1
|
Nygaard TK, Borgogna TR, Pallister KB, Predtechenskaya M, Burroughs OS, Gao A, Lubick EG, Voyich JM. The Relative Importance of Cytotoxins Produced by Methicillin-Resistant Staphylococcus aureus Strain USA300 for Causing Human PMN Destruction. Microorganisms 2024; 12:1782. [PMID: 39338457 PMCID: PMC11434515 DOI: 10.3390/microorganisms12091782] [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: 08/06/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is a prominent Gram-positive bacterial pathogen that expresses numerous cytotoxins known to target human polymorphonuclear leukocytes (PMNs or neutrophils). These include leukocidin G/H (LukGH, also known as LukAB), the Panton-Valentine leukocidin (PVL), γ-hemolysin A/B (HlgAB), γ-hemolysin B/C (HlgBC), leukocidin E/D (LukED), α-hemolysin (Hla), and the phenol-soluble modulin-α peptides (PSMα). However, the relative contribution of each of these cytotoxins in causing human PMN lysis is not clear. In this study, we used a library of cytotoxin deletion mutants in the clinically relevant methicillin-resistant S. aureus (MRSA) isolate LAC (strain ST8:USA300) to determine the relative importance of each for causing human PMN lysis upon exposure to extracellular components as well as following phagocytosis. Using flow cytometry to examine plasma membrane permeability and assays quantifying lactose dehydrogenase release, we found that PVL was the dominant extracellular factor causing human PMN lysis produced by USA300. In contrast, LukGH was the most important cytotoxin causing human PMN lysis immediately following phagocytosis with contributions from the other bicomponent leukocidins only observed at later time points. These results not only clarify the relative importance of different USA300 cytotoxins for causing human PMN destruction but also demonstrate how two apparently redundant virulence factors play distinctive roles in promoting S. aureus pathogenesis.
Collapse
Affiliation(s)
- Tyler K Nygaard
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Timothy R Borgogna
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Kyler B Pallister
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Maria Predtechenskaya
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Owen S Burroughs
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Annika Gao
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Evan G Lubick
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Jovanka M Voyich
- Department of Microbiology Cell Biology, Montana State University, Bozeman, MT 59718, USA
| |
Collapse
|
2
|
Ibraheim HK, Madhi KS, Baqer GK, Gharban HAJ. Effectiveness of raw bacteriocin produced from lactic acid bacteria on biofilm of methicillin-resistant Staphylococcus aureus. Vet World 2023; 16:491-499. [PMID: 37041833 PMCID: PMC10082751 DOI: 10.14202/vetworld.2023.491-499] [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: 12/06/2022] [Accepted: 01/18/2023] [Indexed: 04/13/2023] Open
Abstract
Background and Aim Probiotics are proven beneficial to health since they enhance immunity against dangerous pathogens and increase resistance to illness. Bacteriocin produced by lactic acid bacteria (LAB), demonstrates a broad inhibitory spectrum and therapeutic potential. This study aimed to isolate LAB-producing bacteriocin and investigate the effect of crude bacteriocin on biofilm from methicillin-resistant Staphylococcus aureus (MRSA). Materials and Methods This study used randomly collected 80 white soft local cheeses (40 each from cows and sheep) from different supermarkets in Basrah Province. The obtained samples were cultured and the bacterial suspension of S. aureus was prepared at 1.5 × 108 cells/mL. The crude bacteriocin extracted from LAB was obtained, and the tube was dried and inverted to detect the biofilm loss at the bottom. Results There were 67 (83.75%) LAB isolates. Among 40 milk samples collected directly and indirectly, there were 36 (83.33%). Staphylococcus aureus isolates based on conventional bacteriological analysis and biochemical tests. Molecular testing was conducted to identify LAB and MRSA. Depending on genotypic results, the effect of white soft local cheese (cows and sheep) and the amplification results of the 16S rRNA gene were detected in 46 LAB isolates from white soft local cheese from cows and sheep. Based on the molecular identification of the mecA, results on Staphylococcus determined that only 2 of 36 isolates of S. aureus carried the mecA. Moreover, there were 26 (86.66%) isolates (MRSA) from samples of raw milk from local markets and subclinical mastitis in cows. The ability of LAB isolates was tested. The effects of bacteriocin production on preventing biofilm growth and formation were investigated. Results demonstrated that bacteriocin has high activity. Microtiter plates applied to investigate the ability of S. aureus to produce biofilms revealed that all isolates were either weak or moderate biofilm producers, with neither non-biofilm nor strong biofilm producers found among the tested isolates. Conclusion Lactic acid bacteria demonstrate a high ability to produce bacteriocin. Crude bacteriocin from LAB has a restrictive effect on biofilms produced by MRSA; thus, it can be used to reduce the pathogenicity of this bacterium.
Collapse
Affiliation(s)
- Hanaa Khaleel Ibraheim
- Department of Microbiology, College of Veterinary Medicine, University of Basrah, Basrah, Iraq
- Corresponding author: Hanaa Khaleel Ibraheim, e-mail: Co-authors: KSM: , GKB: , HAJG:
| | - Khadeeja S. Madhi
- Department of Microbiology, College of Medicine, University of Basrah, Basrah, Iraq
| | - Gaida K. Baqer
- Department of Microbiology, College of Medicine, University of Basrah, Basrah, Iraq
| | - Hasanain A. J. Gharban
- Department of Internal and Preventive Veterinary Medicine, College of Veterinary Medicine, University of Wasit, Wasit, Iraq
| |
Collapse
|
3
|
Aljghami ME, Barghash MM, Majaesic E, Bhandari V, Houry WA. Cellular functions of the ClpP protease impacting bacterial virulence. Front Mol Biosci 2022; 9:1054408. [PMID: 36533084 PMCID: PMC9753991 DOI: 10.3389/fmolb.2022.1054408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/15/2022] [Indexed: 09/28/2023] Open
Abstract
Proteostasis mechanisms significantly contribute to the sculpting of the proteomes of all living organisms. ClpXP is a central AAA+ chaperone-protease complex present in both prokaryotes and eukaryotes that facilitates the unfolding and subsequent degradation of target substrates. ClpX is a hexameric unfoldase ATPase, while ClpP is a tetradecameric serine protease. Substrates of ClpXP belong to many cellular pathways such as DNA damage response, metabolism, and transcriptional regulation. Crucially, disruption of this proteolytic complex in microbes has been shown to impact the virulence and infectivity of various human pathogenic bacteria. Loss of ClpXP impacts stress responses, biofilm formation, and virulence effector protein production, leading to decreased pathogenicity in cell and animal infection models. Here, we provide an overview of the multiple critical functions of ClpXP and its substrates that modulate bacterial virulence with examples from several important human pathogens.
Collapse
Affiliation(s)
- Mazen E. Aljghami
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Marim M. Barghash
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Emily Majaesic
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Vaibhav Bhandari
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Walid A. Houry
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Yang K, Zhou X, Li Z, Wang Z, Luo Y, Deng L, He D. Ultrastretchable, Self-Healable, and Tissue-Adhesive Hydrogel Dressings Involving Nanoscale Tannic Acid/Ferric Ion Complexes for Combating Bacterial Infection and Promoting Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2022; 14:43010-43025. [PMID: 36108772 DOI: 10.1021/acsami.2c13283] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Preventing bacterial infections and accelerating wound closure are essential in the process of wound healing. Current wound dressings lack enough mechanical properties, self-healing ability, and tissue adhesiveness, and the bacterial killing also relies on the use of antibiotic drugs. Herein, a well-designed hybrid hydrogel dressing is constructed by simple copolymerization of acrylamide (AM), 3-acrylamido phenylboronic acid (AAPBA), chitosan (CS), and the nanoscale tannic acid (TA)/ferric ion (Fe3+) complex (TFe). The resulting hydrogel possesses lots of free catechol, phenylboronic acid, amine, and hydroxyl groups and contains many reversible and dynamic bonds such as multiple hydrogen bonds and boronate ester bonds, thereby showing satisfactory mechanical properties, fast self-healing ability, and desirable tissue-adhesive performance. Benefiting from the high photothermal conversion efficiency of the TFe, the hydrogel exhibits satisfactory antibacterial activity against both Gram-positive and Gram-negative bacteria. Moreover, the embedded TFe also endows the hydrogel with good antioxidant activity, anti-inflammatory property, and cell proliferation to promote tissue regeneration. Remarkably, in vivo animal assays reveal that the hybrid hydrogel effectively eliminates biofilm bacteria in the wound sites and accelerates the healing process of infected wounds. Taken together, the developed versatile hydrogels overcome the shortcomings of traditional wound dressings and are expected to become potential antibacterial dressings for future biomedical applications.
Collapse
Affiliation(s)
- Ke Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Xueyao Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Zhaoli Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Zefeng Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yuze Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Le Deng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Dinggeng He
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
5
|
Song W, Wang B, Sui L, Shi Y, Ren X, Wang X, Kong X, Hou J, Wang L, Wei L, Luan Y, Guan J, Zhao Y. Tamarixetin Attenuated the Virulence of Staphylococcus aureus by Directly Targeting Caseinolytic Protease P. JOURNAL OF NATURAL PRODUCTS 2022; 85:1936-1944. [PMID: 35833867 DOI: 10.1021/acs.jnatprod.2c00138] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Staphylococcus aureus, especially drug-resistant S. aureus infections, is a worldwide healthcare challenge. There is a growing focus on antivirulence therapy against S. aureus. Caseinolytic protease p (ClpP) is a protein hydrolase essential for pathogenicity in S. aureus. A flavonoid compound, tamarixetin, which was screened in this work, was specifically able to inhibit the hydrolytic activity of ClpP on the fluorescent substrate Suc-LY-AMC with an IC50 of 49.73 μM, without affecting the growth of methicillin-resistant S. aureus strain USA300 and was without obvious cytotoxicity. Further assays found that tamarixetin inhibited the transcription of hla, agr, RNAIII, pvl, PSM-α, and spa genes as well as suppressed the protein expression levels of Hla and PVL. Moreover, tamarixetin was observed to dramatically inhibit the hemolytic activity of hla in S. aureus. Consistent with that of S. aureus USA300-ΔclpP, tamarixetin was shown to increase urease expression. The thermal shift and cellular thermal shift assays showed that tamarixetin markedly changed the thermal stability of ClpP. The dissociation constant (KD) value of tamarixetin with ClpP was 2.52 × 10-6 M measured by surface plasmon resonance. The molecular docking and ClpP point mutation results also demonstrated that tamarixetin had a strong interaction with ClpP. In vivo study showed that tamarixetin was effective in protecting mice from S. aureus pneumonia by increasing survival, reducing lung tissue load, and slowing down the infiltration of inflammatory factors. In addition, tamarixetin was able to enhance the antibacterial activity of cefotaxime in combination. In conclusion, tamarixetin was promising as a ClpP inhibitor for S. aureus infections.
Collapse
Affiliation(s)
- Wu Song
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Bingmei Wang
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Liyan Sui
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
| | - Yan Shi
- School of Pharmacy, Jilin University, Changchun 130021, China
| | - Xinran Ren
- School of Pharmacy, Jilin University, Changchun 130021, China
| | - Xingye Wang
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Xiangri Kong
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Juan Hou
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Li Wang
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Lin Wei
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Yanhe Luan
- Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yicheng Zhao
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
6
|
Ong ZX, Kannan B, Becker DL. Exploiting transposons in the study of Staphylococcus aureus pathogenesis and virulence. Crit Rev Microbiol 2022; 49:297-317. [PMID: 35438613 DOI: 10.1080/1040841x.2022.2052794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The opportunistic pathogen Staphylococcus aureus has an extremely complex relationship with humans. While the bacteria can exist as a commensal in many, it can cause a wide range of diseases and infections when turned pathogenic. Its presence is a determinant of chronicity and poor prognosis in numerous diseases, and its genomic plasticity causes S. aureus antimicrobial resistance to be one of the most dire contemporary medical problems to solve. Genetic manipulation of S. aureus has led to numerous findings that are vital in the fight against its pathogenesis. The utilisation of transposon mutant libraries for the systematic inspection of the S. aureus genome led to many landmark discoveries pertaining to the bacteria's pathogenicity, antimicrobial resistance acquisition, and virulence regulation. In this review, we describe mutant libraries, and their significant contributions, from various S. aureus strains created with commonly used transposons. The general workflow for the construction of libraries will be presented, along with a discussion of the challenges of undertaking the task of large-scale library construction. As the accessibility of transposon mutant library construction, screening, and analysis increases, this genetic tool could be further exploited in the study of the S. aureus genome.
Collapse
Affiliation(s)
- Zi Xin Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Skin Research Institute, Singapore.,Nanyang Institute of Technology in Health and Medicine, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore
| | - Bavani Kannan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Skin Research Institute, Singapore
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Skin Research Institute, Singapore
| |
Collapse
|
7
|
Weiss A, Jackson JK, Shaw LN, Skaar EP. Screening transcriptional connections in Staphylococcus aureus using high-throughput transduction of bioluminescent reporter plasmids. MICROBIOLOGY (READING, ENGLAND) 2022; 168:001174. [PMID: 35446249 PMCID: PMC10233262 DOI: 10.1099/mic.0.001174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/17/2022] [Indexed: 12/14/2022]
Abstract
Characterization of transcriptional networks is one of the main strategies used to understand how bacteria interact with their environment. To reveal novel regulatory elements in the human pathogen Staphylococcus aureus, we adapted a traditional transduction protocol to be used in a high-throughput format in combination with the publicly available S. aureus Nebraska Transposon Mutant Library. Specifically, plasmid transductions are performed in 96-well format, so that a single plasmid can be simultaneously transferred into numerous recipient strains. When used in conjunction with bioluminescent reporter constructs, this strategy enables parallel and continuous monitoring of downstream transcriptional effects of hundreds of defined mutations. Here, we use this workflow in a proof-of-concept study to identify novel regulators of the staphylococcal metalloprotease aureolysin. Importantly, this strategy can be utilized with any other bacterium where plasmid transduction is possible, making it a versatile and efficient tool to probe transcriptional regulatory connections.
Collapse
Affiliation(s)
- Andy Weiss
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jessica K. Jackson
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Lindsey N. Shaw
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Eric P. Skaar
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
8
|
Tang H, Qu X, Zhang W, Chen X, Zhang S, Xu Y, Yang H, Wang Y, Yang J, Yuan WE, Yue B. Photosensitizer Nanodot Eliciting Immunogenicity for Photo-Immunologic Therapy of Postoperative Methicillin-Resistant Staphylococcus aureus Infection and Secondary Recurrence. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107300. [PMID: 34865257 DOI: 10.1002/adma.202107300] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/11/2021] [Indexed: 06/13/2023]
Abstract
The treatment of postoperative infection caused by multidrug-resistant bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), has become an intractable clinical challenge owing to its low therapeutic efficacy and high risk of recurrence. Apart from imperfect antibacterial therapies, induction of insufficient immunogenicity, required for the successful clearance of a pathogen, may also contribute to the problem. Herein, an ultra-micro photosensitizer, AgB nanodots, using photothermal therapy, photodynamic therapy, and Ag+ ion sterilization, are utilized to efficiently clear invading MRSA both in vitro and in vivo. AgB nanodots are also found to upregulate host immunogenicity in a murine model and establish immunological memory by promoting the upregulated expression of danger signals that are commonly induced by stress-related responses, including sudden temperature spikes or excess reactive oxygen production. These stimulations boost the antibacterial effects of macrophages, dendritic cells, T cells, or even memory B cells, which is usually defined as infection-related immunogenic cell death. Hence, the proposed AgB nanodot strategy may offer a novel platform for the effective treatment of postoperative infection while providing a systematic immunotherapeutic strategy to combat persistent infections, thereby markedly reducing the incidence of recurrence following recovery from primary infections.
Collapse
Affiliation(s)
- Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Wenkai Zhang
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Xuan Chen
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Yang Xu
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Hongtao Yang
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus, OH, 43210, USA
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, P. R. China
| | - You Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Jianping Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Wei-En Yuan
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| |
Collapse
|
9
|
Xiao X, Li Y, Li L, Xiong YQ. Identification of Methicillin-Resistant Staphylococcus aureus (MRSA) Genetic Factors Involved in Human Endothelial Cells Damage, an Important Phenotype Correlated with Persistent Endovascular Infection. Antibiotics (Basel) 2022; 11:antibiotics11030316. [PMID: 35326779 PMCID: PMC8944730 DOI: 10.3390/antibiotics11030316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 01/18/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of life-threatening endovascular infections. Endothelial cell (EC) damage is a key factor in the pathogenesis of these syndromes. However, genetic factors related to the EC damage have not been well studied. This study aims to identify genetic determinants that impact human EC damage by screening the genome-wide Nebraska Transposon Mutant Library (NTML). A well-established MTT assay was used to test the in vitro damage of human EC cell line (HMEC-1) caused by each mutant strain in the NTML. We first confirmed some global regulators and genes positively impact the EC damage, which is consistent with published results. These data support the utility of the high-throughput approach. Importantly, we demonstrated 317 mutants significantly decreased the EC damage, while only 6 mutants enhanced the EC damage vs. parental JE2 strain. The majority of these genes have not been previously defined to affect human EC damage. Interestingly, many of these newly identified genes are involved in metabolism, genetic and environmental information processing, and cellular processes. These results advance our knowledge of staphylococcal genetic factors related to human EC damage which may provide novel targets for the development of effective agents against MRSA endovascular infection.
Collapse
Affiliation(s)
- Xia Xiao
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (X.X.); (Y.L.); (L.L.)
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yi Li
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (X.X.); (Y.L.); (L.L.)
| | - Liang Li
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (X.X.); (Y.L.); (L.L.)
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Yan Q. Xiong
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (X.X.); (Y.L.); (L.L.)
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +1-310-222-3545
| |
Collapse
|
10
|
Further Insight into the Mechanism of Human PMN Lysis following Phagocytosis of Staphylococcus aureus. Microbiol Spectr 2021; 9:e0088821. [PMID: 34704790 PMCID: PMC8549732 DOI: 10.1128/spectrum.00888-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Staphylococcus aureus is an important human pathogen that can cause a variety of diseases ranging from mild superficial skin infections to life-threatening conditions like necrotizing pneumonia, endocarditis, and septicemia. Polymorphonuclear leukocytes (PMNs; neutrophils in particular herein) are essential for host defense against S. aureus infections, and the microbe is phagocytosed readily. Most ingested bacteria are killed, but some S. aureus strains—such as the epidemic USA300 strain—have an enhanced ability to cause PMN lysis after phagocytosis. Although progress has been made, the mechanism for lysis after phagocytosis of S. aureus remains incompletely determined. Here, we tested the hypothesis that disruption of phagosome integrity and escape of S. aureus from the PMN phagosome into the cytoplasm precedes PMN lysis. We used USA300 wild-type and isogenic deletion strains to evaluate and/or verify the role of selected S. aureus molecules in this cytolytic process. Compared to the wild-type USA300 strain, Δagr, Δhla, ΔlukGH, and Δpsm strains each caused significantly less lysis of human PMNs 3 h and/or 6 h after phagocytosis, consistent with previous studies. Most notably, confocal microscopy coupled with selective permeabilization assays demonstrated that phagosome membrane integrity is largely maintained prior to PMN lysis after S. aureus phagocytosis. We conclude that PMN lysis does not require escape of S. aureus from the phagosome to the cytoplasm and that these are independent phenomena. The findings are consistent with the ability of S. aureus (via selected molecules) to trigger lysis of human PMNs by an undetermined signaling mechanism. IMPORTANCES. aureus strain USA300 has the ability to cause rapid lysis of human neutrophils after phagocytosis. Although this phenomenon likely contributes to the success of USA300 as a human pathogen, our knowledge of the mechanism remains incomplete. Here, we used a selective permeabilization assay coupled with confocal microscopy to demonstrate that USA300 is contained within human neutrophil phagosomes until the point of host cell lysis. Thus, consistent with a process in macrophages, S. aureus fails to escape into the neutrophil cytoplasm prior to cytolysis.
Collapse
|
11
|
Rasheed S, Fries F, Müller R, Herrmann J. Zebrafish: An Attractive Model to Study Staphylococcus aureus Infection and Its Use as a Drug Discovery Tool. Pharmaceuticals (Basel) 2021; 14:594. [PMID: 34205723 PMCID: PMC8235121 DOI: 10.3390/ph14060594] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
Non-mammalian in vivo disease models are particularly popular in early drug discovery. Zebrafish (Danio rerio) is an attractive vertebrate model, the success of which is driven by several advantages, such as the optical transparency of larvae, the small and completely sequenced genome, the small size of embryos and larvae enabling high-throughput screening, and low costs. In this review, we highlight zebrafish models of Staphyloccoccus aureus infection, which are used in drug discovery and for studying disease pathogenesis and virulence. Further, these infection models are discussed in the context of other relevant zebrafish models for pharmacological and toxicological studies as part of early drug profiling. In addition, we examine key differences to commonly applied models of S.aureus infection based on invertebrate organisms, and we compare their frequency of use in academic research covering the period of January 2011 to January 2021.
Collapse
Affiliation(s)
- Sari Rasheed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
| | - Franziska Fries
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
| |
Collapse
|
12
|
Growth and Stress Tolerance Comprise Independent Metabolic Strategies Critical for Staphylococcus aureus Infection. mBio 2021; 12:e0081421. [PMID: 34101490 PMCID: PMC8262855 DOI: 10.1128/mbio.00814-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Staphylococcus aureus is an important pathogen that leads to high morbidity and mortality. Although S. aureus produces many factors important for pathogenesis, few have been validated as playing a role in the pathogenesis of S. aureus pneumonia. To gain a better understanding of the genetic elements required for S. aureus pathogenesis in the airway, we performed an unbiased genome-wide transposon sequencing (Tn-seq) screen in a model of acute murine pneumonia. We identified 136 genes important for bacterial survival during infection, with a high proportion involved in metabolic processes. Phenotyping 80 individual deletion mutants through diverse in vitro and in vivo assays demonstrated that metabolism is linked to several processes, which include biofilm formation, growth, and resistance to host stressors. We further validated the importance of 23 mutations in pneumonia. Multivariate and principal-component analyses identified two key metabolic mechanisms enabling infection in the airway, growth (e.g., the ability to replicate and form biofilms) and resistance to host stresses. As deep validation of these hypotheses, we investigated the role of pyruvate carboxylase, which was important across multiple infection models and confirmed a connection between growth and resistance to host cell killing. Pathogenesis is conventionally understood in terms of the host-pathogen interactions that enable a pathogen to neutralize a host’s immune response. We demonstrate with the important bacterial pathogen S. aureus that microbial metabolism influences key traits important for in vivo infection, independent from host immunomodulation.
Collapse
|
13
|
Kim GL, Akoolo L, Parker D. The ClpXP Protease Contributes to Staphylococcus aureus Pneumonia. J Infect Dis 2021; 222:1400-1404. [PMID: 32386322 DOI: 10.1093/infdis/jiaa251] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/05/2020] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is a leading cause of pneumonia. We show here that the ClpXP protease involved in protein turnover is important for pathogenesis in a murine model of acute pneumonia. Staphylococcus aureus lacking this protease is attenuated in vivo, being rapidly cleared from the airway and leading to decreased immune cell influx and inflammation. Characterization of defined mutations in vitro identified defects in intracellular survival and protection against neutrophil killing. Our results further expand on what is known about ClpXP in the pathogenesis of S. aureus to include the respiratory tract.
Collapse
Affiliation(s)
- Gyu-Lee Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Lavoisier Akoolo
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
14
|
Li L, Bayer AS, Cheung A, Lu L, Abdelhady W, Donegan NP, Hong JI, Yeaman MR, Xiong YQ. The Stringent Response Contributes to Persistent Methicillin-Resistant Staphylococcus aureus Endovascular Infection Through the Purine Biosynthetic Pathway. J Infect Dis 2021; 222:1188-1198. [PMID: 32333768 DOI: 10.1093/infdis/jiaa202] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/21/2020] [Indexed: 02/02/2023] Open
Abstract
Persistent methicillin-resistant Staphylococcus aureus (MRSA) endovascular infections represent a significant clinical-therapeutic challenge. Of particular concern is antibiotic treatment failure in infections caused by MRSA that are "susceptible" to antibiotic in vitro. In the current study, we investigate specific purine biosynthetic pathways and stringent response mechanism(s) related to this life-threatening syndrome using genetic matched persistent and resolving MRSA clinical bacteremia isolates (PB and RB, respectively), and isogenic MRSA strain sets. We demonstrate that PB isolates (vs RB isolates) have significantly higher (p)ppGpp production, phenol-soluble-modulin expression, polymorphonuclear leukocyte lysis and survival, fibronectin/endothelial cell (EC) adherence, and EC damage. Importantly, an isogenic strain set, including JE2 parental, relP-mutant and relP-complemented strains, translated the above findings into significant outcome differences in an experimental endocarditis model. These observations indicate a significant regulation of purine biosynthesis on stringent response, and suggest the existence of a previously unknown adaptive genetic mechanism in persistent MRSA infection.
Collapse
Affiliation(s)
- Liang Li
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Arnold S Bayer
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA.,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA.,David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ambrose Cheung
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Lou Lu
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA.,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA.,Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Wessam Abdelhady
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Niles P Donegan
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Jong-In Hong
- Department of Chemistry, Seoul National University, Seoul, Korea
| | - Michael R Yeaman
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA.,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA.,David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Yan Q Xiong
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA.,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA.,David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
15
|
Ge C, Monk IR, Monard SC, Bedford JG, Braverman J, Stinear TP, Wakim LM. Neutrophils play an ongoing role in preventing bacterial pneumonia by blocking the dissemination of
Staphylococcus aureus
from the upper to the lower airways. Immunol Cell Biol 2020; 98:577-594. [DOI: 10.1111/imcb.12343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Chenghao Ge
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC 3000 Australia
- School of Medicine Tsinghua University Beijing China
| | - Ian R Monk
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC 3000 Australia
| | - Sarah C Monard
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC 3000 Australia
| | - James G Bedford
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC 3000 Australia
| | - Jessica Braverman
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC 3000 Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC 3000 Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC 3000 Australia
| |
Collapse
|
16
|
Ma Y, Wang C, Li Y, Li J, Wan Q, Chen J, Tay FR, Niu L. Considerations and Caveats in Combating ESKAPE Pathogens against Nosocomial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901872. [PMID: 31921562 PMCID: PMC6947519 DOI: 10.1002/advs.201901872] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Indexed: 05/19/2023]
Abstract
ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) are among the most common opportunistic pathogens in nosocomial infections. ESKAPE pathogens distinguish themselves from normal ones by developing a high level of antibiotic resistance that involves multiple mechanisms. Contemporary therapeutic strategies which are potential options in combating ESKAPE bacteria need further investigation. Herein, a broad overview of the antimicrobial research on ESKAPE pathogens over the past five years is provided with prospective clinical applications.
Collapse
Affiliation(s)
- Yu‐Xuan Ma
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Chen‐Yu Wang
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Yuan‐Yuan Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Jing Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Ji‐Hua Chen
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Franklin R. Tay
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| | - Li‐Na Niu
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| |
Collapse
|