1
|
Sharifi A, Mahmoudi P, Sobhani K, Ashengroph M. The Prevalence and Comparative Analysis of Adhesion and Biofilm-Related Genes in Staphylococcus aureus Isolates: A Network Meta-Analysis. Microbiol Immunol 2025; 69:104-113. [PMID: 39639432 DOI: 10.1111/1348-0421.13189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/26/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Staphylococcus aureus is a versatile pathogen capable of causing a wide range of infections, from minor skin infections to life-threatening invasive diseases. The pathogenicity of S. aureus is attributed to its ability to produce various virulence factors, including adhesion and biofilm-related proteins. Understanding the prevalence and distribution of these genes among S. aureus isolates from different sources is crucial for devising effective strategies to combat biofilm-associated contamination. In this study, we conducted a comprehensive network meta-analysis to assess the prevalence of adhesion and biofilm-related genes in S. aureus isolates and investigate the impact of the isolate source on their occurrence. A systematic search of multiple databases was performed, and a total of 53 relevant studies were included. The prevalence of adhesion and biofilm-related genes in S. aureus isolates was determined, with the highest prevalence observed for clfB (p-estimate = 85.4, 95% confidence interval [CI] 78-90.6), followed by eno (p-estimate = 81.1, 95% CI 61.7-91.9), and icaD (p-estimate = 77, 95% CI 68.6-83.6). Conversely, bap and bbp genes exhibited the lowest prevalence rates (p-estimate = 6.7 and 18.7, respectively). The network meta-analysis allowed us to examine the pairwise co-study of adhesion and biofilm-related genes in S. aureus isolates. The most frequently co-studied gene pairs were icaA-icaD (30 times) and fnbA-fnbB (25 times). Subgroup analysis showed that the occurrence of icaC and icaB genes was significantly lower in animal isolates compared to human and food isolates (p < 0.05). It is worth noting that there was limited data available for the analysis of sasG, bbp, bap, eno, and fib genes. In conclusion, the study revealed varying prevalence rates of adhesion and biofilm-related genes in S. aureus isolates. Genes such as clfB, eno, and icaD were found to be highly prevalent, while bap and bbp were less common. Limited existing data on the prevalence of genes like sasG, bbp, bap, eno, and fib highlights the need for further research to determine their exact prevalence rates. Our results contribute to a better understanding of S. aureus pathogenesis and can facilitate the development of effective strategies for the prevention and treatment of S. aureus infections.
Collapse
Affiliation(s)
- Aram Sharifi
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj, Kurdistan, Iran
| | - Peyman Mahmoudi
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj, Kurdistan, Iran
| | - Keyvan Sobhani
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj, Kurdistan, Iran
| | - Morahem Ashengroph
- Department of Biology, Faculty of Science, University of Kurdistan, Sanandaj, Kurdistan, Iran
| |
Collapse
|
2
|
Canè C, Gallucci N, Amoresano A, Fontanarosa C, Paduano L, De Gregorio E, Duilio A, Di Somma A. The antimicrobial peptide Temporin-L induces vesicle formation and reduces the virulence in S. aureus. Biochem Biophys Rep 2024; 39:101808. [PMID: 39238505 PMCID: PMC11375239 DOI: 10.1016/j.bbrep.2024.101808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 06/06/2024] [Accepted: 08/04/2024] [Indexed: 09/07/2024] Open
Abstract
The evolution of methicillin-resistant Staphylococcus aureus (MRSA) has required the development of new antimicrobial agents and new approaches to prevent and overcome drug resistance. AntiMicrobial Peptides (AMPs) represent promising alternatives due to their rapid bactericidal activity and their broad-spectrum of action against a wide range of microorganisms. The amphibian Temporins constitute a well-known family of AMPs with high antibacterial properties against both Gram-positive and Gram-negative bacteria. In this paper, we evaluated the in vivo effect of Temp-L on S. aureus performing morphological studies using Transmission Electron Microscopy (TEM) that revealed the occurrence of protrusions from the cell surface. The formation of vesicle-like structure was confirmed by Dynamic Light Scattering (DLS). The global effect of Temp-L on Staphylococcus aureus (S. aureus) was deeply investigated by differential proteomics leading to the identification of up-regulated proteins involved in the synthesis of the cell membrane and fatty acids, and down-regulated virulence factors. GC-MS analysis suggested a possible protective response mechanism implemented by the bacterium after treatment with Temp-L, as the synthesis of fatty acids was increased. Adhesion and invasion assays on eukaryotic cells confirmed a reduced virulence of S. aureus following treatment with Temp-L. These results suggested the targeting of virulence factors as novel strategy to replace traditional antimicrobial agents that can be used to treat infections, especially infections caused by the resistant pathogen S. aureus.
Collapse
Affiliation(s)
- Carolina Canè
- Department of Chemical Sciences, University of Naples "Federico II" Via Cinthia 4, 80126, Napoli, Italy
| | - Noemi Gallucci
- Department of Chemical Sciences, University of Naples "Federico II" Via Cinthia 4, 80126, Napoli, Italy
| | - Angela Amoresano
- Department of Chemical Sciences, University of Naples "Federico II" Via Cinthia 4, 80126, Napoli, Italy
- National Institute of Biostructures and Biosystems (INBB), Viale Medaglie d'Oro 305, 00136, Rome, Italy
| | - Carolina Fontanarosa
- Department of Chemical Sciences, University of Naples "Federico II" Via Cinthia 4, 80126, Napoli, Italy
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples "Federico II" Via Cinthia 4, 80126, Napoli, Italy
| | - Eliana De Gregorio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131, Naples, Italy
| | - Angela Duilio
- Department of Chemical Sciences, University of Naples "Federico II" Via Cinthia 4, 80126, Napoli, Italy
- National Institute of Biostructures and Biosystems (INBB), Viale Medaglie d'Oro 305, 00136, Rome, Italy
| | - Angela Di Somma
- Department of Chemical Sciences, University of Naples "Federico II" Via Cinthia 4, 80126, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80126, Napoli, Italy
| |
Collapse
|
3
|
Huang K, Cai S, Fu T, Zhu Q, Liu L, Yao Z, Rao P, Lan X, Li Q, Xiao J. Wnt10b regulates osteogenesis of adipose-derived stem cells through Wnt/β-catenin signalling pathway in osteoporosis. Cell Prolif 2024; 57:e13522. [PMID: 37340715 PMCID: PMC10771102 DOI: 10.1111/cpr.13522] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/24/2023] [Accepted: 06/07/2023] [Indexed: 06/22/2023] Open
Abstract
Our previous finding revealed that the Wnt10b RNA expression of osteoporotic adipose-derived stem cells (OP-ASCs) with impaired osteogenic capacity was significantly reduced than that of ASCs. There are no ideas that the relationship between the OP-ASCs' impaired osteogenic potential and Wnt10b expression. This study aimed to indicate the potential molecular mechanisms and functional role of Wnt10b in OP-ASCs, as well as to investigate a potential application to reverse the OP-ASCs' impaired osteogenic differentiation potential. The OP-ASCs and ASCs were harvested from the inguinal fat of osteoporosis (OP) mice with bilateral ovariectomy (OVX) and normal mice. qPCR and WB were used to detect the different levels of the expression of the Wnt10b RNA in both OP-ASCs and ASCs. Lentiviral-mediated regulation of Wnt10b expression was employed for OP-ASCs, and the detection of the expression levels of key molecules in the Wnt signalling pathway and key osteogenic factors was performed through qPCR and WB in vitro experiments. The capacity of OP-ASCs to osteogenesis was determined using alizarin red staining. Lastly, the repair effect of the BCP scaffolds incorporating modified OP-ASCs on the critical-sized calvarial defects (CSCDs) in OP mice was scanned and detected by micro-computed tomography, haematoxylin and eosin staining, Masson's trichrome staining and immunohistochemistry. First, we discovered that both the RNA and protein expression levels of Wnt10b were significantly lower in OP-ASCs than that in ASCs. In vitro experiments, upregulation of Wnt10b could activate the Wnt signalling pathway, and increase expression of β-catenin, Lef1, Runx2 and osteopontin (Opn), thereby enhancing the osteogenic ability of OP-ASCs. In addition, the OP-ASCs with Wnt10b-overexpressing could promote the repair of CSCD in osteoporotic mice with increasing new bone volume, bone mineral density, and increased expression of Opn in new bone in vivo. Taken together, overexpression of Wnt10b could partially facilitate the differentiation of OP-ASCs towards osteogenesis and accelerated the healing of bone defects by activating the Wnt/β-catenin signalling pathway in vitro and in vivo experiments. This study confirmed the important role of Wnt10b in regulating the osteogenic differentiation capability of OP-ASCs and indicated Wnt10b could be a potential therapeutic target for reversing the impaired osteogenic capabilities of OP-ASCs to therapy bone defects of OP patients.
Collapse
Affiliation(s)
- Kui Huang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationLuzhouChina
| | - Shuyu Cai
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationLuzhouChina
- Department of Oral ImplantologyThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Ting Fu
- Department of Oral ImplantologyThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Qiang Zhu
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationLuzhouChina
| | - Lin Liu
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationLuzhouChina
| | - Zhihao Yao
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Pengcheng Rao
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationLuzhouChina
| | - Qing Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationLuzhouChina
| | - Jingang Xiao
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationLuzhouChina
- Department of Oral ImplantologyThe Affiliated Stomatological Hospital of Southwest Medical UniversityLuzhouChina
- Department of Oral and Maxillofacial SurgeryThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
4
|
Kawasuji H, Ikezawa Y, Morita M, Sugie K, Somekawa M, Ezaki M, Koshiyama Y, Takegoshi Y, Murai Y, Kaneda M, Kimoto K, Nagaoka K, Niimi H, Morinaga Y, Yamamoto Y. High Incidence of Metastatic Infections in Panton-Valentine Leucocidin-Negative, Community-Acquired Methicillin-Resistant Staphylococcus aureus Bacteremia: An 11-Year Retrospective Study in Japan. Antibiotics (Basel) 2023; 12:1516. [PMID: 37887217 PMCID: PMC10604685 DOI: 10.3390/antibiotics12101516] [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: 08/28/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Panton-Valentine leucocidin (PVL)-negative community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) was originally disseminated in Japan and has since replaced healthcare-associated MRSA (HA-MRSA). However, the clinical characteristics of CA-MRSA bacteremia (CA-MRSAB) compared with those of HA-MRSA bacteremia (HA-MRSAB) are unknown. We aim to clarify differences and investigate associations between the clinical manifestations and virulence genes associated with plasma-biofilm formation in PVL-negative CA-MRSA. From 2011 to 2021, when CA-MRSA dramatically replaced HA-MRSA, 79 MRSA strains were collected from blood cultures and analyzed via SCCmec typing and targeted virulence gene (lukSF-PV, cna, and fnbB) detection. The incidence of metastatic infection was significantly higher in CA-MRSAB than in HA-MRSAB. PVL genes were all negative, although cna and fnbB were positive in 55.6% (20/36) and 50% (18/36) of CA-MRSA strains and 3.7% (1/27) and 7.4% (2/27) of HA-MRSA strains, respectively. cna and fnbB carriage were not associated with the development of metastatic infections in MRSAB; however, the bacteremia duration was significantly longer in CA-MRSAB harboring cna. CA-MRSAB may be more likely to cause metastatic infections than HA-MRSAB. Since CA-MRSA is dominant in Japan, suspected metastatic infection foci should be identified by computed tomography, magnetic resonance imaging, and echocardiography when treating MRSAB.
Collapse
Affiliation(s)
- Hitoshi Kawasuji
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yoshihiro Ikezawa
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Mika Morita
- Department of Clinical Laboratory and Molecular Pathology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Kazushige Sugie
- Department of Clinical Laboratory and Molecular Pathology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Mayu Somekawa
- Department of Microbiology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Masayoshi Ezaki
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yuki Koshiyama
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yusuke Takegoshi
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yushi Murai
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Makito Kaneda
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Kou Kimoto
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Kentaro Nagaoka
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Hideki Niimi
- Department of Clinical Laboratory and Molecular Pathology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yoshitomo Morinaga
- Department of Microbiology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yoshihiro Yamamoto
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| |
Collapse
|
5
|
Molecular Characteristics and Distribution of Virulence Genes among Staphylococcus aureus Complex Isolates Derived from Vascular Access Infections. CANADIAN JOURNAL OF INFECTIOUS DISEASES AND MEDICAL MICROBIOLOGY 2022; 2022:3196545. [PMCID: PMC9616667 DOI: 10.1155/2022/3196545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
Abstract
Staphylococcus aureus is a major human pathogen that produces various virulence factors which promote the binding of bacteria to tissues and medical devices such as vascular access devices, thereby developing a wide range of invasive infections. Vascular access serves as an entry site for S. aureus and elevates the risk of infection in the hemodialysis population. Nevertheless, the distribution of virulence genes in Staphylococcus spp. associated with vascular access infections (VAIs) has not been studied previously. In this study, we determined the relationship between the molecular characteristics and virulence profiles of S. aureus isolates obtained from VAIs. We collected isolates from patients with VAIs between August 2017 and December 2020 and further analyzed the molecular characteristics, antimicrobial resistance profiles, and virulence gene distribution in the isolates. Overall, 15 sequence types (STs), including a new ST (ST6892) and 19 spa types, were identified among the 56 isolates. Of the 53 S. aureus isolates, ST8, ST239, ST45, and ST59 were the predominant STs, whereas ST2250 was the only ST in 3 S. argenteus isolates. ST45-SCCmecIV-t026 (abbreviated as ST45-IV-t026), ST59-V-t437, and ST8-IV-t008 were the predominant clones that belonged to agr type I. All isolates harbored clfB and eno, whereas all S. aureus isolates harbored clfA. In addition, 10 Panton-Valentine leucocidin-positive isolates belonged to ST8 and ST59, with ST8-IV-t008 and ST59-V-t437 being the predominant clones. In brief, the distribution of virulence genes associated with STs may assist in the spread of molecular types of Staphylococcus spp.
Collapse
|
6
|
Prencipe F, Alsibaee A, Khaddem Z, Norton P, Towell AM, Ali AFM, Reid G, Fleury OM, Foster TJ, Geoghegan JA, Rozas I, Brennan MP. Allantodapsone is a Pan-Inhibitor of Staphylococcus aureus Adhesion to Fibrinogen, Loricrin, and Cytokeratin 10. Microbiol Spectr 2022; 10:e0117521. [PMID: 35647689 PMCID: PMC9241669 DOI: 10.1128/spectrum.01175-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/17/2022] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus infections have become a major challenge in health care due to increasing antibiotic resistance. We aimed to design small molecule inhibitors of S. aureus surface proteins to be developed as colonization inhibitors. We identified allantodapsone in an initial screen searching for inhibitors of clumping factors A and B (ClfA and ClfB). We used microbial adhesion assays to investigate the effect of allantodapsone on extracellular matrix protein interactions. Allantodapsone inhibited S. aureus Newman adhesion to fibrinogen with an IC50 of 21.3 μM (95% CI 4.5-102 μM), minimum adhesion inhibitory concentration (MAIC) of 100 μM (40.2 μg/mL). Additionally, allantodapsone inhibited adhesion of Lactococcus lactis strains exogenously expressing the clumping factors to fibrinogen (L. lactis ClfA, IC50 of 3.8 μM [95% CI 1.0-14.3 μM], MAIC 10 μM, 4.0 μg/mL; and L. lactis ClfB, IC50 of 11.0 μM [95% CI 0.9-13.6 μM], MAIC 33 μM, 13.3 μg/mL), indicating specific inhibition. Furthermore, the dapsone and alloxan fragments of allantodapsone did not have any inhibitory effect. Adhesion of S. aureus Newman to L2v loricrin is dependent on the expression of ClfB. Allantodapsone caused a dose dependent inhibition of S. aureus adhesion to the L2v loricrin fragment, with full inhibition at 40 μM (OD600 0.11 ± 0.01). Furthermore, recombinant ClfB protein binding to L2v loricrin was inhibited by allantodapsone (P < 0.0001). Allantodapsone also demonstrated dose dependent inhibition of S. aureus Newman adhesion to cytokeratin 10 (CK10). Allantodapsone is the first small molecule inhibitor of the S. aureus clumping factors with potential for development as a colonization inhibitor. IMPORTANCE S. aureus colonization of the nares and the skin provide a reservoir of bacteria that can be transferred to wounds that can ultimately result in systemic infections. Antibiotic resistance can make these infections difficult to treat with significant associated morbidity and mortality. We have identified and characterized a first-in-class small molecule inhibitor of the S. aureus clumping factors A and B, which has the potential to be developed further as a colonization inhibitor.
Collapse
Affiliation(s)
- Filippo Prencipe
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aishah Alsibaee
- School of Pharmacy and Biomedical Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Zainab Khaddem
- School of Pharmacy and Biomedical Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Padraig Norton
- School of Pharmacy and Biomedical Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Aisling M. Towell
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Afnan F. M. Ali
- School of Pharmacy and Biomedical Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gerard Reid
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Orla M. Fleury
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Timothy J. Foster
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Joan A. Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Isabel Rozas
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Marian P. Brennan
- School of Pharmacy and Biomedical Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
7
|
ALBA MARYILORENASEGURA, DURÁN-RODRIGUEZ ANDREATATIANA, PULIDO LUZMARYSALAZAR, ESCOBAR-PÉREZ JAVIER, GUTIÉRREZ SERGIOALEJANDRO, OSPINA JEANNETTENAVARRETE, BERMÚDEZ GLADYSPINILLA, MOLINA LILIANACONSTANZAMUÑOZ. Peptides DLL37-1 and LL37-1, an alternative to inhibit biofilm formation in clinical isolates of Staphylococcus aureus and Staphylococcus epidermidis. AN ACAD BRAS CIENC 2022; 94:e20210848. [DOI: 10.1590/0001-3765202220210848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/08/2021] [Indexed: 11/09/2022] Open
|
8
|
Schwarz C, Töre Y, Hoesker V, Ameling S, Grün K, Völker U, Schulze PC, Franz M, Faber C, Schaumburg F, Niemann S, Hoerr V. Host-pathogen interactions of clinical S. aureus isolates to induce infective endocarditis. Virulence 2021; 12:2073-2087. [PMID: 34490828 PMCID: PMC8425731 DOI: 10.1080/21505594.2021.1960107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
To evaluate potential pathomechanisms in the induction of infective endocarditis (IE), 34 Staphylococcus aureus (S. aureus) isolates, collected from patients with S. aureus endocarditis and from healthy individuals were investigated both in vitro and in vivo. S. aureus isolates were tested in vitro for their cytotoxicity, invasion and the association with platelets. Virulence factor expression profiles and cellular response were additionally investigated and tested for correlation with the ability of S. aureus to induce vegetations on the aortic valves in vivo. In an animal model of IE valvular conspicuity was assessed by in vivo magnetic resonance imaging at 9.4 T, histology and enrichment gene expression analysis. All S. aureus isolates tested in vivo caused a reliable infection and inflammation of the aortic valves, but could not be differentiated and categorized according to the measured in vitro virulence profiles and cytotoxicity. Results from in vitro assays did not correlate with the severity of IE. However, the isolates differed substantially in the activation and inhibition of pathways connected to the extracellular matrix and inflammatory response. Thus, comprehensive approaches of host-pathogen interactions and corresponding immune pathways are needed for the evaluation of the pathogenic capacity of bacteria. An improved understanding of the interaction between virulence factors and immune response in S. aureus infective endocarditis would offer novel possibilities for the development of therapeutic strategies and specific diagnostic imaging markers.
Collapse
Affiliation(s)
- Christian Schwarz
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Yasemin Töre
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Vanessa Hoesker
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Sabine Ameling
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Katja Grün
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | | | - Marcus Franz
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Cornelius Faber
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Muenster, Muenster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Muenster, Muenster, Germany
| | - Verena Hoerr
- Translational Research Imaging Center, Clinic for Radiology, University Hospital Muenster, Muenster, Germany.,Institute of Medical Microbiology, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany.,Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
9
|
Molecular epidemiology of clinical isolates of methicillin-resistant Staphylococcus aureus by multilocus sequence typing in northwestern (Tabriz) and southern (Kerman) of Iran: The emergence of MRSA ST4848-SCCmec III. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
10
|
Biofilm Formation of Multidrug-Resistant MRSA Strains Isolated from Different Types of Human Infections. Pathogens 2021; 10:pathogens10080970. [PMID: 34451434 PMCID: PMC8400568 DOI: 10.3390/pathogens10080970] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main pathogens causing chronic infections, mainly due to its capacity to form biofilms. However, the mechanisms underlying the biofilm formation of MRSA strains from different types of human infections are not fully understood. MRSA strains isolated from distinct human infections were characterized aiming to determine their biofilm-forming capacity, the biofilm resistance to conventional antibiotics and the prevalence of biofilm-related genes, including, icaA, icaB, icaC, icaD, fnbA, fnbB, clfA, clfB, cna, eno, ebpS, fib and bbp. Eighty-three clinical MRSA strains recovered from bacteremia episodes, osteomyelitis and diabetic foot ulcers were used. The biofilm-forming capacity was evaluated by the microtiter biofilm assay and the biofilm structure was analyzed via confocal scanning laser microscopy. The antimicrobial susceptibility of 24-h-old biofilms was assessed against three antibiotics and the biomass reduction was measured. The metabolic activity of biofilms was evaluated by the XTT assay. The presence of biofilm-related genes was investigated by whole-genome sequencing and by PCR. Despite different intensities, all strains showed the capacity to form biofilms. Most strains had also a large number of biofilm-related genes. However, strains isolated from osteomyelitis showed a lower capacity to form biofilms and also a lower prevalence of biofilm-associated genes. There was a significant reduction in the biofilm biomass of some strains tested against antibiotics. Our results provide important information on the biofilm-forming capacity of clinical MRSA strains, which may be essential to understand the influence of different types of infections on biofilm production and chronic infections.
Collapse
|
11
|
Sharabiani HR, Sadeghi J, Pirzade T, Rezaee MA, Ghotaslou R, Laghousi D, Sefidan FY, Kafil HS, Nikbakht M, Mazraeh FN, Hematyar Y. Comparison of superantigens and attachment factors genes of Staphylococcus aureus in clinical isolates and nasal colonizers in the same patients. Microb Pathog 2021; 154:104860. [PMID: 33771631 DOI: 10.1016/j.micpath.2021.104860] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Staphylococcus aureus (S. aureus) is a bacterial pathogen can cause a wide range of nosocomial infections. Nasal colonization by S.aureus plays important role both in the epidemiology and pathogenesis of infection. OBJECTIVES The purpose of this study was to investigate the association of clinical isolates and nasal colonizers of S. aureus in the same patients by molecular methods, and their antibiotic susceptibility pattern. METHODS A total of 181 S. aureus isolates were collected from 100 patients admitted that including 100 clinical isolates and 81 nasal swabs from the same patients (19 cases were found as noncarriers). Superantigens and adhesion genes were identified by PCR. Molecular typing of the isolates was performed by repetitive element polymerase chain reaction (Rep-PCR). Antimicrobial susceptibility pattern of the isolates was conducted by disk diffusion. MIC of the isolates to vancomycin was determined by microbroth dilution. The ability of S. aureus isolates to form biofilm was determined by microtiter plate assay. RESULTS The most frequent adhesion gene in both clinical isolates and nasal colonizer was clfA with 93% and 76%, respectively. Staphylococcal enterotoxin A (SEA) was the most commonly superantigen (68%) in both nasal colonizers (71.6%) and clinical isolates (65%). The highest resistance rate was to erythromycin (45.3%) with 36% and 56.8% in clinical and nasal colonizer isolates, respectively. All S. aureus isolates were susceptible to linezolid and vancomycin. Multiple drug resistance (MDR) was detected in 36% (n = 65) of the isolates. Biofilm formation was identified in 160 (88.4%) isolates with 87% and 90% in clinical isolates and nasal colonizers, respectively. Repetitive element polymerase chain reaction (Rep-PCR) typing divided 181 S. aureus isolates into six clusters. Twelve isolates from clinical isolates and nasal carriers were closely related. CONCLUSION There is a high concordance rate between colonizing and clinical isolates of S. aureus in terms of adhesion factors and superantigen genes. It is suggested that nasal decolonization could be effective in the preventing of S. aureus infections.
Collapse
Affiliation(s)
- Hamideh Richi Sharabiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadeghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Tahere Pirzade
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Ahangarzadeh Rezaee
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Ghotaslou
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Delara Laghousi
- Social Determinants of Health Research Center, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fateme Yeghane Sefidan
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Nikbakht
- Meshginshahr Health Center Laboratory, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Fariba Naeimi Mazraeh
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Hematyar
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Preda M, Mihai MM, Popa LI, Dițu LM, Holban AM, Manolescu LSC, Popa GL, Muntean AA, Gheorghe I, Chifiriuc CM, Popa MI. Phenotypic and genotypic virulence features of staphylococcal strains isolated from difficult-to-treat skin and soft tissue infections. PLoS One 2021; 16:e0246478. [PMID: 33529240 PMCID: PMC7853507 DOI: 10.1371/journal.pone.0246478] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/19/2021] [Indexed: 12/04/2022] Open
Abstract
Chronic infections represent an important burden on the healthcare system and have a significant impact on the patients’ quality of life. While Staphylococcus spp. are commensal bacteria, they can become pathogenic, leading to various types of infections. In this study we aimed to characterize the virulence profiles of staphylococcal strains involved in difficult-to-treat skin and soft tissue infections, from both phenotypic and genotypic points of view. Phenotypic ability of the strains to secrete soluble virulence factors was assessed by a culturing dependent assay and their capacity to develop biofilms on inert substrate was screened by an adapted crystal violet microtiter method. We also tested the presence of several virulence genes by PCR. Most of the studied strains were isolated from purulent secretions of acne lesions and frequently secreted two or three soluble virulence factors. Most frequently secreted soluble virulence factors were caseinase (89%), lipase (71%) and lecithinase (67%). Almost half of the strains produced a well-represented biofilm. The molecular characterization showed the presence of the genes cna, hlg, clfA, and clfB. Staphylococcal strains that produce difficult-to-treat skin and soft tissue infections seem to be characterized by an enhanced ability to produce different soluble virulence factors and to develop biofilms in vitro. Further studies need to be developed in other Staphylococcus spp. infections in order to confirm this hypothesis.
Collapse
Affiliation(s)
- Mădălina Preda
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
- ‘Cantacuzino’ National Medico-Military Research and Development Institute, Bucharest, Romania
| | - Mara Mădălina Mihai
- Department of Oncologic Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
- Department of Dermatology, ‘Elias’ University Emergency Hospital, Bucharest, Romania
- * E-mail: (MMM); (LIP)
| | - Laura Ioana Popa
- Department of Bioinformatics, The National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- * E-mail: (MMM); (LIP)
| | - Lia-Mara Dițu
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Alina Maria Holban
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Loredana Sabina Cornelia Manolescu
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| | - Gabriela-Loredana Popa
- Department of Microbiology, Faculty of Dental Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Irina Gheorghe
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Carmen Mariana Chifiriuc
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Mircea-Ioan Popa
- ‘Cantacuzino’ National Medico-Military Research and Development Institute, Bucharest, Romania
- Department of Microbiology, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
13
|
Fu W, He W, Ren Y, Li Z, Liu J, Liu Y, Xie Z, Xu J, Bi Q, Kong M, Lee CC, Daiss JL, Muthukrishnan G, Owen JR, Kates SL, Peng J, Xie C. Distinct expression trend of signature antigens of Staphylococcus aureus osteomyelitis correlated with clinical outcomes. J Orthop Res 2021; 39:265-273. [PMID: 33336817 PMCID: PMC7946439 DOI: 10.1002/jor.24961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 02/04/2023]
Abstract
The major limitations of clinical outcome predictions of osteomyelitis mediated by Staphylococcus aureus (S. aureus) are not specific and definitive. To this end, current studies aim to investigate host immune responses of trend changes of the iron-regulated surface determinant (Isd) of IsdA, IsdB, IsdH, cell wall-modifying proteins of amidase (Amd) and glucosaminidase (Gmd), and secreted virulence factor of chemotaxis inhibitory protein S. aureus (CHIPS) and staphylococcal complement inhibitor (SCIN) longitudinally to discover their correlationship with clinical outcomes. A total of 55 patients with confirmed S. aureus infection of the long bone by clinical and laboratory methods were recruited for the study. Whole blood was collected at 0, 6, 12 months for the serum that was used to test IsdA, IsdB, IsdH, Gmd, Amd, CHIPS, and SCIN using a customized Luminex assay after clinical standard care parameters were collected. The patients were then divided into two groups: (1) infection controlled versus (2) adverse outcome based on clinical criteria for statistical analysis. We found that standard clinical parameters were unable to distinguish therapeutic outcomes. Significant overexpression of all antigens was confirmed in infection patients at 0-, 6-, and 12-month time points. A distinct expression trend and dynamic changes of IsdB, Amd, Gmd, and CHIPS were observed between infection controlled and adverse outcome patients, while the IsdA, IsdH, SCIN remained demonstrated no statistical significance. We conclude that dynamic changes of specific antigens could predict clinical outcomes of S. aureus osteomyelitis. Clinical Relevance: The trend changes of host immune responses to S. aureus specific antigens of IsdB, Gmd, Amd, and CHIPS could predict clinical outcomes of S. aureus osteomyelitis.
Collapse
Affiliation(s)
- Wei Fu
- Department of Orthopaedics, Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,The authors contributed equally
| | - Wenbin He
- Department of Orthopaedics, Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,The authors contributed equally
| | - Youliang Ren
- Department of Orthopaedics, Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China
| | - Zhengdao Li
- Department of Orthopaedics, Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China
| | - Jinyue Liu
- Department of Orthopaedics, Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China
| | - Yi Liu
- Department of Orthopaedics, Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China
| | - Zhao Xie
- Department of Orthopaedic, Joint Orthopaedic Research Center of Southwest Hospital of Third Military Medical University & University of Rochester Medical Center (JORC – SHTMMU &URMC), Southwest Hospital of Third Military Medical University, Chongqing, 400038 China,Joint Orthopaedic, Research Center of Southwest Hospital of Third Military Medical University & University of Rochester Medical Center (JORC – SHTMMU &URMC), Southwest Hospital of Third Military Medical University, Chongqing, 400038 China
| | - Jianzhong Xu
- Department of Orthopaedic, Joint Orthopaedic Research Center of Southwest Hospital of Third Military Medical University & University of Rochester Medical Center (JORC – SHTMMU &URMC), Southwest Hospital of Third Military Medical University, Chongqing, 400038 China,Joint Orthopaedic, Research Center of Southwest Hospital of Third Military Medical University & University of Rochester Medical Center (JORC – SHTMMU &URMC), Southwest Hospital of Third Military Medical University, Chongqing, 400038 China
| | - Qing Bi
- Department of Orthopaedic, Joint Orthopaedic Research Center of Zhejiang Provincial People’s Hospital & University of Rochester Medical Center (JORC – ZPPH &URMC), Zhejiang Provincial Hospital, Hangzhou, 310024 China,Joint Orthopaedic, Research Center of Zhejiang Provincial People’s Hospital & University of Rochester Medical Center (JORC – ZPPH &URMC), Zhejiang Provincial Hospital, Hangzhou, 310024 China
| | - Mingxiang Kong
- Department of Orthopaedic, Joint Orthopaedic Research Center of Zhejiang Provincial People’s Hospital & University of Rochester Medical Center (JORC – ZPPH &URMC), Zhejiang Provincial Hospital, Hangzhou, 310024 China,Joint Orthopaedic, Research Center of Zhejiang Provincial People’s Hospital & University of Rochester Medical Center (JORC – ZPPH &URMC), Zhejiang Provincial Hospital, Hangzhou, 310024 China
| | - Charles C. Lee
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - John L. Daiss
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA 23298
| | - Stephen L. Kates
- Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA 23298
| | - Jiachen Peng
- Department of Orthopaedics, Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,To whom correspondence should be addressed: Dr. Chao Xie, The Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, Phone 585-275-0818, FAX 585-276-2177, or Dr. Jiachen Peng, Department of Orthopaedics First Affiliated Hospital of Zunyi Medical University Zunyi, 563003 China,
| | - Chao Xie
- Joint Orthopaedic, Research Center of Zunyi Medical University & University of Rochester Medical Center (JORC – ZMU&URMC), First Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 China,Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642,To whom correspondence should be addressed: Dr. Chao Xie, The Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, Phone 585-275-0818, FAX 585-276-2177, or Dr. Jiachen Peng, Department of Orthopaedics First Affiliated Hospital of Zunyi Medical University Zunyi, 563003 China,
| |
Collapse
|
14
|
Jean SS, Chang LW, Hsueh PR. Tentative clinical breakpoints and epidemiological cut-off values of nemonoxacin for Streptococcus pneumoniae and Staphylococcus aureus isolates associated with community-acquired pneumonia. J Glob Antimicrob Resist 2020; 23:388-393. [PMID: 33207229 DOI: 10.1016/j.jgar.2020.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES To determine the minimum inhibitory concentration (MIC) distribution, epidemiological cut-off (ECOFF) values and clinical breakpoints (CBPs) of nemonoxacin, a non-fluorinated quinolone, for community-acquired pneumonia (CAP)-related Streptococcus pneumoniae and Staphylococcus aureus. METHODS We pooled the susceptibility and clinical data of CAP patients enrolled in five clinical trials conducted in three countries from 2006 to 2017. Published pharmacokinetic (PK) profiles of oral (500 mg) and intravenous (IV) (500, 650 and 750 mg) nemonoxacin formulations and pharmacodynamic (PD) parameters of the two aforementioned CAP-related Gram-positive cocci (GPC) were used to determine plausible CBPs. Moreover, nemonoxacin MIC distributions of CAP-relatedS. pneumoniae (n = 1800) and S. aureus (n = 2000) isolates were obtained to evaluate ECOFF values using a visual estimation approach and ECOFFinder. RESULTS More than 92% of patients with CAP caused byS. pneumoniae or S. aureus with nemonoxacin MICs ≤ 0.25 mg/L presented positive clinical and microbiological outcomes. The ECOFF, MIC90 and MIC99 values of nemonoxacin were, respectively, 0.06, 0.125 and 1 mg/L for S. pneumoniae and 0.125, 1 and 8 mg/L for S. aureus. Based on differences in the PK profiles of oral and IV formulations, PD parameters of nemonoxacin for these CAP-GPC and clinical in vivo efficacy data, tentative CBPs of 0.5, 0.5 and 1 mg/L, respectively, were established for the 500 mg oral and 500 mg and 750 mg IV nemonoxacin formulations for S. pneumoniae, and 0.25, 0.5 and 1 mg/L for S. aureus. CONCLUSION This study provides plausible nemonoxacin CBPs for two important CAP-GPC.
Collapse
Affiliation(s)
- Shio-Shin Jean
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Emergency Medicine, Department of Emergency Medicine and Critical Care Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | | | - Po-Ren Hsueh
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
15
|
Moura MC, Procópio TF, Ferreira GRS, Alves RRV, Sá RA, Paiva PMG, Ingmer H, Coelho LCBB, Napoleão TH. Anti-staphylococcal effects of Myracrodruon urundeuva lectins on nonresistant and multidrug resistant isolates. J Appl Microbiol 2020; 130:745-754. [PMID: 32750211 DOI: 10.1111/jam.14811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022]
Abstract
AIMS To evaluate the anti-staphylococcal effects of lectins isolated from bark (MuBL), heartwood (MuHL) and leaves (MuLL) of Myracrodruon urundeuva. METHODS AND RESULTS The lectins were evaluated for: effects on growth, aggregation, haemolytic activity and biofilm-forming ability of Staphylococcus aureus clinical isolates nonresistant (8325-4) and multidrug resistant (LAC USA300); interference with the expression of virulence genes (hla, rnaIII and spa) of the Agr system of S. aureus; and synergistic effect with the antibiotics cefoxitin and cefotaxime. MuBL, MuHL and MuLL reduced growth (minimal inhibitory concentration (MIC): 12·5-50 µg ml-1 ) and viability (minimal bactericidal concentration (MBC): 100 µg ml-1 ) of 8325-4 and LAC USA300 cells. MuLL (at ½MIC and MIC) reduced LAC USA300 agglutination. The lectins did not interfere with haemolytic activity and expression of hla, rnaIII and spa genes. Only MuHL was able to reduce the biofilm production by 8325-4 (50-400 µg ml-1 ) and LAC USA300 (400 µg ml-1 ). CONCLUSION The M. urundeuva lectins showed antibacterial activity against nonresistant and resistant clinical isolates of S. aureus and synergistic effects with antibiotics in reducing growth and biofilm formation. SIGNIFICANCE AND IMPACT OF THE STUDY This work reports bioactive molecules capable of acting as anti-staphylococcal agents, since there are increasing reports of multiresistant isolates of this bacterium.
Collapse
Affiliation(s)
- M C Moura
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - T F Procópio
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - G R S Ferreira
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - R R V Alves
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - R A Sá
- Centro Acadêmico do Agreste, Universidade Federal de Pernambuco, Caruaru, Pernambuco, Brazil
| | - P M G Paiva
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - H Ingmer
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - L C B B Coelho
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - T H Napoleão
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
16
|
Jean SS, Ko WC, Hsueh PR. Susceptibility of clinical isolates of meticillin-resistant Staphylococcus aureus and phenotypic non-extended-spectrum β-lactamase-producing Klebsiella pneumoniae to ceftaroline in Taiwan: Results from Antimicrobial Testing Leadership and Surveillance (ATLAS) in 2012-2018 and Surveillance of Multicentre Antimicrobial Resistance in Taiwan (SMART) in 2018-2019. Int J Antimicrob Agents 2020; 56:106016. [PMID: 32422316 DOI: 10.1016/j.ijantimicag.2020.106016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/21/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023]
Abstract
Data on ceftaroline (CPT) susceptibility amongst clinical isolates of meticillin-resistant Staphylococcus aureus (MRSA, n=1284) and phenotypic non-extended-spectrum β-lactamase-producing (non-ESBL-P) Klebsiella pneumoniae (n=466), obtained from the Antimicrobial Testing Leadership and Surveillance (ATLAS) programme from 2012 to 2018, and selected MRSA isolates from patients with bloodstream infections (BSIs) (n=95) from the Surveillance of Multicentre Antimicrobial Resistance in Taiwan (SMART) programme from 2018 to 2019 were analysed. The minimum inhibitory concentrations (MICs) of ATLAS isolates were determined using the broth microdilution method, whereas the MICs of SMART BSI-MRSA isolates were determined using the Etest and MicroScan system. The pharmacokinetic profiles and pharmacodynamic parameters of CPT were applied to explore the optimal dosage against infections caused by Taiwanese MRSA and K. pneumoniae isolates. Approximately 7.1% of ATLAS MRSA isolates were susceptible-dose dependent (S-DD) to CPT, and 19.7% of the non-ESBL-P K. pneumoniae isolates were not susceptible to CPT. Amongst the ATLAS MRSA isolates, the S-DD rates to CPT amongst isolates causing lower respiratory tract infections were 11.9% and 8.5% for isolates from intensive care units (ICUs) and general wards (GWs), and those causing skin and soft tissue infections (SSTIs) were 20% and 5.3% for isolates from ICUs and GWs, respectively (P=0.015). Of the SSTI MRSA isolates from GWs, 22.7% displayed vancomycin MICs >1 mg/L. Amongst 95 SMART BSI MRSA isolates, 28 (46.7%) isolates exhibited lower CPT MICs by the Etest compared with 60 isolates with CPT MICs of 1-2 mg/L by the MicroScan system. CPT 600 mg as a 2-h intravenous infusion every 8 h is suggested for treatment of infections caused by MRSA and phenotypic non-ESBL-P K. pneumoniae in Taiwan.
Collapse
Affiliation(s)
- Shio-Shin Jean
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wen-Chien Ko
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Infectious Diseases and Centre for Infection Control, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Po-Ren Hsueh
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
17
|
Moosavian M, Baratian Dehkordi P, Hashemzadeh M. Characterization of SCCmec, Spa Types and Multidrug Resistant of Methicillin-Resistant Staphylococcus aureus Isolates in Ahvaz, Iran. Infect Drug Resist 2020; 13:1033-1044. [PMID: 32308445 PMCID: PMC7154036 DOI: 10.2147/idr.s244896] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/28/2020] [Indexed: 11/23/2022] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is one of the most pathogens associated with health care. Molecular typing methods are vital for outbreak investigations of MRSA. The aim of this study was characterization of SCCmec, spa types and multidrug resistant of methicillin-resistant Staphylococcus aureus isolates in Ahvaz, Iran. Methods A total of 50 MRSA isolates were determined by using the phenotypic method and mecA gene. Antibiotic resistance profile and SCCmec types were screened using disc diffusion method and PCR, respectively. For spa typing of MRSA isolates, two molecular typing methods including the PCR-sequencing and high-resolution melting (HRM) analysis were used. Results In the present study, the highest sensitivity of MRSA was to vancomycin and linezolid and the lowest to clindamycin. In the MRSA isolates, 22% were XDR and 78% were MDR. SCCmec type III was found commonly among MRSA. Based on PCR-sequencing and HRM results, 10 different spa types were identified. The spa types t037 and t030 were the most common in this study. Conclusion This study emphasizes the spa variation among MRSA isolates, which may be considered as an important criterion when treating staphylococcal infections. Accurate and early detection of MDR, XDR, or even PDR MRSA isolates strains must be commenced by all clinical microbiology laboratories to reduce the menace of antimicrobial resistance.
Collapse
Affiliation(s)
- Mojtaba Moosavian
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Paria Baratian Dehkordi
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Hashemzadeh
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
18
|
Chang YW, Huang WC, Lin CY, Wang WH, Hung LC, Chen YH. Tellimagrandin II, A Type of Plant Polyphenol Extracted from Trapa bispinosa Inhibits Antibiotic Resistance of Drug-Resistant Staphylococcus aureus. Int J Mol Sci 2019; 20:ijms20225790. [PMID: 31752109 PMCID: PMC6888525 DOI: 10.3390/ijms20225790] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/09/2019] [Accepted: 11/13/2019] [Indexed: 01/25/2023] Open
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) has become a critical global concern. Identifying new candidates of anti-S. aureus agents is urgently required because the therapeutic strategies for infected patients are limited currently. Therefore, the present study investigated whether Tellimagrandin II (TGII), a pure compound extracted from the shells of Trapa bispinosa, exhibits antibacterial effects against MRSA. We first showed that TGII exerted potent inhibitory activity against MRSA with a minimum inhibitory concentration of 128 μg/mL. The obtained fractional inhibitory concentration suggested that TGII could alone exert antistaphylococcal activity, and TGII combined with low doses of antibiotics displayed synergistic effects against MRSA. Moreover, we found that TGII exerted bactericidal activity by reducing the expression of mecA followed by the negative regulation of the penicillin-binding protein 2a (PBP2a) of MRSA. Transmission electron microscopy (TEM) images further confirmed that TGII destroyed the integrity of the cell wall of MRSA and caused the loss of cytoplasm content. In conclusion, we evidenced the antibacterial effects of TGII against MRSA, which enables the effective dose of current antibiotics to be reduced and the predicament of drug-resistant S. aureus isolates to be overcome.
Collapse
Affiliation(s)
- Yu-Wei Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-W.C.); (C.-Y.L.)
- Department of Laboratory, Taitung Hospital, Ministry of Health and Welfare, Taitung 95043, Taiwan
| | - Wan-Chun Huang
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Tropical Medicine and Infectious diseases, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-C.H.); (W.-H.W.); (L.-C.H.)
| | - Chun-Yu Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-W.C.); (C.-Y.L.)
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Tropical Medicine and Infectious diseases, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-C.H.); (W.-H.W.); (L.-C.H.)
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Wen-Hung Wang
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Tropical Medicine and Infectious diseases, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-C.H.); (W.-H.W.); (L.-C.H.)
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Ling-Chien Hung
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Tropical Medicine and Infectious diseases, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-C.H.); (W.-H.W.); (L.-C.H.)
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Yen-Hsu Chen
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Tropical Medicine and Infectious diseases, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (W.-C.H.); (W.-H.W.); (L.-C.H.)
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, HsinChu 30010, Taiwan
- Correspondence: ; Tel.: +886-7-312-1101 (ext. 5677)
| |
Collapse
|
19
|
Liao CH, Chen CS, Chen YC, Jiang NE, Farn CJ, Shen YS, Hsu ML, Chang CH. Vancomycin-loaded oxidized hyaluronic acid and adipic acid dihydrazide hydrogel: Bio-compatibility, drug release, antimicrobial activity, and biofilm model. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 53:525-531. [PMID: 31607570 DOI: 10.1016/j.jmii.2019.08.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/08/2019] [Accepted: 08/13/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Prosthesis infection is a difficult-to-treat situation. Hydrogel is a novel biomaterial, which can be applied by simply spraying or by coating on implants before surgery and can be easily mixed with antibiotics. METHODS In order to evaluate the potential use of antibiotic-loaded hydrogel, we incorporated vancomycin into oxidized hyaluronic acid (HA) and adipic acid dihydrazide and evaluated the drug release and antimicrobial activity against methicillin-resistant Staphylococcus aureus (ATCC 29213). RESULTS The average release percentage of vancomycin on day 3 was about 86%. The antibiotic-loaded gel was biocompatible with mesenchymal stem cell, MC3T3, and L929 cell lines. The in vitro inhibition zones of vancomycin-loaded hydrogel [500X minimal inhibition concentration (MIC), 50X MIC, 10X MIC, and blank hydrogel] were 21, 13, 9, and 5 mm, respectively. In the Ti6Al4V implant biofilm model, 0.01-1% vancomycin-loaded gel exhibited significant anti-biofilm activity, measured by the MTT assay. CONCLUSIONS Vancomycin could be loaded onto oxidized HA and adipic acid dihydrazide, which exhibited excellent drug release and in vitro antimicrobial activity with minimal cell toxicity.
Collapse
Affiliation(s)
- Chun-Hsing Liao
- Department of Medicine, Yang-Ming University, Taiwan; Division of Infectious Disease, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Chiang Sang Chen
- Department of Material and Fiber, Oriental Institute of Technology, New Taipei City, Taiwan; Department of Orthopaedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Yu-Chun Chen
- Department of Orthopaedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan; College of General Studies, Yuan Ze University, Taoyuan City, Taiwan
| | - Ni-En Jiang
- Department of Orthopaedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Chui Jia Farn
- Department of Orthopaedic Surgery, National Taiwan University Hospital, Taiwan
| | - Yi-Shan Shen
- Department of Orthopaedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Ming-Lun Hsu
- School of Dentistry, National Yang-Ming University, Taipei, Taiwan.
| | - Chih-Hung Chang
- Department of Orthopaedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, Taiwan.
| |
Collapse
|