes a novel perspective on the immune escape of S. aureus in bone infections, based on circRNA.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinqi Zhong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jijie Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfeng Wang
- Department of Clinical Laboratory, The Nuclear Industry 417 Hospital, Xi’an, China
| | - Xiaoying Tang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Clinical Laboratory, Kunshan Municipal Third People’s Hospital, Suzhou, China
| | - Mingxiao Han
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ru Li
- Department of Clinical Laboratory, The Nuclear Industry 417 Hospital, Xi’an, China
| | - Ziyuan Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zelei Tong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
13
|
Reslane I, Watson GF, Handke LD, Fey PD. Regulatory dynamics of arginine metabolism in Staphylococcus aureus. Biochem Soc Trans 2024; 52:2513-2523. [PMID: 39656074 PMCID: PMC11668279 DOI: 10.1042/bst20240710] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024]
Abstract
Staphylococcus aureus is a highly significant pathogen with several well studied and defined virulence factors. However, the metabolic pathways that are required to facilitate infection are not well described. Previous data have documented that S. aureus requires glucose catabolism during initial stages of infection. Therefore, certain nutrients whose biosynthetic pathway is under carbon catabolite repression and CcpA, including arginine, must be acquired from the host. However, even though S. aureus encodes pathways to synthesize arginine, biosynthesis of arginine is repressed even in the absence of glucose. Why is S. aureus a functional arginine auxotroph? This review discusses recently described regulatory mechanisms that are linked to repression of arginine biosynthesis using either proline or glutamate as substrates. In addition, recent studies are discussed that shed insight into the ultimate mechanisms linking arginine auxotrophy and infection persistence.
Collapse
Affiliation(s)
- Itidal Reslane
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Gabrielle F. Watson
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Luke D. Handke
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| | - Paul D. Fey
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, U.S.A
| |
Collapse
|
14
|
Zomorodi AR, Motamedifar M, Rahmanian K, Shakeri M, Hajikhani B, Heidari H, Mansury D, Jahromi AS. Investigation of integron classes 1, 2, and 3 among multi-drug resistant Staphylococcus aureus isolates in Iran: a multi-center study. BMC Infect Dis 2024; 24:1430. [PMID: 39696000 DOI: 10.1186/s12879-024-10311-5] [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: 09/01/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Rising methicillin-resistant Staphylococcus aureus (MRSA) poses a global health threat, contributing to serious infections with high mortality rates. Integrons are recognized as significant genetic elements in disseminating multidrug-resistant (MDR) strains. This study focuses on assessing the prevalence of integron classes 1, 2, and 3 in S. aureus strains from four major cities in Iran. METHODS This cross-sectional study analyzed 183 S. aureus isolates from Shiraz, Tehran, Isfahan, and Yazd in Iran. The isolates were identified using specific biochemical and molecullar tests. The Kirby-Bauer disc diffusion method and microbroth dilution method were employed to determine the susceptibility of the isolates to relevant antibiotics and vancomycin, respectively. The macrolide-lincosamide-streptogramin B (MLSB) resistance phenotype was also evaluated using the D-test. All isolates were sought for presence of the intI1, intI2, and intI3 genes. RESULTS Among 183 S. aureus isolates, high resistance rates were noted: 86.3% for erythromycin, 66.1% for ciprofloxacin, and 61.7% for clindamycin, while all isolates were susceptible to linezolid and vancomycin. Of the 183 isolates, 59.6% were identified as MRSA and 78.1% as MDR. According to the D-test results, 112/183 (61.2%), 29/183 (15.8%), 25/183 (13.7%), and 17/183 (9.7%) of S. aureus isolates showed constitutive resistance-MLSB, inducible resistance-MLSB, sensitive, and resistance to macrolide-streptogramin B (MS) phenotypes, respectively. The intI1 gene was found in 14 out of 183 S. aureus isolates (7.6%), while none were positive for the intI2 or intI3 genes. Notably, 11/14 (78.5%) and 13/14 (92.8%) intI1-positive isolates were MRSA and MDR, respectively. CONCLUSIONS The distribution of MRSA and MDR S. aureus isolates in Iran seems concerning. Although the prevalence of intI1 was not as high as in prior studies, almost all S. aureus harbored the intI1 gene were MRSA and MDR.
Collapse
Affiliation(s)
- Abolfazl Rafati Zomorodi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Motamedifar
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Masihollah Shakeri
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Heidari
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Davood Mansury
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
15
|
Botros M, de Mesy Bentley KL, Schloemann DT, Saito M, Constantine R, Ricciardi BF, Muthukrishnan G. Cutibacterium acnes invades submicron osteocyte lacuno-canalicular networks following implant-associated osteomyelitis. J Orthop Res 2024; 42:2593-2603. [PMID: 39044717 DOI: 10.1002/jor.25929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024]
Abstract
Cutibacterium acnes, part of normal skin flora, is increasingly recognized as an opportunistic pathogen capable of causing chronic prosthetic joint infections (PJI) associated with total hip and knee arthroplasty. However, there is a paucity of literature examining the pathogenesis of C. acnes during PJI. To study this, we developed an implant-associated osteomyelitis murine model in which 8-10-week-old C57BL6 mice were subjected to transtibial implantation of titanium or stainless-steel L-shaped pins contaminated with C. acnes. Postsurgery, mice were killed on Days 14 and 28 for terminal assessments of (1) bacterial load in bone, implant, and internal organs (heart, spleen, kidney, and liver), (2) bone osteolysis (micro-CT), (3) abscess formation (histology), and (4) systematic electron microscopy (EM). In vitro scanning EM (SEM) confirmed that C. acnes can form biofilms on stainless-steel and titanium implants. In mice, C. acnes could persist for 28 days in the tibia. Also, we observed C. acnes dissemination to internal organs. C. acnes chronic osteomyelitis revealed markedly reduced bone osteolysis and abscess formation compared to Staphylococcus aureus infections. Importantly, transmission EM (TEM) investigation revealed the presence of C. acnes within canaliculi, demonstrating that C. acnes can invade the osteocyte lacuno-canalicular networks (OLCN) within bone. Our preliminary pilot study, for the first time, revealed that the OLCN in bone can be a reservoir for C. acnes and potentially provides a novel mechanism of why C. acnes chronic implant-associated bone infections are difficult to treat.
Collapse
Affiliation(s)
- Mina Botros
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York, USA
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Derek T Schloemann
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York, USA
| | - Motoo Saito
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York, USA
| | - Robert Constantine
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York, USA
| | - Benjamin F Ricciardi
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York, USA
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
16
|
Zheng Q, Wang L, Hao L, Wu J, Fu R, Du L, Ren Y, Fang K. Synergetic construction of color and multifunction for sustainable lyocell fabric by Coptis chinensis and BTCA. Int J Biol Macromol 2024; 281:136595. [PMID: 39414195 DOI: 10.1016/j.ijbiomac.2024.136595] [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: 03/21/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
In the context of escalating standards of living, the demand for healthy and multifunctional textiles is increasing. As a kind of cellulose macromolecular-based material, lyocell fiber has low carbon, is environmentally friendly, and demonstrates superb performance. The utilization of some Chinese herb dyes solves the pollution problem in the color and functionality construction of lyocell fabric by synthetic dyes and finishing agents. However, problems such as low dye utilization rate, light apparent color, and weak functionality of dyed fabrics remain, thus limiting the further application of the powerful combination of lyocell fabric and Chinese herb dyes. Here, a color and multifunction construction method of lyocell fabric with Coptis chinensis and 1,2,3,4-butanetetracarboxylic acid was proposed. Under the optimal color construction condition, the color depth increased remarkably, and the dye exhaustion rate of the modified fabric enhanced by 332.3 % compared with the unmodified one. The multifunction construction imparted outstanding fuzz and pilling inhibition, fibrillation resistance, and antiwrinkle performance for lyocell fabrics. Moreover, the dyed lyocell fabric exhibited considerable UV protective activity and antibacterial property against Staphylococcus aureus. This work provided an efficient color and multifunction construction technology for lyocell fabric with high value added.
Collapse
Affiliation(s)
- Qiumeng Zheng
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China; Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Lei Wang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
| | - Longyun Hao
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China; Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China; State Key Laboratory of Biofibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Jing Wu
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China; Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Ranran Fu
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China; Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Lixin Du
- Luthai Textile Co., Ltd., Zibo 255100, China
| | - Yanfei Ren
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China; Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China; State Key Laboratory of Biofibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Kuanjun Fang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China; Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China; State Key Laboratory of Biofibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
17
|
Zheng Q, Zhang W, Wang L, Wen X, Wu J, Ren Y, Fu R. Functional dyeing of cellulose macromolecule/synthetic fibers two-component fabrics with sustainable microbial prodigiosins. Int J Biol Macromol 2024; 278:134964. [PMID: 39179072 DOI: 10.1016/j.ijbiomac.2024.134964] [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: 08/10/2023] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
As an important cellulose macromolecular-based material, cotton/polyamide and cotton/polyester fabrics are widely utilized in the textile and garment field due to their combination of the advantages of both cotton and synthetic fibers, such as excellent breathability, hygroscopicity, and abrasion performance. However, the synthetic dyes used in fabric coloration are derived from non-renewable resources, and the long-time dyeing procedure poses large pollution problems. Herein, microbial prodigiosins fermented by Serratia marcescens were employed for cotton/polyamide and cotton/polyester fabric dyeing and functionalizing. The results demonstrated that the prodigiosins suspension exhibited outstanding stability. Synthetic fibers contributed significantly to the overall color of fabrics and provided good dimensional stability and durability. In contrast, cotton fibers imparted relatively lighter color but played an essential role in enhancing the softness and comfort of fabrics. The dyed fabrics presented bright overall color light with good uniformity. Furthermore, the antibacterial rates of the dyed cotton/polyamide and cotton/polyester fabrics were 87.31 % and 89.70 %, respectively. The UPF values of the dyed cotton/polyamide and cotton/polyester fabrics were recorded as 52.3 and 93.5, respectively. This study provided a novel approach for cleaner functional dyeing of cotton/synthetic fiber two-component fabrics using prodigiosins.
Collapse
Affiliation(s)
- Qiumeng Zheng
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Wenjing Zhang
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Lei Wang
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Xue Wen
- Sunvim Group Co., Ltd., Gaomi 261500, China
| | - Jing Wu
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Yanfei Ren
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China; Sunvim Group Co., Ltd., Gaomi 261500, China.
| | - Ranran Fu
- College of Textiles & Clothing, Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
18
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-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: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
19
|
Bhattacharya S, Khanra PK, Dutta A, Gupta N, Aliakbar Tehrani Z, Severová L, Šrédl K, Dvořák M, Fernández-Cusimamani E. Computational Screening of T-Muurolol for an Alternative Antibacterial Solution against Staphylococcus aureus Infections: An In Silico Approach for Phytochemical-Based Drug Discovery. Int J Mol Sci 2024; 25:9650. [PMID: 39273596 PMCID: PMC11395065 DOI: 10.3390/ijms25179650] [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: 08/11/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Staphylococcus aureus infections present a significant threat to the global healthcare system. The increasing resistance to existing antibiotics and their limited efficacy underscores the urgent need to identify new antibacterial agents with low toxicity to effectively combat various S. aureus infections. Hence, in this study, we have screened T-muurolol for possible interactions with several S. aureus-specific bacterial proteins to establish its potential as an alternative antibacterial agent. Based on its binding affinity and interactions with amino acids, T-muurolol was identified as a potential inhibitor of S. aureus lipase, dihydrofolate reductase, penicillin-binding protein 2a, D-Ala:D-Ala ligase, and ribosome protection proteins tetracycline resistance determinant (RPP TetM), which indicates its potentiality against S. aureus and its multi-drug-resistant strains. Also, T-muurolol exhibited good antioxidant and anti-inflammatory activity by showing strong binding interactions with flavin adenine dinucleotide (FAD)-dependent nicotinamide adenine dinucleotide phosphate (NAD(P)H) oxidase, and cyclooxygenase-2. Consequently, molecular dynamics (MD) simulation and recalculating binding free energies elucidated its binding interaction stability with targeted proteins. Furthermore, quantum chemical structure analysis based on density functional theory (DFT) depicted a higher energy gap between the highest occupied molecular orbital and lowest unoccupied molecular orbital (EHOMO-LUMO) with a lower chemical potential index, and moderate electrophilicity suggests its chemical hardness and stability and less polarizability and reactivity. Additionally, pharmacological parameters based on ADMET, Lipinski's rules, and bioactivity score validated it as a promising drug candidate with high activity toward ion channel modulators, nuclear receptor ligands, and enzyme inhibitors. In conclusion, the current findings suggest T-muurolol as a promising alternative antibacterial agent that might be a potential phytochemical-based drug against S. aureus. This study also suggests further clinical research before human application.
Collapse
Affiliation(s)
- Soham Bhattacharya
- Department of Agroecology and Crop Production, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, 16500 Prague, Czech Republic
| | - Pijush Kanti Khanra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Adrish Dutta
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, 16500 Prague, Czech Republic
| | - Neha Gupta
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, 16500 Prague, Czech Republic
| | - Zahra Aliakbar Tehrani
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 25250 Vestec, Czech Republic
| | - Lucie Severová
- Department of Economic Theories, Faculty of Economics and Management, Czech University of Life Sciences Prague, Kamýcká 129, 16500 Prague, Czech Republic
| | - Karel Šrédl
- Department of Economic Theories, Faculty of Economics and Management, Czech University of Life Sciences Prague, Kamýcká 129, 16500 Prague, Czech Republic
| | - Marek Dvořák
- Department of Trade and Finance, Faculty of Economics and Management, Czech University of Life Sciences Prague, Kamýcká 129, 16500 Prague, Czech Republic
| | - Eloy Fernández-Cusimamani
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, 16500 Prague, Czech Republic
| |
Collapse
|
20
|
Yeo HH, Jao YH, Yang FW, Kuo MH, Lee MH, Shiau CW, Chiu HC, Su JC. Discovery of N,N'-diarylurea molecules with activity against multidrug-resistant Staphylococcus aureus. Arch Pharm (Weinheim) 2024; 357:e2400047. [PMID: 38687910 DOI: 10.1002/ardp.202400047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 05/02/2024]
Abstract
The emergence and global spread of methicillin-resistant Staphylococcus aureus (MRSA) pose a serious threat to public health, underscoring the urgent need for novel antibacterial interventions. Here, we screened 18 newly synthesized N,N'-diarylurea derivatives to identify compounds with activity against MRSA. Our investigations led to the discovery of a small molecule, SCB-24, which exhibited promising antimicrobial activity against MRSA USA300. Notably, SCB-24 demonstrated high activity even in the presence of 10% fetal bovine serum and showed excellent selectivity for bacterial over mammalian cells. SCB-24 also showed potent activity against various MRSA strains, including those resistant to second- and third-line antibiotics. Importantly, the efficacy of SCB-24 was inferior to that of vancomycin in MRSA-infected Galleria mellonella larvae. Overall, our findings suggest that SCB-24 has great potential as a new therapeutic for multidrug-resistant S. aureus infections.
Collapse
Affiliation(s)
- Hui-Hui Yeo
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsuan Jao
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fan-Wei Yang
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Min-Hsuan Kuo
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Hsuan Lee
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Wei Shiau
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hao-Chieh Chiu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jung-Chen Su
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
21
|
Weldegebreal F, Urgesa K, Ayele F, Bogale K, Shume T, Ahmed M, Debebe S, Tebeje F, Asmerom H, Tesfa T, Mekonnen S. Nasal carriage rate, associated factors, and antimicrobial susceptibility patterns of methicillin resistance Staphylococcus aureus among pre-clinical undergraduate students at the College of Health and Medical Sciences, Haramaya University, Ethiopia. Front Public Health 2024; 12:1354461. [PMID: 38846602 PMCID: PMC11155450 DOI: 10.3389/fpubh.2024.1354461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/15/2024] [Indexed: 06/09/2024] Open
Abstract
Background Staphylococcus aureus nasal carriage has been linked to higher rates of infection and morbidity. People with Methicillin-resistant Staphylococcus aureus can be a potential source of infection for others. University students living together in crowded conditions increase their risk of acquiring infections. The prevalence of S. aureus, particularly Methicillin-resistant Staphylococcus aureus nasal carriage, in Ethiopian university students is sparse. Objective This study aimed to determine the nasal carriage rate, associated factors, and antimicrobial susceptibility patterns of methicillin-resistant Staphylococcus aureus among pre-clinical students at the College of Health and Medical Sciences, Haramaya University, Ethiopia, from 1 July to 30 August 2022. Methods An institutional-based cross-sectional study was conducted among 270 randomly selected pre-clinical Health and Medical Sciences students. Data on associated factors were collected using pre-tested, structured questionnaires. A nasal swab was taken from each participant and sent to the microbiology laboratory via Amies transport media in a cold chain. There, it was cultivated using conventional techniques. The isolated colonies were found to be S. aureus, and its antimicrobial susceptibility was performed using the Kirby-Bauer disk diffusion method on Muller-Hinton agar. Methicillin-resistant Staphylococcus aureus expressing using cefoxitin based on CLSI breakpoint. Data were entered into Epi-Data version 4.4.2.1 and exported to the Statistical Package for Social Sciences (SPSS) software version 25 for analysis. Pearson's chi-square test was performed to predict the associations between variables. A p-value less than 0.05 was regarded as statistically significant. Result Methicillin-resistant Staphylococcus aureus nasal carriage was 5.9% (95% CI: 3.09-8.7) of cases of S. aureus nasal colonization, which was found to be 12.96% (95% CI: 8.85-16.96). Methicillin-resistant Staphylococcus aureus nasal colonization was significantly associated with the history of cigarette smoking (p = 0.000), intake of khat (p = 0.042), nose-picking habit (p = 0.003), history of sharing personal goods (p = 0.021), and history of hospitalizations (p = 0.00). All of the Methicillin-resistant Staphylococcus aureus isolates were resistant to ampicillin and cefoxitin. Conclusion Based on the findings, a considerable proportion of healthy students harbored Methicillin-resistant Staphylococcus aureus strains associated with behavioral factors. Furthermore, these isolates showed high resistance to cefoxitin and ampicillin. Hence, it is crucial to regularly test pre-clinical students to prevent endogenous infections and the spread of Methicillin-resistant Staphylococcus aureus.
Collapse
Affiliation(s)
- Fitsum Weldegebreal
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
- Laboratory Bacteriology Research, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan, Belgium
| | - Kedir Urgesa
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Firayad Ayele
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Kasahun Bogale
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Taddese Shume
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mohammed Ahmed
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Sileshi Debebe
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Fikru Tebeje
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Haftu Asmerom
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Tewodros Tesfa
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Shambel Mekonnen
- School of Medical Laboratory Sciences, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| |
Collapse
|
22
|
Basta DW, Campbell IW, Sullivan EJ, Hotinger JA, Hullahalli K, Waldor MK. Inducible transposon mutagenesis for genome-scale forward genetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595064. [PMID: 38826325 PMCID: PMC11142078 DOI: 10.1101/2024.05.21.595064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Transposon insertion sequencing (Tn-seq) is a powerful method for genome-scale functional genetics in bacteria. However, its effectiveness is often limited by a lack of mutant diversity, caused by either inefficient transposon delivery or stochastic loss of mutants due to population bottlenecks. Here, we introduce "InducTn-seq", which leverages inducible mutagenesis for temporal control of transposition. InducTn-seq generates millions of transposon mutants from a single colony, enabling the sensitive detection of subtle fitness defects and transforming binary classifications of gene essentiality into a quantitative fitness measurement across both essential and non-essential genes. Using a mouse model of infectious colitis, we show that InducTn-seq bypasses a highly restrictive host bottleneck to generate a diverse transposon mutant population from the few cells that initiate infection, revealing the role of oxygen-related metabolic plasticity in pathogenesis. Overall, InducTn-seq overcomes the limitations of traditional Tn-seq, unlocking new possibilities for genome-scale forward genetic screens in bacteria.
Collapse
Affiliation(s)
- David W. Basta
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ian W. Campbell
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Emily J. Sullivan
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Julia A Hotinger
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Karthik Hullahalli
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
23
|
Moss CE, Johnston SA, Kimble JV, Clements M, Codd V, Hamby S, Goodall AH, Deshmukh S, Sudbery I, Coca D, Wilson HL, Kiss-Toth E. Aging-related defects in macrophage function are driven by MYC and USF1 transcriptional programs. Cell Rep 2024; 43:114073. [PMID: 38578825 DOI: 10.1016/j.celrep.2024.114073] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/15/2024] [Accepted: 03/21/2024] [Indexed: 04/07/2024] Open
Abstract
Macrophages are central innate immune cells whose function declines with age. The molecular mechanisms underlying age-related changes remain poorly understood, particularly in human macrophages. We report a substantial reduction in phagocytosis, migration, and chemotaxis in human monocyte-derived macrophages (MDMs) from older (>50 years old) compared with younger (18-30 years old) donors, alongside downregulation of transcription factors MYC and USF1. In MDMs from young donors, knockdown of MYC or USF1 decreases phagocytosis and chemotaxis and alters the expression of associated genes, alongside adhesion and extracellular matrix remodeling. A concordant dysregulation of MYC and USF1 target genes is also seen in MDMs from older donors. Furthermore, older age and loss of either MYC or USF1 in MDMs leads to an increased cell size, altered morphology, and reduced actin content. Together, these results define MYC and USF1 as key drivers of MDM age-related functional decline and identify downstream targets to improve macrophage function in aging.
Collapse
Affiliation(s)
- Charlotte E Moss
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Simon A Johnston
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Joshua V Kimble
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Martha Clements
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Veryan Codd
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Stephen Hamby
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alison H Goodall
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Sumeet Deshmukh
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Ian Sudbery
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Daniel Coca
- Healthy Lifespan Institute, University of Sheffield, Sheffield, UK; Department of Autonomic Control and Systems Engineering, University of Sheffield, Sheffield, UK
| | - Heather L Wilson
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK.
| | - Endre Kiss-Toth
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK; Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
24
|
Maan M, Goyal H, Joshi S, Barman P, Sharma S, Kumar R, Saini A. DP1, a multifaceted synthetic peptide: Mechanism of action, activity and clinical potential. Life Sci 2024; 340:122458. [PMID: 38266815 DOI: 10.1016/j.lfs.2024.122458] [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: 07/28/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
AIMS Microbial infections remain a leading cause of mortality worldwide, with Staphylococcus aureus (S. aureus) being a prominent etiological agent, responsible for causing persistent bacterial infections in humans. It is a nosocomial, opportunistic pathogen, capable to propagate within the bloodstream and withstand therapeutic interventions. In the current study, a novel, indigenously designed synthetic antimicrobial peptide (sAMP) has been evaluated for its antimicrobial potential to inhibit the growth and proliferation of S. aureus. MAIN METHODS The sAMP, designed peptide (DP1) was evaluated for its minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) against a panel of pathogenic bacterial strains. Membrane mechanistic studies were performed by measuring membrane conductivity via dielectric spectroscopy and visualizing changes in bacterial membrane structure through field emission scanning electron microscopy (FE-SEM). Further, DP1 was tested for its in vivo antimicrobial potential in an S. aureus-induced systemic infection model. KEY FINDINGS The results indicated that DP1 has the potential to inhibit the growth and proliferation of a broad spectrum of Gram-positive, Gram-negative and multidrug-resistant (MDR) bacterial strains. Strong bactericidal effect attributed to change in electrical conductivity of the bacterial cells leading to membrane disruption was observed through dielectric spectroscopy and FE-SEM micrographs. Further, in the in vivo murine systemic infection study, 50 % reduction in S. aureus bioburden was observed within 1 day of the administration of DP1. SIGNIFICANCE The results indicate that DP1 is a multifaceted peptide with potent bactericidal, antioxidant and therapeutic properties. It holds significance as a novel drug candidate to effectively combat S. aureus-mediated systemic infections.
Collapse
Affiliation(s)
- Mayank Maan
- Department of Biophysics, Panjab University, Chandigarh, U.T. 160014, India
| | - Hemant Goyal
- Department of Biophysics, Panjab University, Chandigarh, U.T. 160014, India
| | - Shubhi Joshi
- Department of Biophysics, Panjab University, Chandigarh, U.T. 160014, India
| | - Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Chandigarh, U.T. 160014, India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Chandigarh, U.T. 160014, India
| | - Rajesh Kumar
- Department of Physics, Panjab University, Chandigarh, U.T. 160014, India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Chandigarh, U.T. 160014, India.
| |
Collapse
|
25
|
Yamazaki Y, Ito T, Tamai M, Nakagawa S, Nakamura Y. The role of Staphylococcus aureus quorum sensing in cutaneous and systemic infections. Inflamm Regen 2024; 44:9. [PMID: 38429810 PMCID: PMC10905890 DOI: 10.1186/s41232-024-00323-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of human bacterial infections worldwide. It is the most common causative agent of skin and soft tissue infections, and can also cause various other infections, including pneumonia, osteomyelitis, as well as life-threatening infections, such as sepsis and infective endocarditis. The pathogen can also asymptomatically colonize human skin, nasal cavity, and the intestine. S. aureus colonizes approximately 20-30% of human nostrils, being an opportunistic pathogen for subsequent infection. Its strong ability to silently spread via human contact makes it difficult to eradicate S. aureus. A major concern with S. aureus is its capacity to develop antibiotic resistance and adapt to diverse environmental conditions. The variability in the accessory gene regulator (Agr) region of the genome contributes to a spectrum of phenotypes within the bacterial population, enhancing the likelihood of survival in different environments. Agr functions as a central quorum sensing (QS) system in S. aureus, allowing bacteria to adjust gene expression in response to population density. Depending on Agr expression, S. aureus secretes various toxins, contributing to virulence in infectious diseases. Paradoxically, expressing Agr may be disadvantageous in certain situations, such as in hospitals, causing S. aureus to generate Agr mutants responsible for infections in healthcare settings. MAIN BODY This review aims to demonstrate the molecular mechanisms governing the diverse phenotypes of S. aureus, ranging from a harmless colonizer to an organism capable of infecting various human organs. Emphasis will be placed on QS and its role in orchestrating S. aureus behavior across different contexts. SHORT CONCLUSION The pathophysiology of S. aureus infection is substantially influenced by phenotypic changes resulting from factors beyond Agr. Future studies are expected to give the comprehensive understanding of S. aureus overall profile in various settings.
Collapse
Affiliation(s)
- Yuriko Yamazaki
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka, University, Osaka, 565-0871, Japan
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Tomoka Ito
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Masakazu Tamai
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Seitaro Nakagawa
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Yuumi Nakamura
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka, University, Osaka, 565-0871, Japan.
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan.
| |
Collapse
|
26
|
Vozza EG, Daly CM, O'Rourke SA, Fitzgerald HK, Dunne A, McLoughlin RM. Staphylococcus aureus suppresses the pentose phosphate pathway in human neutrophils via the adenosine receptor A2aR to enhance intracellular survival. mBio 2024; 15:e0257123. [PMID: 38108639 PMCID: PMC10790693 DOI: 10.1128/mbio.02571-23] [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: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
IMPORTANCE Staphylococcus aureus is one of the leading causes of antimicrobial-resistant infections whose success as a pathogen is facilitated by its massive array of immune evasion tactics, including intracellular survival within critical immune cells such as neutrophils, the immune system's first line of defense. In this study, we describe a novel pathway by which intracellular S. aureus can suppress the antimicrobial capabilities of human neutrophils by using the anti-inflammatory adenosine receptor, adora2a (A2aR). We show that signaling through A2aR suppresses the pentose phosphate pathway, a metabolic pathway used to fuel the antimicrobial NADPH oxidase complex that generates reactive oxygen species (ROS). As such, neutrophils show enhanced ROS production and reduced intracellular S. aureus when treated with an A2aR inhibitor. Taken together, we identify A2aR as a potential therapeutic target for combatting intracellular S. aureus infection.
Collapse
Affiliation(s)
- Emilio G. Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Clíodhna M. Daly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Sinead A. O'Rourke
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Hannah K. Fitzgerald
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Aisling Dunne
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Rachel M. McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Goormaghtigh F, Van Bambeke F. Understanding Staphylococcus aureus internalisation and induction of antimicrobial tolerance. Expert Rev Anti Infect Ther 2024; 22:87-101. [PMID: 38180805 DOI: 10.1080/14787210.2024.2303018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
INTRODUCTION Staphylococcus aureus, a human commensal, is also one of the most common and serious pathogens for humans. In recent years, its capacity to survive and replicate in phagocytic and non-phagocytic cells has been largely demonstrated. In these intracellular niches, bacteria are shielded from the immune response and antibiotics, turning host cells into long-term infectious reservoirs. Moreover, neutrophils carry intracellular bacteria in the bloodstream, leading to systemic spreading of the disease. Despite the serious threat posed by intracellular S. aureus to human health, the molecular mechanisms behind its intracellular survival and subsequent antibiotic treatment failure remain elusive. AREA COVERED We give an overview of the killing mechanisms of phagocytes and of the impressive arsenal of virulence factors, toxins and stress responses deployed by S. aureus as a response. We then discuss the different barriers to antibiotic activity in this intracellular niche and finally describe innovative strategies to target intracellular persisting reservoirs. EXPERT OPINION Intracellular niches represent a challenge in terms of diagnostic and treatment. Further research using ad-hoc in-vivo models and single cell approaches are needed to better understand the molecular mechanisms underlying intracellular survival and tolerance to antibiotics in order to identify strategies to eliminate these persistent bacteria.
Collapse
Affiliation(s)
- Frédéric Goormaghtigh
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
28
|
El-Emam MMA, El-Demerdash AS, Abdo SA, Abd-Elfatah EB, El-Sayed MM, Qelliny MR, Eldin ZE, Shehata AA. The ameliorative role of Aloe vera-loaded chitosan nanoparticles on Staphylococcus aureus induced acute lung injury: Targeting TLR/NF-κB signaling pathways. Open Vet J 2024; 14:416-427. [PMID: 38633182 PMCID: PMC11018431 DOI: 10.5455/ovj.2024.v14.i1.38] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/15/2023] [Indexed: 04/19/2024] Open
Abstract
Background Acute lung injury (ALI) is a severe condition distinguished by inflammation and impaired gas exchange in the lungs. Staphylococcus aureus, a common bacterium, can cause ALI through its virulence factors. Aloe vera is a medicinal plant that has been traditionally used to treat a variety of illnesses due to its anti-inflammatory properties. Chitosan nanoparticles are biocompatible and totally biodegradable materials that have shown potential in drug delivery systems. Aim To explore the antibacterial activity of Aloe vera-loaded chitosan nanoparticles (AV-CS-NPs) against S. aureus in vitro and in vivo with advanced techniques. Methods The antibacterial efficacy of AV-CS-NPs was evaluated through a broth microdilution assay. In addition, the impact of AV-CS-NPs on S. aureus-induced ALI in rats was examined by analyzing the expression of genes linked to inflammation, oxidative stress, and apoptosis. Furthermore, rat lung tissue was scanned histologically. The rats were divided into three groups: control, ALI, and treatment with AV-CS-NPs. Results The AV-CS-NPs that were prepared exhibited clustered semispherical and spherical forms, having an average particle size of approximately 60 nm. These nanoparticles displayed a diverse structure with an uneven distribution of particle sizes. The maximum entrapment efficiency of 95.5% ± 1.25% was achieved. The obtained findings revealed that The minimum inhibitory concentration and minimum bactericidal concentration values were determined to be 5 and 10 ug/ml, respectively, indicating the potent bactericidal effect of the NPs. Also, S. aureus infected rats explored upregulation in the mRNA expression of TLR2 and TLR4 compared to healthy control groups. AV-CS-NP treatment reverses the case where there was repression in mRNA expression of TLR2 and TLR4 compared to S. aureus-treated rats. Conclusion These NPs can serve as potential candidates for the development of alternative antimicrobial agents.
Collapse
Affiliation(s)
- Mahran M. Abd El-Emam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Azza S. El-Demerdash
- Laboratory of Biotechnology, Agriculture Research Center (ARC), Department of Microbiology, Animal Health Research Institute (AHRI), Zagazig, Egypt
| | - Samar A. Abdo
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Eman B. Abd-Elfatah
- Department of Animal Medicine, Infectious Diseases, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Marwa M. El-Sayed
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Milad R. Qelliny
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Zienab E. Eldin
- Department of Materials Science and Nanotechnology, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | - Ayman A. Shehata
- Department of Animal Medicine, Infectious Diseases, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
29
|
Li X, Hao Y, Yang N, Mao R, Teng D, Wang J. Plectasin: from evolution to truncation, expression, and better druggability. Front Microbiol 2023; 14:1304825. [PMID: 38188573 PMCID: PMC10771296 DOI: 10.3389/fmicb.2023.1304825] [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: 11/01/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Non-computational classical evolution analysis of plectasin and its functional relatives can especially contribute tool value during access to meet requirements for their better druggability in clinical use. Staphylococcus aureus is a zoonotic pathogen that can infect the skin, blood, and other tissues of humans and animals. The impact of pathogens on humans is exacerbated by the crisis of drug resistance caused by the misuse of antibiotics. In this study, we analyzed the evolution of anti-Staphylococcus target functional sequences, designed a series of plectasin derivatives by truncation, and recombinantly expressed them in Pichia pastoris X-33, from which the best recombinant Ple-AB was selected for the druggability study. The amount of total protein reached 2.9 g/L following 120 h of high-density expression in a 5-L fermenter. Ple-AB was found to have good bactericidal activity against gram-positive bacteria, with minimum inhibitory concentration (MIC) values ranging between 2 and 16 μg/mL. It showed good stability and maintained its bactericidal activity during high temperatures, strong acid and alkali environments. Notably, Ple-AB exhibited better druggability, including excellent trypsin resistance, and still possessed approximately 50% of its initial activity following exposure to simulated intestinal fluids for 1 h. In vitro safety testing of Ple-AB revealed low hemolytic activity against mouse erythrocytes and cytotoxicity against murine-derived macrophages. This study successfully realized the high expression of a new antimicrobial peptide (AMP), Ple-AB, in P. pastoris and the establishment of its oral administration as an additive form with high trypsin resistance; the study also revealed its antibacterial properties, indicating that truncation design is a valuable tool for improving druggability and that the candidate Ple-AB may be a novel promising antimicrobial agent.
Collapse
Affiliation(s)
- Xuan Li
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
30
|
Zhu Q, Lin Q, Jiang Y, Chen S, Tian J, Yang S, Li Y, Li M, Wang Y, Shen C, Meng S, Yang L, Feng Y, Qu J. Construction and application of the conditionally essential gene knockdown library in Klebsiella pneumoniae to screen potential antimicrobial targets and virulence genes via Mobile-CRISPRi-seq. Appl Environ Microbiol 2023; 89:e0095623. [PMID: 37815340 PMCID: PMC10617577 DOI: 10.1128/aem.00956-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/09/2023] [Indexed: 10/11/2023] Open
Abstract
Klebsiella pneumoniae is a ubiquitous human pathogen, and its clinical treatment faces two major challenges: multidrug resistance and the pathogenesis of hypervirulent K. pneumoniae. The discovery and study of conditionally essential (CE) genes that can function as potential antimicrobial targets has always been a research concern due to their restriction in the development of novel antibiotics. However, the lack of essential functional genomic data has hampered the study of the mechanisms of essential genes related to antimicrobial susceptibility. In this study, we developed a pooled CE genes mobile clustered regularly interspaced short palindromic repeat (CRISPR) interference screening method (Mobile-CRISPRi-seq) for K. pneumoniae to identify genes that play critical roles in antimicrobial fitness in vitro and host immunity in vivo. Targeting 870 predicted CE genes in K. pneumoniae, Mobile-CRISPRi-seq uncovered the depletion of tetrahydrofolate synthesis pathway genes folB and folP under trimethoprim pressure. Our screening also identified genes waaE and fldA related to polymyxin and β-lactam susceptibility by applying a screening strategy based on Mobile-CRISPRi-seq and comparative genomics. Furthermore, using a mouse infection model and Mobile-CRISPRi-seq, multiple virulence genes were identified, and among these genes, pal, yciS, and ribB were demonstrated to contribute to the pathogenesis of K. pneumoniae. This study provides a simple, rapid, and effective platform for screening potential antimicrobial targets and virulence genes in K. pneumoniae, and this broadly applicable system can be expanded for high-throughput functional gene study in multiple pathogenic bacteria, especially in gram-negative bacteria. IMPORTANCE The discovery and investigation of conditionally essential (CE) genes that can function as potential antimicrobial targets has always been a research concern because of the restriction of antimicrobial targets in the development of novel antibiotics. In this study, we developed a pooled CE gene-wide mobile clustered regularly interspaced short palindromic repeat (CRISPR) interference sequencing (Mobile-CRISPRi-seq) strategy in Klebsiella pneumoniae to identify genes that play critical roles in the fitness of antimicrobials in vitro and host immunity in vivo. The data suggest a robust tool to screen for loss-of-function phenotypes in a pooled gene knockdown library in K. pneumoniae, and Mobile-CRISPRi-seq may be expanded to multiple bacteria for screening and identification of genes with crucial roles in the fitness of antimicrobials and hosts.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Clinical Laboratory, Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Qiang Lin
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong Province, China
| | - Yushan Jiang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shuyan Chen
- Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Junxuan Tian
- Department of Clinical Laboratory, Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Shijin Yang
- Department of Clinical Laboratory, Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Yuanchun Li
- Department of Clinical Laboratory, Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Mengjun Li
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuelin Wang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chenguang Shen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Songdong Meng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liang Yang
- Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Youjun Feng
- Department of Clinical Laboratory, Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Departments of Microbiology and General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jiuxin Qu
- Department of Clinical Laboratory, Shenzhen Third People’s Hospital, National Clinical Research Center for Infectious Diseases, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| |
Collapse
|
31
|
Zaman A, Diago Navarro E, Fries BC, Kim HK, Carpino N. Inactivation of the Sts enzymes promotes resistance to lethal Staphylococcus aureus infection. Infect Immun 2023; 91:e0026023. [PMID: 37725063 PMCID: PMC10580875 DOI: 10.1128/iai.00260-23] [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: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 09/21/2023] Open
Abstract
Staphylococcus aureus is a highly infective Gram-positive bacterial pathogen that causes a wide range of diseases in both healthy and immunocompromised individuals. It can evade host immune defenses by expressing numerous virulence factors and toxins. Coupled with the inability of the human host to develop protective immunity against S. aureus, the emergence of antibiotic-resistant strains complicates treatment options. The non-canonical Sts phosphatases negatively regulate signaling pathways in varied immune cell types. To determine the role of the Sts proteins in regulating host responses to a Gram-positive microorganism, we investigated the response of mice lacking Sts expression to S. aureus infection. Herein, we demonstrate that Sts -/- animals are significantly resistant to lethal intravenous doses of S. aureus strain USA300. Resistance is characterized by significantly enhanced survival and accelerated bacterial clearance in multiple peripheral organs. Infected Sts -/- animals do not display increased levels of cytokines TNFα, IFNγ, and IL-6 in the spleen, liver, and kidney during the early stages of the infection, suggesting that a heightened pro-inflammatory response does not underlie the resistance phenotype. In vivo ablation of mononuclear phagocytes compromises the Sts -/- enhanced CFU clearance phenotype. Additionally, Sts -/- bone marrow-derived macrophages demonstrate significantly enhanced restriction of intracellular S. aureus following ex vivo infection. These results reveal the Sts enzymes to be critical regulators of host immunity to a virulent Gram-positive pathogen and identify them as therapeutic targets for optimizing host anti-microbial responses.
Collapse
Affiliation(s)
- Anika Zaman
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York, USA
| | - Elizabeth Diago Navarro
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Bettina C. Fries
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York, USA
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Veterans Administration Medical Center, Northport, New York, USA
| | - Hwan Keun Kim
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Nick Carpino
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
32
|
Suthi S, Mounika A, Potukuchi VGKS. Elevated acetate kinase (ackA) gene expression, activity, and biofilm formation observed in methicillin-resistant strains of Staphylococcus aureus (MRSA). J Genet Eng Biotechnol 2023; 21:100. [PMID: 37831271 PMCID: PMC10575836 DOI: 10.1186/s43141-023-00555-0] [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: 10/21/2022] [Accepted: 09/20/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Staphylococcus aureus spreads its infections through biofilms. This usually happens in the stationary phase of S. aureus growth where it utilizes accumulated acetate as a carbon source via the phosphotrans-acetylase-acetate kinase (Pta-Ack) pathway. In which acetate kinase (ackA) catalyzes the substrate-level phosphorylation, a vital secondary energy-yielding pathway that promotes biofilms formation aids bacterium survival in hostile environments. In this study, we describe the cloning, sequencing, and expression of S. aureus ackA gene. The expression analysis of ackA gene in methicillin-resistant strains of S. aureus (MRSA) correlates with ackA activity and biofilm units. The uniqueness of ackA was analyzed by using in silico methods. RESULTS Elevated ackA gene expression was observed in MRSA strains, which correlates with increased ackA activity and biofilm units, explaining ackA role in MRSA growth and pathogenicity. The pure recombinant acetate kinase showed a molecular weight of 44 kDa, with enzyme activity of 3.35 ± 0.05 μM/ml/min. The presence of ACKA-1, ACKA-2 sites, one ATP, and five serine/threonine-protein kinase sites in the ackA gene (KC954623.1) indicated that acetyl phosphate production is strongly controlled. The comparative structural analysis of S. aureus ackA with ackA structures of Mycobacterium avium (3P4I) and Salmonella typhimurium (3SLC) exhibited variations as indicated by the RMSD values 1.877 Å and 2.141 Å respectively, explaining why ackA functions are differently placed in bacteria, concurring its involvement in S. aureus pathogenesis. CONCLUSIONS Overall findings of this study highlight the correlation of ackA expression profoundly increases survival capacity through biofilm formation, which is a pathogenic factor in MRSA and plays a pivotal role in infection spreading.
Collapse
Affiliation(s)
- Subbarayudu Suthi
- Microbial Genetics Laboratory, Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences, Alipiri Road, Tirupati, 517501, Andhra Pradesh, India
| | - A Mounika
- Microbial Genetics Laboratory, Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences, Alipiri Road, Tirupati, 517501, Andhra Pradesh, India
| | | |
Collapse
|
33
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
34
|
Hullahalli K, Dailey KG, Waldor MK. Innate immune responses yield tissue-specific bottlenecks that scale with pathogen dose. Proc Natl Acad Sci U S A 2023; 120:e2309151120. [PMID: 37669395 PMCID: PMC10500177 DOI: 10.1073/pnas.2309151120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
To cause infection, pathogens must overcome bottlenecks imposed by the host immune system. These bottlenecks restrict the inoculum and largely determine whether pathogen exposure results in disease. Infection bottlenecks therefore quantify the effectiveness of immune barriers. Here, using a model of Escherichia coli systemic infection, we identify bottlenecks that tighten or widen with higher inoculum sizes, revealing that the efficacy of innate immune responses can increase or decrease with pathogen dose. We term this concept "dose scaling". During E. coli systemic infection, dose scaling is tissue specific, dependent on the lipopolysaccharide (LPS) receptor TLR4, and can be recapitulated by mimicking high doses with killed bacteria. Scaling therefore depends on sensing of pathogen molecules rather than interactions between the host and live bacteria. We propose that dose scaling quantitatively links innate immunity with infection bottlenecks and is a valuable framework for understanding how the inoculum size governs the outcome of pathogen exposure.
Collapse
Affiliation(s)
- Karthik Hullahalli
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Disease, Brigham & Women’s Hospital, Boston, MA02115
| | - Katherine G. Dailey
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Disease, Brigham & Women’s Hospital, Boston, MA02115
| | - Matthew K. Waldor
- Department of Microbiology, Harvard Medical School, Boston, MA02115
- Division of Infectious Disease, Brigham & Women’s Hospital, Boston, MA02115
- HHMI, Boston, MA02115
| |
Collapse
|
35
|
Lao J, Song XP, Wang HS, Jiang RS, Luo Y, Sun J, Li ZH, Li C, Deng JK, Wang B, Ma XP, Wang JY. Severe congenital neutropenia and liver abscess: Surgical treatment breaks the vicious cycle. Heliyon 2023; 9:e19880. [PMID: 37810153 PMCID: PMC10559282 DOI: 10.1016/j.heliyon.2023.e19880] [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: 11/06/2022] [Revised: 08/28/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Here, we present a case with genetically confirmed SCN. The main symptom of the child was recurring fever. The combination of antibiotics combined with G-CSF injection was proved to be insufficient, and the patient developed "solid" liver abscess. After undergoing surgical anatomical hepatic lobectomy, the child's infection symptoms showed improvement. The postoperative culture of the purulent material from the liver infection lesion revealed an infection with Staphylococcus aureus. Our case raises the possibility of pathogen sources and routes of infection, clinical characteristics, and effective treatment for SCN patients with concomitant liver abscess.
Collapse
Affiliation(s)
- Jing Lao
- Shenzhen Children's Hospital of China Medical University, Shenzhen 518026, Guangdong Province, China
| | - Xin-Ping Song
- Shenzhen Children's Hospital of China Medical University, Shenzhen 518026, Guangdong Province, China
| | - Huan-Sheng Wang
- Shenzhen Children's Hospital of China Medical University, Shenzhen 518026, Guangdong Province, China
| | - Ren-Sen Jiang
- Shenzhen Children's Hospital of Shantou University, Shenzhen 518026, Guangdong Province, China
| | - Yu Luo
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| | - Jun Sun
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| | - Zhi-Han Li
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| | - Chi Li
- Department of Infection, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| | - Ji-Kui Deng
- Department of Infection, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| | - Bin Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| | - Xiao-Peng Ma
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| | - Jian-Yao Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China
| |
Collapse
|
36
|
Torres NJ, Rizzo DN, Reinberg MA, Jobson ME, Totzke BC, Jackson JK, Yu W, Shaw LN. The identification of two M20B family peptidases required for full virulence in Staphylococcus aureus. Front Cell Infect Microbiol 2023; 13:1176769. [PMID: 37538308 PMCID: PMC10394242 DOI: 10.3389/fcimb.2023.1176769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/13/2023] [Indexed: 08/05/2023] Open
Abstract
We have previously demonstrated that deletion of an intracellular leucine aminopeptidase results in attenuated virulence of S. aureus. Herein we explore the role of 10 other aminopeptidases in S. aureus pathogenesis. Using a human blood survival assay we identified mutations in two enzymes from the M20B family (PepT1 and PepT2) as having markedly decreased survival compared to the parent. We further reveal that pepT1, pepT2 and pepT1/2 mutant strains are impaired in their ability to resist phagocytosis by, and engender survival within, human macrophages. Using a co-infection model of murine sepsis, we demonstrate impairment of dissemination and survival for both single mutants that is even more pronounced in the double mutant. We show that these enzymes are localized to the cytosol and membrane but are not necessary for peptide-based nutrition, a hallmark of cell-associated aminopeptidases. Furthermore, none of the survival defects appear to be the result of altered virulence factor production. An exploration of their regulation reveals that both are controlled by known regulators of the S. aureus virulence process, including Agr, Rot and/or SarA, and that this cascade may be mediated by FarR. Structural modeling of PepT1 reveals it bears all the hallmarks of a tripeptidase, whilst PepT2 differs significantly in its catalytic pocket, suggesting a broader substrate preference. In sum, we have identified two M20B aminopeptidases that are integral to S. aureus pathogenesis. The future identification of protein and/or peptide targets for these proteases will be critical to understanding their important virulence impacting functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lindsey N. Shaw
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, United States
| |
Collapse
|
37
|
Seravalli J, Portugal F. Putrescine Detected in Strains of Staphylococcus aureus. Pathogens 2023; 12:881. [PMID: 37513728 PMCID: PMC10386481 DOI: 10.3390/pathogens12070881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/13/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Most forms of life, including the archaea, bacteria, and eukaryotes synthesize the polyamine putrescine. Although putrescine is widely distributed, several Gram-positive bacteria, including Staphylococcus aureus (S. aureus), appear to be the exceptions. We report here that strains of S. aureus can produce the polyamine putrescine, as well as the derivative N-acetyl-putrescine. Three strains of S. aureus from the American Type Culture Collection (ATCC), one strain listed in the National Center for Biotechnology Information (NCBI) database, whose genomic sequence is well defined, and well as eight strains from S. aureus-induced brain abscesses of individual patients from multiple geographic locations were evaluated. Each strain was grown in complete chemically defined medium (CDM) under stringent conditions, after which the partially purified conditioned medium (CM) was analyzed by mass spectroscopy (MS), and the data were reported as the ratio of experimental results to controls. We confirmed the synthesis of putrescine by S. aureus by using 13C/15N-labeled arginine as a tracer. We found that agmatine, N-acetyl-putrescine, ornithine, citrulline, proline, and NH3 were all labeled with heavy isotope derived from 13C/15N-labeled arginine. None of the strains examined produced spermine or spermidine, but strains from either ATCC or human brain abscesses produced putrescine and/or its derivative N-acetyl-putrescine.
Collapse
Affiliation(s)
- Javier Seravalli
- Redox Biology Center and Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Frank Portugal
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
38
|
Alsolami A, ALGhasab NS, Alharbi MSM, Bashir AI, Saleem M, Syed Khaja AS, Aldakheel DF, Rakha E, Alshammari JA, Taha TE, Melibari Z, Alharbi YH, Almutlag AA, Said KB. Community-Acquired Methicillin-Resistant Staphylococcus aureus in Hospitals: Age-Specificity and Potential Zoonotic-Zooanthroponotic Transmission Dynamics. Diagnostics (Basel) 2023; 13:2089. [PMID: 37370983 PMCID: PMC10297644 DOI: 10.3390/diagnostics13122089] [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: 05/08/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) lineages are a devastating clinical and public health issue. Data on local lineage profiles are limited. We report on the frequency of community-acquired and hospital-acquired cases (CA-MRSA, HA-MRSA). We studied 147 isolates from King Khalid tertiary care hospitals (KKH), each from a case in a patient and including 33 patients at the Maternity and Children's Hospital (MCH). Of the 147 isolates, 87 males (59%) and 60 females (41%) were in KKH. The overwhelming majority (80%; n = 119/147) were CA-MRSA in KKH. Intriguingly, despite significant differences between males (70%) and females (53%), lineage-acquisition remained age-specific around 58-60 years in both genders. However, while CA-MRSA dominated early in life (0-20, 70% MCH), it increased with age in KKH adults; 21-50 (28%), >50 (59%) until the overall 80% (n = 144/180). Major specimens included skin-wounds, surgeries (70.3%), blood (13.5%), sputum (8.8%), very rarely urine (4.1%), and nasal (3.4%), albeit most patients showed severe enteritis and necrotizing pneumonia. Antibiograms showed high beta lactam resistances, including amoxicillin-clavulanate (83%), oxacillin (84%), cefoxitin FOX (100%), penicillin and ampicillin (~100%), as well as high resistance (82%) to carbapenem. Fortunately, high susceptibility was seen to non-beta lactams and, to a lesser extent, gentamicin, erythromycin, and fusidic acid; 33%, 34%, and 38%, respectively, in KKH. A similar pattern was seen in MCH except for a low resistance pattern to gentamicin CN, clindamycin CD, erythromycin E, and tobramycin TOB; 34%, 31%, 39%, and 41%, respectively, except for fusidic acid. These findings have significant clinical implications for MRSA patient management strategies. Clinical- and lineage-profiles imply host-selection and zoonotic-zooanthroponotic transmission dynamics. Future molecular typing, sequencing, and characterization of dominant clone(s) is imperative.
Collapse
Affiliation(s)
- Ahmed Alsolami
- Department of Internal Medicine, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (A.A.); (M.S.M.A.)
| | - Naif Saad ALGhasab
- Department of Cardiology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia;
| | - Mohammed S. M. Alharbi
- Department of Internal Medicine, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (A.A.); (M.S.M.A.)
| | - Abdelhafiz I. Bashir
- Department of Physiology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia
| | - Mohd Saleem
- Department of Pathology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (M.S.); (A.S.S.K.); (J.A.A.); (Z.M.); (Y.H.A.); (A.A.A.)
| | - Azharuddin Sajid Syed Khaja
- Department of Pathology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (M.S.); (A.S.S.K.); (J.A.A.); (Z.M.); (Y.H.A.); (A.A.A.)
| | | | - Ehab Rakha
- Departments of Microbiology, King Khalid Hospital, Ha’il 55421, Saudi Arabia;
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Jabar Aziz Alshammari
- Department of Pathology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (M.S.); (A.S.S.K.); (J.A.A.); (Z.M.); (Y.H.A.); (A.A.A.)
| | - Taha E. Taha
- Department of Epidemiology, John Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Ziyad Melibari
- Department of Pathology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (M.S.); (A.S.S.K.); (J.A.A.); (Z.M.); (Y.H.A.); (A.A.A.)
| | - Yaseer H. Alharbi
- Department of Pathology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (M.S.); (A.S.S.K.); (J.A.A.); (Z.M.); (Y.H.A.); (A.A.A.)
| | - Ali A. Almutlag
- Department of Pathology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (M.S.); (A.S.S.K.); (J.A.A.); (Z.M.); (Y.H.A.); (A.A.A.)
| | - Kamaleldin B. Said
- Department of Pathology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia; (M.S.); (A.S.S.K.); (J.A.A.); (Z.M.); (Y.H.A.); (A.A.A.)
- Genomics, Bioinformatics and Systems Biology, Carleton University, 1125 Colonel-By Drive, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
39
|
Hullahalli K, Dailey KG, Waldor MK. Innate immune responses yield tissue-specific bottlenecks that scale with pathogen dose. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.09.543079. [PMID: 37333208 PMCID: PMC10274871 DOI: 10.1101/2023.06.09.543079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
To cause infection, pathogens must overcome bottlenecks imposed by the host immune system. These bottlenecks restrict the inoculum and largely determine whether pathogen exposure results in disease. Infection bottlenecks therefore quantify the effectiveness of immune barriers. Here, using a model of Escherichia coli systemic infection, we identify bottlenecks that tighten or widen with higher inoculum sizes, revealing that the efficacy of innate immune responses can increase or decrease with pathogen dose. We term this concept "dose scaling". During E. coli systemic infection, dose scaling is tissue specific, dependent on the LPS receptor TLR4, and can be recapitulated by mimicking high doses with killed bacteria. Scaling is therefore due to sensing of pathogen molecules rather than interactions between the host and live bacteria. We propose that dose scaling quantitatively links innate immunity with infection bottlenecks and is a valuable framework for understanding how the inoculum size governs the outcome of pathogen exposure.
Collapse
Affiliation(s)
- Karthik Hullahalli
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| | - Katherine G Dailey
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| | - Matthew K Waldor
- Department of Microbiology, Harvard Medical School, Boston, MA 02115; Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA 02115
| |
Collapse
|
40
|
Hachani A, Giulieri SG, Guérillot R, Walsh CJ, Herisse M, Soe YM, Baines SL, Thomas DR, Cheung SD, Hayes AS, Cho E, Newton HJ, Pidot S, Massey RC, Howden BP, Stinear TP. A high-throughput cytotoxicity screening platform reveals agr-independent mutations in bacteraemia-associated Staphylococcus aureus that promote intracellular persistence. eLife 2023; 12:e84778. [PMID: 37289634 PMCID: PMC10259494 DOI: 10.7554/elife.84778] [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: 11/08/2022] [Accepted: 05/23/2023] [Indexed: 06/10/2023] Open
Abstract
Staphylococcus aureus infections are associated with high mortality rates. Often considered an extracellular pathogen, S. aureus can persist and replicate within host cells, evading immune responses, and causing host cell death. Classical methods for assessing S. aureus cytotoxicity are limited by testing culture supernatants and endpoint measurements that do not capture the phenotypic diversity of intracellular bacteria. Using a well-established epithelial cell line model, we have developed a platform called InToxSa (intracellular toxicity of S. aureus) to quantify intracellular cytotoxic S. aureus phenotypes. Studying a panel of 387 S. aureus bacteraemia isolates, and combined with comparative, statistical, and functional genomics, our platform identified mutations in S. aureus clinical isolates that reduced bacterial cytotoxicity and promoted intracellular persistence. In addition to numerous convergent mutations in the Agr quorum sensing system, our approach detected mutations in other loci that also impacted cytotoxicity and intracellular persistence. We discovered that clinical mutations in ausA, encoding the aureusimine non-ribosomal peptide synthetase, reduced S. aureus cytotoxicity, and increased intracellular persistence. InToxSa is a versatile, high-throughput cell-based phenomics platform and we showcase its utility by identifying clinically relevant S. aureus pathoadaptive mutations that promote intracellular residency.
Collapse
Affiliation(s)
- Abderrahman Hachani
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Stefano G Giulieri
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Romain Guérillot
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Calum J Walsh
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Marion Herisse
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ye Mon Soe
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Sarah L Baines
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - David R Thomas
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Infection and Immunity Program, Department of Microbiology and Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Shane Doris Cheung
- Biological Optical Microscopy Platform, University of MelbourneMelbourneAustralia
| | - Ashleigh S Hayes
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ellie Cho
- Biological Optical Microscopy Platform, University of MelbourneMelbourneAustralia
| | - Hayley J Newton
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Infection and Immunity Program, Department of Microbiology and Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Sacha Pidot
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ruth C Massey
- School of Microbiology, University College CorkCorkIreland
- School of Medicine, University College CorkCorkIreland
- APC Microbiome Ireland, University College CorkCorkIreland
- School of Cellular and Molecular Medicine, University of BristolBristolUnited Kingdom
| | - Benjamin P Howden
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| |
Collapse
|
41
|
Magoch M, McEwen AG, Napolitano V, Władyka B, Dubin G. Crystal Structure of Staphopain C from Staphylococcus aureus. Molecules 2023; 28:molecules28114407. [PMID: 37298883 DOI: 10.3390/molecules28114407] [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: 03/29/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Staphylococcus aureus is a common opportunistic pathogen of humans and livestock that causes a wide variety of infections. The success of S. aureus as a pathogen depends on the production of an array of virulence factors including cysteine proteases (staphopains)-major secreted proteases of certain strains of the bacterium. Here, we report the three-dimensional structure of staphopain C (ScpA2) of S. aureus, which shows the typical papain-like fold and uncovers a detailed molecular description of the active site. Because the protein is involved in the pathogenesis of a chicken disease, our work provides the foundation for inhibitor design and potential antimicrobial strategies against this pathogen.
Collapse
Affiliation(s)
- Malgorzata Magoch
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Alastair G McEwen
- CNRS, INSERM, Université de Strasbourg, IGBMC UMR 7104-UMR-S 1258, F-67400 Illkirch, France
| | - Valeria Napolitano
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Benedykt Władyka
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| |
Collapse
|
42
|
Pérez-Delgado O, Espinoza-Culupú AO, López-López E. Antimicrobial Activity of Apis mellifera Bee Venom Collected in Northern Peru. Antibiotics (Basel) 2023; 12:antibiotics12040779. [PMID: 37107142 PMCID: PMC10135115 DOI: 10.3390/antibiotics12040779] [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: 02/08/2023] [Revised: 04/10/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
Due to the emergence of microorganisms resistant to antibiotics and the failure of antibiotic therapies, there is an urgent need to search for new therapeutic options, as well as new molecules with antimicrobial potential. The objective of the present study was to evaluate the in vitro antibacterial activity of Apis mellifera venom collected in the beekeeping areas of the city of Lambayeque in northern Peru against Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus. Bee venom extraction was performed by electrical impulses and separated using the Amicon ultra centrifugal filter. Subsequently, the fractions were quantified by spectrometric 280 nm and evaluated under denaturant conditions in SDS-PAGE. The fractions were pitted against Escherichia coli ATCC 25922, Staphylococcus aureus ATCC 29213, and Pseudomonas aeruginosa ATCC 27853. A purified fraction (PF) of the venom of A. mellifera and three low molecular weight bands of 7 KDa, 6 KDa, and 5 KDa were identified that showed activity against E. coli with a MIC of 6.88 µg/mL, while for P. aeruginosa and S. aureus, it did not present a MIC. No hemolytic activity at a concentration lower than 15.6 µg/mL and no antioxidant activity. The venom of A. mellifera contains a potential presence of peptides and a predilection of antibacterial activity against E. coli.
Collapse
Affiliation(s)
- Orlando Pérez-Delgado
- Health Science Research Laboratory, Universidad Señor de Sipán, Chiclayo 14001, Peru
| | | | - Elmer López-López
- Faculty of Health Sciences, Universidad Señor de Sipán, Chiclayo 14001, Peru
| |
Collapse
|
43
|
Idrees MM, Saeed K, Shahid MA, Akhtar M, Qammar K, Hassan J, Khaliq T, Saeed A. Prevalence of mecA- and mecC-Associated Methicillin-Resistant Staphylococcus aureus in Clinical Specimens, Punjab, Pakistan. Biomedicines 2023; 11:biomedicines11030878. [PMID: 36979857 PMCID: PMC10045897 DOI: 10.3390/biomedicines11030878] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a clinically prevalent bacterium and is resistant to many drugs. Genetic factors such as mec genes are considered to be responsible for this resistance. Recently, Staphylococcal Cassette Chromosome mec (SCCmec) element mutations produced mecC, a new genetic variant that encodes a transpeptidase enzyme (63% similarity with mecA-encoded PBP2a). This cross-sectional study was conducted to establish the prevalence of the mecA and mecC genes among phenotypically identified MRSA and their effectiveness against different antibiotics in clinical specimens. The prevalence of Staphylococcus aureus was 10.2% (n = 102) in the total number of clinical specimens collected (n = 1000). However, the prevalence of MRSA was 6.3% (n = 63) of the total samples collected, while it was 61.8% among total Staphylococcus aureus isolates. mec genes were confirmed in 96.8% (n = 61) isolates of MRSA, while 3.2% (n = 2) were found to be negative for mec genes. The combination of mecA and mecC was detected in 57.1% (n = 36) of the MRSA isolates. The prevalence of lone mecA was 31.8% (n = 20) and that of lone mecC was 7.9% (n = 5) among all the MRSA samples. Penicillin and amoxicillin/clavulanic acid were the most resistant antibiotics followed by norfloxacin (91.2%), levofloxacin (87.1%), ciprofloxacin (83.9%), azithromycin (78.6%), erythromycin (77.4%), moxifloxacin (69.8%), and sulfamethoxazole/trimethoprim (54.9%). On the other hand, vancomycin and teicoplanin (98.4%) were more effective drugs against MRSA followed by linezolid (96.7%), clindamycin (84.6%), chloramphenicol (83.7%), fusidic acid (70.6%), gentamicin (67.7%), and tetracycline (56.8%). In conclusion, a significant prevalence of mecA and mecC has been found among MRSA isolated from clinical specimens, which is likely responsible for antibiotic resistance in MRSA in our clinical settings. However, vancomycin, teicoplanin, and linezolid were found the top three most effective drugs against MRSA in our clinical settings. Thus, MRSA endemics in local areas require routine molecular and epidemiological investigation.
Collapse
Affiliation(s)
- Muhammad Mubashar Idrees
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Punjab, Pakistan
- Department of Medical Laboratory Technology, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan
- Multan Institute of Kidney Diseases (MIKD), Multan 60800, Punjab, Pakistan
| | - Khadija Saeed
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Punjab, Pakistan
| | - Muhammad Akbar Shahid
- Department of Pathobiology, Faculty of Veterinary Sciences, Bahauddin Zakariya University, Multan 60800, Punjab, Pakistan
| | - Muhammad Akhtar
- Department of Medical Laboratory Technology, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan
| | - Khadija Qammar
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Punjab, Pakistan
| | - Javariya Hassan
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Punjab, Pakistan
| | - Tayyaba Khaliq
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Punjab, Pakistan
| | - Ali Saeed
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Punjab, Pakistan
- Department of Pediatric Oncology & Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Correspondence: ; Tel.: +92-(0)-3226616269
| |
Collapse
|
44
|
Giuliano CJ, Wei KJ, Harling FM, Waldman BS, Farringer MA, Boydston EA, Lan TCT, Thomas RW, Herneisen AL, Sanderlin AG, Coppens I, Dvorin JD, Lourido S. Functional profiling of the Toxoplasma genome during acute mouse infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.05.531216. [PMID: 36945434 PMCID: PMC10028831 DOI: 10.1101/2023.03.05.531216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Within a host, pathogens encounter a diverse and changing landscape of cell types, nutrients, and immune responses. Examining host-pathogen interactions in animal models can therefore reveal aspects of infection absent from cell culture. We use CRISPR-based screens to functionally profile the entire genome of the model apicomplexan parasite Toxoplasma gondii during mouse infection. Barcoded gRNAs were used to track mutant parasite lineages, enabling detection of bottlenecks and mapping of population structures. We uncovered over 300 genes that modulate parasite fitness in mice with previously unknown roles in infection. These candidates span multiple axes of host-parasite interaction, including determinants of tropism, host organelle remodeling, and metabolic rewiring. We mechanistically characterized three novel candidates, including GTP cyclohydrolase I, against which a small-molecule inhibitor could be repurposed as an antiparasitic compound. This compound exhibited antiparasitic activity against T. gondii and Plasmodium falciparum, the most lethal agent of malaria. Taken together, we present the first complete survey of an apicomplexan genome during infection of an animal host, and point to novel interfaces of host-parasite interaction that may offer new avenues for treatment.
Collapse
Affiliation(s)
| | - Kenneth J. Wei
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | - Faye M. Harling
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | | | - Madeline A. Farringer
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Biological Sciences in Public Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | | | - Raina W. Thomas
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | - Alice L. Herneisen
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | | | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Jeffrey D. Dvorin
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sebastian Lourido
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| |
Collapse
|
45
|
A Sequalae of Lineage Divergence in Staphylococcus aureus from Community-Acquired Patterns in Youth to Hospital-Associated Profiles in Seniors Implied Age-Specific Host-Selection from a Common Ancestor. Diagnostics (Basel) 2023; 13:diagnostics13050819. [PMID: 36899963 PMCID: PMC10001379 DOI: 10.3390/diagnostics13050819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
The rapidly changing epidemiology of Staphylococcus aureus and evolution of strains with enhanced virulence is a significant issue in global healthcare. Hospital-associated methicillin-resistant S. aureus (HA-MRSA) lineages are being completely replaced by community-associated S. aureus (CA-MRSA) in many regions. Surveillance programs tracing the reservoirs and sources of infections are needed. Using molecular diagnostics, antibiograms, and patient demographics, we have examined the distributions of S. aureus in Ha'il hospitals. Out of 274 S. aureus isolates recovered from clinical specimens, 181 (66%, n = 181) were MRSA, some with HA-MRSA patterns across 26 antimicrobials with almost full resistances to all beta-lactams, while the majority were highly susceptible to all non-beta-lactams, indicating the CA-MRSA type. The rest of isolates (34%, n = 93) were methicillin-susceptible, penicillin-resistant MSSA lineages (90%). The MRSA in men was over 56% among total MRSA (n = 181) isolates and 37% of overall isolates (n = 102 of 274) compared to MSSA in total isolates (17.5%, n = 48), respectively. However, these were 28.4% (n = 78) and 12.4% (n = 34) for MRSA and MSSA infections in women, respectively. MRSA rates per age groups of 0-20, 21-50, and >50 years of age were 15% (n = 42), 17% (n = 48), and 32% (n = 89), respectively. However, MSSA in the same age groups were 13% (n = 35), 9% (n = 25), and 8% (n = 22). Interestingly, MRSA increased proportional to age, while MSSA concomitantly decreased, implying dominance of the latter ancestors early in life and then gradual replacement by MRSA. The dominance and seriousness of MRSA despite enormous efforts in place is potentially for the increased use of beta-lactams known to enhance virulence. The Intriguing prevalence of the CA-MRSA patterns in young otherwise healthy individuals replaced by MRSA later in seniors and the dominance of penicillin-resistant MSSA phenotypes imply three types of host- and age-specific evolutionary lineages. Thus, the decreasing MSSA trend by age with concomitant increase and sub-clonal differentiation into HA-MRSA in seniors and CA-MRSA in young and otherwise healthy patients strongly support the notion of subclinal emergences from a resident penicillin-resistant MSSA ancestor. Future vertical studies should focus on the surveillance of invasive CA-MRSA rates and phenotypes.
Collapse
|
46
|
Oliveira-Tintino CDDM, Tintino SR, Justino de Araújo AC, dos Santos Barbosa CR, Ramos Freitas P, de Araújo Neto JB, Begnini IM, Rebelo RA, da Silva LE, Mireski SL, Nasato MC, Krautler MIL, Barreto HM, Ribeiro-Filho J, de Menezes IRA, Coutinho HDM. Efflux Pump (QacA, QacB, and QacC) and β-Lactamase Inhibitors? An Evaluation of 1,8-Naphthyridines against Staphylococcus aureus Strains. Molecules 2023; 28:molecules28041819. [PMID: 36838807 PMCID: PMC9961278 DOI: 10.3390/molecules28041819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
The bacterial species Staphylococcus aureus presents a variety of resistance mechanisms, among which the expression of β-lactamases and efflux pumps stand out for providing a significant degree of resistance to clinically relevant antibiotics. The 1,8-naphthyridines are nitrogen heterocycles with a broad spectrum of biological activities and, as such, are promising research targets. However, the potential roles of these compounds on bacterial resistance management remain to be better investigated. Therefore, the present study evaluated the antibacterial activity of 1,8-naphthyridine sulfonamides, addressing their ability to act as inhibitors of β-lactamases and efflux pump (QacA/B and QacC) against the strains SA-K4414 and SA-K4100 of S. aureus. All substances were prepared at an initial concentration of 1024 μg/mL, and their minimum inhibitory concentrations (MIC) were determined by the broth microdilution method. Subsequently, their effects on β-lactamase- and efflux pump-mediated antibiotic resistance was evaluated from the reduction of the MIC of ethidium bromide (EtBr) and β-lactam antibiotics, respectively. The 1,8-naphthyridines did not present direct antibacterial activity against the strains SA-K4414 and SA-K4100 of S. aureus. On the other hand, when associated with antibiotics against both strains, the compounds reduced the MIC of EtBr and β-lactam antibiotics, suggesting that they may act by inhibiting β-lactamases and efflux pumps such as QacC and QacA/B. However, further research is required to elucidate the molecular mechanisms underlying these observed effects.
Collapse
Affiliation(s)
| | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri (URCA), Crato 63105-000, CE, Brazil
- Correspondence: (S.R.T.); (I.R.A.d.M.)
| | - Ana Carolina Justino de Araújo
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri (URCA), Crato 63105-000, CE, Brazil
| | - Cristina Rodrigues dos Santos Barbosa
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri (URCA), Crato 63105-000, CE, Brazil
| | - Priscilla Ramos Freitas
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri (URCA), Crato 63105-000, CE, Brazil
| | - José Bezerra de Araújo Neto
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri (URCA), Crato 63105-000, CE, Brazil
| | - Iêda Maria Begnini
- Department of Chemistry, Regional University of Blumenau (FURB), Itoupava Seca, Blumenau 89030-903, SC, Brazil
| | - Ricardo Andrade Rebelo
- Department of Chemistry, Regional University of Blumenau (FURB), Itoupava Seca, Blumenau 89030-903, SC, Brazil
| | - Luiz Everson da Silva
- Postgraduate Program in Sustainable Territorial Development, Coastal Sector, Federal University of Paraná (UFPR), Curitiba 81531-990, PR, Brazil
| | - Sandro Lucio Mireski
- Department of Chemistry, Regional University of Blumenau (FURB), Itoupava Seca, Blumenau 89030-903, SC, Brazil
| | - Michele Caroline Nasato
- Department of Chemistry, Regional University of Blumenau (FURB), Itoupava Seca, Blumenau 89030-903, SC, Brazil
| | | | | | - Jaime Ribeiro-Filho
- Oswaldo Cruz Foundation (Fiocruz), Fiocruz Ceará, Eusébio 60180-900, CE, Brazil
| | - Irwin Rose Alencar de Menezes
- Laboratory of Pharmacology and Molecular Chemistry (LFQM), Department of Biological Chemistry, Regional University of Cariri (URCA), Crato 63105-000, CE, Brazil
- Correspondence: (S.R.T.); (I.R.A.d.M.)
| | - Henrique Douglas Melo Coutinho
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri (URCA), Crato 63105-000, CE, Brazil
| |
Collapse
|
47
|
Ersanli C, Tzora A, Skoufos I, Fotou K, Maloupa E, Grigoriadou K, Voidarou C(C, Zeugolis DI. The Assessment of Antimicrobial and Anti-Biofilm Activity of Essential Oils against Staphylococcus aureus Strains. Antibiotics (Basel) 2023; 12:384. [PMID: 36830295 PMCID: PMC9952819 DOI: 10.3390/antibiotics12020384] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
The increase in antimicrobial resistance and tolerance over the years has become a serious public health problem, leading to the inevitable development of alternative antimicrobial agents as substitutes for industrial pharmaceutical antibiotics targeting humans and animals under the concept of one health. Essential oils (EOs) extracted from aromatic and pharmaceutical plants incorporate several bioactive compounds (phytochemicals) that positively affect human and animal health. Herein, this work aimed to examine a standardized chemical composition and screen the antimicrobial and anti-biofilm activity of Thymus sibthorpii, Origanum vulgare, Salvia fruticosa, and Crithmum maritimum EOs against three different Staphylococcus aureus strains by gold-standard disc diffusion, broth microdilution, and microtiter plate biofilm assays. Therefore, the evaluation of the above-mentioned EOs were considered as substitutes for antibiotics to combat the ever-mounting antimicrobial resistance problem. The observed bacterial growth inhibition varied significantly depending on the type and concentration of the antimicrobials. Thymus sibthorpii was determined as the strongest antimicrobial, with 0.091 mg/mL minimum inhibitory concentration (MIC) and a 14-33 mm diameter inhibition zone at 5% (v/v) concentration. All tested EOs indicated almost 95% inhibition of biofilm formation at their half MIC, while gentamicin sulfate did not show sufficient anti-biofilm activity. None of the methicillin-resistant strains showed resistance to the EOs compared to methicillin-sensitive strains. Thymus sibthorpii and Origanum vulgare could be potential alternatives as antimicrobial agents to overcome the problem of microbial resistance. The tested EOs might be incorporated into antimicrobial products as safe and potent antimicrobial and anti-biofilm agents.
Collapse
Affiliation(s)
- Caglar Ersanli
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, 47100 Arta, Greece
- Laboratory of Animal Health, Food Hygiene, and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Athina Tzora
- Laboratory of Animal Health, Food Hygiene, and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, 47100 Arta, Greece
| | - Konstantina Fotou
- Laboratory of Animal Health, Food Hygiene, and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece
| | - Eleni Maloupa
- Laboratory of Conservation and Evaluation of Native and Floricultural Species, Institute of Plant Breeding; and Genetic Resources, Hellenic Agricultural Organization Demeter, Thermi, 57001 Thessaloniki, Greece
| | - Katerina Grigoriadou
- Laboratory of Conservation and Evaluation of Native and Floricultural Species, Institute of Plant Breeding; and Genetic Resources, Hellenic Agricultural Organization Demeter, Thermi, 57001 Thessaloniki, Greece
| | - Chrysoula (Chrysa) Voidarou
- Laboratory of Animal Health, Food Hygiene, and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
48
|
Pyae NYL, Maiuthed A, Phongsopitanun W, Ouengwanarat B, Sukma W, Srimongkolpithak N, Pengon J, Rattanajak R, Kamchonwongpaisan S, Ei ZZ, Chunhacha P, Wilasluck P, Deetanya P, Wangkanont K, Hengphasatporn K, Shigeta Y, Rungrotmongkol T, Chamni S. N-Containing α-Mangostin Analogs via Smiles Rearrangement as the Promising Cytotoxic, Antitrypanosomal, and SARS-CoV-2 Main Protease Inhibitory Agents. Molecules 2023; 28:molecules28031104. [PMID: 36770770 PMCID: PMC9919084 DOI: 10.3390/molecules28031104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
New N-containing xanthone analogs of α-mangostin were synthesized via one-pot Smiles rearrangement. Using cesium carbonate in the presence of 2-chloroacetamide and catalytic potassium iodide, α-mangostin (1) was subsequently transformed in three steps to provide ether 2, amide 3, and amine 4 in good yields at an optimum ratio of 1:3:3, respectively. The evaluation of the biological activities of α-mangostin and analogs 2-4 was described. Amine 4 showed promising cytotoxicity against the non-small-cell lung cancer H460 cell line fourfold more potent than that of cisplatin. Both compounds 3 and 4 possessed antitrypanosomal properties against Trypanosoma brucei rhodesiense at a potency threefold stronger than that of α-mangostin. Furthermore, ether 2 gave potent SARS-CoV-2 main protease inhibition by suppressing 3-chymotrypsinlike protease (3CLpro) activity approximately threefold better than that of 1. Fragment molecular orbital method (FMO-RIMP2/PCM) indicated the improved binding interaction of 2 in the 3CLpro active site regarding an additional ether moiety. Thus, the series of N-containing α-mangostin analogs prospectively enhance druglike properties based on isosteric replacement and would be further studied as potential biotically active chemical entries, particularly for anti-lung-cancer, antitrypanosomal, and anti-SARS-CoV-2 main protease applications.
Collapse
Affiliation(s)
- Nan Yadanar Lin Pyae
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok 10330, Thailand
| | - Arnatchai Maiuthed
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Wongsakorn Phongsopitanun
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Bongkot Ouengwanarat
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok 10330, Thailand
| | - Warongrit Sukma
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok 10330, Thailand
| | - Nitipol Srimongkolpithak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Jutharat Pengon
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Roonglawan Rattanajak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Zin Zin Ei
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Preedakorn Chunhacha
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Patcharin Wilasluck
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence for Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Peerapon Deetanya
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence for Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kittikhun Wangkanont
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence for Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kowit Hengphasatporn
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
| | - Thanyada Rungrotmongkol
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Supakarn Chamni
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: ; Tel.: +662-218-8357
| |
Collapse
|
49
|
Pidwill GR, Pyrah JF, Sutton JAF, Best A, Renshaw SA, Foster SJ. Clonal population expansion of Staphylococcus aureus occurs due to escape from a finite number of intraphagocyte niches. Sci Rep 2023; 13:1188. [PMID: 36681703 PMCID: PMC9867732 DOI: 10.1038/s41598-023-27928-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus aureus is a human commensal and also an opportunist pathogen causing life threatening infections. During S. aureus disease, the abscesses that characterise infection can be clonal, whereby a large bacterial population is founded by a single or few organisms. Our previous work has shown that macrophages are responsible for restricting bacterial growth such that a population bottleneck occurs and clonality can emerge. A subset of phagocytes fail to control S. aureus resulting in bacterial division, escape and founding of microabscesses that can seed other host niches. Here we investigate the basis for clonal microabscess formation, using in vitro and in silico models of S. aureus macrophage infection. Macrophages that fail to control S. aureus are characterised by formation of intracellular bacterial masses, followed by cell lysis. High-resolution microscopy reveals that most macrophages had internalised only a single S. aureus, providing a conceptual framework for clonal microabscess generation, which was supported by a stochastic individual-based, mathematical model. Once a threshold of masses was reached, increasing the number of infecting bacteria did not result in greater mass numbers, despite enhanced phagocytosis. This suggests a finite number of permissive, phagocyte niches determined by macrophage associated factors. Increased understanding of the parameters of infection dynamics provides avenues for development of rational control measures.
Collapse
Affiliation(s)
- Grace R Pidwill
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Josie F Pyrah
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK
| | - Joshua A F Sutton
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Alex Best
- School of Mathematics & Statistics, University of Sheffield, Sheffield, S3 7RH, UK.
| | - Stephen A Renshaw
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK.
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK.
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, S10 2RX, UK.
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK.
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK.
| |
Collapse
|
50
|
Abdelaziz R, Tartor YH, Barakat AB, EL-Didamony G, Gado MM, Berbecea A, Radulov HDI. Bioactive metabolites of Streptomyces misakiensis display broad-spectrum antimicrobial activity against multidrug-resistant bacteria and fungi. Front Cell Infect Microbiol 2023; 13:1162721. [PMID: 37168394 PMCID: PMC10165089 DOI: 10.3389/fcimb.2023.1162721] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/07/2023] [Indexed: 05/13/2023] Open
Abstract
Background Antimicrobial resistance is a serious threat to public health globally. It is a slower-moving pandemic than COVID-19, so we are fast running out of treatment options. Purpose Thus, this study was designed to search for an alternative biomaterial with broad-spectrum activity for the treatment of multidrug-resistant (MDR) bacterial and fungal pathogen-related infections. Methods We isolated Streptomyces species from soil samples and identified the most active strains with antimicrobial activity. The culture filtrates of active species were purified, and the bioactive metabolite extracts were identified by thin-layer chromatography (TLC), preparative high-performance liquid chromatography (HPLC), nuclear magnetic resonance (NMR) spectroscopy, and gas chromatography-mass spectrometry (GC-MS). The minimum inhibitory concentrations (MICs) of the bioactive metabolites against MDR bacteria and fungi were determined using the broth microdilution method. Results Preliminary screening revealed that Streptomyces misakiensis and S. coeruleorubidus exhibited antimicrobial potential. The MIC50 and MIC90 of S. misakiensis antibacterial bioactive metabolite (ursolic acid methyl ester) and antifungal metabolite (tetradecamethylcycloheptasiloxane) against all tested bacteria and fungi were 0.5 μg/ml and 1 μg/mL, respectively, versus S. coeruleorubidus metabolites: thiocarbamic acid, N,N-dimethyl, S-1,3-diphenyl-2-butenyl ester against bacteria (MIC50: 2 μg/ml and MIC90: 4 μg/mL) and fungi (MIC50: 4 μg/ml and MIC90: 8 μg/mL). Ursolic acid methyl ester was active against ciprofloxacin-resistant strains of Streptococcus pyogenes, S. agalactiae, Escherichia coli, Klebsiella pneumoniae, and Salmonella enterica serovars, colistin-resistant Aeromonas hydrophila and K. pneumoniae, and vancomycin-resistant Staphylococcus aureus. Tetradecamethylcycloheptasiloxane was active against azole- and amphotericin B-resistant Candida albicans, Cryptococcus neoformans, C. gattii, Aspergillus flavus, A. niger, and A. fumigatus. Ursolic acid methyl ester was applied in vivo for treating S. aureus septicemia and K. pneumoniae pneumonia models in mice. In the septicemia model, the ursolic acid methyl ester-treated group had a significant 4.00 and 3.98 log CFU/g decrease (P < 0.05) in liver and spleen tissue compared to the infected, untreated control group. Lung tissue in the pneumonia model showed a 2.20 log CFU/g significant decrease in the ursolic acid methyl ester-treated group in comparison to the control group. The haematological and biochemical markers in the ursolic acid methyl ester-treated group did not change in a statistically significant way. Moreover, no abnormalities were found in the histopathology of the liver, kidneys, lungs, and spleen of ursolic acid methyl ester-treated mice in comparison with the control group. Conclusion S. misakiensis metabolite extracts are broad-spectrum antimicrobial biomaterials that can be further investigated for the potential against MDR pathogen infections. Hence, it opens up new horizons for exploring alternative drugs for current and reemerging diseases.
Collapse
Affiliation(s)
- Rewan Abdelaziz
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Yasmine H. Tartor
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- *Correspondence: Yasmine H. Tartor, ;
| | - Ahmed B. Barakat
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Gamal EL-Didamony
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Marwa M. Gado
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Adina Berbecea
- Department of Soil Science, University of Life Science”King Mihai I” from, Timioara, Romania
| | | |
Collapse
|