1
|
Silva-Santana G. Staphylococcus aureus: Dynamics of pathogenicity and antimicrobial-resistance in hospital and community environments - Comprehensive overview. Res Microbiol 2025; 176:104267. [PMID: 39805330 DOI: 10.1016/j.resmic.2025.104267] [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: 10/24/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
This study reviews Staphylococcus aureus, a significant pathogen in both hospital and community-acquired infections, addressing its epidemiology, pathogenesis, and antimicrobial resistance. It highlights virulence mechanisms, such as adhesion factors, toxins, enzymes, and biofilms, which contribute to survival and immune evasion. The spread of resistance occurs through the transfer of mobile genetic elements like SCCmec and genetic mutations. The analysis also compares hospital and community strains, including multidrug-resistant lineages like MRSA, VISA, and VRSA. The study concludes that S. aureus presents a major public health challenge, requiring new therapeutic approaches and preventive strategies.
Collapse
Affiliation(s)
- Giorgio Silva-Santana
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro (RJ), Brazil.
| |
Collapse
|
2
|
Fathi P, Pan D. Current trends in pyrrole and porphyrin-derived nanoscale materials for biomedical applications. Nanomedicine (Lond) 2020; 15:2493-2515. [PMID: 32975469 PMCID: PMC7610151 DOI: 10.2217/nnm-2020-0125] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/14/2020] [Indexed: 02/01/2023] Open
Abstract
This article is written to provide an up-to-date review of pyrrole-based biomedical materials. Porphyrins and other tetrapyrrolic molecules possess unique magnetic, optical and other photophysical properties that make them useful for bioimaging and therapy. This review touches briefly on some of the synthetic strategies to obtain porphyrin- and tetrapyrrole-based nanoparticles, as well as the variety of applications in which crosslinked, self-assembled, porphyrin-coated and other nanoparticles are utilized. We explore examples of these nanoparticles' applications in photothermal therapy, drug delivery, photodynamic therapy, stimuli response, fluorescence imaging, photoacoustic imaging, magnetic resonance imaging, computed tomography and positron emission tomography. We anticipate that this review will provide a comprehensive summary of pyrrole-derived nanoparticles and provide a guideline for their further development.
Collapse
Affiliation(s)
- Parinaz Fathi
- Departments of Bioengineering, Materials Science & Engineering & Beckman Institute, University of Illinois, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
| | - Dipanjan Pan
- Departments of Bioengineering, Materials Science & Engineering & Beckman Institute, University of Illinois, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
- Departments of Diagnostic Radiology & Nuclear Medicine & Pediatrics, University of Maryland Baltimore, Health Sciences Facility III, 670 W Baltimore St., Baltimore, MD 21201, USA
- Department of Chemical, Biochemical & Environmental Engineering, University of Maryland Baltimore County, Interdisciplinary Health Sciences Facility, 1000 Hilltop Circle Baltimore, MD 21250, USA
| |
Collapse
|
3
|
Shivaee A, Sadeghi Kalani B, Talebi M, Darban-Sarokhalil D. Does biofilm formation have different pathways in Staphylococcus aureus?. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 22:1147-1152. [PMID: 31998455 PMCID: PMC6885392 DOI: 10.22038/ijbms.2019.34888.8281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Objective(s): Biofilm formation is one of the most important factors in the development of infections caused by Staphylococcus aureus. In this study, the expression levels of genes responsible for biofilm formation were studied in methicillin sensitive and methicillin resistant S. aureus. Materials and Methods: A total of 100 meticillin-resistant s.aureus (MRSA) and meticillin-sensetive s.aureus (MSSA) isolates were studied. Bacterial biofilm formation was evaluated phenotypically using microtiter plate method. Real-time PCR tests were conducted to determine the expression levels of genes involved in biofilm formation. Results: Quantitative biofilm formation test was repeated three times for each specimen. The prevalence of weak, medium, and strong biofilm producers were 16%, 49%, and 35%, respectively. In MSSA isolates, expression levels of ica genes increased compared to the fnbA, fnbB, clfA and clfB genes. These results were different in MRSA isolates, and ica genes showed a decreased gene expression levels compared to the aforementioned genes. Conclusion: Considering the results of this study, clf genes probably contribute to the same extent in both MRSA and MSSA isolates, and there is probably no significant difference in the role of these genes in these isolates. In addition, the results of this study indicated that MRSA may not use the conventional route for biofilm formation and may use independent pathways through Polysaccharide intercellular adhesion (PIA).
Collapse
Affiliation(s)
- Ali Shivaee
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behrooz Sadeghi Kalani
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Carruthers NJ, Stemmer PM, Media J, Swartz K, Wang X, Aube N, Hamann MT, Valeriote F, Shaw J. The anti-MRSA compound 3-O-alpha-L-(2″,3″-di-p-coumaroyl)rhamnoside (KCR) inhibits protein synthesis in Staphylococcus aureus. J Proteomics 2019; 210:103539. [PMID: 31629958 DOI: 10.1016/j.jprot.2019.103539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/05/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Methicillin-resistant S aureus (MRSA) contributes to patient mortality and extended hospital stays. 3-O-alpha-L-(2″,3″-di-p-coumaroyl)rhamnoside (KCR) is a natural product antibiotic that is effective against MRSA but has no known mechanism of action (MOA). We used proteomics to identify the MOA for KCR. Methicillin sensitive S aureus and a mixture of four KCR stereoisomers were tested. A time-kill assay was used to choose a 4 h treatment using KCR at 5× its MIC for proteomic analysis. S aureus was treated in triplicate with KCR, oxacillin or vehicle and quantitative proteomic analysis was carried out using isobaric tags and mass spectrometry. 1190 proteins were identified and 552 were affected by KCR (q < 0.01). Ontology analysis identified 6 distinct translation-related categories that were affected by KCR (PIANO, 10% false-discovery rate) including structural constituent of ribosome, translation, rRNA binding, tRNA binding, tRNA processing and aminoacyl-tRNA ligase activity. Median fold changes (KCR vs Control) for small and large ribosomal components were 1.46 and 1.43 respectively. KCR inhibited the production of luciferase protein in an in vitro assay (IC50 39.6 μg/ml). Upregulation of translation-related proteins in response to KCR indicates that KCR acts to disrupt S aureus protein synthesis. This was confirmed with an in vitro transcription/translation assay. SIGNIFICANCE: Methicillin-resistant S aureus (MRSA) contributes to patient mortality and extended hospital stays. 3-O-alpha-L-(2″,3″-di-p-coumaroyl)rhamnoside (KCR) is a natural product antibiotic that is effective against MRSA but has no known mechanism of action (MOA). Using proteomic analysis we determined that KCR acts by inhibiting protein synthesis. KCR is an exciting novel antibiotic and this work represents an important step in its development towards clinical use.
Collapse
Affiliation(s)
- Nicholas J Carruthers
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201, USA; Wayne State University, Institute of Environmental Health Sciences, 2309 Scott Hall, 540 E Canfield Ave, Detroit, MI 48202, United States of America.
| | - Paul M Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Joe Media
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48201, USA.
| | - Ken Swartz
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48201, USA.
| | - Xiaojuan Wang
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nicholas Aube
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Mark T Hamann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Frederick Valeriote
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48201, USA.
| | - Jiajiu Shaw
- Henry Ford Health System, Detroit, MI, USA; 21st Century Therapeutics, Detroit, MI 48201, USA
| |
Collapse
|
5
|
Shivaee A, Sadeghi Kalani B, Talebi M, Darban-Sarokhalil D. Does biofilm formation have different pathways in Staphylococcus aureus?. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019. [PMID: 31998455 DOI: 10.22038/ijbms2019348888281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
OBJECTIVES Biofilm formation is one of the most important factors in the development of infections caused by Staphylococcus aureus. In this study, the expression levels of genes responsible for biofilm formation were studied in methicillin sensitive and methicillin resistant S. aureus. MATERIALS AND METHODS A total of 100 meticillin-resistant s.aureus (MRSA) and meticillin-sensetive s.aureus (MSSA) isolates were studied. Bacterial biofilm formation was evaluated phenotypically using microtiter plate method. Real-time PCR tests were conducted to determine the expression levels of genes involved in biofilm formation. RESULTS Quantitative biofilm formation test was repeated three times for each specimen. The prevalence of weak, medium, and strong biofilm producers were 16%, 49%, and 35%, respectively. In MSSA isolates, expression levels of ica genes increased compared to the fnbA, fnbB, clfA and clfB genes. These results were different in MRSA isolates, and ica genes showed a decreased gene expression levels compared to the aforementioned genes. CONCLUSION Considering the results of this study, clf genes probably contribute to the same extent in both MRSA and MSSA isolates, and there is probably no significant difference in the role of these genes in these isolates. In addition, the results of this study indicated that MRSA may not use the conventional route for biofilm formation and may use independent pathways through Polysaccharide intercellular adhesion (PIA).
Collapse
Affiliation(s)
- Ali Shivaee
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behrooz Sadeghi Kalani
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Microbial Biotechnology Research Center, Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Distinct virulent network between healthcare- and community-associated Staphylococcus aureus based on proteomic analysis. Clin Proteomics 2018; 15:2. [PMID: 29321722 PMCID: PMC5757299 DOI: 10.1186/s12014-017-9178-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/22/2017] [Indexed: 12/18/2022] Open
Abstract
Background Staphylococcus aureus (S. aureus or SA) is a leading cause of healthcare-associated (HA-) and community-associated (CA) infection. HA-SA isolates usually cause nosocomial pneumonia, bloodstream infections, catheter-related urinary tract infections, etc. On the other hand, CA-SA isolates usually cause highly fatal diseases, such as SSTIs as well as post influenza necrotic hemorrhagic pneumonia. The differences of the infection types are partially due to the unique characteristics between HA-SA and CA-SA isolates. For example, HA-SA isolates showed strong adherence to host epithelial cells, while CA-SA isolates displayed higher virulence due to the increased activity of the important quorum-sensing system accessory gene regulator (agr). Thus, the aim of this study was to characterize the proteomic difference between HA-SA and CA-SA lineage. Methods In this study, the extracted peptides from those representative strains were analyzed by LC-MS/MS. The protein-protein interaction network was constructed by bioinformatics and their expressions were verified by RT-PCR and Western blot. Results We demonstrated that Agr system (AgrA and AgrC) and its interactive factors (PhoP, SrrB, YycG, SarX, SigB and ClpP) based on the protein–protein interaction network were expressed significantly higher in the epidemic Chinese CA-SA lineage ST398 compared to HA-SA lineage ST239 by LC-MS/MS. We further verified the increased transcription of all these genes in ST398 by RT-PCR, suggesting that the higher expression of these genes/proteins probably play role in the acute infection of CA-SA. Moreover, surface-related proteins (FnbpA, SpA, Atl, ClfA, IsaA, IsaB, LtaS, SsaA and Cna) that are repressed by the Agr system have significantly higher expression in the epidemic Chinese HA-SA clone ST239 in comparison to CA-SA lineage ST398 by LC-MS/MS. Furthermore, we confirmed the significantly increased expression of two important adhesive proteins (Atl and ClfA) in ST239 by Western blot, which may contribute to the durative infection of HA-SA. Conclusion The results suggest that the different proteomic profile, at least partially, contribute to the pathogenic differences between HA-SA and CA-SA. Electronic supplementary material The online version of this article (10.1186/s12014-017-9178-5) contains supplementary material, which is available to authorized users.
Collapse
|
7
|
Liu PF, Cheng JS, Sy CL, Huang WC, Yang HC, Gallo RL, Huang CM, Shu CW. IsaB Inhibits Autophagic Flux to Promote Host Transmission of Methicillin-Resistant Staphylococcus aureus. J Invest Dermatol 2015; 135:2714-2722. [PMID: 26134948 PMCID: PMC4641007 DOI: 10.1038/jid.2015.254] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 06/17/2015] [Indexed: 12/19/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has emerged as a major nosocomial pathogen that is widespread in both health-care facilities and in the community at large, as a result of direct host-to-host transmission. Several virulence factors are associated with pathogen transmission to naive hosts. Immunodominant surface antigen B (IsaB) is a virulence factor that helps Staphylococcus aureus to evade the host defense system. However, the mechanism of IsaB on host transmissibility remains unclear. We found that IsaB expression was elevated in transmissible MRSA. Wild-type isaB strains inhibited autophagic flux to promote bacterial survival and elicit inflammation in THP-1 cells and mouse skin. MRSA isolates with increased IsaB expression showed decreased autophagic flux, and the MRSA isolate with the lowest IsaB expression showed increased autophagic flux. In addition, recombinant IsaB rescued the virulence of the isaB deletion strain and increased the group A streptococcus (GAS) virulence in vivo. Together, these results reveal that IsaB diminishes autophagic flux, thereby allowing MRSA to evade host degradation. These findings suggest that IsaB is a suitable target for preventing or treating MRSA infection.
Collapse
Affiliation(s)
- Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Medicine, Division of Dermatology, University of California, San Diego, USA
| | - Jin-Shiung Cheng
- Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Cheng-Len Sy
- Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Chun Huang
- Cardiovascular Medical Center, Kaohsiung Veterans General Hospital, Kaohsiung City
- School of Medicine, National Yang-Ming University, Taipei City
| | - Hsiu-Chen Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Richard L. Gallo
- Department of Medicine, Division of Dermatology, University of California, San Diego, USA
- VA San Diego Healthcare Center, San Diego, USA
| | - Chun-Ming Huang
- Department of Medicine, Division of Dermatology, University of California, San Diego, USA
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| |
Collapse
|
8
|
Zhang Y, Valeriote F, Swartz K, Chen B, Hamann MT, Rodenburg DL, McChesney JD, Shaw J. HPLC Plasma Assay of a Novel Anti-MRSA Compound, Kaempferol-3-O-Alpha-L-(2",3"-di-p-coumaroyl)rhamnoside, from Sycamore Leaves. Nat Prod Commun 2015; 10:1383-1386. [PMID: 26434123 PMCID: PMC4895204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a serious pathogen that is resistant to current antibiotic therapy. Thus, there is an urgent need for novel antimicrobial agents that can effectively combat these new strains of drug-resistant "superbugs". Recently, fractionation of an extract from Platanus occidentalis (American sycamore) leaves produced an active kaempferol molecule, 3-O-alpha-L-(2",3"-di-p-coumaroyl)rhamnoside (KCR), in four isomeric forms; all four isomers exhibit potent anti-MRSA activity. In order to further the preclinical development of KCR as a new antibiotic class, we developed and validated a simple analytical method for assaying KCR plasma concentration. Because KCR will be developed as a new drug, although comprising four stereoisomers, the analytical method was devised to assay the total amount of all four isomers. In the present work, both a plasma processing procedure and an HPLC method have been developed and validated. Mouse plasma containing KCR was first treated with ethanol and then centrifuged. The supernatant was dried, suspended in ethanol, centrifuged, and the supernatant was injected into an HPLC system comprising a Waters C18, a mobile phase composing methanol, acetonitrile, and trifluoroacetic acid and monitored at 313 nm. The method was validated by parameters including a good linear correlation, a limit of quantification of 0.27 microg/mL, and high accuracy. In summary, this method allows a rapid analysis of KCR in the plasma samples for pharmacokinetics studies.
Collapse
Affiliation(s)
- Yiguan Zhang
- Henry Ford Health System, 440 Burroughs St, Detroit, MI 48202, USA
| | | | - Kenneth Swartz
- Henry Ford Health System, 440 Burroughs St, Detroit, MI 48202, USA
| | - Ben Chen
- 21st Century Therapeutics, 1366 Hilton Rd, Ferndale, MI 48220, USA
| | - Mark T. Hamann
- Oxford Pharmaceutical Development, Oxford, MS 38655, USA
| | | | | | - Jiajiu Shaw
- Henry Ford Health System, 440 Burroughs St, Detroit, MI 48202, USA
- 21st Century Therapeutics, 1366 Hilton Rd, Ferndale, MI 48220, USA
| |
Collapse
|
9
|
Land AD, Hogan P, Fritz S, Levin PA. Phenotypic Variation Is Almost Entirely Independent of the Host-Pathogen Relationship in Clinical Isolates of S. aureus. PLoS One 2015; 10:e0129670. [PMID: 26098551 PMCID: PMC4476556 DOI: 10.1371/journal.pone.0129670] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 05/12/2015] [Indexed: 11/18/2022] Open
Abstract
Background A key feature of Staphylococcus aureus biology is its ability to switch from an apparently benign colonizer of ~30% of the population to a cutaneous pathogen, to a deadly invasive pathogen. Little is known about the mechanisms driving this transition or the propensity of different S. aureus strains to engender different types of host-pathogen interactions. At the same time, significant weight has been given to the role of specific in vitro phenotypes in S. aureus virulence. Biofilm formation, hemolysis and pigment formation have all been associated with virulence in mice. Design To determine if there is a correlation between in vitro phenotype and the three types of host-pathogen relationships commonly exhibited by S. aureus in the context of its natural human host, we assayed 300 clinical isolates for phenotypes implicated in virulence including hemolysis, sensitivity to autolysis, and biofilm formation. For comparative purposes, we also assayed phenotype in 9 domesticated S. aureus strains routinely used for analysis of virulence determinants in laboratory settings. Results Strikingly, the clinical strains exhibited significant phenotypic uniformity in each of the assays evaluated in this study. One exception was a small, but significant, correlation between an increased propensity for biofilm formation and isolation from skin and soft tissue infections (SSTIs). In contrast, we observed a high degree of phenotypic variation between common laboratory strains that exhibit virulence in mouse models. These data suggest the existence of significant evolutionary pressure on the S. aureus genome and highlight a role for host factors as a strong determinant of the host-pathogen relationship. In addition, the high degree of variation between laboratory strains emphasizes the need for caution when applying data obtained in one lab strain to the analysis of another.
Collapse
Affiliation(s)
- Adrian D. Land
- Department of Biology, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Patrick Hogan
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Stephanie Fritz
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Petra Anne Levin
- Department of Biology, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
10
|
Kamenyeva O, Boularan C, Kabat J, Cheung GYC, Cicala C, Yeh AJ, Chan JL, Periasamy S, Otto M, Kehrl JH. Neutrophil recruitment to lymph nodes limits local humoral response to Staphylococcus aureus. PLoS Pathog 2015; 11:e1004827. [PMID: 25884622 PMCID: PMC4401519 DOI: 10.1371/journal.ppat.1004827] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 03/23/2015] [Indexed: 12/25/2022] Open
Abstract
Neutrophils form the first line of host defense against bacterial pathogens. They are rapidly mobilized to sites of infection where they help marshal host defenses and remove bacteria by phagocytosis. While splenic neutrophils promote marginal zone B cell antibody production in response to administered T cell independent antigens, whether neutrophils shape humoral immunity in other lymphoid organs is controversial. Here we investigate the neutrophil influx following the local injection of Staphylococcus aureus adjacent to the inguinal lymph node and determine neutrophil impact on the lymph node humoral response. Using intravital microscopy we show that local immunization or infection recruits neutrophils from the blood to lymph nodes in waves. The second wave occurs temporally with neutrophils mobilized from the bone marrow. Within lymph nodes neutrophils infiltrate the medulla and interfollicular areas, but avoid crossing follicle borders. In vivo neutrophils form transient and long-lived interactions with B cells and plasma cells, and their depletion augments production of antigen-specific IgG and IgM in the lymph node. In vitro activated neutrophils establish synapse- and nanotube-like interactions with B cells and reduce B cell IgM production in a TGF- β1 dependent manner. Our data reveal that neutrophils mobilized from the bone marrow in response to a local bacterial challenge dampen the early humoral response in the lymph node. Highly antibiotic resistant Staphylococcus aureus (S. aureus) are an important human pathogen and major cause of hospital acquired infections. An early host defense mechanism against bacterial infection is neutrophil recruitment, which helps eliminate the bacteria at the site of invasion. However, unless quickly neutralized, pathogens such as S. aureus can gain access to nearby lymph nodes via draining lymphatics. Lymph nodes protect the host by mobilizing additional resources that limit further pathogen dissemination. These include recruitment of neutrophils to the lymph node to directly target pathogens and the initiation of adaptive immune mechanisms, such as the humoral immune response, which transforms B lymphocytes capable of making pathogen specific antibodies into antibody producing plasma cells. Using a mouse model that allows direct visualization of lymphocytes, neutrophils, and fluorescently-labeled S. aureus in lymph nodes, we document the rapid appearance of bacteria in the lymph node following local S. aureus infection. We characterize the dynamic influx of neutrophils that occurs as a consequence and reveal direct B cell-neutrophil interactions within the lymph node parenchyma. We find that while lymph node neutrophils rapidly engage bacteria, they limit the subsequent humoral immune response likely by producing Transforming Growth Factor-β1, a factor known to limit B cell responses. These finding have important implication for our understanding of B cell responses against potent pathogens such as S. aureus and for the design of effective vaccines.
Collapse
Affiliation(s)
- Olena Kamenyeva
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cedric Boularan
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gordon Y C Cheung
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Claudia Cicala
- Immunopathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anthony J Yeh
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - June L Chan
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Saravanan Periasamy
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
11
|
Yadav MK, Chae SW, Im GJ, Chung JW, Song JJ. Eugenol: a phyto-compound effective against methicillin-resistant and methicillin-sensitive Staphylococcus aureus clinical strain biofilms. PLoS One 2015; 10:e0119564. [PMID: 25781975 PMCID: PMC4364371 DOI: 10.1371/journal.pone.0119564] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 01/14/2015] [Indexed: 12/29/2022] Open
Abstract
Background Inhibition and eradication of Staphylococcus aureus biofilms with conventional antibiotic is difficult, and the treatment is further complicated by the rise of antibiotic resistance among staphylococci. Consequently, there is a need for novel antimicrobials that can treat biofilm-related infections and decrease antibiotics burden. Natural compounds such as eugenol with anti-microbial properties are attractive agents that could reduce the use of conventional antibiotics. In this study we evaluated the effect of eugenol on MRSA and MSSA biofilms in vitro and bacterial colonization in vivo. Methods and Results Effect of eugenol on in vitro biofilm and in vivo colonization were studied using microtiter plate assay and otitis media-rat model respectively. The architecture of in vitro biofilms and in vivo colonization of bacteria was viewed with SEM. Real-time RT-PCR was used to study gene expression. Check board method was used to study the synergistic effects of eugenol and carvacrol on established biofilms. Eugenol significantly inhibited biofilms growth of MRSA and MSSA in vitro in a concentration-dependent manner. Eugenol at MIC or 2×MIC effectively eradicated the pre-established biofilms of MRSA and MSSA clinical strains. In vivo, sub-MIC of eugenol significantly decreased 88% S. aureus colonization in rat middle ear. Eugenol was observed to damage the cell-membrane and cause a leakage of the cell contents. At sub-inhibitory concentration, it decreases the expression of biofilm-and enterotoxin-related genes. Eugenol showed a synergistic effect with carvacrol on the eradication of pre-established biofilms. Conclusion/Major Finding This study demonstrated that eugenol exhibits notable activity against MRSA and MSSA clinical strains biofilms. Eugenol inhibited biofilm formation, disrupted the cell-to-cell connections, detached the existing biofilms, and killed the bacteria in biofilms of both MRSA and MSSA with equal effectiveness. Therefore, eugenol may be used to control or eradicate S. aureus biofilm-related infections.
Collapse
Affiliation(s)
- Mukesh Kumar Yadav
- Institute for Medical Device Clinical Trials, Korea University College of Medicine, Seoul, South Korea
- Department of Otolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Sung-Won Chae
- Department of Otolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Gi Jung Im
- Department of Otolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Jae-Woo Chung
- Laboratory of Medicine, Dongguk University Ilsan Hospital, Goyang, Gyeonggi, South Korea
| | - Jae-Jun Song
- Department of Otolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
- * E-mail:
| |
Collapse
|
12
|
Simonetti O, Cirioni O, Mocchegiani F, Cacciatore I, Silvestri C, Baldassarre L, Orlando F, Castelli P, Provinciali M, Vivarelli M, Fornasari E, Giacometti A, Offidani A. The efficacy of the quorum sensing inhibitor FS8 and tigecycline in preventing prosthesis biofilm in an animal model of staphylococcal infection. Int J Mol Sci 2013; 14:16321-16332. [PMID: 23965956 PMCID: PMC3759913 DOI: 10.3390/ijms140816321] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/01/2013] [Accepted: 07/26/2013] [Indexed: 12/15/2022] Open
Abstract
We investigated the efficacy of tigecycline and FS8, alone or combined, in preventing prosthesis biofilm in a rat model of staphylococcal vascular graft infection. Graft infections were established in the back subcutaneous tissue of adult male Wistar rats by implantation of Dacron prostheses followed by topical inoculation with 2 x 107 colony-forming units of Staphylococcus aureus, strain Smith diffuse. The study included a control group, a contaminated group that did not receive any antibiotic prophylaxis, and three contaminated groups that received: (i) intraperitoneal tigecycline, (ii) FS8-soaked graft, and (iii) tigecycline plus FS8-soaked graft, respectively. Each group included 15 animals. The infection burden was evaluated by using sonication and quantitative agar culture. Moreover, an in vitro binding-study was performed to quantify the how much FS8 was coated to the surface of the prosthesis. Tigecycline, combined with FS8, against the adherent bacteria showed MICs (2.00 mg/L) and MBCs (4.00 mg/L) four-fold lower with respect to tigecycline alone in in vitro studies. The rat groups treated with tigecycline showed the lowest bacterial numbers (4.4 x 104 ± 1.2 x 104 CFU/mL). The FS8-treated group showed a good activity and significant differences compared to control group with bacterial numbers of 6.8 x 104 ± 2.0 x 104 CFU/mL. A stronger inhibition of bacterial growth was observed in rats treated with a combined FS8 and tigecycline therapy than in those that were singly treated with bacterial numbers of 101 CFU/mL graft. In conclusion, the ability to affect biofilm formation as well, its property to be an antibiotic enhancer suggests FS8 as alternative or additional agent to use in conjunction with conventional antimicrobial for prevention of staphylococcal biofilm related infection.
Collapse
Affiliation(s)
- Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mail:
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Federico Mocchegiani
- Centre for Abdominal Surgery and Organ Transplant, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (F.M.); (M.V.)
| | - Ivana Cacciatore
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara 66013, Italy; E-Mails: (I.C.); (L.B.); (E.F.)
| | - Carmela Silvestri
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Leonardo Baldassarre
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara 66013, Italy; E-Mails: (I.C.); (L.B.); (E.F.)
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona 60100, Italy; E-Mails: (F.O.); (M.P.)
| | - Pamela Castelli
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Mauro Provinciali
- Experimental Animal Models for Aging Units, Research Department, I.N.R.C.A. I.R.R.C.S., Ancona 60100, Italy; E-Mails: (F.O.); (M.P.)
| | - Marco Vivarelli
- Centre for Abdominal Surgery and Organ Transplant, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (F.M.); (M.V.)
| | - Erika Fornasari
- Department of Pharmacy, Università degli Studi G. D’Annunzio, Chieti-Pescara 66013, Italy; E-Mails: (I.C.); (L.B.); (E.F.)
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mails: (O.C.); (C.S.); (P.C.); (A.G.)
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche–Ospedali Riuniti, Ancona 60020, Italy; E-Mail:
| |
Collapse
|
13
|
Yadav PK, Singh G, Gautam B, Singh S, Yadav M, Srivastav U, Singh B. Molecular modeling, dynamics studies and virtual screening of Fructose 1, 6 biphosphate aldolase-II in community acquired- methicillin resistant Staphylococcus aureus (CA-MRSA). Bioinformation 2013; 9:158-64. [PMID: 23423142 PMCID: PMC3569604 DOI: 10.6026/97320630009158] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 01/15/2013] [Indexed: 11/23/2022] Open
Abstract
Community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) has recently emerged as a nosocomial pathogen to the
community which commonly causes skin and soft-tissue infections (SSTIs). This strain (MW2) has now become resistant to the
most of the beta-lactam antibiotics; therefore it is the urgent need to identify the novel drug targets. Recently fructose 1,6
biphosphate aldolase-II (FBA) has been identified as potential drug target in CA-MRSA. The FBA catalyses the retro-ketolic
cleavage of fructose-1,6-bisphosphate (FBP) to yield dihydroxyacetone phosphate (DHAP) and glyceraldehyde-3-phosphate (G3P)
in glycolytic pathway. In the present research work the 3D structure of FBA was predicted using the homology modeling method
followed by validation. The molecular dynamics simulation (MDS) of the predicted model was carried out using the 2000 ps time
scale and 1000000 steps. The MDS results suggest that the modeled structure is stable. The predicted model of FBA was used for
virtual screening against the NCI diversity subset-II ligand databases which contain 1364 compounds. Based on the docking energy
scores, it was found that top four ligands i.e. ZINC01690699, ZINC13154304, ZINC29590257 and ZINC29590259 were having lower
energy scores which reveal higher binding affinity towards the active site of FBA. These ligands might act as potent inhibitors for
the FBA so that the menace of antimicrobial resistance in CA-MRSA can be conquered. However, pharmacological studies are
required to confirm the inhibitory activity of these ligands against the FBA in CA-MRSA.
Collapse
Affiliation(s)
- Pramod Kumar Yadav
- Department of Computational Biology & Bioinformatics, Sam Higginbottom Institute of Agriculture, Technology & Sciences (Deemed University), Allahabad-211007, India
| | | | | | | | | | | | | |
Collapse
|
14
|
Ma Y, Xu Y, Yestrepsky BD, Sorenson RJ, Chen M, Larsen SD, Sun H. Novel inhibitors of Staphylococcus aureus virulence gene expression and biofilm formation. PLoS One 2012; 7:e47255. [PMID: 23077578 PMCID: PMC3471953 DOI: 10.1371/journal.pone.0047255] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 09/10/2012] [Indexed: 12/27/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen and one of the more prominent pathogens causing biofilm related infections in clinic. Antibiotic resistance in S. aureus such as methicillin resistance is approaching an epidemic level. Antibiotic resistance is widespread among major human pathogens and poses a serious problem for public health. Conventional antibiotics are either bacteriostatic or bacteriocidal, leading to strong selection for antibiotic resistant pathogens. An alternative approach of inhibiting pathogen virulence without inhibiting bacterial growth may minimize the selection pressure for resistance. In previous studies, we identified a chemical series of low molecular weight compounds capable of inhibiting group A streptococcus virulence following this alternative anti-microbial approach. In the current study, we demonstrated that two analogs of this class of novel anti-virulence compounds also inhibited virulence gene expression of S. aureus and exhibited an inhibitory effect on S. aureus biofilm formation. This class of anti-virulence compounds could be a starting point for development of novel anti-microbial agents against S. aureus.
Collapse
Affiliation(s)
- Yibao Ma
- Department of Internal Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Yuanxi Xu
- Department of Internal Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Bryan D. Yestrepsky
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Roderick J. Sorenson
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Meng Chen
- Nanova, Inc., Columbia, Missouri, United States of America
| | - Scott D. Larsen
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (HS); (SDL)
| | - Hongmin Sun
- Department of Internal Medicine, University of Missouri, Columbia, Missouri, United States of America
- * E-mail: (HS); (SDL)
| |
Collapse
|
15
|
Conserved small protein associates with the multidrug efflux pump AcrB and differentially affects antibiotic resistance. Proc Natl Acad Sci U S A 2012; 109:16696-701. [PMID: 23010927 DOI: 10.1073/pnas.1210093109] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The AcrAB-TolC multidrug efflux pump confers resistance to a wide variety of antibiotics and other compounds in Escherichia coli. Here we show that AcrZ (formerly named YbhT), a 49-amino-acid inner membrane protein, associates with the AcrAB-TolC complex. Co-purification of AcrZ with AcrB, in the absence of both AcrA and TolC, two-hybrid assays and suppressor mutations indicate that this interaction occurs through the inner membrane protein AcrB. The highly conserved acrZ gene is coregulated with acrAB through induction by the MarA, Rob, and SoxS transcription regulators. In addition, mutants lacking AcrZ are sensitive to many, but not all, of the antibiotics transported by AcrAB-TolC. This differential antibiotic sensitivity suggests that AcrZ may enhance the ability of the AcrAB-TolC pump to export certain classes of substrates.
Collapse
|
16
|
Rakette S, Donat S, Ohlsen K, Stehle T. Structural analysis of Staphylococcus aureus serine/threonine kinase PknB. PLoS One 2012; 7:e39136. [PMID: 22701750 PMCID: PMC3372466 DOI: 10.1371/journal.pone.0039136] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 05/18/2012] [Indexed: 11/29/2022] Open
Abstract
Effective treatment of infections caused by the bacterium Staphylococcus aureus remains a worldwide challenge, in part due to the constant emergence of new strains that are resistant to antibiotics. The serine/threonine kinase PknB is of particular relevance to the life cycle of S. aureus as it is involved in the regulation of purine biosynthesis, autolysis, and other central metabolic processes of the bacterium. We have determined the crystal structure of the kinase domain of PknB in complex with a non-hydrolyzable analog of the substrate ATP at 3.0 Å resolution. Although the purified PknB kinase is active in solution, it crystallized in an inactive, autoinhibited state. Comparison with other bacterial kinases provides insights into the determinants of catalysis, interactions of PknB with ligands, and the pathway of activation.
Collapse
Affiliation(s)
- Sonja Rakette
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Stefanie Donat
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Knut Ohlsen
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
17
|
Morino H, Fukuda T, Miura T, Shibata T. Effect of low-concentration chlorine dioxide gas against bacteria and viruses on a glass surface in wet environments. Lett Appl Microbiol 2011; 53:628-34. [PMID: 21950421 PMCID: PMC7199474 DOI: 10.1111/j.1472-765x.2011.03156.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aims: To evaluate the efficacy of low‐concentration chlorine dioxide (ClO2) gas against model microbes in the wet state on a glass surface. Methods and Results: We set up a test room (39 m3) and the ClO2 gas was produced by a ClO2 gas generator that continuously releases a constant low‐concentration ClO2 gas. Influenza A virus (Flu‐A), feline calicivirus (FCV), Staphylococcus aureus and Escherichia coli were chosen as the model microbes. The low‐concentration ClO2 gas (mean 0·05 ppmv, 0·14 mg m−3) inactivated Flu‐A and E. coli (>5 log10 reductions) and FCV and S. aureus (>2 log10 reductions) in the wet state on glass dishes within 5 h. Conclusions: The treatment of wet environments in the presence of human activity such as kitchens and bathrooms with the low‐concentration ClO2 gas would be useful for reducing the risk of infection by bacteria and viruses residing on the environmental hard surfaces without adverse effects. Significance and Impact of the Study: This study demonstrates that the low‐concentration ClO2 gas (mean 0·05 ppmv) inactivates various kinds of microbes such as Gram‐positive and Gram‐negative bacteria, enveloped and nonenveloped viruses in the wet state.
Collapse
Affiliation(s)
- H Morino
- Research and Development Department, Taiko Pharmaceutical Co., Ltd, Suita, Osaka, Japan.
| | | | | | | |
Collapse
|
18
|
Otto M. Panton-Valentine leukocidin antibodies for the treatment of MRSA skin infections? Expert Rev Anti Infect Ther 2011; 9:389-92. [PMID: 21504395 PMCID: PMC3106302 DOI: 10.1586/eri.11.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has recently caused epidemic outbreaks of community-associated (CA) skin infections. The infecting strains frequently contain the genes encoding the staphylococcal toxin, Panton-Valentine leukocidin (PVL). On that ground, the use of PVL-based vaccines has been proposed for the treatment of CA-MRSA infections, despite experimental and epidemiological evidence that does not support a major role of the PVL toxin in CA-MRSA skin disease. Hermos et al. show that antibodies to PVL do not protect from CA-MRSA skin infections in children, strongly suggesting that PVL-based immunization is of little benefit for this most frequent disease caused by CA-MRSA.
Collapse
Affiliation(s)
- Michael Otto
- Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, The National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Fehér D, Barlow R, McAtee J, Hemscheidt TK. Highly brominated antimicrobial metabolites from a marine Pseudoalteromonas sp. JOURNAL OF NATURAL PRODUCTS 2010; 73:1963-1966. [PMID: 20973551 PMCID: PMC2993825 DOI: 10.1021/np100506z] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Extracts of a marine Pseudoalteromonas sp. (CMMED 290) isolated from the surface of a nudibranch collected in Kaneohe Bay, Oahu, displayed significant antimicrobial activity against methicillin-resistant Staphylococcus aureus. Bioassay-guided fractionation of the lipophilic extract led to the isolation and structure elucidation of two new highly brominated compounds, 2,3,5,7-tetrabromobenzofuro[3,2-b]pyrrole (1) and 4,4',6-tribromo-2,2'-biphenol (2). In addition, we have identified the known compounds pentabromopseudilin and bromophene. We describe the isolation and structure elucidation of the compounds 1 and 2 together with their antimicrobial activities against methicillin-resistant Staphylococcus aureus.
Collapse
Affiliation(s)
| | | | | | - Thomas K. Hemscheidt
- To whom correspondence may be addressed. Phone: 1-808-956-6401; Fax: 1-808-956-5908;
| |
Collapse
|
20
|
Llarrull LI, Testero SA, Fisher JF, Mobashery S. The future of the β-lactams. Curr Opin Microbiol 2010; 13:551-7. [PMID: 20888287 DOI: 10.1016/j.mib.2010.09.008] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 09/08/2010] [Accepted: 09/09/2010] [Indexed: 11/15/2022]
Abstract
In the 80 years since their discovery the β-lactam antibiotics have progressed through structural generations, each in response to the progressive evolution of bacterial resistance mechanisms. The generational progression was driven by the ingenious, but largely empirical, manipulation of structure by medicinal chemists. Nonetheless, the true creative force in these efforts was Nature, and as the discovery of new β-lactams from Nature has atrophied while at the same time multi-resistant and opportunistic bacterial pathogens have burgeoned, the time for empirical drug discovery has passed. We concisely summarize recent developments with respect to bacterial resistance, the identity of the new β-lactams, and the emerging non-empirical strategies that will ensure that this incredible class of antibiotics has a future.
Collapse
Affiliation(s)
- Leticia I Llarrull
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | |
Collapse
|
21
|
Burnside K, Lembo A, de los Reyes M, Iliuk A, BinhTran NT, Connelly JE, Lin WJ, Schmidt BZ, Richardson AR, Fang FC, Tao WA, Rajagopal L. Regulation of hemolysin expression and virulence of Staphylococcus aureus by a serine/threonine kinase and phosphatase. PLoS One 2010; 5:e11071. [PMID: 20552019 PMCID: PMC2884019 DOI: 10.1371/journal.pone.0011071] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 05/14/2010] [Indexed: 02/02/2023] Open
Abstract
Exotoxins, including the hemolysins known as the alpha (alpha) and beta (beta) toxins, play an important role in the pathogenesis of Staphylococcus aureus infections. A random transposon library was screened for S. aureus mutants exhibiting altered hemolysin expression compared to wild type. Transposon insertions in 72 genes resulting in increased or decreased hemolysin expression were identified. Mutations inactivating a putative cyclic di-GMP synthetase and a serine/threonine phosphatase (Stp1) were found to reduce hemolysin expression, and mutations in genes encoding a two component regulator PhoR, LysR family transcriptional regulator, purine biosynthetic enzymes and a serine/threonine kinase (Stk1) increased expression. Transcription of the hla gene encoding alpha toxin was decreased in a Deltastp1 mutant strain and increased in a Deltastk1 strain. Microarray analysis of a Deltastk1 mutant revealed increased transcription of additional exotoxins. A Deltastp1 strain is severely attenuated for virulence in mice and elicits less inflammation and IL-6 production than the Deltastk1 strain. In vivo phosphopeptide enrichment and mass spectrometric analysis revealed that threonine phosphorylated peptides corresponding to Stk1, DNA binding histone like protein (HU), serine-aspartate rich fibrinogen/bone sialoprotein binding protein (SdrE) and a hypothetical protein (NWMN_1123) were present in the wild type and not in the Deltastk1 mutant. Collectively, these studies suggest that Stk1 mediated phosphorylation of HU, SrdE and NWMN_1123 affects S. aureus gene expression and virulence.
Collapse
Affiliation(s)
- Kellie Burnside
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| | - Annalisa Lembo
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| | - Melissa de los Reyes
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| | - Anton Iliuk
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Nguyen-Thao BinhTran
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| | - James E. Connelly
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| | - Wan-Jung Lin
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| | - Byron Z. Schmidt
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| | - Anthony R. Richardson
- Departments of Laboratory Medicine and Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Ferric C. Fang
- Departments of Laboratory Medicine and Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Weiguo Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Lakshmi Rajagopal
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Hospital Research Institute, Seattle, Washington, United States of America
| |
Collapse
|
22
|
Abstract
Meticillin-resistant Staphylococcus aureus (MRSA) is endemic in hospitals worldwide, and causes substantial morbidity and mortality. Health-care-associated MRSA infections arise in individuals with predisposing risk factors, such as surgery or presence of an indwelling medical device. By contrast, many community-associated MRSA (CA-MRSA) infections arise in otherwise healthy individuals who do not have such risk factors. Additionally, CA-MRSA infections are epidemic in some countries. These features suggest that CA-MRSA strains are more virulent and transmissible than are traditional hospital-associated MRSA strains. The restricted treatment options for CA-MRSA infections compound the effect of enhanced virulence and transmission. Although progress has been made towards understanding emergence of CA-MRSA, virulence, and treatment of infections, our knowledge remains incomplete. Here we review the most up-to-date knowledge and provide a perspective for the future prophylaxis or new treatments for CA-MRSA infections.
Collapse
Affiliation(s)
- Frank R DeLeo
- Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA.
| | | | | | | |
Collapse
|
23
|
Sheen TR, Ebrahimi CM, Hiemstra IH, Barlow SB, Peschel A, Doran KS. Penetration of the blood-brain barrier by Staphylococcus aureus: contribution of membrane-anchored lipoteichoic acid. J Mol Med (Berl) 2010; 88:633-9. [PMID: 20419283 DOI: 10.1007/s00109-010-0630-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 03/19/2010] [Accepted: 03/30/2010] [Indexed: 11/26/2022]
Abstract
Staphylococcus aureus is one of the most prevalent organisms responsible for nosocomial infections, and cases of community-acquired S. aureus infection have continued to increase despite widespread preventative measures. Pathologies attributed to S. aureus infection are diverse; ranging from dermal lesions to bacteremia, abscesses, and endocarditis. Reported cases of S. aureus-associated meningitis and brain abscesses have also increased in recent years, however, the precise mechanism whereby S. aureus leave the bloodstream and gain access to the central nervous system (CNS) are not known. Here we demonstrate for the first time that S. aureus efficiently adheres to and invades human brain microvascular endothelial cells (hBMEC), the single-cell layer which constitutes the blood-brain barrier (BBB). The addition of cytochalasin D, an actin microfilament aggregation inhibitor, strongly reduced bacterial invasion, suggesting an active hBMEC process is required for efficient staphylococcal uptake. Furthermore, mice injected with S. aureus exhibited significant levels of brain bacterial counts and histopathologic evidence of meningeal inflammation and brain abscess formation, indicating that S. aureus was able to breech the BBB in an experimental model of hematogenous meningitis. We found that a YpfP-deficient mutant, defective in lipoteichoic acid (LTA) membrane anchoring, exhibited a decreased ability to invade hBMEC and correlated to a reduced risk for the development of meningitis in vivo. Our results demonstrate that LTA-mediated penetration of the BBB may be a primary step in the pathogenesis of staphylococcal CNS disease.
Collapse
Affiliation(s)
- Tamsin R Sheen
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | | | | | | | | | | |
Collapse
|