1
|
Liu Y, Meng X, Ma Z, Gu H, Luo X, Yin X, Yi H, Chen Y. Hybrid recognition-enabled ratiometric electrochemical sensing of Staphylococcus aureus via in-situ growth of MOF/Ti 3C 2T x-MXene and a self-reporting bacterial imprinted polymer. Food Chem 2025; 463:141496. [PMID: 39378721 DOI: 10.1016/j.foodchem.2024.141496] [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: 07/26/2024] [Revised: 09/07/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
Rapid and effective analysis of foodborne bacteria is crucial for preventing and controlling bacterial infections. Here, we present the synthesis of a self-reporting molecularly imprinted polymer (MIP) as an inner reference probe (IR), and the in-situ growth of metal-organic frameworks on transition metal carbon nitrides (MOF/Ti3C2TX-MXene) as a signaling nanoprobe (SP). These advancements are then applied in a ratiometric electrochemical bioassay for Staphylococcus aureus (S. aureus) using a hybrid recognition mechanism. When S. aureus is present, the aptamer-integrated MIP (MIP@Apt) efficiently captures it, followed by binding with SP to form a sandwich structure. This leads to decreased current response of IR (IIR) and increased current intensity of SP (Isp), enabling quantification through utilization of the ISP to IIR ratio. The biosensor shows a wide detection range (10-108 CFU mL-1) and low detection limit of 1.2 CFU mL-1. Its feasibility for testing complex samples indicates the potential application in food analysis.
Collapse
Affiliation(s)
- Yang Liu
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou 434023, China
| | - Xianzhu Meng
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou 434023, China
| | - Zhongrui Ma
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou 434023, China
| | - Huiwen Gu
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou 434023, China
| | - Xiao Luo
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou 434023, China
| | - Xiaoli Yin
- College of Life Sciences, Yangtze University, Jingzhou 434025, China
| | - Hongchao Yi
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou 434023, China
| | - Ying Chen
- College of Chemistry and Environmental Engineering, Yangtze University, Jingzhou 434023, China.
| |
Collapse
|
2
|
da Cunha IV, da Silva Oliveira DD, Calefi GG, Silva NBS, Martins CHG, Rezende Júnior CDO, Tsubone TM. Photosensitizer associated with efflux pump inhibitors as a strategy for photodynamic therapy against bacterial resistance. Eur J Med Chem 2024; 284:117197. [PMID: 39731789 DOI: 10.1016/j.ejmech.2024.117197] [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: 10/04/2024] [Revised: 12/01/2024] [Accepted: 12/08/2024] [Indexed: 12/30/2024]
Abstract
Antimicrobial resistance is currently one of the biggest challenges in controlling infectious diseases and was listed among the top 10 threats to global health by the World Health Organization (WHO) in 2023. The antibiotics misuse has led to the widespread emergence of antimicrobial resistance, marking the beginning of the alarming increase in antibiotic resistance. In this context, Antimicrobial Photodynamic Therapy (aPDT) has garnered significant attention from the scientific community due to its potential to effectively eliminate multidrug-resistant pathogenic bacteria and its low propensity to induce drug resistance, which bacteria can quickly develop against traditional antibiotic treatments. However, some efflux pumps can expel diverse substrates from inside the cell, including photosensitizers used in aPDT, contributing to multidrug-resistance mechanisms. Efflux Pump Inhibitors are potential solutions to combat resistance mediated by these pumps and can play a crucial role in enhancing aPDT's effectiveness against multidrug-resistant bacteria. Therefore, combining efflux pumps inhibitors with photosensitizers can possible to eliminate the pathogen more efficiently. This review summarizes the mechanisms in which bacteria resist conventional antibiotic treatment, with a particular emphasis on efflux pump-mediated resistance, and present aPDT as a promising strategy to combat antibiotic resistance. Additionally, we highlighted several molecules of photosensitizer associated with efflux pump inhibitors as potential strategies to optimize aPDT, aiming to offer a perspective on future research directions on aPDT for overcoming the limitations of antibiotic resistance.
Collapse
Affiliation(s)
- Ieda Vieira da Cunha
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | - Gabriel Guimarães Calefi
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | | | | | - Tayana Mazin Tsubone
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
3
|
Peng H, Rao Y, Shang W, Yang Y, Tan L, Liu L, Hu Z, Wang Y, Huang X, Liu H, Li M, Guo Z, Chen J, Yang Y, Wu J, Yuan W, Hu Q, Rao X. Vancomycin‐intermediate Staphylococcus aureus employs CcpA‐GlmS metabolism regulatory cascade to resist vancomycin. MEDCOMM – FUTURE MEDICINE 2024; 3. [DOI: 10.1002/mef2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/28/2024] [Indexed: 01/04/2025]
Abstract
AbstractVancomycin (VAN)‐intermediate Staphylococcus aureus (VISA) is a critical cause of VAN treatment failure worldwide. Multiple genetic changes are reportedly associated with VISA formation, whereas VISA strains often present common phenotypes, such as reduced autolysis and thickened cell wall. However, how mutated genes lead to VISA common phenotypes remains unclear. Here, we show a metabolism regulatory cascade (CcpA‐GlmS), whereby mutated two‐component systems (TCSs) link to the common phenotypes of VISA. We found that ccpA deletion decreased VAN resistance in VISA strains with diverse genetic backgrounds. Metabolic alteration in VISA was associated with ccpA upregulation, which was directly controlled by TCSs WalKR and GraSR. RNA‐sequencing revealed the crucial roles of CcpA in changing the carbon flow and nitrogen flux of VISA to promote VAN resistance. A gate enzyme (GlmS) that drives carbon flow to the cell wall precursor biosynthesis was upregulated in VISA. CcpA directly controlled glmS expression. Blocking CcpA sensitized VISA strains to VAN treatment in vitro and in vivo. Overall, this work uncovers a link between the formation of VISA phenotypes and commonly mutated genes. Inhibition of CcpA‐GlmS cascade is a promising strategy to restore the therapeutic efficiency of VAN against VISA infections.
Collapse
Affiliation(s)
- Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Yifan Rao
- Department of Emergency Medicine, Xinqiao Hospital Army Medical University Chongqing China
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Li Tan
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Lu Liu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Yuting Wang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Xiaonan Huang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - He Liu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Mengyang Li
- Department of Microbiology, School of Medicine Chongqing University Chongqing China
| | - Zuwen Guo
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Juan Chen
- Department of Pharmacy, Xinqiao Hospital Army Medical University Chongqing China
| | - Yuhua Yang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Jianghong Wu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Wenchang Yuan
- KingMed School of Laboratory Medicine Guangzhou Medical University Guangzhou China
| | - Qiwen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing Chongqing China
- Department of Microbiology, School of Medicine Chongqing University Chongqing China
| |
Collapse
|
4
|
Cao X, Huang L, Tang M, Liang Y, Liu X, Hou H, Liang S. Antibiotics daptomycin interacts with S protein of SARS-CoV-2 to promote cell invasion of Omicron (B1.1.529) pseudovirus. Virulence 2024; 15:2339703. [PMID: 38576396 PMCID: PMC11057663 DOI: 10.1080/21505594.2024.2339703] [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: 10/30/2023] [Accepted: 04/03/2024] [Indexed: 04/06/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has posed enormous challenges to global public health. The use of antibiotics has greatly increased during the SARS-CoV-2 epidemic owing to the presence of bacterial co-infection and secondary bacterial infections. The antibiotics daptomycin (DAP) is widely used in the treatment of infectious diseases caused by gram-positive bacteria owing to its highly efficient antibacterial activity. It is pivotal to study the antibiotics usage options for patients of coronavirus infectious disease (COVID-19) with pneumonia those need admission to receive antibiotics treatment for bacterial co-infection in managing COVID-19 disease. Herein, we have revealed the interactions of DAP with the S protein of SARS-CoV-2 and the variant Omicron (B1.1.529) using the molecular docking approach and Omicron (B1.1.529) pseudovirus (PsV) mimic invasion. Molecular docking analysis shows that DAP has a certain degree of binding ability to the S protein of SARS-CoV-2 and several derived virus variants, and co-incubation of 1-100 μM DAP with cells promotes the entry of the PsV into human angiotensin-converting enzyme 2 (hACE2)-expressing HEK-293T cells (HEK-293T-hACE2), and this effect is related to the concentration of extracellular calcium ions (Ca2+). The PsV invasion rate in the HEK-293T-hACE2 cells concurrently with DAP incubation was 1.7 times of PsV infection alone. In general, our findings demonstrate that DAP promotes the infection of PsV into cells, which provides certain reference of antibiotics selection and usage optimization for clinicians to treat bacterial coinfection or secondary infection during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xu Cao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Min Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinpeng Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huijin Hou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shufang Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Adeyemi FM, Oyedara OO, Yusuf-Omoloye NA, Ajigbewu OH, Ndaji OL, Adegbite-Badmus MK, Olumakinde TS, Oluokun TE. Guardians of resistance and virulence: detection of mec, femA, Van, pvl, hlg and spa genes in methicillin and vancomycin-resistant Staphylococcus aureus from clinical and food samples in Southwestern Nigeria. BMC Microbiol 2024; 24:498. [PMID: 39592938 PMCID: PMC11590366 DOI: 10.1186/s12866-024-03660-3] [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: 06/30/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Staphylococcus aureus strains are highly virulent and associated with an eclectic range of severe nosocomial and community-acquired infections. OBJECTIVES This study assessed methicillin- and vancomycin-resistant Staphylococcus aureus (MRSA/VRSA) from clinical and ready-to-eat (RTE) food sources, screened for antibiotic resistance; and molecular determinants of antibiotic and virulence genes. METHODS Altogether, 465 clinical and RTE food samples were analyzed via conventional microbiological techniques and S. aureus identification was confirmed by nuc gene detection. Phenotypic screening for methicillin and vancomycin-resistance was by agar-screen cum micro-broth dilution respectively, while antibiotic susceptibility testing was done by the disc-diffusion technique. VanA/vanB/VanC1, femA, mecA/mecC; pvl/hlg and spa gene detection was via Polymerase chain reaction. RESULTS Phenotypically, 211 Staphylococcal isolates were recovered, 138 (65.4%) of them carrying the nuc gene - all 138 (100.0%) were VRSA, while 59/138 (42.8%) were MRSA phenotypically. Overall, 114/138 (82.6%), 7/138 (5.1%), and 6/138 (4.3%) of isolates had the femA, mecA, and mecC genes, while van genes were detected in only 3 (2.2%) isolates, with virulence determinants pvl, hlg, and spa gene carriage in 8 (5.8%), 10 (7.2%), and 77 (55.8%) isolates respectively. In all, 11.6% carried resistance-associated genes, 55.8% carried virulence genes, and co-detection of resistance and virulence genes was observed in 12.3%. Overall, 96/138 (69.6%) were multidrug-resistant (MDR), while one strain was extremely drug-resistant (XDR). MAR Indices ≥ 0.2 was observed in 83.3% of isolates. CONCLUSION This study highlights virulence levels of MRSA and VRSA circulating strains in Osogbo, contributing to their sustained surveillance, and improving available data for successive epidemiology investigations. This study also reports the occurrence of the mecC gene in S. aureus isolates from RTE foods and human samples in Southwestern Nigeria.
Collapse
Affiliation(s)
- Folasade Muibat Adeyemi
- Department of Microbiology, Faculty of Basic and Applied Sciences, Osun State University, P.M.B. 4494, Osogbo, Osun State, 230212, Nigeria.
| | - Omotayo Opemipo Oyedara
- Department of Biotechnology, Faculty of Basic and Applied Sciences, Osun State University, P.M.B. 4494, Osogbo, 230212, Nigeria
| | - Nana Aishat Yusuf-Omoloye
- Department of Microbiology, Faculty of Basic and Applied Sciences, Osun State University, P.M.B. 4494, Osogbo, Osun State, 230212, Nigeria
| | - Olaoniye Habeebat Ajigbewu
- Department of Microbiology, Faculty of Basic and Applied Sciences, Osun State University, P.M.B. 4494, Osogbo, Osun State, 230212, Nigeria
| | - Onyinye Lynda Ndaji
- Department of Microbiology, Faculty of Basic and Applied Sciences, Osun State University, P.M.B. 4494, Osogbo, Osun State, 230212, Nigeria
| | - Maryam Kikelomo Adegbite-Badmus
- Department of Microbiology, Faculty of Basic and Applied Sciences, Osun State University, P.M.B. 4494, Osogbo, Osun State, 230212, Nigeria
| | | | | |
Collapse
|
6
|
Douarre C, David D, Fangazio M, Picard E, Hadji E, Vandenberg O, Barbé B, Hardy L, Marcoux PR. Simple Imaging System for Label-Free Identification of Bacterial Pathogens in Resource-Limited Settings. Int J Biomed Imaging 2024; 2024:6465280. [PMID: 39606275 PMCID: PMC11599477 DOI: 10.1155/2024/6465280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
Fast, accurate, and affordable bacterial identification methods are paramount for the timely treatment of infections, especially in resource-limited settings (RLS). However, today, only 1.3% of the sub-Saharan African diagnostic laboratories are performing clinical bacteriology. To improve this, diagnostic tools for RLS should prioritize simplicity, affordability, and ease of maintenance, as opposed to the costly equipment utilized for bacterial identification in high-income countries, such as matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). In this work, we present a new high-throughput approach based on a simple wide-field (864 mm2) lensless imaging system allowing for the acquisition of a large portion of a Petri dish coupled with a supervised deep learning algorithm for identification at the bacterial colony scale. This wide-field imaging system is particularly well suited to RLS since it includes neither moving mechanical parts nor optics. We validated this approach through the acquisition and the subsequent analysis of a dataset comprising 252 clinical isolates from five species, encompassing some of the most prevalent pathogens. The resulting optical morphotypes exhibited intra- and interspecies variability, a scenario considerably more akin to real-world clinical practice than the one achievable by solely concentrating on reference strains. Despite this variability, high identification performance was achieved with a correct species identification rate of 91.7%. These results open up some new prospects for identification in RLS. We released both the acquired dataset and the trained identification algorithm in publicly available repositories.
Collapse
Affiliation(s)
- Clément Douarre
- Laboratoire d'Électronique et de Technologie de l'Information, French Alternative Energies and Atomic Energy Commission, Grenoble, France
| | - Dylan David
- Institut de Recherche Interdisciplinaire de Grenoble, French Alternative Energies and Atomic Energy Commission, Grenoble, France
| | - Marco Fangazio
- Research and Technology Innovation Unit, Laboratoire Hospitalier Universitaire de Bruxelles-Universitair Laboratorium Brussel (LHUB-ULB), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Center for Environmental Health and Occupational Health, School of Public Health, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Emmanuel Picard
- Institut de Recherche Interdisciplinaire de Grenoble, French Alternative Energies and Atomic Energy Commission, Grenoble, France
| | - Emmanuel Hadji
- Institut de Recherche Interdisciplinaire de Grenoble, French Alternative Energies and Atomic Energy Commission, Grenoble, France
| | - Olivier Vandenberg
- Research and Technology Innovation Unit, Laboratoire Hospitalier Universitaire de Bruxelles-Universitair Laboratorium Brussel (LHUB-ULB), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Center for Environmental Health and Occupational Health, School of Public Health, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Barbara Barbé
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Liselotte Hardy
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Pierre R. Marcoux
- Laboratoire d'Électronique et de Technologie de l'Information, French Alternative Energies and Atomic Energy Commission, Grenoble, France
| |
Collapse
|
7
|
Timofeeva AM, Galyamova MR, Krivosheev DM, Karabanov SY, Sedykh SE. Investigation of Antibiotic Resistance of E. coli Associated with Farm Animal Feces with Participation of Citizen Scientists. Microorganisms 2024; 12:2308. [PMID: 39597696 PMCID: PMC11596788 DOI: 10.3390/microorganisms12112308] [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: 10/14/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
This paper presents the findings of a large-scale study on antibiotic resistance in bacteria found in farm animal feces across Russia. The study included 6578 samples of farm animal manure from 13 regions in Russia, with the help of citizen scientists. Molecular and microbiological methods were used to analyze 1111 samples of E. coli. The microbiological analysis focused on culturing the microorganisms present in the fecal samples on selective media for E. coli and evaluating the sensitivity of the bacteria to different antibiotics, including ampicillin, tetracycline, chloramphenicol, cefotaxime, and ciprofloxacin. The molecular analysis involved isolating the genomic DNA of the bacteria and conducting PCR assays to detect the vanA, vanB, and mcr-1 antibiotic resistance genes. The results demonstrated significant differences in antibiotic sensitivity of the samples that are morphologically identical to E. coli from different regions. For example, 98.0% and 82.5% of E. coli and other fecal bacterial isolates from the Omsk and Vologda regions lacked antibiotic resistance genes, while 97.7% of samples from the Voronezh region possessed three resistance genes simultaneously. The phenotypic antibiotic sensitivity test also revealed regional differences. For instance, 98.1% of fecal bacterial samples from cattle in the Udmurt Republic were sensitive to all five antibiotics tested, whereas 92.8% of samples from the Voronezh region showed resistance to all five antibiotics. The high level of antibiotic resistance observed may be attributed to their use in farming practices. The distinctive feature of our research is that comprehensive geographical coverage was achieved by using a citizen science platform. Citizen scientists, specifically students from colleges and universities, were responsible for the collection and initial analysis of samples. The project attracted 3096 student participants, enabling the collection and analysis of a significant number of samples from various locations in Russia.
Collapse
Affiliation(s)
- Anna M. Timofeeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia;
| | - Maria R. Galyamova
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia;
| | | | | | - Sergey E. Sedykh
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia;
| |
Collapse
|
8
|
Tawfeek CE, Khattab S, Elmaraghy N, Heiba AA, Nageeb WM. Reduced vancomycin susceptibility in Staphylococcus aureus clinical isolates: a spectrum of less investigated uncertainties. BMC Infect Dis 2024; 24:1218. [PMID: 39472820 PMCID: PMC11520445 DOI: 10.1186/s12879-024-10047-2] [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: 07/09/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Staphylococcus aureus clinical isolates with vancomycin MICs of 2 µg/ml have been associated with vancomycin therapeutic failure and the heterogenous vancomycin-intermediate S. aureus (hVISA) phenotype. While carriage of van genes has usually been associated with higher level of MIC and frank vancomycin resistance, the unrecognized risk of hetero-resistance is frequently underestimated. Methods used for assessing vancomycin susceptibility have also shown different concordance and variable performance and accessibility in routine clinical diagnostics posing a challenge to inform treatment selection in hospital settings. METHODS A total of 195 clinical samples were obtained among which 100 S. aureus isolates were identified. Ninety-six MRSA isolates have been identified using cefoxitin disc and mecA gene detection. The vanA and vanB genes have been screened for in the studied isolates using conventional PCR amplification. Examination of reduced vancomycin susceptibility has been performed using vancomycin screen agar, Broth Micro Dilution method (BMD), and VITEK2. Blood isolates were screened for hVISA using PAP-AUC method. RESULTS Vancomycin screening agar applied to 96 MRSA isolates revealed 16 isolates with reduced vancomycin susceptibility. Further MIC testing revealed that 7 isolates were VISA and only 1 isolate was identified as VRSA using both BMD MIC method and VITEK2. Among 24 tested blood isolates, 4 isolates (16.7%) revealed the hVISA phenotype as identified using PAP-AUC method. Using PCR, vanA gene was identified in 5 S. aureus isolates (5%). Three of them were VSSA while the other two isolates were VISA. CONCLUSION In this study, we report the very low prevalence of VRSA among the tested S. aureus clinical isolates (1%) and the existence of hVISA phenotype among studied S. aureus blood isolates at the rate of 16.7% in our setting. Fifty percent (8/16) of isolates that demonstrated reduced vancomycin susceptibility using vancomycin agar screen tested susceptible using both broth dilution method and VITEK2. These finding together with the concerning silent carriage of vanA gene among VSSA and VISA (5%) may underly hidden and uninvestigated factors contributing to vancomycin treatment failure that warrant cautious vancomycin prescription.
Collapse
Affiliation(s)
- Christine E Tawfeek
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Sally Khattab
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Nermine Elmaraghy
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Anwar A Heiba
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Wedad M Nageeb
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
9
|
Hira J, Singh B, Halder T, Mahmutovic A, Ajayi C, Sekh AA, Hegstad K, Johannessen M, Lentz CS. Single-cell phenotypic profiling and backtracing exposes and predicts clinically relevant subpopulations in isogenic Staphylococcus aureus communities. Commun Biol 2024; 7:1228. [PMID: 39354092 PMCID: PMC11445386 DOI: 10.1038/s42003-024-06894-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: 12/04/2023] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
Isogenic bacterial cell populations are phenotypically heterogenous and may include subpopulations of antibiotic tolerant or heteroresistant cells. The reversibility of these phenotypes and lack of biomarkers to differentiate functionally different, but morphologically identical cells is a challenge for research and clinical detection. To overcome this, we present ´Cellular Phenotypic Profiling and backTracing (CPPT)´, a fluorescence-activated cell sorting platform that uses fluorescent probes to visualize and quantify cellular traits and connects this phenotypic profile with a cell´s experimentally determined fate in single cell-derived growth and antibiotic susceptibility analysis. By applying CPPT on Staphylococcus aureus we phenotypically characterized dormant cells, exposed bimodal growth patterns in colony-derived cells and revealed different culturability of single cells on solid compared to liquid media. We demonstrate that a fluorescent vancomycin conjugate marks cellular subpopulations of vancomycin-intermediate S. aureus with increased likelihood to survive antibiotic exposure, showcasing the value of CPPT for discovery of clinically relevant biomarkers.
Collapse
Affiliation(s)
- Jonathan Hira
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Bhupender Singh
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Tirthankar Halder
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Anel Mahmutovic
- Early Biometrics & Statistical Innovation Data Science & AI AstraZeneca, Biopharmaceuticals RD AstraZeneca, Mölndal, Sweden
| | - Clement Ajayi
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | | | - Kristin Hegstad
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Mona Johannessen
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Christian S Lentz
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
10
|
Hotz JF, Staudacher M, Schefberger K, Spettel K, Schmid K, Kriz R, Schneider L, Hagemann JB, Cyran N, Schmidt K, Starzengruber P, Lötsch F, Leutzendorff A, Daller S, Ramharter M, Burgmann H, Lagler H. Unraveling novel mutation patterns and morphological variations in two dalbavancin-resistant MRSA strains in Austria using whole genome sequencing and transmission electron microscopy. BMC Infect Dis 2024; 24:899. [PMID: 39223565 PMCID: PMC11367932 DOI: 10.1186/s12879-024-09797-w] [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: 03/29/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The increasing prevalence of methicillin-resistant Staphylococcus aureus (MRSA) strains resistant to non-beta-lactam antimicrobials poses a significant challenge in treating severe MRSA bloodstream infections. This study explores resistance development and mechanisms in MRSA isolates, especially after the first dalbavancin-resistant MRSA strain in our hospital in 2016. METHODS This study investigated 55 MRSA bloodstream isolates (02/2015-02/2021) from the University Hospital of the Medical University of Vienna, Austria. The MICs of dalbavancin, linezolid, and daptomycin were assessed. Two isolates (16-33 and 19-362) resistant to dalbavancin were analyzed via whole-genome sequencing, with morphology evaluated using transmission electron microscopy (TEM). RESULTS S.aureus BSI strain 19-362 had two novel missense mutations (p.I515M and p.A606D) in the pbp2 gene. Isolate 16-33 had a 534 bp deletion in the DHH domain of GdpP and a SNV in pbp2 (p.G146R). Both strains had mutations in the rpoB gene, but at different positions. TEM revealed significantly thicker cell walls in 16-33 (p < 0.05) compared to 19-362 and dalbavancin-susceptible strains. None of the MRSA isolates showed resistance to linezolid or daptomycin. CONCLUSION In light of increasing vancomycin resistance reports, continuous surveillance is essential to comprehend the molecular mechanisms of resistance in alternative MRSA treatment options. In this work, two novel missense mutations (p.I515M and p.A606D) in the pbp2 gene were newly identified as possible causes of dalbavancin resistance.
Collapse
Affiliation(s)
- Julian Frederic Hotz
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
- Department of Internal Medicine III, Division of Infectious Diseases, University Hospital of Ulm, Ulm, 89081, Germany
- Department of Neurology, Hospital St. John's of God, Vienna, 1020, Austria
| | - Moritz Staudacher
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
- Department of Angiology, Medical University of Vienna, Vienna, 1090, Austria
| | - Katharina Schefberger
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Kathrin Spettel
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, 1090, Austria
| | - Katharina Schmid
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Richard Kriz
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Lisa Schneider
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | | | - Norbert Cyran
- Faculty of Life Sciences, Research Support Facilities UBB, University of Vienna, Vienna, 1030, Austria
| | - Katy Schmidt
- Faculty of Life Sciences, Research Support Facilities UBB, University of Vienna, Vienna, 1030, Austria
| | - Peter Starzengruber
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, 1090, Austria
| | - Felix Lötsch
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, 1090, Austria
| | - Amelie Leutzendorff
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, 1090, Austria
| | - Simon Daller
- Department of Respiratory and Lung Diseases, Klinik Penzing, Vienna, 1140, Austria
| | - Michael Ramharter
- Center for Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20359, Germany
| | - Heinz Burgmann
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Heimo Lagler
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, 1090, Austria.
| |
Collapse
|
11
|
Hsu YC, Liu CH, Wu YC, Lai SJ, Lin CJ, Tseng TS. Combatting Antibiotic-Resistant Staphylococcus aureus: Discovery of TST1N-224, a Potent Inhibitor Targeting Response Regulator VraRC, through Pharmacophore-Based Screening and Molecular Characterizations. J Chem Inf Model 2024; 64:6132-6146. [PMID: 39078379 PMCID: PMC11323011 DOI: 10.1021/acs.jcim.4c01046] [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: 06/15/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Staphylococcus aureus (S. aureus) is a major global health concern, causing various infections and presenting challenges due to antibiotic resistance. In particular, methicillin-resistant S. aureus, vancomycin-intermediate S. aureus (VISA), and vancomycin-resistant S. aureus pose significant obstacles in treating S. aureus infections. Therefore, the critical need for novel drugs to counter these resistant forms is pressing. Two-component systems (TCSs), integral to bacterial regulation, offer promising targets for disruption. In this study, a comprehensive approach, involving pharmacophore-based inhibitor screening, along with biochemical and biophysical analyses were conducted to identify, characterize, and validate potential inhibitors targeting the response regulator VraRC of S. aureus. The constructed pharmacophore model, Phar-VRPR-N3, demonstrated effectiveness in identifying a potent inhibitor, TST1N-224 (IC50 = 60.2 ± 4.0 μM), against the formation of the VraRC-DNA complex. Notably, TST1N-224 exhibited strong binding to VraRC (KD = 23.4 ± 1.2 μM) using a fast-on-fast-off binding mechanism. Additionally, NMR-based molecular modeling revealed that TST1N-224 predominantly interacts with the α9- and α10-helixes of the DNA-binding domain of VraR, where the interactive and functionally essential residues (N165, K180, S184, and R195) act as hotspots for structure-based inhibitor optimization. Furthermore, TST1N-224 evidently enhanced the susceptibility of VISA to both vancomycin and methicillin. Importantly, TST1N-224 distinguished by 1,2,5,6-tetrathiocane with the 3 and 8 positions modified with ethanesulfonates holds significant potential as a lead compound for the development of new antimicrobial agents.
Collapse
Affiliation(s)
- Ying-Chu Hsu
- Division
of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation ChiaYi Christian Hospital, Chiayi 600566, Taiwan
| | - Ching-Hui Liu
- Institute
of Molecular Biology, National Chung Hsing University, Taichung 40202, Taiwan
| | - Yi-Chen Wu
- Institute
of Molecular Biology, National Chung Hsing University, Taichung 40202, Taiwan
| | - Shu-Jung Lai
- Graduate
Institute of Biomedical Sciences, China
Medical University, Taichung 404333, Taiwan
- Research
Center for Cancer Biology, China Medical
University, Taichung 404333, Taiwan
| | - Chi-Jan Lin
- Institute
of Molecular Biology, National Chung Hsing University, Taichung 40202, Taiwan
| | - Tien-Sheng Tseng
- Institute
of Molecular Biology, National Chung Hsing University, Taichung 40202, Taiwan
| |
Collapse
|
12
|
van Groesen E, Mons E, Kotsogianni I, Arts M, Tehrani KHME, Wade N, Lysenko V, Stel FM, Zwerus JT, De Benedetti S, Bakker A, Chakraborty P, van der Stelt M, Scheffers DJ, Gooskens J, Smits WK, Holden K, Gilmour PS, Willemse J, Hitchcock CA, van Hasselt JGC, Schneider T, Martin NI. Semisynthetic guanidino lipoglycopeptides with potent in vitro and in vivo antibacterial activity. Sci Transl Med 2024; 16:eabo4736. [PMID: 39110780 DOI: 10.1126/scitranslmed.abo4736] [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: 02/05/2022] [Revised: 02/23/2024] [Accepted: 07/16/2024] [Indexed: 08/13/2024]
Abstract
Gram-positive bacterial infections present a major clinical challenge, with methicillin- and vancomycin-resistant strains continuing to be a cause for concern. In recent years, semisynthetic vancomycin derivatives have been developed to overcome this problem as exemplified by the clinically used telavancin, which exhibits increased antibacterial potency but has also raised toxicity concerns. Thus, glycopeptide antibiotics with enhanced antibacterial activities and improved safety profiles are still necessary. We describe the development of a class of highly potent semisynthetic glycopeptide antibiotics, the guanidino lipoglycopeptides, which contain a positively charged guanidino moiety bearing a variable lipid group. These glycopeptides exhibited enhanced in vitro activity against a panel of Gram-positive bacteria including clinically relevant methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant strains, showed minimal toxicity toward eukaryotic cells, and had a low propensity for resistance selection. Mechanistically, guanidino lipoglycopeptides engaged with bacterial cell wall precursor lipid II with a higher binding affinity than vancomycin. Binding to both wild-type d-Ala-d-Ala lipid II and the vancomycin-resistant d-Ala-d-Lac variant was confirmed, providing insight into the enhanced activity of guanidino lipoglycopeptides against vancomycin-resistant isolates. The in vivo efficacy of guanidino lipoglycopeptide EVG7 was evaluated in a S. aureus murine thigh infection model and a 7-day sepsis survival study, both of which demonstrated superiority to vancomycin. Moreover, the minimal to mild kidney effects at supratherapeutic doses of EVG7 indicate an improved therapeutic safety profile compared with vancomycin. These findings position guanidino lipoglycopeptides as candidates for further development as antibacterial agents for the treatment of clinically relevant multidrug-resistant Gram-positive infections.
Collapse
Affiliation(s)
- Emma van Groesen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Elma Mons
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Kamaleddin H M E Tehrani
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Nicola Wade
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Vladyslav Lysenko
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Florence M Stel
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Jordy T Zwerus
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Stefania De Benedetti
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Alexander Bakker
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Parichita Chakraborty
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Dirk-Jan Scheffers
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Jairo Gooskens
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Wiep Klaas Smits
- Experimental Bacteriology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Kirsty Holden
- Evotec (U.K.) Ltd., Alderley Park, Macclesfield, Cheshire, SK10 4TG UK
| | | | - Joost Willemse
- Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | | | - J G Coen van Hasselt
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, Netherlands
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| |
Collapse
|
13
|
Malachowa N, Sturdevant DE, Porter AR, Martin G, Martens C, Nair V, Hansen B, Ricklefs S, Jenkins SG, Chen L, Kreiswirth BN, DeLeo FR. Insights into the molecular basis of reduced vancomycin susceptibility among three prominent Staphylococcus aureus clonal complexes. Microbiol Spectr 2024; 12:e0048624. [PMID: 38916317 PMCID: PMC11302346 DOI: 10.1128/spectrum.00486-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/21/2024] [Indexed: 06/26/2024] Open
Abstract
Staphylococcus aureus is a leading cause of healthcare-associated infections globally. Vancomycin-resistant S. aureus (VRSA), those with high-level resistance [minimum inhibitory concentration (MIC) of 16-32 µg/mL vancomycin], are uncommon, whereas vancomycin-intermediate S. aureus (VISA; MIC of 4-8 µg/mL), are isolated more frequently and develop during long-term and/or repeated use of the antibiotic. VISA can be difficult to eradicate and infections may persist. Our knowledge of mechanisms that underlie the development of VISA is incomplete. We used a genomics approach to investigate the VISA phenotype in three prominent S. aureus lineages. All VISA clinical isolates tested had increased cell wall thickness compared with vancomycin-susceptible S. aureus strains. Growth rates of clonal complex (CC) 5, CC8, and CC45 clinical isolates were reduced in 2 µg/mL vancomycin compared to media alone. Culture in 2 and 4 µg/mL vancomycin sequentially for two weeks reduced susceptibility to daptomycin, televancin, tigecycline, and vancomycin in a majority of CC5, CC8, and CC45 isolates tested. We identified alleles reported previously to contribute to the VISA phenotype, but unexpectedly, these alleles were unique to each CC. A subtherapeutic concentration of vancomycin elicited changes in the VISA transcriptome-common and unique-among the three CCs tested. Multiple genes, including those encoding a glycerate kinase, an M50 family metallopeptidase, and an uncharacterized membrane protein, were upregulated among all three lineages and not reported previously as associated with VISA. Although there are lineage-specific changes in DNA sequence, our findings suggest changes in the VISA transcriptome constitute a general response to stress that confers reduced susceptibility to multiple antibiotics. IMPORTANCE Our understanding of the mechanisms that underlie the development of vancomycin-intermediate Staphylococcus aureus (VISA) is incomplete. To provide a more comprehensive view of this process, we compared genome sequences of clonal complex (CC) 5, CC8, and CC45 VISA clinical isolates and measured changes in the transcriptomes of these isolates during culture with a subtherapeutic concentration of vancomycin. Notably, we identified differentially expressed genes that were lineage-specific or common to the lineages tested, including genes that have not been previously reported to contribute to a VISA phenotype. Changes in gene expression were accompanied by reduced growth rate, increased cell wall thickness, and reduced susceptibility to daptomycin, televancin, tigecycline, and vancomycin. Our results provide support to the idea that changes in gene expression contribute to the development of VISA among three CCs that are a prominent cause of human infections.
Collapse
Affiliation(s)
- Natalia Malachowa
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Daniel E. Sturdevant
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Adeline R. Porter
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Grant Martin
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Craig Martens
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Vinod Nair
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Bryan Hansen
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Stacy Ricklefs
- Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Stephen G. Jenkins
- Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Liang Chen
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Barry N. Kreiswirth
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Frank R. DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
14
|
Yun JH, Chang E, Bae S, Jung J, Kim MJ, Chong YP, Kim SH, Choi SH, Lee SO, Kim YS. Risk factors for vancomycin treatment failure in heterogeneous vancomycin-intermediate Staphylococcus aureus bacteremia. Microbiol Spectr 2024; 12:e0033324. [PMID: 38916352 PMCID: PMC11302031 DOI: 10.1128/spectrum.00333-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024] Open
Abstract
The incidence of heterogeneous vancomycin-intermediate Staphylococcus aureus (hVISA) infection is increasing and is associated with vancomycin treatment failures. However, studies investigating the risk factors for treatment failure in hVISA infection are limited. Patients with hVISA bacteremia treated with vancomycin over 7 days between August 2008 and June 2020 were enrolled in this study. Clinical and microbiological characteristics were compared between vancomycin treatment failure and success groups to identify the risk factors for vancomycin treatment failure. Among the 180 patients with hVISA bacteremia, 102 patients treated with vancomycin over 7 days were included. Vancomycin treatment failed in 80 (78%) patients. Patients in the vancomycin treatment failure group were older (P < 0.001) and more frequently had solid cancer (P = 0.04) than those in the vancomycin treatment success group. Solid organ transplantation (SOT) was more frequent (P < 0.001) in the vancomycin treatment success group. The Charlson comorbidity index (P = 0.01) and Acute Physiology and Chronic Health Evaluation II scores (P < 0.001) were higher in the vancomycin treatment failure group. In multivariate analysis, independent risk factors for vancomycin treatment failure were old age and severity of bacteremia. SOT and vancomycin minimal inhibitory concentration (MIC) ≤ 1.0 mg/L using the broth microdilution (BMD) method were associated with successful vancomycin treatment. Old age and infection severity were independent risk factors for vancomycin treatment failure. Vancomycin MIC using the BMD method is an important risk factor for vancomycin treatment failure, and its use should be considered in hVISA bacteremia.IMPORTANCEIn this study, we assessed the clinical and microbiological characteristics of heterogeneous vancomycin-intermediated Staphylococcus aureus (hVISA) bacteremia and identified risk factors for vancomycin treatment failure. We found that advanced age and severity of infection were independent risk factors for vancomycin treatment failure. On the other hand, solid organ transplantation and a low vancomycin minimal inhibitory concentration were associated with successful vancomycin treatment. This study highlights the importance of vancomycin minimal inhibitory concentration in hVISA bacteremia.
Collapse
Affiliation(s)
- Ji Hyun Yun
- Division of Infectious Diseases, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| | - Euijin Chang
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seongman Bae
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jiwon Jung
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Min Jae Kim
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yong Pil Chong
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung-Han Kim
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sang-Ho Choi
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sang-Oh Lee
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yang Soo Kim
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
15
|
Chaudhary K, Agrahari B, Biswas B, Chatterjee N, Chaudhary A, Kumar A, Sonker H, Dewan S, Saxena D, Akhir A, Malhotra N, Chopra S, Misra S, Matheswaran S, Singh RG. Pyridine-2,6-Dicarboxamide Proligands and their Cu(II)/Zn(II) Complexes Targeting Staphylococcus Aureus for the Attenuation of In Vivo Dental Biofilm. Adv Healthc Mater 2024; 13:e2400378. [PMID: 38621382 DOI: 10.1002/adhm.202400378] [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: 02/10/2024] [Revised: 04/07/2024] [Indexed: 04/17/2024]
Abstract
In the pursuit to combat stubborn bacterial infections, particularly those stemming from gram-positive bacteria, this study is an attempt to craft a precision-driven platform characterized by unparalleled selectivity, specificity, and synergistic antimicrobial mechanisms. Leveraging remarkable potential of metalloantibiotics in antimicrobial applications, herein, this work rationally designs, synthesizes, and characterizes a new library of Pyridine-2,6-dicarboxamide ligands and their corresponding transition metal Cu(II)/Zn(II) complexes. The lead compound L11 demonstrates robust antibacterial properties against Staphylococcus aureus (Minimum Inhibitory Concentration (MIC) = 2-16 µg mL-1), methicillin and vancomycin-resistant S. aureus (MIC = 2-4 µg mL-1) and exhibit superior antibacterial activity when compared to FDA-approved vancomycin, the drug of last resort. Additionally, the compound exhibits notable antimicrobial efficacy against resistant enterococcus strains (MIC = 2-8 µg mL-1). To unravel mechanistic profile, advanced imaging techniques including SEM and AFM are harnessed, collectively suggesting a mechanistic pathway involving cell wall disruption. Live/dead fluorescence studies further confirm efficacy of L11 and its complexes against S. aureus membranes. This translational exploration extends to a rat model, indicating promising in vivo therapeutic potential. Thus, this comprehensive research initiative has capabilities to transcends the confines of this laboratory, heralding a pivotal step toward combatting antibiotic-resistant pathogens and advancing the frontiers of metalloantibiotics-based therapy with a profound clinical implication.
Collapse
Affiliation(s)
| | | | - Bhumika Biswas
- Department of Biological Sciences and Bioengineering, IIT, Kanpur, 208016, India
| | - Niranjan Chatterjee
- Department of Biological Sciences and Bioengineering, IIT, Kanpur, 208016, India
| | | | | | | | - Sayari Dewan
- Department of Chemistry, IIT, Kanpur, 208016, India
| | - Deepanshi Saxena
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Abdul Akhir
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Nidhi Malhotra
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Budh Nagar, 201314, India
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Santosh Misra
- Department of Biological Sciences and Bioengineering, IIT, Kanpur, 208016, India
| | | | | |
Collapse
|
16
|
Liu X, de Bakker V, Heggenhougen MV, Mårli MT, Frøynes AH, Salehian Z, Porcellato D, Morales Angeles D, Veening JW, Kjos M. Genome-wide CRISPRi screens for high-throughput fitness quantification and identification of determinants for dalbavancin susceptibility in Staphylococcus aureus. mSystems 2024; 9:e0128923. [PMID: 38837392 PMCID: PMC11265419 DOI: 10.1128/msystems.01289-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/01/2024] [Indexed: 06/07/2024] Open
Abstract
Antibiotic resistance and tolerance remain a major problem for the treatment of staphylococcal infections. Identifying genes that influence antibiotic susceptibility could open the door to novel antimicrobial strategies, including targets for new synergistic drug combinations. Here, we developed a genome-wide CRISPR interference library for Staphylococcus aureus, demonstrated its use by quantifying gene fitness in different strains through CRISPRi-seq, and used it to identify genes that modulate susceptibility to the lipoglycopeptide dalbavancin. By exposing the library to sublethal concentrations of dalbavancin using both CRISPRi-seq and direct selection methods, we not only found genes previously reported to be involved in antibiotic susceptibility but also identified genes thus far unknown to affect antibiotic tolerance. Importantly, some of these genes could not have been detected by more conventional transposon-based knockout approaches because they are essential for growth, stressing the complementary value of CRISPRi-based methods. Notably, knockdown of a gene encoding the uncharacterized protein KapB specifically sensitizes the cells to dalbavancin, but not to other antibiotics of the same class, whereas knockdown of the Shikimate pathway showed the opposite effect. The results presented here demonstrate the promise of CRISPRi-seq screens to identify genes and pathways involved in antibiotic susceptibility and pave the way to explore alternative antimicrobial treatments through these insights.IMPORTANCEAntibiotic resistance is a challenge for treating staphylococcal infections. Identifying genes that affect how antibiotics work could help create new treatments. In our study, we made a CRISPR interference library for Staphylococcus aureus and used this to find which genes are critical for growth and also mapped genes that are important for antibiotic sensitivity, focusing on the lipoglycopeptide antibiotic dalbavancin. With this method, we identified genes that altered the sensitivity to dalbavancin upon knockdown, including genes involved in different cellular functions. CRISPRi-seq offers a means to uncover untapped antibiotic targets, including those that conventional screens would disregard due to their essentiality. This paves the way for the discovery of new ways to fight infections.
Collapse
Affiliation(s)
- Xue Liu
- Department of Pathogen, Biology, International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, China
- Department of Fundamental Microbiology, University of Lausanne, , Switzerland
| | - Vincent de Bakker
- Department of Fundamental Microbiology, University of Lausanne, , Switzerland
| | | | - Marita Torrissen Mårli
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Norway
| | - Anette Heidal Frøynes
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Norway
| | - Zhian Salehian
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Norway
| | - Davide Porcellato
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Norway
| | - Danae Morales Angeles
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Norway
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, University of Lausanne, , Switzerland
| | - Morten Kjos
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Norway
| |
Collapse
|
17
|
Lv JX, Huang YH, Kafauit F, Wang YH, Su C, Ma JH, Xu Y, Huang CC, Zhang Q, Su YW. Pharmacokinetics and pharmacodynamics of intravenous delafloxacin in healthy subjects: model-based dose optimization. Antimicrob Agents Chemother 2024; 68:e0042824. [PMID: 38899925 PMCID: PMC11232397 DOI: 10.1128/aac.00428-24] [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] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Delafloxacin, a fluoroquinolone antibiotic to treat skin infections, exhibits a broad-spectrum antimicrobial activity. The first randomized, open-label phase I clinical trial was conducted to assess the safety and pharmacokinetics (PK) of intravenous delafloxacin in the Chinese population. A population pharmacokinetic (PopPK) model based on the clinical trial was conducted by NONMEM software. Monte Carlo simulation was performed to evaluate the antibacterial effects of delafloxacin at different doses in different Chinese populations. The PK characteristics of delafloxacin were best described by a three-compartment model with mixed linear and nonlinear clearance. Body weight was included as a covariate in the model. We simulated the AUC0-24h in a steady state at five doses in patient groups of various weights. The results indicated that for patients weighing 70 kg and treated with methicillin-resistant Staphylococcus aureus (MRSA) infections, a minimum dose of 300 mg achieved a PTA > 90% at MIC90 of 0.25 µg/mL, suggesting an ideal bactericidal effect. For patients weighing less than 60 kg, a dose of 200 mg achieved a PTA > 90% at MIC90 of 0.25 µg/mL, also suggesting an ideal bactericidal effect. Additionally, this trial demonstrated the high safety of delafloxacin in single-dose and multiple-dose groups of Chinese. Delafloxacin (300 mg, q12h, iv) was recommended for achieving optimal efficacy in Chinese bacterial skin infections patients. To ensure optimal efficacy, an individualized dose of 200 mg (q12h, iv) could be advised for patients weighing less than 60 kg, and 300 mg (q12h, iv) for those weighing more than 60 kg.
Collapse
Affiliation(s)
- Jiong-Xian Lv
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi-Huan Huang
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Farah Kafauit
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuan-Hui Wang
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chang Su
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun-Heng Ma
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Xu
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chao-Chao Huang
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Zhang
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu-Wen Su
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Clinical Pharmacology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Long DR, Holmes EA, Lo HY, Penewit K, Almazan J, Hodgson T, Berger NF, Bishop ZH, Lewis JD, Waalkes A, Wolter DJ, Salipante SJ. Clinical and in vitro models identify distinct adaptations enhancing Staphylococcus aureus pathogenesis in human macrophages. PLoS Pathog 2024; 20:e1012394. [PMID: 38991026 PMCID: PMC11265673 DOI: 10.1371/journal.ppat.1012394] [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: 02/16/2024] [Revised: 07/23/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen of human macrophages, which facilitates chronic infection. The genotypes, pathways, and mutations influencing that phenotype remain incompletely explored. Here, we used two distinct strategies to ascertain S. aureus gene mutations affecting pathogenesis in macrophages. First, we analyzed isolates collected serially from chronic cystic fibrosis (CF) respiratory infections. We found that S. aureus strains evolved greater macrophage invasion capacity during chronic human infection. Bacterial genome-wide association studies (GWAS) identified 127 candidate genes for which mutation was significantly associated with macrophage pathogenesis in vivo. In parallel, we passaged laboratory S. aureus strains in vitro to select for increased infection of human THP-1 derived macrophages, which identified 15 candidate genes by whole-genome sequencing. Functional validation of candidate genes using isogenic transposon mutant knockouts and CRISPR interference (CRISPRi) knockdowns confirmed virulence contributions from 37 of 39 tested genes (95%) implicated by in vivo studies and 7 of 10 genes (70%) ascertained from in vitro selection, with one gene in common to the two strategies. Validated genes included 17 known virulence factors (39%) and 27 newly identified by our study (61%), some encoding functions not previously associated with macrophage pathogenesis. Most genes (80%) positively impacted macrophage invasion when disrupted, consistent with the phenotype readily arising from loss-of-function mutations in vivo. This work reveals genes and mechanisms that contribute to S. aureus infection of macrophages, highlights differences in mutations underlying convergent phenotypes arising from in vivo and in vitro systems, and supports the relevance of S. aureus macrophage pathogenesis during chronic respiratory infection in CF. Additional studies will be needed to illuminate the exact mechanisms by which implicated mutations affect their phenotypes.
Collapse
Affiliation(s)
- Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Elizabeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jared Almazan
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Taylor Hodgson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Nova F. Berger
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Zoe H. Bishop
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Daniel J. Wolter
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
19
|
Nguyen A, Roy JJS, Kim JH, Yun KH, Lee W, Kim KK, Kim T, Chaurasia AK. Repeated Exposure of Vancomycin to Vancomycin-Susceptible Staphylococcus aureus (VSSA) Parent Emerged VISA and VRSA Strains with Enhanced Virulence Potentials. J Microbiol 2024; 62:535-553. [PMID: 38814539 DOI: 10.1007/s12275-024-00139-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/31/2024]
Abstract
The emergence of resistance against the last-resort antibiotic vancomycin in staphylococcal infections is a serious concern for human health. Although various drug-resistant pathogens of diverse genetic backgrounds show higher virulence potential, the underlying mechanism behind this is not yet clear due to variability in their genetic dispositions. In this study, we investigated the correlation between resistance and virulence in adaptively evolved isogenic strains. The vancomycin-susceptible Staphylococcus aureus USA300 was exposed to various concentrations of vancomycin repeatedly as a mimic of the clinical regimen to obtain mutation(s)-accrued-clonally-selected (MACS) strains. The phenotypic analyses followed by expression of the representative genes responsible for virulence and resistance of MACS strains were investigated. MACS strains obtained under 2 and 8 µg/ml vancomycin, named Van2 and Van8, respectively; showed enhanced vancomycin minimal inhibitory concentrations (MIC) to 4 and 16 µg/ml, respectively. The cell adhesion and invasion of MACS strains increased in proportion to their MICs. The correlation between resistance and virulence potential was partially explained by the differential expression of genes known to be involved in both virulence and resistance in MACS strains compared to parent S. aureus USA300. Repeated treatment of vancomycin against vancomycin-susceptible S. aureus (VSSA) leads to the emergence of vancomycin-resistant strains with variable levels of enhanced virulence potentials.
Collapse
Affiliation(s)
- An Nguyen
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - J Jean Sophy Roy
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Ji-Hoon Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyung-Hee Yun
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Truc Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| | - Akhilesh Kumar Chaurasia
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| |
Collapse
|
20
|
Quan K, Mao Z, Lu Y, Qin Y, Wang S, Yu C, Bi X, Tang H, Ren X, Chen D, Cheng Y, Wang Y, Zheng Y, Xia D. Composited silk fibroins ensured adhesion stability and magnetic controllability of Fe 3O 4-nanoparticle coating on implant for biofilm treatment. MATERIALS HORIZONS 2024; 11:3157-3165. [PMID: 38629215 DOI: 10.1039/d4mh00097h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Magnetic propulsion of nano-/micro-robots is an effective way to treat implant-associated infections by physically destroying biofilm structures to enhance antibiotic killing. However, it is hard to precisely control the propulsion in vivo. Magnetic-nanoparticle coating that can be magnetically pulled off does not need precise control, but the requirement of adhesion stability on an implant surface restricts its magnetic responsiveness. Moreover, whether the coating has been fully pulled-off or not is hard to ensure in real-time in vivo. Herein, composited silk fibroins (SFMA) are optimized to stabilize Fe3O4 nanoparticles on a titanium surface in a dry environment; while in an aqueous environment, the binding force of SFMA on titanium is significantly reduced due to hydrophilic interaction, making the coating magnetically controllable by an externally-used magnet but still stable in the absence of a magnet. The maximum working distance of the magnet can be calculated using magnetomechanical simulation in which the yielding magnetic traction force is strong enough to pull Fe3O4 nanoparticles off the surface. The pulling-off removes the biofilms that formed on the coating and enhances antibiotic killing both in vitro and in a rat sub-cutaneous implant model by up to 100 fold. This work contributes to the practical knowledge of magnetic propulsion for biofilm treatment.
Collapse
Affiliation(s)
- Kecheng Quan
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Zhinan Mao
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Yupu Lu
- Department of Dental Materials, Peking University School and Hospital of Stomatology, & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| | - Yu Qin
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Shuren Wang
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Chunhao Yu
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Xuewei Bi
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Hao Tang
- Department of Dental Materials, Peking University School and Hospital of Stomatology, & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| | - Yan Cheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Yong Wang
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Dandan Xia
- Department of Dental Materials, Peking University School and Hospital of Stomatology, & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| |
Collapse
|
21
|
Fait A, Silva SF, Abrahamsson JÅH, Ingmer H. Staphylococcus aureus response and adaptation to vancomycin. Adv Microb Physiol 2024; 85:201-258. [PMID: 39059821 DOI: 10.1016/bs.ampbs.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Antibiotic resistance is an increasing challenge for the human pathogen Staphylococcus aureus. Methicillin-resistant S. aureus (MRSA) clones have spread globally, and a growing number display decreased susceptibility to vancomycin, the favoured antibiotic for treatment of MRSA infections. These vancomycin-intermediate S. aureus (VISA) or heterogeneous vancomycin-intermediate S. aureus (hVISA) strains arise from accumulation of a variety of point mutations, leading to cell wall thickening and reduced vancomycin binding to the cell wall building block, Lipid II, at the septum. They display only minor changes in vancomycin susceptibility, with varying tolerance between cells in a population, and therefore, they can be difficult to detect. In this review, we summarize current knowledge of VISA and hVISA. We discuss the role of genetic strain background or epistasis for VISA development and the possibility of strains being 'transient' VISA with gene expression changes mediated by, for example, VraTSR, GraXSR, or WalRK signal transduction systems, leading to temporary vancomycin tolerance. Additionally, we address collateral susceptibility to other antibiotics than vancomycin. Specifically, we estimate how mutations in rpoB, encoding the β-subunit of the RNA polymerase, affect overall protein structure and compare changes with rifampicin resistance. Ultimately, such in-depth analysis of VISA and hVISA strains in terms of genetic and transcriptional changes, as well as changes in protein structures, may pave the way for improved detection and guide antibiotic therapy by revealing strains at risk of VISA development. Such tools will be valuable for keeping vancomycin an asset also in the future.
Collapse
Affiliation(s)
- Anaëlle Fait
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark; Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland
| | - Stephanie Fulaz Silva
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
22
|
Zhou C, Pawline MB, Pironti A, Morales SM, Perault AI, Ulrich RJ, Podkowik M, Lejeune A, DuMont A, Stubbe FX, Korman A, Jones DR, Schluter J, Richardson AR, Fey PD, Drlica K, Cadwell K, Torres VJ, Shopsin B. Microbiota and metabolic adaptation shape Staphylococcus aureus virulence and antimicrobial resistance during intestinal colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593044. [PMID: 38766195 PMCID: PMC11100824 DOI: 10.1101/2024.05.11.593044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Depletion of microbiota increases susceptibility to gastrointestinal colonization and subsequent infection by opportunistic pathogens such as methicillin-resistant Staphylococcus aureus (MRSA). How the absence of gut microbiota impacts the evolution of MRSA is unknown. The present report used germ-free mice to investigate the evolutionary dynamics of MRSA in the absence of gut microbiota. Through genomic analyses and competition assays, we found that MRSA adapts to the microbiota-free gut through sequential genetic mutations and structural changes that enhance fitness. Initially, these adaptations increase carbohydrate transport; subsequently, evolutionary pathways largely diverge to enhance either arginine metabolism or cell wall biosynthesis. Increased fitness in arginine pathway mutants depended on arginine catabolic genes, especially nos and arcC, which promote microaerobic respiration and ATP generation, respectively. Thus, arginine adaptation likely improves redox balance and energy production in the oxygen-limited gut environment. Findings were supported by human gut metagenomic analyses, which suggest the influence of arginine metabolism on colonization. Surprisingly, these adaptive genetic changes often reduced MRSA's antimicrobial resistance and virulence. Furthermore, resistance mutation, typically associated with decreased virulence, also reduced colonization fitness, indicating evolutionary trade-offs among these traits. The presence of normal microbiota inhibited these adaptations, preserving MRSA's wild-type characteristics that effectively balance virulence, resistance, and colonization fitness. The results highlight the protective role of gut microbiota in preserving a balance of key MRSA traits for long-term ecological success in commensal populations, underscoring the potential consequences on MRSA's survival and fitness during and after host hospitalization and antimicrobial treatment.
Collapse
Affiliation(s)
- Chunyi Zhou
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Miranda B. Pawline
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
- Microbial Computational Genomic Core Lab, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Sabrina M. Morales
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Andrew I. Perault
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Robert J. Ulrich
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY 10016, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alannah Lejeune
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ashley DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | | | - Aryeh Korman
- Metabolomics Core Resource Laboratory, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Drew R. Jones
- Metabolomics Core Resource Laboratory, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jonas Schluter
- Institute for Systems Genetics, Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony R. Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Paul D. Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Karl Drlica
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07102, USA
- Department of Microbiology, Biochemistry & Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ 07102, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY 10016, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
23
|
Porchera BR, da Silva CM, Miranda RP, Gomes ARQ, Fernandes PHDS, de Menezes CGO, Laurindo PDSDODC, Dolabela MF, Brígido HPC. Linezolid and vancomycin for nosocomial infections in pediatric patients: a systematic review. J Pediatr (Rio J) 2024; 100:242-249. [PMID: 38145631 PMCID: PMC11065658 DOI: 10.1016/j.jped.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/13/2023] [Accepted: 08/30/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVE To investigate the effectiveness of linezolid and vancomycin for the treatment of nosocomial infections in children under 12 years old. DATA SOURCES This is a systematic review in which five randomized clinical trials about the effectiveness of linezolid and vancomycin, involving a total of 429 children with nosocomial infections, were evaluated. They were searched in scientific databases: PubMed, Bvs, and SciELO. SUMMARY OF FINDINGS The main nosocomial infections that affected children were bacteremia, skin, and soft tissue infections followed by nosocomial pneumonia. Most infections were caused by Gram-positive bacteria, which all studies showed infections caused by Staphylococcus aureus, with methicillin-resistant S. aureus (MRSA) and methicillin-resistant coagulase-negative staphylococci strains being isolated. Both linezolid and vancomycin showed high therapeutic efficacy against different types of nosocomial infections, ranging from 84.4% to 94% for linezolid and 76.9% to 90% for vancomycin. Patients receiving linezolid had lower rates of rash and red man syndrome compared to those receiving vancomycin. However, despite the adverse reactions, antimicrobials can be safely administered to children to treat nosocomial infections caused by resistant Gram-positive bacteria. CONCLUSION Both linezolid and vancomycin showed good efficacy in the treatment of bacterial infections caused by resistant Gram-positive bacteria in hospitalized children. However, linezolid stands out regarding its pharmacological safety. Importantly, to strengthen this conclusion, further clinical trials are needed to provide additional evidence.
Collapse
Affiliation(s)
- Bruno Russo Porchera
- Centro Universitário do Estado do Pará (CESUPA), Curso de Medicina, Belém, PA, Brazil
| | | | | | - Antônio Rafael Quadros Gomes
- Universidade Federal do Pará (UFPA), Programa de Pós-graduação em Inovação Farmacêutica (PPGIF), Belém, PA, Brazil
| | | | | | | | - Maria Fani Dolabela
- Universidade Federal do Pará (UFPA), Programa de Pós-graduação em Inovação Farmacêutica (PPGIF), Belém, PA, Brazil
| | - Heliton Patrick Cordovil Brígido
- Universidade Federal do Pará (UFPA), Programa de Pós-graduação em Inovação Farmacêutica (PPGIF), Belém, PA, Brazil; Centro Universitário Metropolitano da Amazônia (UNIFAMAZ), Curso de Medicina, Belém, PA, Brazil.
| |
Collapse
|
24
|
Luqman A, Saising J, Prasetya YA, Ammanath AV, Andini, Amala SN, Zulaika E, Kuswytasari ND, Goetz F, Wibowo AT. Detection of Vancomycin Resistant Genes in Intrinsically Antibiotic Resistant Bacteria from the Gut Microbiota of Indonesian Individuals. IRANIAN JOURNAL OF MEDICAL SCIENCES 2024; 49:302-312. [PMID: 38751872 PMCID: PMC11091268 DOI: 10.30476/ijms.2023.98767.3087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/27/2023] [Accepted: 07/14/2023] [Indexed: 05/18/2024]
Abstract
Background Antibiotic resistance is a global public health concern that has been exacerbated by the overuse and misuse of antibiotics, leading to the emergence of resistant bacteria. The gut microbiota, often influenced by antibiotic usage, plays a crucial role in overall health. Therefore, this study aimed to investigate the prevalence of antibiotic resistant genes in the gut microbiota of Indonesian coastal and highland populations, as well as to identify vancomycin-resistant bacteria and their resistant genes. Methods Stool samples were collected from 22 individuals residing in Pacet, Mojokerto, and Kenjeran, Surabaya Indonesia in 2022. The read count of antibiotic resistant genes was analyzed in the collected samples, and the bacterium concentration was counted by plating on the antibiotic-containing agar plate. Vancomycin-resistant strains were further isolated, and the presence of vancomycin-resistant genes was detected using a multiplex polymerase chain reaction (PCR). Results The antibiotic resistant genes for tetracycline, aminoglycosides, macrolides, beta-lactams, and vancomycin were found in high frequency in all stool samples (100%) of the gut microbiota. Meanwhile, those meant for chloramphenicol and sulfonamides were found in 86% and 16% of the samples, respectively. Notably, vancomycin-resistant genes were found in 16 intrinsically resistant Gram-negative bacterial strains. Among the detected vancomycin-resistant genes, vanG was the most prevalent (27.3%), while vanA was the least prevalent (4.5%). Conclusion The presence of multiple vancomycin resistance genes in intrinsically resistant Gram-negative bacterial strains demonstrated the importance of the gut microbiota as a reservoir and hub for the horizontal transfer of antibiotic resistant genes.
Collapse
Affiliation(s)
- Arif Luqman
- Department of Biology, Institute Teknologi Sepuluh Nopember, Surabaya, Indonesia
- Institute for Molecular Infection Biology (IMIB), Julius Maximilians University of Wuerzburg, Wuerzburg, Germany
| | - Jongkon Saising
- School of Health Science and Biomedical Technology Research Group for Vulnerable Populations, Mae Fah Luang University, Chiang Rai, Thailand
| | | | | | - Andini
- Department of Biology, Institute Teknologi Sepuluh Nopember, Surabaya, Indonesia
| | - Siti Nur Amala
- Department of Biology, Institute Teknologi Sepuluh Nopember, Surabaya, Indonesia
| | - Enny Zulaika
- Department of Biology, Institute Teknologi Sepuluh Nopember, Surabaya, Indonesia
| | | | - Friedrich Goetz
- Microbial Genetics, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Anjar Tri Wibowo
- Department of Biology, Faculty of Science and Technology, Airlangga University, Surabaya, Indonesia
| |
Collapse
|
25
|
Mediati DG, Dan W, Lalaouna D, Dinh H, Pokhrel A, Rowell KN, Michie KA, Stinear TP, Cain AK, Tree JJ. The 3' UTR of vigR is required for virulence in Staphylococcus aureus and has expanded through STAR sequence repeat insertions. Cell Rep 2024; 43:114082. [PMID: 38583155 DOI: 10.1016/j.celrep.2024.114082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 01/17/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are alarmingly common, and treatment is confined to last-line antibiotics. Vancomycin is the treatment of choice for MRSA bacteremia, and treatment failure is often associated with vancomycin-intermediate S. aureus isolates. The regulatory 3' UTR of the vigR mRNA contributes to vancomycin tolerance and upregulates the autolysin IsaA. Using MS2-affinity purification coupled with RNA sequencing, we find that the vigR 3' UTR also regulates dapE, a succinyl-diaminopimelate desuccinylase required for lysine and peptidoglycan synthesis, suggesting a broader role in controlling cell wall metabolism and vancomycin tolerance. Deletion of the 3' UTR increased virulence, while the isaA mutant is completely attenuated in a wax moth larvae model. Sequence and structural analyses of vigR indicated that the 3' UTR has expanded through the acquisition of Staphylococcus aureus repeat insertions that contribute sequence for the isaA interaction seed and may functionalize the 3' UTR.
Collapse
Affiliation(s)
- Daniel G Mediati
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia; Australian Institute for Microbiology and Infection, University of Technology Sydney, Ultimo, NSW, Australia.
| | - William Dan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - David Lalaouna
- Université de Strasbourg, CNRS, ARN UPR 9002, Strasbourg, France
| | - Hue Dinh
- School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Alaska Pokhrel
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Ultimo, NSW, Australia; School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Keiran N Rowell
- Structural Biology Facility, University of New South Wales, Sydney, NSW, Australia
| | - Katharine A Michie
- Structural Biology Facility, University of New South Wales, Sydney, NSW, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Amy K Cain
- School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Jai J Tree
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
26
|
Wu W, Pang CNI, Mediati DG, Tree JJ. The functional small RNA interactome reveals targets for the vancomycin-responsive sRNA RsaOI in vancomycin-tolerant Staphylococcus aureus. mSystems 2024; 9:e0097123. [PMID: 38534138 PMCID: PMC11019875 DOI: 10.1128/msystems.00971-23] [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: 09/11/2023] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Small RNAs have been found to control a broad range of bacterial phenotypes including tolerance to antibiotics. Vancomycin tolerance in multidrug resistance Staphylococcus aureus is correlated with dysregulation of small RNAs although their contribution to antibiotic tolerance is poorly understood. RNA-RNA interactome profiling techniques are expanding our understanding of sRNA-mRNA interactions in bacteria; however, determining the function of these interactions for hundreds of sRNA-mRNA pairs is a major challenge. At steady-state, protein and mRNA abundances are often highly correlated and lower than expected protein abundance may indicate translational repression of an mRNA. To identify sRNA-mRNA interactions that regulate mRNA translation, we examined the correlation between gene transcript abundance, ribosome occupancy, and protein levels. We used the machine learning technique self-organizing maps (SOMs) to cluster genes with similar transcription and translation patterns and identified a cluster of mRNAs that appeared to be post-transcriptionally repressed. By integrating our clustering with sRNA-mRNA interactome data generated in vancomycin-tolerant S. aureus by RNase III-CLASH, we identified sRNAs that may be mediating translational repression. We have confirmed sRNA-dependant post-transcriptional repression of several mRNAs in this cluster. Two of these interactions are mediated by RsaOI, a sRNA that is highly upregulated by vancomycin. We demonstrate the regulation of HPr and the cell-wall autolysin Atl. These findings suggest that RsaOI coordinates carbon metabolism and cell wall turnover during vancomycin treatment. IMPORTANCE The emergence of multidrug-resistant Staphylococcus aureus (MRSA) is a major public health concern. Current treatment is dependent on the efficacy of last-line antibiotics like vancomycin. The most common cause of vancomycin treatment failure is strains with intermediate resistance or tolerance that arise through the acqusition of a diverse repertoire of point mutations. These strains have been shown to altered small RNA (sRNA) expression in response to antibiotic treatment. Here, we have used a technique termed RNase III-CLASH to capture sRNA interactions with their target mRNAs. To understand the function of these interactions, we have looked at RNA and protein abundance for mRNAs targeted by sRNAs. Messenger RNA and protein levels are generally well correlated and we use deviations from this correlation to infer post-transcriptional regulation and the function of individual sRNA-mRNA interactions. Using this approach we identify mRNA targets of the vancomycin-induced sRNA, RsaOI, that are repressed at the translational level. We find that RsaOI represses the cell wall autolysis Atl and carbon transporter HPr suggestion a link between vancomycin treatment and suppression of cell wall turnover and carbon metabolism.
Collapse
Affiliation(s)
- Winton Wu
- School of Biotechnology and Biomolecular Sciences, Sydney, New South Wales, Australia
| | | | - Daniel G. Mediati
- School of Biotechnology and Biomolecular Sciences, Sydney, New South Wales, Australia
| | - Jai Justin Tree
- School of Biotechnology and Biomolecular Sciences, Sydney, New South Wales, Australia
| |
Collapse
|
27
|
van Hal SJ, Jensen SO, Tong SYC, Bentley S, Holden MT. Unravelling the complex interplay between antibiotic consumption and adaptive changes in methicillin-resistant Staphylococcus aureus. J Antimicrob Chemother 2024; 79:891-896. [PMID: 38412336 DOI: 10.1093/jac/dkae048] [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: 10/19/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
OBJECTIVES This study aims to elucidate the genomic dynamics driving the emergence of antimicrobial resistance (AMR), with a specific focus on the interplay between AMR and antimicrobial usage. METHODS We conducted a comprehensive analysis using a ST239 methicillin-resistant Staphylococcus aureus (MRSA) dataset over a continuous 12-year period from a single hospital. Genomic analyses were performed tracking the changes in MRSA populations, particularly the emergence of reduced vancomycin susceptibility, and assessing the impact of glycopeptide use on these emergence events. RESULTS Our findings reveal a significant correlation between hospital glycopeptide usage and the selection of MRSA strains with reduced vancomycin susceptibility. Genomic analyses provided insights into the molecular mechanisms driving resistance emergence, including the slowing of the molecular clock rate in response to heightened antimicrobial consumption. CONCLUSIONS In conclusion, this study the highlights the complex dynamics between AMR and antimicrobial use at the hospital level. The observed correlation between antimicrobial consumption and the development of less susceptible MRSA strains underscores the importance of antimicrobial stewardship programmes and the establishment of optimal consumption thresholds for mitigating AMR effectively.
Collapse
Affiliation(s)
- Sebastiaan J van Hal
- Department of Microbiology and Infectious Diseases, Royal Prince Alfred Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Antimicrobial Resistance and Mobile Elements Group, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Slade O Jensen
- Antimicrobial Resistance and Mobile Elements Group, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- Microbiology and Infectious Diseases, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Stephen Y C Tong
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephen Bentley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Matthew T Holden
- School of Medicine, University of St Andrews, St Andrews, Fife KY16 9TF, UK
| |
Collapse
|
28
|
Zhao R, Du B, Luo Y, Xue F, Wang H, Qu D, Han S, Heilbronner S, Zhao Y. Antimicrobial and anti-biofilm activity of a thiazolidinone derivative against Staphylococcus aureus in vitro and in vivo. Microbiol Spectr 2024; 12:e0232723. [PMID: 38329365 PMCID: PMC10913468 DOI: 10.1128/spectrum.02327-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 01/11/2024] [Indexed: 02/09/2024] Open
Abstract
Staphylococcus aureus (S. aureus) causes many infections with significant morbidity and mortality. S. aureus can form biofilms, which can cause biofilm-associated diseases and increase resistance to many conventional antibiotics, resulting in chronic infection. It is critical to develop novel antibiotics against staphylococcal infections, particularly those that can kill cells embedded in biofilms. This study aimed to investigate the bacteriocidal and anti-biofilm activities of thiazolidinone derivative (TD-H2-A) against S. aureus. A total of 40 non-duplicate strains were collected, and the minimum inhibitory concentrations (MICs) of TD-H2-A were determined. The effect of TD-H2-A on established S. aureus mature biofilms was examined using a confocal laser scanning microscope (CLSM). The antibacterial effects of the compound on planktonic bacteria and bacteria in mature biofilms were investigated. Other characteristics, such as cytotoxicity and hemolytic activity, were researched. A mouse skin infection model was used, and a routine hematoxylin and eosin (H&E) staining was used for histological examination. The MIC values of TD-H2-A against the different S. aureus strains were 6.3-25.0 µg/mL. The 5 × MIC TD-H2-A killed almost all planktonic S. aureus USA300. The derivative was found to have strong bacteriocidal activity against cells in mature biofilms meanwhile having low cytotoxicity and hemolytic activity against Vero cells and human erythrocytes. TD-H2-A had a good bacteriocidal effect on S. aureus SA113-infected mice. In conclusion, TD-H2-A demonstrated good bacteriocidal and anti-biofilm activities against S. aureus, paving the way for the development of novel agents to combat biofilm infections and multidrug-resistant staphylococcal infections.IMPORTANCEStaphylococcus aureus, a notorious pathogen, can form a stubborn biofilm and develop drug resistance. It is crucial to develop new anti-infective therapies against biofilm-associated infections. The manuscript describes the new antibiotic to effectively combat multidrug-resistant and biofilm-associated diseases.
Collapse
Affiliation(s)
- Rui Zhao
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Bingyu Du
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Luo
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Fen Xue
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Huanhuan Wang
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Di Qu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shiqing Han
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Simon Heilbronner
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Yanfeng Zhao
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
29
|
Wagner TM, Pöntinen AK, Al Rubaye M, Sundsfjord A, Hegstad K. Adaptive cell wall thickening in Enterococcus faecalis is associated with decreased vancomycin susceptibility. Clin Microbiol Infect 2024; 30:396.e1-396.e5. [PMID: 38065364 DOI: 10.1016/j.cmi.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/25/2023]
Abstract
OBJECTIVES Enterococcus faecalis can adopt both a commensal and a nosocomial lifestyle, resisting numerous antibiotics. In this study, we aim to investigate the relationship between the cell wall (CW) thickness and decreased susceptibility to vancomycin (VD) in van-gene negative clinical isolates of E. faecalis (nMIC 8 = 2, nMIC 4 = 3, ST30, ST40, and ST59). METHODS The CW thickness was assessed in VD strains and compared with vancomycin susceptible isolates of the same sequence type (ST) (Vancomycin susceptible [VS]; nMIC 2 = 5). The VD and VS strains were subjected to serial passage (evolved [ev]) with and without vancomycin selection. Subsequent measurements of CW thickness and vancomycin MICs were performed. RESULTS The VD strains exhibited increased CW thickness when compared with ST-related VS strains (ΔCW thickness VD vs. VS ST30 25 nm, ST59 15 nm, and ST40 1 nm). Serial passages without vancomycin selection led to a decrease in CW thickness and vancomycin MIC in VD strains (ΔCW thickness VD vs. evVD ST30 22 nm, ST59 3 nm, and ST40 2 nm). Serial passages with vancomycin selection caused an increase in CW thickness and vancomycin MIC in ST-related VS strains (ΔCW thickness VS vs. evVS ST30 22 nm, ST59 16 nm, and ST40 1 nm). DISCUSSION Adaptive changes in CW thickness were observed in response to vancomycin exposure. Increased CW thickness correlated with decreased vancomycin susceptibility, whereas decreased CW thickness correlated with increased vancomycin susceptibility. Core single nucleotide polymorphisms in the evolved mutants were mostly found in genes encoding proteins associated with the cytoplasm or the cytoplasmic membrane. The potential relevance of these adaptive changes is underlined by the observed phenotypes in clinical isolates. Our findings emphasize the importance of monitoring adaptive changes, as vancomycin-resistant enterococci infections are a growing concern.
Collapse
Affiliation(s)
- Theresa Maria Wagner
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Anna K Pöntinen
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway; Department of Biostatistics, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mushtaq Al Rubaye
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Arnfinn Sundsfjord
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway; Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Kristin Hegstad
- Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway; Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway.
| |
Collapse
|
30
|
Gulone L, Di Gregorio S, Morales M, Haim MS, García S, Perazzi B, Famiglietti A, Mollerach M. The Changing Epidemiology and Antimicrobial Susceptibility of Staphylococcus aureus Isolated from Blood Cultures in a University Hospital from Argentina. Microb Drug Resist 2024; 30:109-117. [PMID: 38133499 DOI: 10.1089/mdr.2023.0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Staphylococcus aureus bacteremia (SAB) is one of the most common serious bacterial infections worldwide. In this study, we demonstrated changes in SAB epidemiology in an Argentinean University Hospital during an 8-year period (2009-2016). A total of 326 S. aureus clinical isolates were recovered in three periods: P1: 2009-2010, P2: 2012-2014, and P3: 2015-2016. Among these, 127 were methicillin-resistant S. aureus (MRSA) and were characterized by phenotypic and molecular methods. We hereby report a significant decline in multiple drug resistance among MRSA isolates associated with an increase in SCCmec IV between the three periods. A diversity of MRSA-IV clones (mainly ST30-MRSA-IV, ST5-MRSA-IV, and ST8-MRSA-IV) replaced between 2009 and 2016 the previous prevalent MRSA clone causing bloodstream infections at this hospital (ST5-MRSA-I). MRSA population structure continued to diversify between P2 and P3. Notably, ST8-MRSA-IV-t008 related to USA300 was first detected during P2, and ST8-MRSA-IV together with ST30-MRSA-IV related to the Southwest Pacific clone were the more prevalent MRSA genotypes circulating during P3.
Collapse
Affiliation(s)
- Lucía Gulone
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Sabrina Di Gregorio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Maia Morales
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Sol Haim
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Susana García
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Beatriz Perazzi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Angela Famiglietti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Hospital de Clínicas José de San Martín, Laboratorio de Bacteriología Clínica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Marta Mollerach
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
31
|
Crozier D, Gray JM, Maltas JA, Bonomo RA, Burke ZDC, Card KJ, Scott JG. The evolution of diverse antimicrobial responses in vancomycin-intermediate Staphylococcus aureus and its therapeutic implications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.30.569373. [PMID: 38077036 PMCID: PMC10705500 DOI: 10.1101/2023.11.30.569373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Staphylococcus aureus causes endocarditis, osteomyelitis, and bacteremia. Clinicians often prescribe vancomycin as an empiric therapy to account for methicillin-resistant S. aureus (MRSA) and narrow treatment based on culture susceptibility results. However, these results reflect a single time point before empiric treatment and represent a limited subset of the total bacterial population within the patient. Thus, while they may indicate that the infection is susceptible to a particular drug, this recommendation may no longer be accurate during therapy. Here, we addressed how antibiotic susceptibility changes over time by accounting for evolution. We evolved 18 methicillin-susceptible S. aureus (MSSA) populations under increasing vancomycin concentrations until they reached intermediate resistance levels. Sequencing revealed parallel mutations that affect cell membrane stress response and cell-wall biosynthesis. The populations exhibited repeated cross-resistance to daptomycin and varied responses to meropenem, gentamicin, and nafcillin. We accounted for this variability by deriving likelihood estimates that express a population's probability of exhibiting a drug response following vancomycin treatment. Our results suggest antistaphylococcal penicillins are preferable first-line treatments for MSSA infections but also highlight the inherent uncertainty that evolution poses to effective therapies. Infections may take varied evolutionary paths; therefore, considering evolution as a probabilistic process should inform our therapeutic choices.
Collapse
|
32
|
Chagas RA, Silva CDS, de Cássia Teixeira Birro J, Dos Santos KV. Effects of Vancomycin Subtherapeutic Concentration on Staphylococcus aureus Isolated from Hemodialysis Patients with Low Serum Trough Concentrations. Curr Microbiol 2024; 81:65. [PMID: 38231442 DOI: 10.1007/s00284-023-03588-3] [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: 08/14/2023] [Accepted: 12/12/2023] [Indexed: 01/18/2024]
Abstract
Blood bactericidal activity and antimicrobial therapy are crucial against catheter-related infection in patients undergoing hemodialysis (HD). It is well-known that catheters colonized by biofilm-producing bacteria are a risk factor for bacteremia in HD-patients. Methicillin-resistant S. aureus bacteremia in HD-patients justify the use of vancomycin as empiric therapy. The recommended vancomycin target for therapeutic efficacy is a minimum serum concentration of 10 µg mL-1 to avoid resistance. However, subtherapeutic concentrations of vancomycin have frequently occurred in HD-patients. Thus, we aim to investigate the effect of subtherapeutic vancomycin concentration on S. aureus growth, susceptibility to antimicrobials, resistance to whole blood activity, and biofilm formation. Seventeen S. aureus strains isolated from bacteremia in HD-patients and two reference strains were exposed to a vancomycin-gradient (0-10 µg mL-1) for five consecutive days to mimic the dosing interval of vancomycin in HD-patients. After that, we observed the following: no effect on growth curve; decreased susceptibility to vancomycin and daptomycin; increased S. aureus survival to whole blood bactericidal action; and a strain-dependent biofilm production after drug exposure. In conclusion, our findings suggest that the subtherapeutic concentration of vancomycin decrease S. aureus susceptibility to vancomycin and daptomycin and increases its survival to whole blood bactericidal action.
Collapse
Affiliation(s)
- Rodrigo Altoé Chagas
- Department of Pathology, Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Cristiana de Souza Silva
- Department of Pathology, Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | | | - Kênia Valéria Dos Santos
- Department of Pathology, Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil.
| |
Collapse
|
33
|
Kohl MP, Chane-Woon-Ming B, Bahena-Ceron R, Jaramillo-Ponce J, Antoine L, Herrgott L, Romby P, Marzi S. Ribosome Profiling Methods Adapted to the Study of RNA-Dependent Translation Regulation in Staphylococcus aureus. Methods Mol Biol 2024; 2741:73-100. [PMID: 38217649 DOI: 10.1007/978-1-0716-3565-0_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] [Indexed: 01/15/2024]
Abstract
Noncoding RNAs, including regulatory RNAs (sRNAs), are instrumental in regulating gene expression in pathogenic bacteria, allowing them to adapt to various stresses encountered in their host environments. Staphylococcus aureus is a well-studied model for RNA-mediated regulation of virulence and pathogenicity, with sRNAs playing significant roles in shaping S. aureus interactions with human and animal hosts. By modulating the translation and/or stability of target mRNAs, sRNAs regulate the synthesis of virulence factors and regulatory proteins required for pathogenesis. Moreover, perturbation of the levels of RNA modifications in two other classes of noncoding RNAs, rRNAs, and tRNAs, has been proposed to contribute to stress adaptation. However, the study of how these various factors affect translation regulation has often been restricted to specific genes, using in vivo reporters and/or in vitro translation systems. Genome-wide sequencing approaches offer novel perspectives for studying RNA-dependent regulation. In particular, ribosome profiling methods provide a powerful resource for characterizing the overall landscape of translational regulation, contributing to a better understanding of S. aureus physiopathology. Here, we describe protocols that we have adapted to perform ribosome profiling in S. aureus.
Collapse
Affiliation(s)
- Maximilian P Kohl
- Architecture et Réactivité de l'ARN, CNRS 9002, Université de Strasbourg, Strasbourg, France
| | | | - Roberto Bahena-Ceron
- Architecture et Réactivité de l'ARN, CNRS 9002, Université de Strasbourg, Strasbourg, France
| | - Jose Jaramillo-Ponce
- Architecture et Réactivité de l'ARN, CNRS 9002, Université de Strasbourg, Strasbourg, France
| | - Laura Antoine
- Architecture et Réactivité de l'ARN, CNRS 9002, Université de Strasbourg, Strasbourg, France
| | - Lucas Herrgott
- Architecture et Réactivité de l'ARN, CNRS 9002, Université de Strasbourg, Strasbourg, France
| | - Pascale Romby
- Architecture et Réactivité de l'ARN, CNRS 9002, Université de Strasbourg, Strasbourg, France
| | - Stefano Marzi
- Architecture et Réactivité de l'ARN, CNRS 9002, Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
34
|
Jiang JH, Cameron DR, Nethercott C, Aires-de-Sousa M, Peleg AY. Virulence attributes of successful methicillin-resistant Staphylococcus aureus lineages. Clin Microbiol Rev 2023; 36:e0014822. [PMID: 37982596 PMCID: PMC10732075 DOI: 10.1128/cmr.00148-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of severe and often fatal infections. MRSA epidemics have occurred in waves, whereby a previously successful lineage has been replaced by a more fit and better adapted lineage. Selection pressures in both hospital and community settings are not uniform across the globe, which has resulted in geographically distinct epidemiology. This review focuses on the mechanisms that trigger the establishment and maintenance of current, dominant MRSA lineages across the globe. While the important role of antibiotic resistance will be mentioned throughout, factors which influence the capacity of S. aureus to colonize and cause disease within a host will be the primary focus of this review. We show that while MRSA possesses a diverse arsenal of toxins including alpha-toxin, the success of a lineage involves more than just producing toxins that damage the host. Success is often attributed to the acquisition or loss of genetic elements involved in colonization and niche adaptation such as the arginine catabolic mobile element, as well as the activity of regulatory systems, and shift metabolism accordingly (e.g., the accessory genome regulator, agr). Understanding exactly how specific MRSA clones cause prolonged epidemics may reveal targets for therapies, whereby both core (e.g., the alpha toxin) and acquired virulence factors (e.g., the Panton-Valentine leukocidin) may be nullified using anti-virulence strategies.
Collapse
Affiliation(s)
- Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - David R Cameron
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Cara Nethercott
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Marta Aires-de-Sousa
- Laboratory of Molecular Genetics, Institutode Tecnologia Químicae Biológica António Xavier (ITQB-NOVA), Universidade Nova de Lisboa, Oeiras, Portugal
- Escola Superior de Saúde da Cruz Vermelha Portuguesa-Lisboa (ESSCVP-Lisboa), Lisbon, Portugal
| | - Anton Y Peleg
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Kim YH, Park J, Chung HS. Genetic characterization of tetracycline-resistant Staphylococcus aureus with reduced vancomycin susceptibility using whole-genome sequencing. Arch Microbiol 2023; 206:24. [PMID: 38103051 DOI: 10.1007/s00203-023-03760-0] [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: 07/21/2023] [Revised: 11/09/2023] [Accepted: 11/19/2023] [Indexed: 12/17/2023]
Abstract
This study aimed to analyze the genetic characteristics of Staphylococcus aureus with reduced vancomycin susceptibility (RVS-SA). Whole-genome sequencing was performed on 27 RVS-SA clinical isolates, and comparative genomic analysis was performed using S. aureus reference strains. Pan-genome orthologous groups (POGs) were identified that were present in RVS-SA but absent in the reference strains, but further analysis showed that the presence of these POGs was influenced by tetracycline resistance rather than vancomycin resistance. Therefore, we restricted our analysis to tetracycline-resistant (tetR) RVS-SA and tetR vancomycin-susceptible S. aureus (VSSA). Phylogenomic analysis showed them to be closely related, and further analysis revealed the presence of an uncharacterized protein SAB0394 and the absence of lytA in tetR RVS-SA, which are involved in cell wall thickening. In summary, using whole-genome sequencing we identified gain or loss of genes in tetR RVS-SA strains. These findings provide insights into the investigation of mechanisms associated with reduced vancomycin susceptibility and have the potential to contribute to the development of molecular biomarkers for the rapid and efficient detection of RVS-SA.
Collapse
Affiliation(s)
- Yu-Hee Kim
- Advanced Biomedical Research Institute, Ewha Womans University Seoul Hospital, Seoul, Korea
- Ewha Education and Research Center for Infection, Ewha Womans University Medical Center, Seoul, Korea
| | - Junghun Park
- Ewha Education and Research Center for Infection, Ewha Womans University Medical Center, Seoul, Korea
| | - Hae-Sun Chung
- Ewha Education and Research Center for Infection, Ewha Womans University Medical Center, Seoul, Korea.
- Department of Laboratory Medicine, Ewha Womans University College of Medicine, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea.
| |
Collapse
|
36
|
Douglas EA, Marshall B, Alghamadi A, Joseph EA, Duggan S, Vittorio S, De Luca L, Serpi M, Laabei M. Improved Antibacterial Activity of 1,3,4-Oxadiazole-Based Compounds That Restrict Staphylococcus aureus Growth Independent of LtaS Function. ACS Infect Dis 2023; 9:2141-2159. [PMID: 37828912 PMCID: PMC10644342 DOI: 10.1021/acsinfecdis.3c00250] [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: 05/31/2023] [Indexed: 10/14/2023]
Abstract
The lipoteichoic acid (LTA) biosynthesis pathway has emerged as a promising antimicrobial therapeutic target. Previous studies identified the 1,3,4 oxadiazole compound 1771 as an LTA inhibitor with activity against Gram-positive pathogens. We have succeeded in making six 1771 derivatives and, through subsequent hit validation, identified the incorporation of a pentafluorosulfanyl substituent as central in enhancing activity. Our newly described derivative, compound 13, showed a 16- to 32-fold increase in activity compared to 1771 when tested against a cohort of multidrug-resistant Staphylococcus aureus strains while simultaneously exhibiting an improved toxicity profile against mammalian cells. Molecular techniques were employed in which the assumed target, lipoteichoic acid synthase (LtaS), was both deleted and overexpressed. Neither deletion nor overexpression of LtaS altered 1771 or compound 13 susceptibility; however, overexpression of LtaS increased the MIC of Congo red, a previously identified LtaS inhibitor. These data were further supported by comparing the docking poses of 1771 and derivatives in the LtaS active site, which indicated the possibility of an additional target(s). Finally, we show that both 1771 and compound 13 have activity that is independent of LtaS, extending to cover Gram-negative species if the outer membrane is first permeabilized, challenging the classification that these compounds are strict LtaS inhibitors.
Collapse
Affiliation(s)
| | - Brandon Marshall
- School
of Chemistry, Cardiff University, Cardiff CF10 3AT, Wales, U.K.
| | - Arwa Alghamadi
- School
of Chemistry, Cardiff University, Cardiff CF10 3AT, Wales, U.K.
| | - Erin A. Joseph
- School
of Chemistry, Cardiff University, Cardiff CF10 3AT, Wales, U.K.
| | - Seána Duggan
- Medical
Research Council Centre for Medical Mycology at the University of
Exeter, University of Exeter, Exeter EX4 4DQ, U.K.
| | - Serena Vittorio
- Department
of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina I-98125, Italy
| | - Laura De Luca
- Department
of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina I-98125, Italy
| | - Michaela Serpi
- School
of Chemistry, Cardiff University, Cardiff CF10 3AT, Wales, U.K.
| | - Maisem Laabei
- Department
of Life Sciences, University of Bath, Bath BA2 7AY, U.K.
| |
Collapse
|
37
|
Szekat C, Josten M, Rickmeyer J, Crüsemann M, Bierbaum G. A Staphylococcus capitis strain with unusual bacteriocin production. Microb Biotechnol 2023; 16:2181-2193. [PMID: 37850940 PMCID: PMC10616647 DOI: 10.1111/1751-7915.14356] [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: 07/21/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023] Open
Abstract
Staphylococcus capitis is a member of the human and mammal skin microbiomes and is considered less harmful than Staphylococcus aureus. S. capitis subsp. urealyticus BN2 was isolated from a cat and expressed strong antibacterial activity against a range of Gram-positive species, most notably including S. aureus strains with resistance to methicillin (MRSA) and strains with intermediate resistance to vancomycin (VISA). These latter strains are normally relatively resistant to bacteriocins, due to cell wall and cell membrane modifications. Genomic sequencing showed that the strain harboured at least two complete gene clusters for biosynthesis of antagonistic substances. The complete biosynthetic gene cluster of the well-known lantibiotic gallidermin was encoded on a large plasmid and the mature peptide was present in isopropanol cell extracts. In addition, a chromosomal island contained a novel non-ribosomal peptide synthetase (NRPS) gene cluster. Accidental deletion of two NRPS modules and partial purification of the anti-VISA activity showed that this novel bacteriocin represents a complex of differently decorated, non-ribosomal peptides. Additionally, a number of phenol-soluble modulins (PSMs) was detected by mass spectrometry of whole cells. Producing these compounds, the strain was able to outcompete several S. aureus strains, including MRSA and VISA, in tube cultures.
Collapse
Affiliation(s)
- Christiane Szekat
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Michaele Josten
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Jasmin Rickmeyer
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Max Crüsemann
- Institute of Pharmaceutical BiologyUniversity of BonnBonnGermany
| | - Gabriele Bierbaum
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
38
|
Sreejisha M, Shenoy MS, Shenoy MS, Dhanashree B, Chakrapani M, Bhat KG. Molecular and Clinical Features of Heterogeneous Vancomycin-Intermediate Staphylococcus aureus in Tertiary Care Hospitals in South India. Sultan Qaboos Univ Med J 2023; 23:447-454. [PMID: 38090245 PMCID: PMC10712385 DOI: 10.18295/squmj.3.2023.018] [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: 01/03/2023] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
Objectives This study aimed to detect heterogeneous vancomycin-intermediate Staphylococcus aureus (hVISA) among methicillin-resistant S. aureus (MRSA) isolated from healthcare-associated infections and identify staphylococcal cassette chromosome mec (SCCmec) types. Methods This study was conducted from February 2019 to March 2020 and included patients admitted in 4 tertiary care hospitals in Karnataka, India. Isolation and identification of MRSA were done using standard bacteriological methods. Antimicrobial susceptibility testing was done using Kirby-Bauer disc diffusion; macrolide-lincosamide-streptogramin B phenotypes were identified using the D test. The minimum inhibitory concentration (MIC) of vancomycin was determined using agar dilution. hVISA were confirmed by the modified population analysis profile-area under the curve test. SCCmec types and the Panton-Valentine leukocidin (pvl) gene were detected using multiplex polymerase chain reaction. Results Of 220 MRSA stains, 14 (6.4%) were hVISA. None of the MRSA isolates was vancomycin-intermediate or -resistant and all hVISA were susceptible to linezolid and teicoplanin. The macrolide-streptogramin B phenotype was present in 42.9% of hVISA; 92.9% of the hVISA strains had vancomycin MIC in the range of 1-2 μg/mL. Majority of the hVISA and vancomycin-susceptible MRSA were isolated from patients with skin and soft tissue infections. SCCmec III and IV were present in 50% and 35.7% of hVISA, respectively; 14.3% of the hVISA harboured SCCmec V. Conclusion The prevalence rate of hVISA among MRSA was 6.4%. Therefore, MRSA strains should be tested for hVISA before starting vancomycin treatment. None of the isolates was vancomycin-intermediate or -resistant and all the hVISA strains were susceptible to linezolid and teicoplanin. The majority of the hVISA were isolated from patients with skin and soft tissue infections and harboured SCCmec III and IV.
Collapse
Affiliation(s)
- M. Sreejisha
- Department of Microbiology, Kasturba Medical College, Mangalore, (A constituent unit of Manipal Academy of Higher Education, Manipal), Karnataka, India
| | - M. Shalini Shenoy
- Department of Microbiology, Kasturba Medical College, Mangalore, (A constituent unit of Manipal Academy of Higher Education, Manipal), Karnataka, India
| | - M. Suchitra Shenoy
- Department of Microbiology, Kasturba Medical College, Mangalore, (A constituent unit of Manipal Academy of Higher Education, Manipal), Karnataka, India
| | - B. Dhanashree
- Department of Microbiology, Kasturba Medical College, Mangalore, (A constituent unit of Manipal Academy of Higher Education, Manipal), Karnataka, India
| | - M. Chakrapani
- Department of Medicine, Kasturba Medical College, Mangalore, (A constituent unit of Manipal Academy of Higher Education, Manipal), Karnataka, India
| | - K. Gopalakrishna Bhat
- Department of Microbiology, Kasturba Medical College, Mangalore, (A constituent unit of Manipal Academy of Higher Education, Manipal), Karnataka, India
| |
Collapse
|
39
|
Giulieri SG. Case Commentary: The hidden side of oxacillin resistance in Staphylococcus aureus. Antimicrob Agents Chemother 2023; 67:e0071623. [PMID: 37655923 PMCID: PMC10583679 DOI: 10.1128/aac.00716-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
Acquisition of PBP2a (encoded by the mec gene) is the key resistance mechanism to β-lactams in Staphylococcus aureus. The mec gene can be easily detected by PCR assays; however, these tools will miss mec-independent oxacillin resistance. This phenotype is mediated by mutations in cell wall metabolism genes that can be acquired during persistent infections under prolonged antibiotic exposure. The complex case presented by Hess et al. (Antimicrob Agents Chemother 67:e00437-23, 2023, https://doi.org/10.1128/aac.00437-23) highlights the diagnostic and therapeutic challenges in the management of mec-independent oxacillin resistance.
Collapse
Affiliation(s)
- Stefano G. Giulieri
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Disease Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, Austin Health, Heidelberg, Australia
| |
Collapse
|
40
|
Habib G, Gul H, Ahmad P, Hayat A, Rehman MU, Mohamed Moussa I, Elansary HO. Teicoplanin associated gene tcaA inactivation increases persister cell formation in Staphylococcus aureus. Front Microbiol 2023; 14:1241995. [PMID: 37901830 PMCID: PMC10611510 DOI: 10.3389/fmicb.2023.1241995] [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: 06/18/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Staphylococcus aureus is part of normal human flora and is widely associated with hospital-acquired bacteremia. S. aureus has shown a diverse array of resistance to environmental stresses and antibiotics. Methicillin-resistant S. aureus (MRSA) is on the high priority list of new antibiotics discovery and glycopeptides are considered the last drug of choice against MRSA. S. aureus has developed resistance against glycopeptides and the emergence of vancomycin-intermediate-resistant, vancomycin-resistant, and teicoplanin-resistant strains is globally reported. Teicoplanin-associated genes tcaR-tcaA-tcaB (tcaRAB) is known as the S. aureus glycopeptide resistance operon that is associated with glycopeptide resistance. Here, for the first time, the role of tcaRAB in S. aureus persister cells formation, and ΔtcaA dependent persisters' ability to resuscitate the bacterial population was explored. We recovered a clinical strain of MRSA from a COVID-19 patient which showed a high level of resistance to teicoplanin, vancomycin, and methicillin. Whole genome RNA sequencing revealed that the tcaRAB operon expression was altered followed by high expression of glyS and sgtB. The RNA-seq data revealed a significant decrease in tcaA (p = 0.008) and tcaB (p = 0.04) expression while tcaR was not significantly altered. We knocked down tcaA, tcaB, and tcaR using CRISPR-dCas9 and the results showed that when tcaA was suppressed by dCas9, a significant increase was witnessed in persister cells while tcaB suppression did not induce persistence. The results were further evaluated by creating a tcaA mutant that showed ΔtcaA formed a significant increase in persisters in comparison to the wild type. Based on our findings, we concluded that tcaA is the gene that increases persister cells and glycopeptide resistance and could be a potential therapeutic target in S. aureus.
Collapse
Affiliation(s)
- Gul Habib
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Haji Gul
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Faculty of Veterinary and Animal Sciences, Gomal University, Dera Ismail Khan, Pakistan
| | - Prevez Ahmad
- Department of Physics, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Azam Hayat
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Mujaddad Ur Rehman
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Ihab Mohamed Moussa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hosam O. Elansary
- Plant Production Department, College of Food & Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
41
|
Kim YK, Eom Y, Kim E, Chang E, Bae S, Jung J, Kim MJ, Chong YP, Kim SH, Choi SH, Lee SO, Kim YS. Molecular Characteristics and Prevalence of Rifampin Resistance in Staphylococcus aureus Isolates from Patients with Bacteremia in South Korea. Antibiotics (Basel) 2023; 12:1511. [PMID: 37887212 PMCID: PMC10604019 DOI: 10.3390/antibiotics12101511] [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: 08/31/2023] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
Rifampin resistance (RIF-R) in Staphylococcus aureus (S. aureus) with rpoB mutations as one of its resistance mechanisms has raised concern about clinical treatment and infection prevention strategies. Data on the prevalence and molecular epidemiology of RIF-R S. aureus blood isolates in South Korea are scarce. We used broth microdilution to investigate RIF-R prevalence and analyzed the rpoB gene mutation in 1615 S. aureus blood isolates (772 methicillin-susceptible and 843 methicillin-resistant S. aureus (MRSA)) from patients with bacteremia, between 2008 and 2017. RIF-R prevalence and antimicrobial susceptibility were determined. Multilocus sequence typing was used to characterize the isolate's molecular epidemiology; Staphylococcus protein A (spa), staphylococcal cassette chromosome mec (SCCmec), and rpoB gene mutations were detected by PCR. Among 52 RIF-R MRSA isolates out of 57 RIF-R S. aureus blood isolates (57/1615, 0.4%; 5 methicillin-susceptible and 52 MRSA), ST5 (44/52, 84.6%), SCCmec IIb (40/52, 76.9%), and spa t2460 (27/52, 51.9%) were predominant. rpoB gene mutations with amino acid substitutions showed that A477D (17/48, 35.4%) frequently conferred high-level RIF resistance (MIC > 128 mg/L), followed by H481Y (4/48, 8.3%). RIF-R S. aureus blood isolates in South Korea have unique molecular characteristics and are closely associated with rpoB gene mutations. RIF-R surveillance through S. aureus-blood isolate epidemiology could enable effective therapeutic management.
Collapse
Affiliation(s)
- Yong Kyun Kim
- Department of Internal Medicine, Division of Infectious Diseases, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea;
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Yewon Eom
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (Y.E.); (E.K.); (S.B.)
- Asan Medical Center, Asan Institute for Life Science, Seoul 05505, Republic of Korea
| | - Eunsil Kim
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (Y.E.); (E.K.); (S.B.)
- Asan Medical Center, Asan Institute for Life Science, Seoul 05505, Republic of Korea
| | - Euijin Chang
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Seongman Bae
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (Y.E.); (E.K.); (S.B.)
| | - Jiwon Jung
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Min Jae Kim
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Yong Pil Chong
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Sung-Han Kim
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Sang-Ho Choi
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Sang-Oh Lee
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
| | - Yang Soo Kim
- Division of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (E.C.); (J.J.); (M.J.K.); (Y.P.C.); (S.-H.K.); (S.-H.C.); (S.-O.L.)
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (Y.E.); (E.K.); (S.B.)
| |
Collapse
|
42
|
Yan Y, Liu Y, Li J, Li Y, Wu H, Li H, Ma X, Tang Y, Tong Y, Yi K, Liang Q, Liu Z. A Molecular Switch-Integrated Nanoplatform Enables Photo-Unlocked Antibacterial Drug Delivery for Synergistic Abscess Therapy. Adv Healthc Mater 2023; 12:e2301157. [PMID: 37392145 DOI: 10.1002/adhm.202301157] [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: 04/13/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/03/2023]
Abstract
Drug delivery systems (DDSs) capable of sequential multistage drug release are urgently needed for antibacterial applications. Herein, a molecular switch-integrated, photo-responsive nanoplatform is reported based on hollow mesoporous silica nanospheres (HMSN) loaded with silver nanoparticles (Ag NPs), vancomycin (Van), and hemin (HAVH) for bacteria elimination and abscess therapy. Upon near-infrared (NIR) light irradiation, the molecular switch, hemin, can effuse from the mesopores of HMSN, triggering the release of pre-loaded Ag+ and Van, which enables photothermal-modulated drug release and synergistic photothermal-chemo therapy (PTT-CHT). The HAVH_NIR irreversibly disrupts the bacterial cell membrane, facilitating the penetration of Ag+ and Van. It is found that these compounds restrain the transcription and translation of ribosomes and lead to rapid bacterial death. Furthermore, hemin can effectively inhibit excessive inflammatory responses associated with the treatment, promoting accelerated wound healing in a murine abscess model. This work presents a new strategy for antibacterial drug delivery with high controllability and extendibility, which may benefit the development of smart multifunctional nanomedicine for diseases not limited to bacterial infections.
Collapse
Affiliation(s)
- Yunxiang Yan
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| | - Yong Liu
- School of Science, Hainan University, Haikou, 570228, China
| | - Juanjuan Li
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| | - Ye Li
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Haoheng Wu
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| | - Hong Li
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| | - Xiang Ma
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| | - Yanqiong Tang
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| | - Yuan Tong
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| | - Kexian Yi
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Quanfeng Liang
- State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, China
| | - Zhu Liu
- School of Life Sciences, Hainan University, Haikou, 570228, China
- One Health Institute, Hainan University, Haikou, 570228, China
| |
Collapse
|
43
|
Kebriaei R, Abdul-Mutakabbir JC, Stamper KC, Lev KL, Rybak MJ. Targeting Dalbavancin Inoculum Effect: Adjunctive Single Dose of Daptomycin. Infect Dis Ther 2023; 12:2485-2494. [PMID: 37798469 PMCID: PMC10600059 DOI: 10.1007/s40121-023-00875-1] [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: 08/11/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Daptomycin (DAP) has proven to be a viable alternative amid vancomycin resistance; however, the use of DAP post vancomycin treatment has led to the development of DAP non-susceptible (DNS) strains. Dalbavancin (DAL), a novel single-dosed lipoglycopeptide, has shown enhanced activity against highly resistant Staphylococcus aureus strains. However, on the basis of previous reports and our observations, DAL does not demonstrate similar activity at high versus low inoculum levels. Therefore, we hypothesized that addition of DAP even at minimal concentrations (single dose on day 1) will lower the inoculum to the level that can be cleared by dalbavancin. METHODS Isolates from methicillin-resistant S. aureus (MRSA)-infected patients with varying susceptibility profiles were evaluated using broth microdilution methods. Two DNS-VISA strains (vancomycin intermediate resistant S. aureus) and one MRSA strain were further evaluated in a one-compartment PK/PD model using a high starting initial inoculum of 109 CFU/mL as well as low initial inoculum of 107 CFU/mL over 168 h to assess the activity of DAL and DAP monotherapy and in combination. RESULTS Single therapies were not bactericidal when evaluated in the 168 h in vitro one-compartment model with an initial inoculum of 109; however, the combination of DAL plus single dose of DAP resulted in enhanced killing at the end of the 168-h exposure. DAL single therapy caused reduction in colony counts down to detection limit (2 log10 CFU/ml) at a lower inoculum but did not show enhancement (< 2 log10 CFU/ml) at higher initial inoculums (P < 0.01) for all three strains. Similarly, DAP caused initial bacterial reduction up to 4 log10 CFU/ml with regrowth at about 32 h of exposure, which stayed at initial inoculum levels for the duration of the model for all three strains. CONCLUSIONS Dalbavancin inoculum effect is a major issue in bacterial infections with high bacterial loads and the combination of DAL plus single dose of DAP showed promise in eradicating resistant S. aureus strains at high inoculums.
Collapse
Affiliation(s)
- Razieh Kebriaei
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, Ohio State University, Columbus, USA
| | - Jacinda C Abdul-Mutakabbir
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- Division of Clinical Pharmacy, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Kyle C Stamper
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- Prism Labs, LLC, Walled Lake, MI, USA
| | - Katherine L Lev
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA
| | - Michael J Rybak
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA.
- Division of Infectious Diseases, School of Medicine, Wayne State University, Detroit, MI, USA.
- Detroit Receiving Hospital, Detroit, MI, USA.
| |
Collapse
|
44
|
Periferakis AT, Periferakis A, Periferakis K, Caruntu A, Badarau IA, Savulescu-Fiedler I, Scheau C, Caruntu C. Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients 2023; 15:4097. [PMID: 37836381 PMCID: PMC10574431 DOI: 10.3390/nu15194097] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Capsaicin is a phytochemical derived from plants of the genus Capsicum and subject of intensive phytochemical research due to its numerous physiological and therapeutical effects, including its important antimicrobial properties. Depending on the concentration and the strain of the bacterium, capsaicin can exert either bacteriostatic or even bactericidal effects against a wide range of both Gram-positive and Gram-negative bacteria, while in certain cases it can reduce their pathogenicity by a variety of mechanisms such as mitigating the release of toxins or inhibiting biofilm formation. Likewise, capsaicin has been shown to be effective against fungal pathogens, particularly Candida spp., where it once again interferes with biofilm formation. The parasites Toxoplasma gondi and Trypanosoma cruzi have been found to be susceptible to the action of this compound too while there are also viruses whose invasiveness is significantly dampened by it. Among the most encouraging findings are the prospects for future development, especially using new formulations and drug delivery mechanisms. Finally, the influence of capsaicin in somatostatin and substance P secretion and action, offers an interesting array of possibilities given that these physiologically secreted compounds modulate inflammation and immune response to a significant extent.
Collapse
Affiliation(s)
- Aristodemos-Theodoros Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Argyrios Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs (P.O.E.P), 17236 Athens, Greece
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, ‘Prof. N.C. Paulescu’ National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| |
Collapse
|
45
|
Muraglia M, Schiavone BIP, Rosato A, Clodoveo ML, Corbo F. Antimicrobial Synergistic Effects of Linezolid and Vancomycin with a Small Synthesized 2-Mercaptobenzothiazole Derivative: A Challenge for MRSA Solving. Molecules 2023; 28:6348. [PMID: 37687173 PMCID: PMC10489997 DOI: 10.3390/molecules28176348] [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: 07/03/2023] [Revised: 08/11/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) emerged as one of the leading causes of persistent human infections and makes it difficult to treat bacteremia, especially with biofilm formation. In this work, we investigated the in vitro synergism between Linezolid (LNZ) and Vancomycin (VAN) with a 2-mercaptobenzothiazole derivative, resulting in a new small-molecule antibacterial compound that we named BTZ2e, on several clinical MRSA, MRSE (methicillin-resistant Staphylococcus epidermidis) and control (ATCC Collection) strains in their planktonic and biofilms cultures. The broth microdilution method evaluated the susceptibility of planktonic cells to each investigated antibiotic combined with BTZ2e. The biofilm's metabolic activity was studied with the XTT reduction assay. As a result, in this study, biofilm formation was significantly suppressed by the BTZ2e treatment. In terms of minimal biofilm inhibitory concentration (MBIC), BTZ2e revealed an MBIC50 value of 32 μg/mL against methicillin-susceptible S. aureus (MSSA) and 16 μg/mL against methicillin-resistant S. aureus ATCC 43300 biofilms. An inhibition range of 32 μg/mL and 256 μg/mL was registered for the clinical isolates. Interestingly, a synergistic effect (FICI ≤ 0.5) was encountered for the combination of BTZ2e with LNZ and VAN on several planktonic and sessile strains. In particular, the best result against planktonic cells emerged as a result of the synergistic association between LNZ and BTZ2e, while against sessile cells, the best synergistic association resulted from VAN and BTZ2e. The consistent results indicate BTZ2e as a promising adjuvant against multi-resistant strains such as MRSA and MRSE.
Collapse
Affiliation(s)
- Marilena Muraglia
- Dipartimento di Farmacia-Scienze del Farmaco, Università Degli Studi di Bari “A. Moro”, Campus Universitario E. Quagliarello Via Orabona 4, 70125 Bari, Italy; (B.I.P.S.); (A.R.); (F.C.)
| | - Brigida Immacolata Pia Schiavone
- Dipartimento di Farmacia-Scienze del Farmaco, Università Degli Studi di Bari “A. Moro”, Campus Universitario E. Quagliarello Via Orabona 4, 70125 Bari, Italy; (B.I.P.S.); (A.R.); (F.C.)
| | - Antonio Rosato
- Dipartimento di Farmacia-Scienze del Farmaco, Università Degli Studi di Bari “A. Moro”, Campus Universitario E. Quagliarello Via Orabona 4, 70125 Bari, Italy; (B.I.P.S.); (A.R.); (F.C.)
| | - Maria Lisa Clodoveo
- Dipartimento Interdisciplinare di Medicina, Università degli Studi di Bari “A. Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Filomena Corbo
- Dipartimento di Farmacia-Scienze del Farmaco, Università Degli Studi di Bari “A. Moro”, Campus Universitario E. Quagliarello Via Orabona 4, 70125 Bari, Italy; (B.I.P.S.); (A.R.); (F.C.)
| |
Collapse
|
46
|
Almuhayawi MS, Alruhaili MH, Gattan HS, Alharbi MT, Nagshabandi M, Al Jaouni S, Selim S, Alanazi A, Alruwaili Y, Faried OA, Elnosary ME. Staphylococcus aureus Induced Wound Infections Which Antimicrobial Resistance, Methicillin- and Vancomycin-Resistant: Assessment of Emergence and Cross Sectional Study. Infect Drug Resist 2023; 16:5335-5346. [PMID: 37605760 PMCID: PMC10440082 DOI: 10.2147/idr.s418681] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
Background Wound infection is a prevalent concern in the medical field, being is a multi-step process involving several biological processes. Methicillin-resistant S. aureus (MRSA) and vancomycin-resistant S. aureus (VRSA) infections often occur in areas of damaged skin, such as abrasions and open wounds. Methods This research aims to light the incidence of MRSA and VRSA in wound swabs, the antimicrobial susceptibility configuration of isolated S. aureus patterns in pus/wound samples collected from Saudi Arabian tertiary hospital. The cross section study, β- lactamase detection, VRSA genotyping, MAR index, D-test and VRSA genotyping are methods, which used for completed this research. Results Patients of several ages and genders delivered specimens from two hospitals in the Al jouf area, in the northern province of Saudi Arabia. S. aureus was found in 188 (34.7%) of the 542 wounds. The traumatized wounds provided 71 isolates (38.8%), surgical wound provided 49 isolates (26.8%) and abscess were represented 16 by isolates (8.7%). In the study, 123 (65.4%) out of 188 were MRSA, 60 (31.9%) were MSSA, and five (2.7%) were VRSA. Linezolid and rifampin were found to be the most effective antimicrobials with 100% in vitro antibacterial activity against S. aureus isolates. The Multiple antimicrobials resistance (MAR) index revealed 73 isolates (38.9%) with a MAR index greater than 0.2, and 115 (61.1%) less than 0.2. The D-test showed that of MLSb phenotypes among S. aureus, 22 (11.7%) strains were D-test positive (MLSbi phenotype), 53 (28.2%) strains were constitutive MLSc phenotypes, and 17 (9%) strains were shown to have MSb phenotypes. All VRSA isolates (n=5) were found to be positive for vanA, and no vanB positive isolates were detected in the study. Conclusion Regular monitoring and an antimicrobials stewardship program should be in place to provide critical information that can be utilized for empirical therapy and future prevention strategies.
Collapse
Affiliation(s)
- Mohammed S Almuhayawi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Mohammed H Alruhaili
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King AbdulAziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahad Medical Research Center, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Hattan S Gattan
- Special Infectious Agents Unit, King Fahad Medical Research Center, King AbdulAziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohanned Talal Alharbi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, University of Jeddah, Jeddah, 23218, Saudi Arabia
| | - Mohammed Nagshabandi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, University of Jeddah, Jeddah, 23218, Saudi Arabia
| | - Soad Al Jaouni
- Department of Hematology/Oncology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Awadh Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Yasir Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Osama Ahmed Faried
- Medical Microbiology and Immunology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed E Elnosary
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
47
|
Tabaraki R, Nazari F. Vancomycin-modified nitrogen and chloride doped carbon dots and their application as a Staphylococcus aureus probe. Anal Chim Acta 2023; 1268:341311. [PMID: 37268336 DOI: 10.1016/j.aca.2023.341311] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/30/2023] [Accepted: 05/01/2023] [Indexed: 06/04/2023]
Abstract
In this research, N, Cl-doped carbon dots (N, Cl-CDs) were prepared in choline chloride-glycerol deep eutectic solvent (DES) by microwave method. N, Cl-CDs surface was modified with vancomycin for detection of Staphylococcus aureus (S. aureus) bacteria in the range of 102-107 colony-forming unit per milliliter (CFU/mL). The detection limit was 101 CFU/mL. Morphology and structure of N, Cl-CDs were characterized by transmission electron microscopy (TEM), X-ray photon spectroscopy (XPS), photoluminescence spectroscopy, FT-IR spectroscopy, energy dispersive X-ray spectroscopy (EDXS) and zeta potential. The prepared N, Cl-CDs had excellent dispersion in water, particle size range of 2-3 nm, and quantum yield of 38.75%. Speed, wide linear range and more convenient were advantages of new probe with respect to other methods.
Collapse
Affiliation(s)
- Reza Tabaraki
- Department of Chemistry, Faculty of Science, Ilam University, Ilam, Iran.
| | - Fereshteh Nazari
- Department of Chemistry, Faculty of Science, Ilam University, Ilam, Iran
| |
Collapse
|
48
|
Heinzinger LR, Pugh AR, Wagner JA, Otto M. Evaluating the Translational Potential of Bacteriocins as an Alternative Treatment for Staphylococcus aureus Infections in Animals and Humans. Antibiotics (Basel) 2023; 12:1256. [PMID: 37627676 PMCID: PMC10451987 DOI: 10.3390/antibiotics12081256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Antibiotic resistance remains a global threat to human and animal health. Staphylococcus aureus is an opportunistic pathogen that causes minor to life-threatening infections. The widespread use of antibiotics in the clinical, veterinary, and agricultural setting combined with the increasing prevalence of antibiotic-resistant S. aureus strains makes it abundantly clear that alternatives to antibiotics are urgently needed. Bacteriocins represent one potential alternative therapeutic. They are antimicrobial peptides that are produced by bacteria that are generally nontoxic and have a relatively narrow target spectrum, and they leave many commensals and most mammalian cells unperturbed. Multiple studies involving bacteriocins (e.g., nisin, epidermicin, mersacidin, and lysostaphin) have demonstrated their efficacy at eliminating or treating a wide variety of S. aureus infections in animal models. This review provides a comprehensive and updated evaluation of animal studies involving bacteriocins and highlights their translational potential. The strengths and limitations associated with bacteriocin treatments compared with traditional antibiotic therapies are evaluated, and the challenges that are involved with implementing novel therapeutics are discussed.
Collapse
Affiliation(s)
| | | | | | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA; (L.R.H.); (A.R.P.); (J.A.W.)
| |
Collapse
|
49
|
Kaur H, Modgil V, Chaudhary N, Mohan B, Taneja N. Computational Guided Drug Targets Identification against Extended-Spectrum Beta-Lactamase-Producing Multi-Drug Resistant Uropathogenic Escherichia coli. Biomedicines 2023; 11:2028. [PMID: 37509666 PMCID: PMC10377140 DOI: 10.3390/biomedicines11072028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Urinary tract infections (UTIs) are one of the most frequent bacterial infections in the world, both in the hospital and community settings. Uropathogenic Escherichia coli (UPEC) are the predominant etiological agents causing UTIs. Extended-spectrum beta-lactamase (ESBL) production is a prominent mechanism of resistance that hinders the antimicrobial treatment of UTIs caused by UPEC and poses a substantial danger to the arsenal of antibiotics now in use. As bacteria have several methods to counteract the effects of antibiotics, identifying new potential drug targets may help in the design of new antimicrobial agents, and in the control of the rising trend of antimicrobial resistance (AMR). The public availability of the entire genome sequences of humans and many disease-causing organisms has accelerated the hunt for viable therapeutic targets. Using a unique, hierarchical, in silico technique using computational tools, we discovered and described potential therapeutic drug targets against the ESBL-producing UPEC strain NA114. Three different sets of proteins (chokepoint, virulence, and resistance genes) were explored in phase 1. In phase 2, proteins shortlisted from phase 1 were analyzed for their essentiality, non-homology to the human genome, and gut flora. In phase 3, the further shortlisted putative drug targets were qualitatively characterized, including their subcellular location, broad-spectrum potential, and druggability evaluations. We found seven distinct targets for the pathogen that showed no similarity to the human proteome. Thus, possibilities for cross-reactivity between a target-specific antibacterial and human proteins were minimized. The subcellular locations of two targets, ECNA114_0085 and ECNA114_1060, were predicted as cytoplasmic and periplasmic, respectively. These proteins play an important role in bacterial peptidoglycan biosynthesis and inositol phosphate metabolism, and can be used in the design of drugs against these bacteria. Inhibition of these proteins will be helpful to combat infections caused by MDR UPEC.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Vinay Modgil
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Naveen Chaudhary
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Balvinder Mohan
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Neelam Taneja
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
50
|
Kim JW, Lee KJ. Development of a Single-nucleotide Polymorphism Genotyping Assay for the Rapid Detection of Vancomycin-intermediate Resistance in Staphylococcus aureus Epidemic Lineage ST5. Ann Lab Med 2023; 43:355-363. [PMID: 36843404 PMCID: PMC9989536 DOI: 10.3343/alm.2023.43.4.355] [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: 09/02/2022] [Revised: 10/25/2022] [Accepted: 01/04/2023] [Indexed: 02/28/2023] Open
Abstract
Background Vancomycin is a treatment option for patients with severe methicillin-resistant Staphylococcus aureus (MRSA) infection. Unfortunately, reduced susceptibility to vancomycin in S. aureus is becoming increasingly common. We developed a method for the rapid and accurate diagnosis of vancomycin-intermediate resistant S. aureus (VISA). Methods We performed a microbial genome-wide association study to discriminate between VISA and vancomycin-susceptible S. aureus (VSSA) using 42 whole-genome sequences. A TaqMan single-nucleotide polymorphism (SNP) genotyping assay was developed to detect target SNPs in VISA strains. Results Four SNPs in the VISA strains resulting in nonsynonymous amino-acid substitutions were associated with reduced susceptibility to vancomycin: SA_RS01235 (G203S), SA_RS09725 (V171A), SA_RS12250 (I48F), and SA_RS12550 (G478A). These four SNPs were mainly detected in the typical hospital-associated sequence type (ST)5 clonal lineage. The TaqMan assay successfully detected all four SNPs using as little as 0.2 ng DNA per reaction. Using 10 VSSA and VISA clinical strains each, we validated that the assay accurately discriminates between VISA and VSSA. Conclusions The TaqMan SNP genotyping assay targeting four SNPs may be an alternative to current standard methods for the rapid detection of vancomycin-intermediate resistance in S. aureus epidemic lineage ST5.
Collapse
Affiliation(s)
- Jung Wook Kim
- Division of Antimicrobial Resistance Research, Center for Infectious Diseases Research, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Kwang Jun Lee
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Diseases Research, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| |
Collapse
|