1
|
Esemu SN, Nya'Nying SF, Ndip LM, Bessong PO, Tanih NF, Smith SI, Ndip RN. Isolation and characterization of methicillin-resistant Staphylococcus aureus from bovine mastitis in North West Cameroon: public health implications. BMC Res Notes 2024; 17:389. [PMID: 39731175 DOI: 10.1186/s13104-024-07073-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024] Open
Abstract
OBJECTIVES Methicillin-resistant Staphylococcus aureus (MRSA) is a zoonotic pathogen that poses a serious threat to veterinary and public health worldwide. We investigated mastitis milk samples for contamination with MRSA and also characterized the MRSA isolates by investigating antimicrobial resistance and virulence factors. RESULT We confirmed MRSA in 69 of 201 (34.3%) S. aureus isolates recovered from a total of 300 samples. Of the 69 MRSA, 19 (27.5%) were from subclinical cases, while 50 (72.5%) were from clinical cases. The MRSA showed high resistance to penicillin (100%), ampicillin (100%), trimethoprim (69.6%), and tetracycline (69.6%) while susceptibility was observed for gentamicin (100%), vancomycin (95.7%), and ciprofloxacin (91.3%). Most isolates (65.2%, 45/69) were multidrug resistant. Thirteen antibiotypes (A1-A13) were identified and the most prevalent was A8 (TMPRERTETRAMPRPR)). All MRSA produced haemolysins, caseinase, and coagulase. Lipase, gelatinase and lecithinase were found in 97.1%, 94.2% and 91.3% of isolates respectively. Genotyping revealed coa (100%) and spa (68.1%) genes. We recommend educating dairy farmers on the public health implications of consuming unpasteurized raw milk and the implementation of proper hygiene practices in dairy farms.
Collapse
Affiliation(s)
- Seraphine Nkie Esemu
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Box 63, Buea, Cameroon.
- Laboratory for Emerging Infectious Diseases, University of Buea, Box 63, Buea, Cameroon.
| | - Shantal Fuenbi Nya'Nying
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Box 63, Buea, Cameroon
| | - Lucy Mande Ndip
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Box 63, Buea, Cameroon
- Laboratory for Emerging Infectious Diseases, University of Buea, Box 63, Buea, Cameroon
| | - Pascal Obong Bessong
- South African Medical Research Council - University of Venda Antimicrobial Resistance and Global Health Research Unit, Thohoyandou, 0950, South Africa
- Center for Global Health Equity, University of Virginia, Charlottesville, USA
| | - Nicoline Fri Tanih
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Buea, Box 63, Buea, Cameroon
| | - Stella Ifeanyi Smith
- Department of Molecular Biology and Biotechnology, Nigerian Institute of Medical Research, Lagos, Nigeria
| | - Roland Ndip Ndip
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Box 63, Buea, Cameroon
- Laboratory for Emerging Infectious Diseases, University of Buea, Box 63, Buea, Cameroon
| |
Collapse
|
2
|
Shi J, Xiao Y, Shen L, Wan C, Wang B, Zhou P, Zhang J, Han W, Yu F. Phenotypic and genomic analysis of the hypervirulent methicillin-resistant Staphylococcus aureus ST630 clone in China. mSystems 2024; 9:e0066424. [PMID: 39158330 PMCID: PMC11406941 DOI: 10.1128/msystems.00664-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/11/2024] [Indexed: 08/20/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) sequence type 630 (ST630) is a rarely reported lineage worldwide. This study aimed to trace the dissemination of the emerging MRSA ST630 clones in China and investigate their virulence potential. We collected 22 ST630-MRSA isolates from across China and performed whole-genome sequencing analysis and virulence characterization on these isolates. Epidemiological results showed that MRSA ST630 isolates were primarily isolated from pus/wound secretions, mainly originating from Jiangxi province, and carried diverse virulence and drug resistance genes. Staphylococcal cassette chromosome mec type V (SCCmec V) predominated (11/22, 50.0%) among the MRSA ST630 isolates. Interestingly, nearly half (45.5%) of the 22 ST630-MRSA isolates tested lacked intact SCCmec elements. Phylogenetic analysis demonstrated that ST630-MRSA could be divided into two distinct clades, with widespread dissemination mainly in Chinese regions. Five representative isolates were selected for phenotypic assays, including hemolysin activity, real-time fluorescence quantitative PCR, western blot analysis, hydrogen peroxide killing assay, blood killing assay, cell adhesion and invasion assay, and mouse skin abscess model. The results showed that, compared to the USA300-LAC strain, ST630 isolates exhibited particularly strong invasiveness and virulence in the aforementioned phenotypic assays. This study described the emergence of a highly virulent ST630-MRSA lineage and improved our insight into the molecular epidemiology of ST630 clones in China.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA) sequence type 630 (ST630) is an emerging clone with an increasing isolation rate in China. This study raises awareness of the hypervirulent MRSA ST630 clones in China and alerts people to their widespread dissemination. ST630-staphylococcal cassette chromosome mec V is a noteworthy clone in China, and we present the first comprehensive genetic and phenotypic analysis of this lineage. Our findings provide valuable insights for the prevention and control of infections caused by this emerging MRSA clone.
Collapse
Affiliation(s)
- Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cailing Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
3
|
Beadell B, Nehra S, Gusenov E, Huse H, Wong-Beringer A. Machine Learning with Alpha Toxin Phenotype to Predict Clinical Outcome in Patients with Staphylococcus aureus Bloodstream Infection. Toxins (Basel) 2023; 15:417. [PMID: 37505686 PMCID: PMC10467129 DOI: 10.3390/toxins15070417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/17/2023] [Accepted: 06/25/2023] [Indexed: 07/29/2023] Open
Abstract
Staphylococcus aureus bloodstream (SAB) infection remains a leading cause of sepsis-related mortality. Yet, current treatment does not account for variable virulence traits that mediate host dysregulated immune response, such as SA α-toxin (Hla)-mediated thrombocytopenia. Here, we applied machine learning (ML) to bacterial growth images combined with platelet count data to predict patient outcomes. We profiled Hla phenotypes of SA isolates collected from patients with bacteremia by taking smartphone images of beta-hemolytic growth on sheep blood agar (SBA). Electronic medical records were reviewed to extract relevant laboratory and clinical data. A convolutional neural network was applied to process the plate image data for input along with day 1 patient platelet count to generate ML-based models that predict thrombocytopenia on day 4 and mortality. A total of 229 patients infected with SA strains exhibiting varying zone sizes of beta-hemolysis on SBA were included. A total of 539 images of bacterial growth on SBA were generated as inputs for model development. One-third of patients developed thrombocytopenia at onset, with an overall mortality rate of 18.8%. The models developed from the ML algorithm showed strong performance (AUC 0.92) for predicting thrombocytopenia on day 4 of infection and modest performance (AUC 0.711) for mortality. Our findings support further development and validation of a proof-of-concept ML application in digital microbiology, with a measure of bacterial virulence factor production that carries prognostic significance and can help guide treatment selection.
Collapse
Affiliation(s)
- Brent Beadell
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (B.B.); (E.G.)
| | - Surya Nehra
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA;
| | - Elizabeth Gusenov
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (B.B.); (E.G.)
| | - Holly Huse
- Department of Microbiology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
| | - Annie Wong-Beringer
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (B.B.); (E.G.)
- Department of Pharmacy, Huntington Hospital, Pasadena, CA 91105, USA
| |
Collapse
|
4
|
Gui Y, Zeng Y, Chen B, Yang Y, Ma J, Li C. A smart pathogen detector engineered from intracellular hydrogelation of DNA-decorated macrophages. Nat Commun 2023; 14:2927. [PMID: 37217531 DOI: 10.1038/s41467-023-38733-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
Bacterial infection is a major threat to global public health, which urgently requires useful tools to rapidly analyze pathogens in the early stages of infection. Herein, we develop a smart macrophage (Mø)-based bacteria detector, which can recognize, capture, enrich and detect different bacteria and their secreted exotoxins. We transform the fragile native Møs into robust gelated cell particles (GMøs) using photo-activated crosslinking chemistry, which retains membrane integrity and recognition capacity for different microbes. Meanwhile, these GMøs equipped with magnetic nanoparticles and DNA sensing elements can not only respond to an external magnet for facile bacteria collection, but allow the detection of multiple types of bacteria in a single assay. Additionally, we design a propidium iodide-based staining assay to rapidly detect pathogen-associated exotoxins at ultralow concentrations. Overall, these nanoengineered cell particles have broad applicability in the analysis of bacteria, and could potentially be used for the management and diagnosis of infectious diseases.
Collapse
Affiliation(s)
- Yueyue Gui
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China
| | - Yujing Zeng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, 210023, Nanjing, P. R. China
| | - Binrui Chen
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China
| | - Yueping Yang
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China
| | - Jiehua Ma
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, Shanghai, P. R. China
| | - Chao Li
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China.
| |
Collapse
|
5
|
Ni L, Li Y, Zhang H, Ma Y, Song Y, Tang X, Fan J, Shi JY, Cui X, Xu H, Zhou H, Shen K, Guo W, Yu L. The combination of insulin and linezolid ameliorates Staphylococcus aureus pneumonia in individuals with diabetes via the TLR2/MAPKs/NLRP3 pathway. Int J Biol Macromol 2023; 242:124750. [PMID: 37160172 DOI: 10.1016/j.ijbiomac.2023.124750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 04/10/2023] [Accepted: 05/01/2023] [Indexed: 05/11/2023]
Abstract
Diabetes mellitus (DM) complicated with Staphylococcus aureus (S. aureus) infection lacks effective treatment strategies. In this study, we found that insulin combined with linezolid has potential to deal with the thorny problem. In vitro, our drug sensitivity assay, bacterial growth curve and hemolytic tests showed that a combination of insulin and linezolid exerted good antibacterial and anti-α-hemolysin activity, CCK8 experiment, glucose content and glycogen content determination showed that the combination of insulin and linezolid increased murine macrophage survival rate and reduced the extracellular glucose level of high glucose-treated MH-S cells and intracellular glycogen level, and Western blot showed that the combination inhibited TLR2/MAPKs/NLRP3-related inflammatory pathways in MH-S cells. The results of in vivo experiments showed that the combination therapy stabilized glucose level, remained body weight, ameliorated lung injury including improving pulmonary edema and decreasing lung wet/dry weight ratio, reduced the CFUs and inflammation in the lung tissue in a mouse model of diabetes with S. aureus pneumonia, and inhibited the expression of TLR2, MAPKs and NLRP3 inflammatory pathway. Overall, the combination of insulin and linezolid as autolytic inhibitor exhibited the effects of significant antibacterial and improving glucose level in vitro and in vivo, and also has an anti-inflammation activity via the TLR2/MAPKs/NLRP3 pathway, this paves the way for new treatments for diabetes mellitus complicated with S. aureus infection.
Collapse
Affiliation(s)
- Lihui Ni
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Yuan Li
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Haifeng Zhang
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Yunxiao Ma
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Yuli Song
- Shenzhen Liyunde Biotechnology Co., Ltd., Shenzhen 518057, China
| | - Xudong Tang
- Key Lab for New Drug Research of TCM, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Junwen Fan
- Beijing Center for Animal Disease Control and Prevention, Beijing 102629, China
| | - Jin Yang Shi
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Xinhua Cui
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Hongyue Xu
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Hong Zhou
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China
| | - Keshu Shen
- Jilin Hepatobiliary Hospital, Changchun 130062, China
| | - Weiying Guo
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China.
| | - Lu Yu
- State Key Laboratory of Human-Animal Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Changchun 130062, China.
| |
Collapse
|
6
|
Pivard M, Bastien S, Macavei I, Mouton N, Rasigade JP, Couzon F, Youenou B, Tristan A, Carrière R, Moreau K, Lemoine J, Vandenesch F. Targeted proteomics links virulence factor expression with clinical severity in staphylococcal pneumonia. Front Cell Infect Microbiol 2023; 13:1162617. [PMID: 37077532 PMCID: PMC10106754 DOI: 10.3389/fcimb.2023.1162617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Introduction The bacterial pathogen Staphylococcus aureus harbors numerous virulence factors that impact infection severity. Beyond virulence gene presence or absence, the expression level of virulence proteins is known to vary across S. aureus lineages and isolates. However, the impact of expression level on severity is poorly understood due to the lack of high-throughput quantification methods of virulence proteins. Methods We present a targeted proteomic approach able to monitor 42 staphylococcal proteins in a single experiment. Using this approach, we compared the quantitative virulomes of 136 S. aureus isolates from a nationwide cohort of French patients with severe community-acquired staphylococcal pneumonia, all requiring intensive care. We used multivariable regression models adjusted for patient baseline health (Charlson comorbidity score) to identify the virulence factors whose in vitro expression level predicted pneumonia severity markers, namely leukopenia and hemoptysis, as well as patient survival. Results We found that leukopenia was predicted by higher expression of HlgB, Nuc, and Tsst-1 and lower expression of BlaI and HlgC, while hemoptysis was predicted by higher expression of BlaZ and HlgB and lower expression of HlgC. Strikingly, mortality was independently predicted in a dose-dependent fashion by a single phage-encoded virulence factor, the Panton-Valentine leucocidin (PVL), both in logistic (OR 1.28; 95%CI[1.02;1.60]) and survival (HR 1.15; 95%CI[1.02;1.30]) regression models. Discussion These findings demonstrate that the in vitro expression level of virulence factors can be correlated with infection severity using targeted proteomics, a method that may be adapted to other bacterial pathogens.
Collapse
Affiliation(s)
- Mariane Pivard
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR5308, École Normale Supérieure (ENS) de Lyon, Lyon, France
| | - Sylvère Bastien
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR5308, École Normale Supérieure (ENS) de Lyon, Lyon, France
| | - Iulia Macavei
- Institut des Sciences Analytiques, Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR 5280, Villeurbanne, France
| | - Nicolas Mouton
- Institut des Sciences Analytiques, Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR 5280, Villeurbanne, France
| | - Jean-Philippe Rasigade
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR5308, École Normale Supérieure (ENS) de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| | - Florence Couzon
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR5308, École Normale Supérieure (ENS) de Lyon, Lyon, France
| | - Benjamin Youenou
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| | - Anne Tristan
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR5308, École Normale Supérieure (ENS) de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| | - Romain Carrière
- Institut des Sciences Analytiques, Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR 5280, Villeurbanne, France
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR5308, École Normale Supérieure (ENS) de Lyon, Lyon, France
| | - Jérôme Lemoine
- Institut des Sciences Analytiques, Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR 5280, Villeurbanne, France
| | - François Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), UMR5308, École Normale Supérieure (ENS) de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
7
|
Merghni A, Hamdi H, Ben Abdallah M, Al-Hasawi ZM, Al-Quwaie DA, Abid-Essefi S. Detection of Methicillin-Resistant Staphylococcus aureus among Foodborne Pathogenic Strains and Assessment of Their Adhesion Ability and Cytotoxic Effects in HCT-116 Cells. Foods 2023; 12:foods12050974. [PMID: 36900491 PMCID: PMC10001405 DOI: 10.3390/foods12050974] [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: 01/25/2023] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Staphylococcus aureus is one of the high-threat pathogens equipped with a repertoire of virulence factors making it responsible for many infections in humans, including foodborne diseases. The present study aims to characterize antibiotic resistance and virulence factors in foodborne S. aureus isolates, and to investigate their cytotoxic effects in human intestinal cells (HCT-116). Our results revealed methicillin resistance phenotypes (MRSA) along with the detection of mecA gene (20%) among tested foodborne S. aureus strains. Furthermore, 40% of tested isolates showed a strong ability for adhesion and biofilm formation. A high rate of exoenzymes production by tested bacteria was also registered. Additionally, treatment with S. aureus extracts leads to a significant decrease in HCT-116 cell viability, accompanied by a reduction in the mitochondrial membrane potential (MMP), as a result of reactive oxygen species (ROS) generation. Thereby, S. aureus food poisoning remains daunting and needs particular concern to prevent foodborne illness.
Collapse
Affiliation(s)
- Abderrahmen Merghni
- Laboratory of Antimicrobial Resistance LR99ES09, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis 1007, Tunisia
- Correspondence:
| | - Hiba Hamdi
- Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Faculty of Dental Medicine, University of Monastir, Monastir 5000, Tunisia
| | - Marwa Ben Abdallah
- Laboratory of Transmissible Diseases and Biologically Active Substances, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Zaki M. Al-Hasawi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Biological Sciences Department, College of Science & Arts, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| | - Diana A. Al-Quwaie
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Salwa Abid-Essefi
- Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Faculty of Dental Medicine, University of Monastir, Monastir 5000, Tunisia
| |
Collapse
|
8
|
Microbiological and Molecular Features Associated with Persistent and Relapsing Staphylococcus aureus Prosthetic Joint Infection. Antibiotics (Basel) 2022; 11:antibiotics11081119. [PMID: 36009988 PMCID: PMC9405193 DOI: 10.3390/antibiotics11081119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/07/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Persistent and relapsing prosthetic joint infection (PJI) due to Staphylococcus aureus presents a clinical challenge. This study aimed to provide an extensive description of phenotypic and genomic changes that could be related to persistence or relapse. Methods: Initial and second S. aureus isolates from 6 cases of persistent and relapsing PJI, along with clinical isolates from 8 cases, with favorable outcome were included. All isolates were studied by phenotypic and genotypic approaches. Results: Recurrent S. aureus isolates exhibited a significant increase in adhesive capacity, invasion and persistence compared to resolved isolates. No association was found for the presence or absence of certain genes with the persistence or relapse of PJI. All sequential isolates showed identical sequence type (ST). Resistance gene loss during the infection and a great diversity of variants in different virulence genes between the pair of strains, mainly in genes encoding adhesins such as fnbA, were observed. Conclusions: S. aureus-caused relapse and persistence PJI is associated with bacterial phenotypical and genotypical adaptation. The main paths of adaptation were persistence in the intracellular compartment, and the loss of antibiotic resistance genes and variant acquisition, especially in genes encoding adhesins.
Collapse
|
9
|
Spaan AN, Neehus AL, Laplantine E, Staels F, Ogishi M, Seeleuthner Y, Rapaport F, Lacey KA, Van Nieuwenhove E, Chrabieh M, Hum D, Migaud M, Izmiryan A, Lorenzo L, Kochetkov T, Heesterbeek DAC, Bardoel BW, DuMont AL, Dobbs K, Chardonnet S, Heissel S, Baslan T, Zhang P, Yang R, Bogunovic D, Wunderink HF, Haas PJA, Molina H, Van Buggenhout G, Lyonnet S, Notarangelo LD, Seppänen MRJ, Weil R, Seminario G, Gomez-Tello H, Wouters C, Mesdaghi M, Shahrooei M, Bossuyt X, Sag E, Topaloglu R, Ozen S, Leavis HL, van Eijk MMJ, Bezrodnik L, Blancas Galicia L, Hovnanian A, Nassif A, Bader-Meunier B, Neven B, Meyts I, Schrijvers R, Puel A, Bustamante J, Aksentijevich I, Kastner DL, Torres VJ, Humblet-Baron S, Liston A, Abel L, Boisson B, Casanova JL. Human OTULIN haploinsufficiency impairs cell-intrinsic immunity to staphylococcal α-toxin. Science 2022; 376:eabm6380. [PMID: 35587511 PMCID: PMC9233084 DOI: 10.1126/science.abm6380] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.
Collapse
Affiliation(s)
- András N Spaan
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Institute of Experimental Hematology, REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Emmanuel Laplantine
- Centre d'Immunologie et des Maladies Infectieuses, INSERM U1135, CNRS ERL8255, Sorbonne University, 75724 Paris, France
- Institut de Recherche St. Louis, Hôpital St. Louis, INSERM U944, CNRS U7212, Paris Cité University, 75010 Paris, France
| | - Frederik Staels
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Franck Rapaport
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Erika Van Nieuwenhove
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Maya Chrabieh
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - David Hum
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Araksya Izmiryan
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Laboratory of Genetic Skin Diseases, INSERM U1163, 75015 Paris, France
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Tatiana Kochetkov
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Dani A C Heesterbeek
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Bart W Bardoel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Ashley L DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20852, USA
| | - Solenne Chardonnet
- Plateforme Post-génomique de la Pitié-Salpêtrière, P3S, UMS Production et Analyse de données en Sciences de la vie et en Santé, PASS, INSERM, Sorbonne University, 75013 Paris, France
| | - Søren Heissel
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Dusan Bogunovic
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Herman F Wunderink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Pieter-Jan A Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Griet Van Buggenhout
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Stanislas Lyonnet
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Laboratory Embryology and Genetics of Malformations, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20852, USA
| | - Mikko R J Seppänen
- Rare Disease and Pediatric Research Centers, Children and Adolescents, University of Helsinki and HUS Helsinki University Hospital, 00260 Helsinki, Finland
| | - Robert Weil
- Centre d'Immunologie et des Maladies Infectieuses, INSERM U1135, CNRS ERL8255, Sorbonne University, 75724 Paris, France
| | - Gisela Seminario
- Center for Clinical Immunology, Immunology Group Children's Hospital Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Héctor Gomez-Tello
- Immunology Department, Poblano Children's Hospital, 72190 Puebla, Mexico
| | - Carine Wouters
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Mehrnaz Mesdaghi
- Department of Allergy and Clinical Immunology, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, 15468-155514 Tehran, Iran
| | - Mohammad Shahrooei
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Specialized Immunology Laboratory of Dr. Shahrooei, Sina Medical Complex, 15468-155514 Ahvaz, Iran
| | - Xavier Bossuyt
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Erdal Sag
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
| | - Rezan Topaloglu
- Department of Pediatric Nephrology, Hacettepe University School of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Seza Ozen
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
| | - Helen L Leavis
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Maarten M J van Eijk
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Liliana Bezrodnik
- Center for Clinical Immunology, Immunology Group Children's Hospital Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | | | - Alain Hovnanian
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Laboratory of Genetic Skin Diseases, INSERM U1163, 75015 Paris, France
- Department of Genetics, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
| | - Aude Nassif
- Centre Médical, Institut Pasteur, 75724 Paris, France
| | - Brigitte Bader-Meunier
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Pediatric Immunology, Hematology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmunity, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
| | - Bénédicte Neven
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Pediatric Immunology, Hematology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmunity, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
| | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, Jeffrey Modell Diagnostic and Research Network Center, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Rik Schrijvers
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Daniel L Kastner
- Inflammatory Disease Section, National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Stéphanie Humblet-Baron
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Adrian Liston
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Brain and Disease Research, Leuven 3000, Belgium
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
10
|
Fernandez JS, Tuttobene MR, Montaña S, Subils T, Cantera V, Iriarte A, Tuchscherr L, Ramirez MS. Staphylococcus aureus α-Toxin Effect on Acinetobacter baumannii Behavior. BIOLOGY 2022; 11:biology11040570. [PMID: 35453769 PMCID: PMC9028598 DOI: 10.3390/biology11040570] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/02/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022]
Abstract
Polymicrobial infections are more challenging to treat and are recognized as responsible for significant morbidity and mortality. It has been demonstrated that multiple Gram-negative organisms take advantage of the effects of Staphylococcus aureus α-toxin on mucosal host defense, resulting in proliferation and dissemination of the co-infecting pathogens. Through phenotypic approaches, we observed a decrease in the motility of A. baumannii A118 after exposure to cell-free conditioned media (CFCM) of S. aureus strains, USA300 and LS1. However, the motility of A. baumannii A118 was increased after exposure to the CFCM of S. aureus strains USA300 Δhla and S. aureus LSI ΔagrA. Hemolytic activity was seen in A118, in the presence of CFCM of S. aureus LS1. Further, A. baumannii A118 showed an increase in biofilm formation and antibiotic resistance to tetracycline, in the presence of CFCM of S. aureus USA300. Transcriptomic analysis of A. baumannii A118, with the addition of CFCM from S. aureus USA300, was carried out to study A. baumannii response to S. aureus’ released molecules. The RNA-seq data analysis showed a total of 463 differentially expressed genes, associated with a wide variety of functions, such as biofilm formation, virulence, and antibiotic susceptibility, among others. The present results showed that A. baumannii can sense and respond to molecules secreted by S. aureus. These findings demonstrate that A. baumannii may perceive and respond to changes in its environment; specifically, when in the presence of CFCM from S. aureus.
Collapse
Affiliation(s)
- Jennifer S. Fernandez
- Center for Applied Biotechnology Studies, Department of Biological Science, California State University Fullerton, Fullerton, CA 92831, USA; (J.S.F.); (M.R.T.)
| | - Marisel R. Tuttobene
- Center for Applied Biotechnology Studies, Department of Biological Science, California State University Fullerton, Fullerton, CA 92831, USA; (J.S.F.); (M.R.T.)
- Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Rosario S2000, Argentina
| | - Sabrina Montaña
- Laboratorio de Bacteriología Clínica, Departamento de Bioquímica Clínica, Hospital de Clínicas José de San Martín, Facultad de Farmacia y Bioquímica, Buenos Aires C1113, Argentina;
| | - Tomás Subils
- Instituto de Procesos Biotecnológicos y Químicos de Rosario (IPROBYQ, CONICET-UNR), Rosario S2000, Argentina;
| | - Virginia Cantera
- Laboratorio de Biología Computacional, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Universidad de la República, Montevideo 11200, Uruguay; (V.C.); (A.I.)
| | - Andrés Iriarte
- Laboratorio de Biología Computacional, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Universidad de la República, Montevideo 11200, Uruguay; (V.C.); (A.I.)
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany;
| | - Maria Soledad Ramirez
- Center for Applied Biotechnology Studies, Department of Biological Science, California State University Fullerton, Fullerton, CA 92831, USA; (J.S.F.); (M.R.T.)
- Correspondence: ; Tel.: +1-657-278-4562
| |
Collapse
|
11
|
Montagut EJ, Acosta G, Albericio F, Royo M, Godoy-Tena G, Lacoma A, Prat C, Salvador JP, Marco MP. Direct Quantitative Immunochemical Analysis of Autoinducer Peptide IV for Diagnosing and Stratifying Staphylococcus aureus Infections. ACS Infect Dis 2022; 8:645-656. [PMID: 35175740 PMCID: PMC8922274 DOI: 10.1021/acsinfecdis.1c00670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An immunochemical strategy to detect and quantify AIP-IV, the quorum sensing (QS) signaling molecule produced by Staphylococcus aureus agr type IV, is reported here for the first time. Theoretical calculations and molecular modeling studies have assisted on the design and synthesis of a suitable peptide hapten (AIPIVS), allowing to obtain high avidity and specific antibodies toward this peptide despite its low molecular weight. The ELISA developed achieves an IC50 value of 2.80 ± 0.17 and an LOD of 0.19 ± 0.06 nM in complex media such as 1/2 Tryptic Soy Broth. Recognition of other S. aureus AIPs (I-III) is negligible (cross-reactivity below 0.001%), regardless of the structural similarities. A pilot study with a set of clinical isolates from patients with airways infection or colonization demonstrates the potential of this ELISA to perform biomedical investigations related to the role of QS in pathogenesis and the association between dysfunctional agr or the agr type with unfavorable clinical outcomes. The AIP-IV levels could be quantified in the low nanomolar range in less than 1 h after inoculating agr IV-genotyped isolates in the culture broth, while those genotyped as I-III did not show any immunoreactivity after a 48 h growth, pointing to the possibility to use this technology for phenotyping S. aureus. The research strategy here reported can be extended to the rest of the AIP types of S. aureus, allowing the development of powerful multiplexed chips or point-of-care (PoC) diagnostic devices to unequivocally identify its presence and its agr type on samples from infected patients.
Collapse
Affiliation(s)
- Enrique-J. Montagut
- Nanobiotechnology for Diagnostics (Nb4D), Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), 08750 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid Spain
| | - Gerardo Acosta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid Spain
- Multivalent Systems for Nanomedicine (MS4N), Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), 08750 Barcelona, Spain
| | - Fernando Albericio
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid Spain
- Multivalent Systems for Nanomedicine (MS4N), Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), 08750 Barcelona, Spain
- Department of Organic Chemistry, Faculty of Chemistry, University of Barcelona, 08028 Barcelona, Spain
- School of Chemistry and Physics, University of KwaZulu-Natal, 4000 Durban, South Africa
| | - Miriam Royo
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid Spain
- Multivalent Systems for Nanomedicine (MS4N), Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), 08750 Barcelona, Spain
| | - Gerard Godoy-Tena
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Alicia Lacoma
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Cristina Prat
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 Utrecht, the Netherlands
| | - Juan-Pablo Salvador
- Nanobiotechnology for Diagnostics (Nb4D), Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), 08750 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid Spain
| | - María-Pilar Marco
- Nanobiotechnology for Diagnostics (Nb4D), Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), 08750 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid Spain
| |
Collapse
|
12
|
Save J, Que YA, Entenza JM, Kolenda C, Laurent F, Resch G. Bacteriophages Combined With Subtherapeutic Doses of Flucloxacillin Act Synergistically Against Staphylococcus aureus Experimental Infective Endocarditis. J Am Heart Assoc 2022; 11:e023080. [PMID: 35043655 PMCID: PMC9238497 DOI: 10.1161/jaha.121.023080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background The potential of phage therapy for the treatment of endovascular Staphylococcus aureus infections remains to be evaluated. Methods and Results The efficacy of a phage cocktail combining Herelleviridae phage vB_SauH_2002 and Podoviriae phage 66 was evaluated against a methicillin‐sensitive S. aureus strain in vitro and in vivo in a rodent model of experimental endocarditis. Six hours after bacterial challenge, animals were treated with (1) the phage cocktail. (2) subtherapeutic flucloxacillin dosage, (3) combination of the phage cocktail and flucloxacillin, or (4) saline. Bacterial loads in cardiac vegetations at 30 hours were the primary outcome. Secondary outcomes were phage loads at 30 hours in cardiac vegetations, blood, spleen, liver, and kidneys. We evaluated phage resistance 30 hours post infection in vegetations of rats under combination treatment. In vitro, phages synergized against S. aureus planktonic cells and the cocktail synergized with flucloxacillin to eradicated biofilms. In infected animals, the phage cocktail achieved bacteriostatic effect. The addition of low‐dose flucloxacillin elevated bacterial suppression (∆ of −5.25 log10 colony forming unit/g [CFU/g] versus treatment onset, P<0.0001) and synergism was confirmed (∆ of −2.15 log10 CFU/g versus low‐dose flucloxacillin alone, P<0.01). Importantly, 9/12 rats given the combination treatment had sterile vegetations at 30 hours. In vivo phage replication was partially suppressed by the antibiotic and selection of resistance to the Podoviridae component of the phage cocktail occurred. Plasma‐mediated inhibition of phage killing activity was observed in vitro. Conclusions Combining phages with a low‐dose standard of care antibiotic represents a promising strategy for the treatment of S. aureus infective endocarditis.
Collapse
Affiliation(s)
- Jonathan Save
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences Lausanne University Hospital Lausanne Switzerland.,Department of Intensive Care Medicine Inselspital, Bern University Hospital, University of Bern Bern Switzerland
| | - Yok-Ai Que
- Department of Intensive Care Medicine Inselspital, Bern University Hospital, University of Bern Bern Switzerland
| | - José M Entenza
- Department of Fundamental Microbiology University of Lausanne Lausanne Switzerland
| | - Camille Kolenda
- Bacteriology Department, Institute for Infectious Agents French National Reference Centre for Staphylococci, Croix-Rousse University Hospital Hospices Civils de Lyon, Lyon France.,National Centre of Research in Infectiology, Team "Staphylococcal Pathogenesis", INSERM U1111, CNRS UMR5308, ENS Lyon, University of Lyon Lyon France
| | - Frédéric Laurent
- Bacteriology Department, Institute for Infectious Agents French National Reference Centre for Staphylococci, Croix-Rousse University Hospital Hospices Civils de Lyon, Lyon France.,National Centre of Research in Infectiology, Team "Staphylococcal Pathogenesis", INSERM U1111, CNRS UMR5308, ENS Lyon, University of Lyon Lyon France
| | - Grégory Resch
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences Lausanne University Hospital Lausanne Switzerland
| |
Collapse
|
13
|
Increased Risk of Thrombocytopenia and Death in Patients with Bacteremia Caused by High Alpha Toxin-Producing Methicillin-Resistant Staphylococcus aureus. Toxins (Basel) 2021; 13:toxins13100726. [PMID: 34679019 PMCID: PMC8537302 DOI: 10.3390/toxins13100726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 01/25/2023] Open
Abstract
Alpha toxin (Hla) is a major virulence factor of Staphylococcus aureus that targets platelets but clinical data on Hla pathogenesis in bacteremia (SAB) is limited. We examined the link between in vitro Hla activity and outcome. Study isolates obtained from 100 patients with SAB (50 survivors; 50 non-survivors) were assessed for in vitro Hla production by Western immunoblotting in a subset of isolates and Hla activity by hemolysis assay in all isolates. Relevant demographics, laboratory and clinical data were extracted from patients' medical records to correlate Hla activity of the infecting isolates with outcome. Hla production strongly correlated with hemolytic activity (rs = 0.93) in vitro. A trend towards higher hemolytic activity was observed for MRSA compared to MSSA and with high-risk source infection. Significantly higher hemolytic activity was noted for MRSA strains isolated from patients who developed thrombocytopenia (median 52.48 vs. 16.55 HU/mL in normal platelet count, p = 0.012) and from non survivors (median 30.96 vs. 14.87 HU/mL in survivors, p = 0.014) but hemolytic activity of MSSA strains did not differ between patient groups. In vitro Hla activity of MRSA strains obtained from patients with bacteremia is significantly associated with increased risk for thrombocytopenia and death which supports future studies to evaluate feasibility of bedside phenotyping and therapeutic targeting.
Collapse
|
14
|
Abstract
Staphylococcus aureus is both a commensal and a pathogenic bacterium for humans. Its ability to induce severe infections is based on a wide range of virulence factors. S. aureus community-acquired pneumonia (SA-CAP) is rare and severe, and the contribution of certain virulence factors in this disease has been recognized over the past 2 decades. First, the factors involved in metabolism adaptation are crucial for S. aureus survival in the lower respiratory tract, and toxins and enzymes are required for it to cross the pulmonary epithelial barrier. S. aureus subsequently faces host defense mechanisms, including the epithelial barrier, but most importantly the immune system. Here, again, S. aureus uses myriad virulence factors to successfully escape from the host's defenses and takes advantage of them. The impact of S. aureus virulence, combined with the collateral damage caused by an overwhelming immune response, leads to severe tissue damage and adverse clinical outcomes. In this review, we summarize step by step all of the S. aureus factors implicated in CAP and described to date, and we provide an outlook for future research.
Collapse
Affiliation(s)
- Mariane Pivard
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - François Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
15
|
Virulence Factors Found in Nasal Colonization and Infection of Methicillin-Resistant Staphylococcus aureus (MRSA) Isolates and Their Ability to Form a Biofilm. Toxins (Basel) 2020; 13:toxins13010014. [PMID: 33375552 PMCID: PMC7823648 DOI: 10.3390/toxins13010014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/10/2020] [Accepted: 10/27/2020] [Indexed: 11/23/2022] Open
Abstract
Hospitalizations related to Methicillin-resistant Staphylococcus aureus (MRSA) are frequent, increasing mortality and health costs. In this way, this study aimed to compare the genotypic and phenotypic characteristics of MRSA isolates that colonize and infect patients seen at two hospitals in the city of Niterói—Rio de Janeiro, Brazil. A total of 147 samples collected between March 2013 and December 2015 were phenotyped and genotyped to identify the protein A (SPA) gene, the mec staphylococcal chromosomal cassette (SCCmec), mecA, Panton-Valentine Leucocidin (PVL), icaC, icaR, ACME, and hla virulence genes. The strength of biofilm formation has also been exploited. The prevalence of SCCmec type IV (77.1%) was observed in the colonization group; however, in the invasive infection group, SCCmec type II was prevalent (62.9%). The Multilocus Sequence Typing (MLST), ST5/ST30, and ST5/ST239 analyses were the most frequent clones in colonization, and invasive infection isolates, respectively. Among the isolates selected to assess the ability to form a biofilm, 51.06% were classified as strong biofilm builders. Surprisingly, we observed that isolates other than the Brazilian Epidemic Clone (BEC) have appeared in Brazilian hospitals. The virulence profile has changed among these isolates since the ACME type I and II genes were also identified in this collection.
Collapse
|
16
|
Muñoz-Gallego I, Viedma E, Esteban J, Mancheño-Losa M, García-Cañete J, Blanco-García A, Rico A, García-Perea A, Ruiz Garbajosa P, Escudero-Sánchez R, Sánchez Somolinos M, Marín Arriaza M, Romanyk J, Barbero JM, Arribi Vilela A, González Romo F, Pérez-Jorge C, M. Arana D, Monereo A, Domingo D, Cordero J, Sánchez Romero MI, García Viejo MÁ, Lora-Tamayo J, Chaves F. Genotypic and Phenotypic Characteristics of Staphylococcus aureus Prosthetic Joint Infections: Insight on the Pathogenesis and Prognosis of a Multicenter Prospective Cohort. Open Forum Infect Dis 2020; 7:ofaa344. [PMID: 33005695 PMCID: PMC7519778 DOI: 10.1093/ofid/ofaa344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is the leading cause of prosthetic joint infection (PJI). Beyond the antibiogram, little attention has been paid to the influence of deep microbiological characteristics on patient prognosis. Our aim was to investigate whether microbiological genotypic and phenotypic features have a significant influence on infection pathogenesis and patient outcome. METHODS A prospective multicenter study was performed, including all S. aureus PJIs (2016-2017). Clinical data and phenotypic (agr functionality, β-hemolysis, biofilm formation) and genotypic characteristics of the strains were collected. Biofilm susceptibility to antimicrobials was investigated (minimal biofilm eradication concentration [MBEC] assay). RESULTS Eighty-eight patients (39.8% men, age 74.7 ± 14.1 years) were included. Forty-five had early postoperative infections (EPIs), 21 had chronic infections (CPIs), and 19 had hematogenous infections (HIs). Twenty (22.7%) were caused by methicillin-resistant S. aureus. High genotypic diversity was observed, including 16 clonal complexes (CCs), with CC5 being the most frequent (30.7%). agr activity was greater in EPI than CPI (55.6% vs 28.6%; P = .041). Strains causing EPI were phenotypically and genotypically similar, regardless of symptom duration. Treatment failure (36.5%) occurred less frequently among cases treated with implant removal. In cases treated with debridement and implant retention, there were fewer failures among those who received combination therapy with rifampin. No genotypic or phenotypic characteristics predicted failure, except vancomycin minimal inhibitory concentration ≥1.5 mg/L (23.1% failure vs 3.4%; P = .044). MBEC50 was >128 mg/L for all antibiotics tested and showed no association with prognosis. CONCLUSIONS S. aureus with different genotypic backgrounds is capable of causing PJI, showing slight differences in clinical presentation and pathogenesis. No major microbiological characteristics were observed to influence the outcome, including MBEC.
Collapse
Affiliation(s)
- Irene Muñoz-Gallego
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Esther Viedma
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
| | - Jaime Esteban
- Servicio de Microbiología, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Mikel Mancheño-Losa
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - Joaquín García-Cañete
- Servicio de Medicina Interna-Urgencias, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Antonio Blanco-García
- Servicio de Medicina Interna-Urgencias, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Alicia Rico
- Servicio de Medicina Interna, Hospital Universitario La Paz, Madrid, Spain
| | | | - Patricia Ruiz Garbajosa
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Rosa Escudero-Sánchez
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Enfermedades Infecciosas, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Mar Sánchez Somolinos
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Mercedes Marín Arriaza
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Juan Romanyk
- Servicio de Microbiología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - José María Barbero
- Servicio de Medicina Interna, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Ana Arribi Vilela
- Servicio de Microbiología, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Conchita Pérez-Jorge
- Servicio de Microbiología, Hospital Universitario Rey Juan Carlos, Móstoles, Madrid, Spain
| | - David M. Arana
- Servicio de Microbiología, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | - Alfonso Monereo
- Servicio de Medicina Interna, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | - Diego Domingo
- Servicio de Microbiología, Hospital Universitario de La Princesa, Madrid, Spain
| | - José Cordero
- Servicio de Traumatología, Hospital Universitario de La Princesa, Madrid, Spain
| | | | | | - Jaime Lora-Tamayo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - Fernando Chaves
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
| |
Collapse
|
17
|
Muñoz-Gallego I, Mancheño M, Pérez-Montarelo D, Viedma E, Chaves F, Lora-Tamayo J. Staphylococcus aureus native arthritis over 10 years. Med Mal Infect 2020; 50:257-262. [PMID: 32057526 DOI: 10.1016/j.medmal.2020.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/25/2018] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
|
18
|
Lucas R, Hadizamani Y, Gonzales J, Gorshkov B, Bodmer T, Berthiaume Y, Moehrlen U, Lode H, Huwer H, Hudel M, Mraheil MA, Toque HAF, Chakraborty T, Hamacher J. Impact of Bacterial Toxins in the Lungs. Toxins (Basel) 2020; 12:toxins12040223. [PMID: 32252376 PMCID: PMC7232160 DOI: 10.3390/toxins12040223] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial toxins play a key role in the pathogenesis of lung disease. Based on their structural and functional properties, they employ various strategies to modulate lung barrier function and to impair host defense in order to promote infection. Although in general, these toxins target common cellular signaling pathways and host compartments, toxin- and cell-specific effects have also been reported. Toxins can affect resident pulmonary cells involved in alveolar fluid clearance (AFC) and barrier function through impairing vectorial Na+ transport and through cytoskeletal collapse, as such, destroying cell-cell adhesions. The resulting loss of alveolar-capillary barrier integrity and fluid clearance capacity will induce capillary leak and foster edema formation, which will in turn impair gas exchange and endanger the survival of the host. Toxins modulate or neutralize protective host cell mechanisms of both the innate and adaptive immunity response during chronic infection. In particular, toxins can either recruit or kill central players of the lung's innate immune responses to pathogenic attacks, i.e., alveolar macrophages (AMs) and neutrophils. Pulmonary disorders resulting from these toxin actions include, e.g., acute lung injury (ALI), the acute respiratory syndrome (ARDS), and severe pneumonia. When acute infection converts to persistence, i.e., colonization and chronic infection, lung diseases, such as bronchitis, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF) can arise. The aim of this review is to discuss the impact of bacterial toxins in the lungs and the resulting outcomes for pathogenesis, their roles in promoting bacterial dissemination, and bacterial survival in disease progression.
Collapse
Affiliation(s)
- Rudolf Lucas
- Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Department of Medicine and Division of Pulmonary Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Correspondence: (R.L.); (J.H.); Tel.: +41-31-300-35-00 (J.H.)
| | - Yalda Hadizamani
- Lungen-und Atmungsstiftung, Bern, 3012 Bern, Switzerland;
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012 Bern, Switzerland
| | - Joyce Gonzales
- Department of Medicine and Division of Pulmonary Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Boris Gorshkov
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Thomas Bodmer
- Labormedizinisches Zentrum Dr. Risch, Waldeggstr. 37 CH-3097 Liebefeld, Switzerland;
| | - Yves Berthiaume
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada;
| | - Ueli Moehrlen
- Pediatric Surgery, University Children’s Hospital, Zürich, Steinwiesstrasse 75, CH-8032 Zürch, Switzerland;
| | - Hartmut Lode
- Insitut für klinische Pharmakologie, Charité, Universitätsklinikum Berlin, Reichsstrasse 2, D-14052 Berlin, Germany;
| | - Hanno Huwer
- Department of Cardiothoracic Surgery, Voelklingen Heart Center, 66333 Voelklingen/Saar, Germany;
| | - Martina Hudel
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Mobarak Abu Mraheil
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Haroldo Alfredo Flores Toque
- Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Trinad Chakraborty
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Jürg Hamacher
- Lungen-und Atmungsstiftung, Bern, 3012 Bern, Switzerland;
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012 Bern, Switzerland
- Medical Clinic V-Pneumology, Allergology, Intensive Care Medicine and Environmental Medicine, Faculty of Medicine, Saarland University, University Medical Centre of the Saarland, D-66421 Homburg, Germany
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, D-66421 Homburg, Germany
- Correspondence: (R.L.); (J.H.); Tel.: +41-31-300-35-00 (J.H.)
| |
Collapse
|
19
|
Prazak J, Iten M, Cameron DR, Save J, Grandgirard D, Resch G, Goepfert C, Leib SL, Takala J, Jakob SM, Que YA, Haenggi M. Bacteriophages Improve Outcomes in Experimental Staphylococcus aureus Ventilator-associated Pneumonia. Am J Respir Crit Care Med 2020; 200:1126-1133. [PMID: 31260638 DOI: 10.1164/rccm.201812-2372oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Rationale: Infections caused by multidrug-resistant bacteria are a major clinical challenge. Phage therapy is a promising alternative antibacterial strategy.Objectives: To evaluate the efficacy of intravenous phage therapy for the treatment of ventilator-associated pneumonia due to methicillin-resistant Staphylococcus aureus in rats.Methods: In a randomized, blinded, controlled experimental study, we compared intravenous teicoplanin (3 mg/kg, n = 12), a cocktail of four phages (2-3 × 109 plaque-forming units/ml of 2003, 2002, 3A, and K; n = 12), and a combination of both (n = 11) given 2, 12, and 24 hours after induction of pneumonia, and then once daily for 4 days. The primary outcome was survival at Day 4. Secondary outcomes were bacterial and phage densities in lungs and spleen, histopathological scoring of infection within the lungs, and inflammatory biomarkers in blood.Measurements and Main Results: Treatment with either phages or teicoplanin increased survival from 0% to 58% and 50%, respectively (P < 0.005). The combination of phages and antibiotics did not further improve outcomes (45% survival). Animal survival correlated with reduced bacterial burdens in the lung (1.2 × 106 cfu/g of tissue for survivors vs. 1.2 × 109 cfu/g for nonsurviving animals; P < 0.0001), as well as improved histopathological outcomes. Phage multiplication within the lung occurred during treatment. IL-1β increased in all treatment groups over the course of therapy.Conclusions: Phage therapy was as effective as teicoplanin in improving survival and decreasing bacterial load within the lungs of rats infected with methicillin-resistant S. aureus. Combining antibiotics with phage therapy did not further improve outcomes.
Collapse
Affiliation(s)
- Josef Prazak
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Manuela Iten
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David R Cameron
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jonathan Save
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland; and
| | | | - Gregory Resch
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland; and
| | - Christine Goepfert
- Institute of Animal Pathology, Faculty of Veterinary Medicine, University of Bern, Bern, Switzerland
| | | | - Jukka Takala
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stephan M Jakob
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yok-Ai Que
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matthias Haenggi
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
The utility of endotracheal aspirate bacteriology in identifying mechanically ventilated patients at risk for ventilator associated pneumonia: a single-center prospective observational study. BMC Infect Dis 2019; 19:756. [PMID: 31464593 PMCID: PMC6716855 DOI: 10.1186/s12879-019-4367-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 08/07/2019] [Indexed: 01/08/2023] Open
Abstract
Background Ventilator-associated pneumonia (VAP) is a well-known, life-threatening disease that persists despite preventative measures and approved antibiotic therapies. This prospective observational study investigated bacterial airway colonization, and whether its detection and quantification in the endotracheal aspirate (ETA) is useful for identifying mechanically ventilated ICU patients who are at risk of developing VAP. Methods 240 patients admitted to 3 ICUs at the Lahey Hospital and Medical Center (Burlington, MA) between June 2014 and June 2015 and mechanically ventilated for > 2 days were included. ETA samples and clinical data were collected. Airway colonization was assessed, and subsequently categorized into “heavy” and “light” by semi-quantitative microbiological analysis of ETAs. VAP was diagnosed retrospectively by the study sponsor according to a pre-specified pneumonia definition. Results Pathogenic bacteria were isolated from ETAs of 125 patients. The most common species isolated was S. aureus (56.8%), followed by K. pneumoniae, P. aeruginosa, and E. coli (35.2% combined). VAP was diagnosed in 85 patients, 44 (51.7%) with no bacterial pathogen, 18 associated with S. aureus and 18 Gram-negative-only cases, and 5 associated with other Gram-positive or mixed species. A higher proportion of patients who were heavily colonized with S. aureus developed VAP (32.4%) associated with S. aureus compared to those lightly colonized (17.6%). The same tendency was seen for patients heavily and lightly colonized with Gram-negative pathogens (30.0 and 0.0%, respectively). Detection of S. aureus in the ETA preceded S. aureus VAP by approximately 4 days, while Gram-negative organisms were first detected 2.5 days prior to Gram-negative VAP. VAP was associated with significantly longer duration of mechanical ventilation and hospitalization regardless of microbiologic cause when compared to patients who did not develop VAP. Conclusions The overall VAP rate was 35%. Heavy tracheal colonization supported identification of patients at higher risk of developing a corresponding S. aureus or Gram-negative VAP. Detection of bacterial ETA-positivity tended to precede VAP. Electronic supplementary material The online version of this article (10.1186/s12879-019-4367-7) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
S. aureus Evades Macrophage Killing through NLRP3-Dependent Effects on Mitochondrial Trafficking. Cell Rep 2019; 22:2431-2441. [PMID: 29490278 PMCID: PMC7160668 DOI: 10.1016/j.celrep.2018.02.027] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 01/05/2018] [Accepted: 02/06/2018] [Indexed: 12/28/2022] Open
Abstract
Clinical severity of Staphylococcus aureus respiratory infection correlates with alpha toxin (AT) expression. AT activates the NLRP3 inflammasome; deletion of Nlrp3, or AT neutralization, protects mice from lethal S. aureus pneumonia. We tested the hypothesis that this protection is not due to a reduction in inflammasome-dependent cytokines (IL-1β/IL-18) but increased bactericidal function of macrophages. In vivo, neutralization of AT or NLRP3 improved bacterial clearance and survival, while blocking IL-1β/IL-18 did not. Primary human monocytes were used in vitro to determine the mechanism through which NLRP3 alters bacterial killing. In cells treated with small interfering RNA (siRNA) targeting NLRP3 or infected with AT-null S. aureus, mitochondria co-localize with bacterial-containing phagosomes. Mitochondrial engagement activates caspase-1, a process dependent on complex II of the electron transport chain, near the phagosome, promoting its acidification. These data demonstrate a mechanism utilized by S. aureus to sequester itself from antimicrobial processes within the cell.
Collapse
|
22
|
Gao X, Yan X, Zhang Q, Yin Y, Cao J. CD5L contributes to the pathogenesis of methicillin-resistant Staphylococcus aureus-induced pneumonia. Int Immunopharmacol 2019; 72:40-47. [PMID: 30959370 DOI: 10.1016/j.intimp.2019.03.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 11/25/2022]
Abstract
Staphylococcus aureus is a major causative microorganism in community- and healthcare-acquired pneumonia. CD5L is an important protein in the control of immune homeostasis. In this study, we found that patients with S. aureus pneumonia displayed increased levels of circulating CD5L. Likewise, mice with S. aureus pneumonia had elevated CD5L levels in the lungs. Anti-CD5L antibody protected mice from lethal pneumonia induced by methicillin-resistant S. aureus. The survival benefit obtained with antibody against CD5L was associated with an improvement of bacterial clearance and a reduction of pulmonary inflammatory cytokines and chemokines. Conversely, co-injection of recombinant CD5L and S. aureus markedly increased the lethality of S. aureus pneumonia. These findings suggest that CD5L contributed to the immunopathology of S. aureus pneumonia.
Collapse
Affiliation(s)
- Xun Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Key Laboratory of Diagnostic Medicine designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xingxing Yan
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qun Zhang
- Clinical Laboratories Center, Affiliated Children's Hospital, Chongqing Medical University, Chongqing, China
| | - Yibing Yin
- Key Laboratory of Diagnostic Medicine designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
23
|
Preventing lung pathology and mortality in rabbit Staphylococcus aureus pneumonia models with cytotoxin-neutralizing monoclonal IgGs penetrating the epithelial lining fluid. Sci Rep 2019; 9:5339. [PMID: 30926865 PMCID: PMC6441091 DOI: 10.1038/s41598-019-41826-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 03/19/2019] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus pneumonia is associated with high mortality irrespective of antibiotic susceptibility. Both MRSA and MSSA strains produce powerful cytotoxins: alpha-hemolysin(Hla) and up to five leukocidins - LukSF-PV, HlgAB, HlgCB, LukED and LukGH (LukAB) - to evade host innate defense mechanisms. Neutralizing cytotoxins has been shown to provide survival benefit in rabbit S. aureus pneumonia models. We studied the mechanisms of protection of ASN100, a combination of two human monoclonal antibodies (mAbs), ASN-1 and ASN-2, that together neutralize Hla and the five leukocidins, in rabbit MRSA and MSSA pneumonia models. Upon prophylactic passive immunization, ASN100 displayed dose-dependent increase in survival and was fully protective against all S. aureus strains tested at 5 or 20 mg/kg doses. Macroscopic and microscopic lung pathology, edema rate, and bacterial burden were evaluated 12 hours post infection and reduced by ASN100. Pharmacokinetic analysis of ASN100 in bronchoalveolar-lavage fluid from uninfected animals detected efficient penetration to lung epithelial lining fluid reaching peak levels between 24 and 48 hours post dosing that were comparable to the mAb concentration measured in serum. These data confirm that the ASN100 mAbs neutralize the powerful cytotoxins of S. aureus in the lung and prevent damage to the mucosal barrier and innate immune cells.
Collapse
|
24
|
Adaptation of the Staphylococcus aureus leukocidin LukGH for the rabbit host by protein engineering. Biochem J 2019; 476:275-292. [PMID: 30559327 DOI: 10.1042/bcj20180691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023]
Abstract
Host defense against Staphylococcus aureus greatly depends on bacterial clearance by phagocytic cells. LukGH (or LukAB) is the most potent staphylococcal leukocidin towards human phagocytes in vitro, but its role in pathogenesis is obscured by the lack of suitable small animal models because LukGH has limited or no cytotoxicity towards rodent and rabbit compared with human polymorphonuclear cells (PMNs) likely due to an impaired interaction with its cellular receptor, CD11b. We aimed at adapting LukGH for the rabbit host by improving binding to the rabbit homolog of CD11b, specifically its I-domain (CD11b-I). Targeted amino acid substitutions were introduced into the LukH polypeptide to map its receptor interaction site(s). We found that the binding affinity of LukGH variants to the human and rabbit CD11b-I correlated well with their PMN cytotoxicity. Importantly, we identified LukGH variants with significantly improved cytotoxicity towards rabbit PMNs, when expressed recombinantly (10-15-fold) or by engineered S. aureus strains. These findings support the development of small animal models of S. aureus infection with the potential for demonstrating the importance of LukGH in pathogenesis.
Collapse
|
25
|
Haddad O, Merghni A, Elargoubi A, Rhim H, Kadri Y, Mastouri M. Comparative study of virulence factors among methicillin resistant Staphylococcus aureus clinical isolates. BMC Infect Dis 2018; 18:560. [PMID: 30424731 PMCID: PMC6234561 DOI: 10.1186/s12879-018-3457-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/23/2018] [Indexed: 01/04/2023] Open
Abstract
Background Methicillin resistant Staphylococcus aureus (MRSA) is recognized worldwide as a leading cause of hospital and community infections. Biofilm formation by MRSA is an extremely important virulence factor to be understood. Our aim was to establish phenotypic and genotypic characterization of virulence factors among 43 MRSA clinical isolates in a Tunisian hospital. Methods We investigated enzymatic profiles, biofilm production and prevalences of genes encoding intracellular adhesion molecules (icaA and icaD), Microbial Surface Components Recognizing Adhesive Matrix Molecules genes (fnbA, fnbB and cna) and exoenzymes genes (geh, sspA and sspB). Results Our findings revealed that caseinase, gelatinase, lipase and lecithinase activities were detected in 100%, 100%, 76.6% and 93.3% of cases respectively. This study showed that 23 strains (76.7%) were slime producers on Congo red medium. Furthermore, 46.5% and 53.5% of isolates were respectively highly and moderately biofilm-forming on polystyrene. Significant association was found between both biofilm tests. PCR detection showed that 74.4%, 18.6%, 69.8%, 65.1% and 74.4% of isolates harbored fnbA, fnbB, icaA, icaD and cna genes respectively. In addition, 34.9%, 18.6% and 30.2% of MRSA strains were found positive for sspA, sspB and geh genes respectively. Further, statistical data showed that the presence of the fnbA and fnbB genes was significantly associated with a high biofilm production on polystyrene. However, no statistical association was observed for the icaA, icaD and cna genes. Conclusions This study indicates that the detection of fnbA and fnbB contributing to the first step of biofilm formation has been predictable of high biofilm production. As studied factors contribute to MRSA virulence, this research could be of value in orienting towards the development of new preventive and therapeutic measures.
Collapse
Affiliation(s)
- Ons Haddad
- Laboratoire de Microbiologie, CHU Fatouma Bourguiba de Monastir, Monastir, Tunisie. .,Laboratoire des Maladies Transmissible et Substances Biologiquement Actives, LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisie.
| | - Abderrahmen Merghni
- Laboratoire des Maladies Transmissible et Substances Biologiquement Actives, LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisie
| | - Aida Elargoubi
- Laboratoire de Microbiologie, CHU Fatouma Bourguiba de Monastir, Monastir, Tunisie.,Laboratoire des Maladies Transmissible et Substances Biologiquement Actives, LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisie
| | - Hajer Rhim
- Laboratoire de Microbiologie, CHU Fatouma Bourguiba de Monastir, Monastir, Tunisie.,Laboratoire des Maladies Transmissible et Substances Biologiquement Actives, LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisie
| | - Yosr Kadri
- Laboratoire de Microbiologie, CHU Fatouma Bourguiba de Monastir, Monastir, Tunisie.,Laboratoire des Maladies Transmissible et Substances Biologiquement Actives, LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisie
| | - Maha Mastouri
- Laboratoire de Microbiologie, CHU Fatouma Bourguiba de Monastir, Monastir, Tunisie.,Laboratoire des Maladies Transmissible et Substances Biologiquement Actives, LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisie
| |
Collapse
|
26
|
Alanine Scanning Mutagenesis of the MEDI4893 (Suvratoxumab) Epitope Reduces Alpha Toxin Lytic Activity In Vitro and Staphylococcus aureus Fitness in Infection Models. Antimicrob Agents Chemother 2018; 62:AAC.01033-18. [PMID: 30150481 PMCID: PMC6201083 DOI: 10.1128/aac.01033-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/18/2018] [Indexed: 02/06/2023] Open
Abstract
Alpha toxin (AT) is a cytolytic pore-forming toxin that plays a key role in Staphylococcus aureus pathogenesis; consequently, extensive research was undertaken to understand the AT mechanism of action and its utility as a target for novel prophylaxis and treatment strategies against S. aureus infections. MEDI4893 (suvratoxumab) is a human anti-AT IgG1 monoclonal antibody (MAb) that targets AT and is currently in phase 2 clinical development. Alpha toxin (AT) is a cytolytic pore-forming toxin that plays a key role in Staphylococcus aureus pathogenesis; consequently, extensive research was undertaken to understand the AT mechanism of action and its utility as a target for novel prophylaxis and treatment strategies against S. aureus infections. MEDI4893 (suvratoxumab) is a human anti-AT IgG1 monoclonal antibody (MAb) that targets AT and is currently in phase 2 clinical development. As shown previously, the MEDI4893-binding epitope on AT is comprised of the highly conserved amino acid regions 177 to 200 and 261 to 271, suggesting these amino acids are important for AT function. To test this hypothesis and gain insight into the effect of mutations in the epitope on AT neutralization by MEDI4893, nine MEDI4893 contact residues in AT were individually mutated to alanine. Consistent with our hypothesis, 8 out of 9 mutants exhibited >2-fold loss in lytic activity resulting from a defect in cell binding and pore formation. MEDI4893 binding affinity was reduced >2-fold (2- to 27-fold) for 7 out of 9 mutants, and no binding was detected for the W187A mutant. MEDI4893 effectively neutralized all of the lytic mutants in vitro and in vivo. When the defective mutants were introduced into an S. aureus clinical isolate, the mutant-expressing strains exhibited less severe disease in mouse models and were effectively neutralized by MEDI4893. These results indicate the MEDI4893 epitope is highly conserved due in part to its role in AT pore formation and bacterial fitness, thereby decreasing the likelihood for the emergence of MAb-resistant variants.
Collapse
|
27
|
Gillet Y, Henry T, Vandenesch F. Fulminant Staphylococcal Infections. Microbiol Spectr 2018; 6:10.1128/microbiolspec.gpp3-0036-2018. [PMID: 30291703 PMCID: PMC11633626 DOI: 10.1128/microbiolspec.gpp3-0036-2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Fulminant staphylococcal infection indicates an explosive, intense, and severe infection occurring in a patient whose previous condition and antecedent would never have caused any anticipation of life-threatening development. This includes necrotizing pneumonia, necrotizing fasciitis, and to some extent toxic shock syndrome and infective endocarditis. In the three former diseases, toxin production plays a major role whereas in the latter (fulminant presentation of infective endocarditis), association with any particular toxinic profile has never been demonstrated. This article reviews the clinical, pathophysiological, and therapeutic aspects of these diseases.
Collapse
Affiliation(s)
- Yves Gillet
- Department of Pediatric Emergency, Hospices Civils de Lyon, Hôpital Femme Mère Enfant, F-69677, Bron, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University of Lyon, F-69007, Lyon, France
| | - Francois Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University of Lyon, F-69007, Lyon, France
| |
Collapse
|
28
|
Pérez-Montarelo D, Viedma E, Larrosa N, Gómez-González C, Ruiz de Gopegui E, Muñoz-Gallego I, San Juan R, Fernández-Hidalgo N, Almirante B, Chaves F. Molecular Epidemiology of Staphylococcus aureus Bacteremia: Association of Molecular Factors With the Source of Infection. Front Microbiol 2018; 9:2210. [PMID: 30319561 PMCID: PMC6167439 DOI: 10.3389/fmicb.2018.02210] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/29/2018] [Indexed: 12/16/2022] Open
Abstract
Staphylococcus aureus bacteremia (SAB) is associated with high morbidity and mortality, which varies depending on the source of infection. Nevertheless, the global molecular epidemiology of SAB and its possible association with specific virulence factors remains unclear. Using DNA microarrays, a total of 833 S. aureus strains (785 SAB and 48 colonizing strains) collected in Spain over a period of 15 years (2002–2017) were characterized to determine clonal complex (CC), agr type and repertoire of resistance and virulence genes in order to provide an epidemiological overview of CCs causing bloodstream infection, and to analyze possible associations between virulence genes and the most common sources of bacteremia. The results were also analyzed by acquisition (healthcare-associated [HA] and community-acquired [CA]), methicillin-resistant (MRSA) and methicillin-susceptible (MSSA) strains, and patient age (adults vs. children). Our results revealed high clonal diversity among SAB strains with up to 28 different CCs. The most prevalent CCs were CC5 (30.8%), CC30 (20.3%), CC45 (8.3%), CC8 (8.4%), CC15 (7.5%), and CC22 (5.9%), which together accounted for 80% of all cases. A higher proportion of CC5 was found among HA strains than CA strains (35.6 vs. 20.2%, p < 0.001). CC5 was associated with methicillin resistance (14.7 vs. 79.4%, p < 0.001), whereas CC30, CC45, and CC15 were correlated with MSSA strains (p < 0.001). Pathogen-related molecular markers significantly associated with a specific source of bacteremia included the presence of sea, undisrupted hlb and isaB genes with catheter-related bacteremia; sed, splE, and fib genes with endocarditis; undisrupted hlb with skin and soft tissue infections; and finally, CC5, msrA resistance gene and hla gene with osteoarticular source. Our study suggests an association between S. aureus genotype and place of acquisition, methicillin resistance and sources of bloodstream infection, and provides a valuable starting point for further research insights into intrinsic pathogenic mechanisms involved in the development of SAB.
Collapse
|
29
|
Alpha-Toxin Contributes to Biofilm Formation among Staphylococcus aureus Wound Isolates. Toxins (Basel) 2018; 10:toxins10040157. [PMID: 29659477 PMCID: PMC5923323 DOI: 10.3390/toxins10040157] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 01/03/2023] Open
Abstract
Biofilms complicate treatment of Staphylococcus aureus (SA) wound infections. Previously, we determined alpha-toxin (AT)-promoted SA biofilm formation on mucosal tissue. Therefore, we evaluated SA wound isolates for AT production and biofilm formation on epithelium and assessed the role of AT in biofilm formation. Thirty-eight wound isolates were molecularly typed by pulsed-field gel electrophoresis (PFGE), multilocus sequence typing (ST), and spa typing. We measured biofilm formation of these SA isolates in vitro and ex vivo and quantified ex vivo AT production. We also investigated the effect of an anti-AT monoclonal antibody (MEDI4893*) on ex vivo biofilm formation by methicillin-resistant SA (USA 300 LAC) and tested whether purified AT rescued the biofilm defect of hla mutant SA strains. The predominant PFGE/ST combinations were USA100/ST5 (50%) and USA300/ST8 (33%) for methicillin-resistant SA (MRSA, n = 18), and USA200/ST30 (20%) for methicillin-susceptible SA (MSSA, n = 20). Ex vivo AT production correlated significantly with ex vivo SA wound isolate biofilm formation. Anti-alpha-toxin monoclonal antibody (MEDI4893*) prevented ex vivo biofilm formation by MRSA USA300 strain LAC. Wild-type AT rescued the ex vivo biofilm defect of non-AT producing SA strains. These findings provide evidence that AT plays a role in SA biofilm formation on epithelial surfaces and suggest that neutralization of AT may be useful in preventing and treating SA infections.
Collapse
|
30
|
Prevalence of IgG and Neutralizing Antibodies against Staphylococcus aureus Alpha-Toxin in Healthy Human Subjects and Diverse Patient Populations. Infect Immun 2018; 86:IAI.00671-17. [PMID: 29263109 DOI: 10.1128/iai.00671-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/17/2017] [Indexed: 02/02/2023] Open
Abstract
Staphylococcus aureus causes an array of serious infections resulting in high morbidity and mortality worldwide. This study evaluated naturally occurring serum anti-alpha-toxin (anti-AT) antibody levels in human subjects from various age groups, individuals with S. aureus dialysis and surgical-site infections, and S. aureus-colonized versus noncolonized subjects. Anti-AT immunoglobulin G (IgG) and neutralizing antibody (NAb) levels in infants (aged ≤1 year) were significantly lower than those in other populations. In comparison to adolescent, adult, and elderly populations, young children (aged 2 to 10 years) had equivalent anti-AT IgG levels but significantly lower anti-AT NAb levels. Therefore, the development of anti-AT NAbs appears to occur later than that of AT-specific IgG, suggesting a maturation of the immune response to AT. Anti-AT IgG levels were slightly higher in S. aureus-colonized subjects than in noncolonized subjects. The methicillin susceptibility status of colonizing isolates had no effect on anti-AT antibody levels in S. aureus-colonized subjects. The highest anti-AT IgG and NAb levels were observed in dialysis patients with acute S. aureus infection. Anti-AT IgG and NAb levels were well correlated in subjects aged >10 years, regardless of colonization or infection status. These data demonstrate that AT elicits a robust IgG humoral response in infants and young children that becomes stable prior to adolescence, matures into higher levels of NAbs in healthy adolescents, and becomes elevated during S. aureus infection. These findings may assist in identifying subjects and patient populations that could benefit from vaccination or immunoprophylaxis with anti-AT monoclonal antibodies.
Collapse
|
31
|
Hook JL, Islam MN, Parker D, Prince AS, Bhattacharya S, Bhattacharya J. Disruption of staphylococcal aggregation protects against lethal lung injury. J Clin Invest 2018; 128:1074-1086. [PMID: 29431734 DOI: 10.1172/jci95823] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/04/2018] [Indexed: 01/23/2023] Open
Abstract
Infection by Staphylococcus aureus strain USA300 causes tissue injury, multiorgan failure, and high mortality. However, the mechanisms by which the bacteria adhere to, then stabilize on, mucosal surfaces before causing injury remain unclear. We addressed these issues through the first real-time determinations of USA300-alveolar interactions in live lungs. We found that within minutes, inhaled USA300 established stable, self-associated microaggregates in niches at curved, but not at flat, regions of the alveolar wall. The microaggregates released α-hemolysin toxin, causing localized alveolar injury, as indicated by epithelial dye loss, mitochondrial depolarization, and cytosolic Ca2+ increase. Spread of cytosolic Ca2+ through intercellular gap junctions to adjoining, uninfected alveoli caused pulmonary edema. Systemic pretreatment with vancomycin, a USA300-cidal antibiotic, failed to protect mice infected with inhaled WT USA300. However, vancomycin pretreatment markedly abrogated mortality in mice infected with mutant USA300 that lacked the aggregation-promoting factor PhnD. We interpret USA300-induced mortality as having resulted from rapid bacterial aggregation in alveolar niches. These findings indicate, for the first time to our knowledge, that alveolar microanatomy is critical in promoting the aggregation and, hence, in causing USA300-induced alveolar injury. We propose that in addition to antibiotics, strategies for bacterial disaggregation may constitute novel therapy against USA300-induced lung injury.
Collapse
Affiliation(s)
- Jaime L Hook
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Mohammad N Islam
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | | | - Sunita Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Pediatrics, and
| | - Jahar Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
32
|
Ruzin A, Wu Y, Yu L, Yu XQ, Tabor DE, Mok H, Tkaczyk C, Jensen K, Bellamy T, Roskos L, Esser MT, Jafri HS. Characterisation of anti-alpha toxin antibody levels and colonisation status after administration of an investigational human monoclonal antibody, MEDI4893, against Staphylococcus aureus alpha toxin. Clin Transl Immunology 2018; 7:e1009. [PMID: 29484186 PMCID: PMC5822409 DOI: 10.1002/cti2.1009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/20/2017] [Accepted: 12/29/2017] [Indexed: 01/07/2023] Open
Abstract
Objectives MEDI4893 is a novel, long‐acting human monoclonal antibody targeting Staphylococcus aureus (SA) alpha toxin (AT). This report presents the results of the exploratory analyses from a randomised phase 1 dose‐escalation study in healthy human subjects receiving single intravenous MEDI4893 doses or placebo. Methods Anti‐AT antibodies and AT expression were measured as described previously. Nasal swabs were analysed by culture and PCR. Data were summarised by treatment groups and visits by using SAS System Version 9.3. Results Subjects receiving 2250 or 5000 mg of MEDI4893 had the highest serum anti‐AT neutralising antibody (NAb) levels: approximately 180‐ to 240‐, 70‐ to 100‐ and sevenfold to 10‐fold higher than respective baseline levels at peak, 30 and 360 days, respectively. In these subjects, levels of serum anti‐AT NAbs were >3.2 International Units (IU) mL−1 for at least 211 days. In the upper respiratory tract, anti‐AT NAb levels increased with MEDI4893 dose. No apparent effect of MEDI4893 on SA nasal colonisation, hla gene sequence or AT expression was observed. Five AT variants were detected, their lytic activity was fully neutralised by MEDI4893. Discussion Our results indicate that (1) MEDI4893 administration at 2250 and 5000 mg would provide effective immunoprophylaxis against systemic SA disease; (2) MEDI4983 distributes to the upper respiratory tract and retains neutralising activity against AT; and (3) potential for emergence of MEDI4893 resistance is low. Conclusion Intravenous administration of MEDI4893 maintained levels of anti‐AT NAbs in serum and nasal mucosa that may provide effective immunoprophylaxis against SA disease and support continued clinical development of MEDI4893.
Collapse
Affiliation(s)
| | | | - Li Yu
- MedImmune Gaithersburg MD USA
| | - Xiang-Qing Yu
- MedImmune Gaithersburg MD USA.,Present address: Janssen Pharmaceuticals, Inc. Johnson & Johnson Spring House PA USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rouha H, Weber S, Janesch P, Maierhofer B, Gross K, Dolezilkova I, Mirkina I, Visram ZC, Malafa S, Stulik L, Badarau A, Nagy E. Disarming Staphylococcus aureus from destroying human cells by simultaneously neutralizing six cytotoxins with two human monoclonal antibodies. Virulence 2017; 9:231-247. [PMID: 29099326 PMCID: PMC5955178 DOI: 10.1080/21505594.2017.1391447] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pathogenesis of Staphylococcus aureus is increasingly recognized to be driven by powerful toxins. Staphylococcus aureus employs up to six pore-forming toxins to subvert the human host defense and to promote bacterial invasion: alpha-hemolysin that disrupts epithelial and endothelial barriers and five leukocidins that lyse phagocytes involved in bacterial clearance. Previously, we described two human monoclonal antibodies (mAbs), ASN-1 that neutralizes alpha-hemolysin and four leukocidins (LukSF-PV, LukED, HlgAB, HlgCB), and ASN-2 that inactivates the 5th leukocidin, LukGH. In this study we tested the individual and combined effects of ASN-1 and ASN-2 in multiple in vitro models employing relevant human target cells. We found that diverse S. aureus isolates with different genetic backgrounds (based on MLST- and spa-typing) and antibiotic sensitivity (both MRSA and MSSA) displayed greatly different cytotoxin expression patterns influenced by the type of growth medium used. Both mAbs were required to fully prevent the lysis of human neutrophils exposed to the mixture of recombinant cytotoxins or native toxins present in the culture supernatants of S. aureus isolates. Flow cytometry confirmed the protective effects of ASN-1 + ASN-2 (known as ASN100) on granulocytes, monocytes, NK-cells and T-lymphocytes. ASN-1 alone preserved the integrity of a 3D-primary culture of human tracheal/bronchial mucociliary epithelial tissue infected with S. aureus. We conclude that simultaneous inhibition of alpha-hemolysin and five leukocidins by ASN100 blocks cytolytic activity of S. aureus towards human target cells in vitro.
Collapse
Affiliation(s)
- Harald Rouha
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Susanne Weber
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Philipp Janesch
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Barbara Maierhofer
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Karin Gross
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Ivana Dolezilkova
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Irina Mirkina
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Zehra C Visram
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Stefan Malafa
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Lukas Stulik
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Adriana Badarau
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| | - Eszter Nagy
- a Arsanis Biosciences, Helmut-Qualtinger-Gasse 2, Campus Vienna Biocenter , Vienna , Austria
| |
Collapse
|
34
|
Diep BA, Le VTM, Badiou C, Le HN, Pinheiro MG, Duong AH, Wang X, Dip EC, Aguiar-Alves F, Basuino L, Marbach H, Mai TT, Sarda MN, Kajikawa O, Matute-Bello G, Tkaczyk C, Rasigade JP, Sellman BR, Chambers HF, Lina G. IVIG-mediated protection against necrotizing pneumonia caused by MRSA. Sci Transl Med 2017; 8:357ra124. [PMID: 27655850 DOI: 10.1126/scitranslmed.aag1153] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/02/2016] [Indexed: 12/12/2022]
Abstract
New therapeutic approaches are urgently needed to improve survival outcomes for patients with necrotizing pneumonia caused by Staphylococcus aureus One such approach is adjunctive treatment with intravenous immunoglobulin (IVIG), but clinical practice guidelines offer conflicting recommendations. In a preclinical rabbit model, prophylaxis with IVIG conferred protection against necrotizing pneumonia caused by five different epidemic strains of community-associated methicillin-resistant S. aureus (MRSA) as well as a widespread strain of hospital-associated MRSA. Treatment with IVIG, either alone or in combination with vancomycin or linezolid, improved survival outcomes in this rabbit model. Two specific IVIG antibodies that neutralized the toxic effects of α-hemolysin (Hla) and Panton-Valentine leukocidin (PVL) conferred protection against necrotizing pneumonia in the rabbit model. This mechanism of action of IVIG was uncovered by analyzing loss-of-function mutant bacterial strains containing deletions in 17 genes encoding staphylococcal exotoxins, which revealed only Hla and PVL as having an impact on necrotizing pneumonia. These results demonstrate the potential clinical utility of IVIG in the treatment of severe pneumonia induced by S. aureus.
Collapse
Affiliation(s)
- Binh An Diep
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA.
| | - Vien T M Le
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Cedric Badiou
- INSERM U1111, Université Lyon 1, CNRS UMR5308, ENS Lyon, Lyon, France. Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Bron, France
| | - Hoan N Le
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Marcos Gabriel Pinheiro
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA. Pathology Program, Fluminense Federal University, Niterói, RJ, Brazil
| | - Au H Duong
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Xing Wang
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Etyene Castro Dip
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Fábio Aguiar-Alves
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA. Pathology Program, Fluminense Federal University, Niterói, RJ, Brazil
| | - Li Basuino
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Helene Marbach
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Thuy T Mai
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Marie N Sarda
- Laboratory of Immunology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Osamu Kajikawa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Gustavo Matute-Bello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Christine Tkaczyk
- Department of Infectious Diseases, MedImmune, LLC, Gaithersburg, MD 20878, USA
| | - Jean-Philippe Rasigade
- INSERM U1111, Université Lyon 1, CNRS UMR5308, ENS Lyon, Lyon, France. Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Bron, France
| | - Bret R Sellman
- Department of Infectious Diseases, MedImmune, LLC, Gaithersburg, MD 20878, USA
| | - Henry F Chambers
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Gerard Lina
- INSERM U1111, Université Lyon 1, CNRS UMR5308, ENS Lyon, Lyon, France. Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Bron, France.
| |
Collapse
|
35
|
Future Directions and Molecular Basis of Ventilator Associated Pneumonia. Can Respir J 2017; 2017:2614602. [PMID: 29162982 PMCID: PMC5661065 DOI: 10.1155/2017/2614602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 09/14/2017] [Indexed: 12/26/2022] Open
Abstract
Mechanical ventilation is a lifesaving treatment and has complications such as ventilator associated pneumonia (VAP) that lead to high morbidity and mortality. Moreover VAP is the second most common hospital-acquired infection in pediatric intensive care units. Although it is still not well understood, understanding molecular pathogenesis is essential for preventing and treating pneumonia. A lot of microbes are detected as a causative agent of VAP. The most common isolated VAP pathogens in pediatric patients are Staphylococcus aureus, Pseudomonas aeruginosa, and other gram negative bacteria. All of the bacteria have different pathogenesis due to their different virulence factors and host reactions. This review article focused on mechanisms of VAP with molecular pathogenesis of the causative bacteria one by one from the literature. We hope that we know more about molecular pathogenesis of VAP and we can investigate and focus on the management of the disease in near future.
Collapse
|
36
|
Song Z, Zhang X, Zhang L, Xu F, Tao X, Zhang H, Lin X, Kang L, Xiang Y, Lai X, Zhang Q, Huang K, Dai Y, Yin Y, Cao J. Progranulin Plays a Central Role in Host Defense during Sepsis by Promoting Macrophage Recruitment. Am J Respir Crit Care Med 2017; 194:1219-1232. [PMID: 27149013 DOI: 10.1164/rccm.201601-0056oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
RATIONALE Progranulin, a widely expressed protein, has multiple physiological functions. The functional role of progranulin in the host response to sepsis remains unknown. OBJECTIVES To assess the role of progranulin in the host response to sepsis. METHODS Effects of progranulin on host response to sepsis were determined. MEASUREMENTS AND MAIN RESULTS Progranulin concentrations were significantly elevated in adult (n = 74) and pediatric (n = 26) patients with sepsis relative to corresponding healthy adult (n = 36) and pediatric (n = 17) control subjects, respectively. By using a low-lethality model of nonsevere sepsis, we observed that progranulin deficiency not only increased mortality but also decreased bacterial clearance during sepsis. The decreased host defense to sepsis in progranulin-deficient mice was associated with reduced macrophage recruitment, with correspondingly impaired chemokine CC receptor ligand 2 (CCL2) production in peritoneal lavages during the early phase of sepsis. Progranulin derived from hematopoietic cells contributed to host defense in sepsis. Therapeutic administration of recombinant progranulin not only rescued impaired host defense in progranulin-deficient mice after nonsevere sepsis but also protected wild-type mice against a high-lethality model of severe sepsis. Progranulin-mediated protection against sepsis was closely linked to improved peritoneal macrophage recruitment. In addition, CCL2 treatment of progranulin-deficient mice improved survival and decreased peritoneal bacterial loads during sepsis, at least in part through promotion of peritoneal macrophage recruitment. CONCLUSIONS This proof-of-concept study supports a central role of progranulin-dependent macrophage recruitment in host defense to sepsis, opening new opportunities to host-directed therapeutic strategy that manipulate host immune response in the treatment of sepsis.
Collapse
Affiliation(s)
- Zhixin Song
- 1 Department of Laboratory Medicine.,2 Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, and
| | - Xuemei Zhang
- 2 Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, and
| | | | - Fang Xu
- 3 Department of Emergency and Intensive Care Unit
| | - Xintong Tao
- 2 Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, and
| | - Hua Zhang
- 4 Department of Obstetrics and Gynecology, and
| | - Xue Lin
- 1 Department of Laboratory Medicine
| | - Lihua Kang
- 2 Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, and
| | - Yu Xiang
- 1 Department of Laboratory Medicine
| | | | - Qun Zhang
- 5 Clinical Laboratories Center, Affiliated Children's Hospital of Chongqing Medical University, Chongqing, China; and
| | - Kun Huang
- 6 Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University
| | - Yubing Dai
- 7 Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas
| | - Yibing Yin
- 2 Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, and
| | - Ju Cao
- 1 Department of Laboratory Medicine
| |
Collapse
|
37
|
Stulik L, Hudcova J, Craven DE, Nagy G, Nagy E. Low Efficacy of Antibiotics Against Staphylococcus aureus Airway Colonization in Ventilated Patients. Clin Infect Dis 2017; 64:1081-1088. [PMID: 28158685 DOI: 10.1093/cid/cix055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 02/02/2017] [Indexed: 11/14/2022] Open
Abstract
Background Airway-colonization by Staphylococcus aureus predisposes to the development of ventilator-associated tracheobronchitis (VAT) and ventilator-associated pneumonia (VAP). Despite extensive antibiotic treatment of intensive care unit patients, limited data are available on the efficacy of antibiotics on bacterial airway colonization and/or prevention of infections. Therefore, microbiologic responses to antibiotic treatment were evaluated in ventilated patients. Methods Results of semiquantitative analyses of S. aureus burden in serial endotracheal-aspirate (ETA) samples and VAT/VAP diagnosis were correlated to antibiotic treatment. Minimum inhibitory concentrations of relevant antibiotics using serially collected isolates were evaluated. Results Forty-eight mechanically ventilated patients who were S. aureus positive by ETA samples and treated with relevant antibiotics for at least 2 consecutive days were included in the study. Vancomycin failed to reduce methicillin-resistant S. aureus (MRSA) or methicillin-susceptible S. aureus (MSSA) burden in the airways. Oxacillin was ineffective for MSSA colonization in approximately 30% of the patients, and responders were typically coadministered additional antibiotics. Despite antibiotic exposure, 15 of the 39 patients (approximately 38%) colonized only by S. aureus and treated with appropriate antibiotic for at least 2 days still progressed to VAP. Importantly, no change in antibiotic susceptibility of S. aureus isolates was observed during treatment. Staphylococcus aureus colonization levels inversely correlated with the presence of normal respiratory flora. Conclusions Antibiotic treatment is ineffective in reducing S. aureus colonization in the lower airways and preventing VAT or VAP. Staphylococcus aureus is in competition for colonization with the normal respiratory flora. To improve patient outcomes, alternatives to antibiotics are urgently needed.
Collapse
Affiliation(s)
- Lukas Stulik
- Arsanis Biosciences GmbH, Vienna, Austria.,Arsanis, Inc, Waltham, MA, USA
| | - Jana Hudcova
- Department of Surgical Critical Care, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Donald E Craven
- Infectious Diseases, Lahey Hospital and Medical Center, Burlington, Massachusetts, USA
| | - Gabor Nagy
- Arsanis Biosciences GmbH, Vienna, Austria.,Arsanis, Inc, Waltham, MA, USA
| | - Eszter Nagy
- Arsanis Biosciences GmbH, Vienna, Austria.,Arsanis, Inc, Waltham, MA, USA
| |
Collapse
|
38
|
Gomes-Fernandes M, Laabei M, Pagan N, Hidalgo J, Molinos S, Villar Hernandez R, Domínguez-Villanueva D, Jenkins ATA, Lacoma A, Prat C. Accessory gene regulator (Agr) functionality in Staphylococcus aureus derived from lower respiratory tract infections. PLoS One 2017; 12:e0175552. [PMID: 28410390 PMCID: PMC5391941 DOI: 10.1371/journal.pone.0175552] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/28/2017] [Indexed: 01/09/2023] Open
Abstract
Objective Characterization of Staphylococcus aureus clinical isolates derived from lower respiratory tract infections (LRTIs), and correlation between the functionality of the accessory gene regulator (Agr) and genotypic and phenotypic characteristics, clinical variables and clinical outcome. Methods S aureus isolates derived from LRTIs and control groups (nasal carriage and bacteraemia) were genotyped using StaphyType DNA microarray. Agr activity was evaluated using the CAMP synergistic haemolysis assay and the Vesicle Lysis Test (VLT). Discordant strains were analysed by quantitative reverse-transcriptase real-time PCR (qRT-PCR). Results Agr was functional in 79.7% and 84.5% of strains according to the CAMP and VLT assays respectively. Higher concordance with RNAIII expression measured by qRT-PCR was observed with the VLT assay (76.2%) compared with the CAMP assay (23.8%). No statistically significant differences were observed in Agr functionality between the study groups, nor the phenotypical/genotypical bacterial characteristics. No association between increased mortality/respiratory complications and Agr function was observed. Conclusions Agr activity was high (82.2%) in isolates from LRTIs suggesting the importance of this global regulator in lower respiratory tract colonisation and infection. However, equally high Agr activity was observed in isolates derived from nasal carriage and bacteraemia, contradictory to previous observations. Agr functionality measured by the VLT assay was superior to CAMP assay.
Collapse
Affiliation(s)
- Meissiner Gomes-Fernandes
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Maisem Laabei
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Natalia Pagan
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Jessica Hidalgo
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Sònia Molinos
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Raquel Villar Hernandez
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Dídac Domínguez-Villanueva
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | | | - Alicia Lacoma
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- CIBER Enfermedades Respiratorias, CIBER, Instituto de Salud Carlos III, Badalona, Spain
| | - Cristina Prat
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d’ Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- CIBER Enfermedades Respiratorias, CIBER, Instituto de Salud Carlos III, Badalona, Spain
- * E-mail:
| |
Collapse
|
39
|
Targeting Alpha Toxin To Mitigate Its Lethal Toxicity in Ferret and Rabbit Models of Staphylococcus aureus Necrotizing Pneumonia. Antimicrob Agents Chemother 2017; 61:AAC.02456-16. [PMID: 28115346 DOI: 10.1128/aac.02456-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/15/2017] [Indexed: 11/20/2022] Open
Abstract
The role broad-spectrum antibiotics play in the spread of antimicrobial resistance, coupled with their effect on the healthy microbiome, has led to advances in pathogen-specific approaches for the prevention or treatment of serious bacterial infections. One approach in clinical testing is passive immunization with a monoclonal antibody (MAb) targeting alpha toxin for the prevention or treatment of Staphylococcus aureus pneumonia. Passive immunization with the human anti-alpha toxin MAb, MEDI4893*, has been shown to improve disease outcome in murine S. aureus pneumonia models. The species specificity of some S. aureus toxins necessitates testing anti-S. aureus therapeutics in alternate species. We developed a necrotizing pneumonia model in ferrets and utilized an existing rabbit pneumonia model to characterize MEDI4893* protective activity in species other than mice. MEDI4893* prophylaxis reduced disease severity in ferret and rabbit pneumonia models against both community-associated methicillin-resistant S. aureus (MRSA) and hospital-associated MRSA strains. In addition, adjunctive treatment of MEDI4893* with either vancomycin or linezolid provided enhanced protection in rabbits relative to the antibiotics alone. These results confirm that MEDI4893 is a promising candidate for immunotherapy against S. aureus pneumonia.
Collapse
|
40
|
Cohen TS, Hilliard JJ, Jones-Nelson O, Keller AE, O'Day T, Tkaczyk C, DiGiandomenico A, Hamilton M, Pelletier M, Wang Q, Diep BA, Le VTM, Cheng L, Suzich J, Stover CK, Sellman BR. Staphylococcus aureus α toxin potentiates opportunistic bacterial lung infections. Sci Transl Med 2016; 8:329ra31. [PMID: 26962155 DOI: 10.1126/scitranslmed.aad9922] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Broad-spectrum antibiotic use may adversely affect a patient's beneficial microbiome and fuel cross-species spread of drug resistance. Although alternative pathogen-specific approaches are rationally justified, a major concern for this precision medicine strategy is that co-colonizing or co-infecting opportunistic bacteria may still cause serious disease. In a mixed-pathogen lung infection model, we find that the Staphylococcus aureus virulence factor α toxin potentiates Gram-negative bacterial proliferation, systemic spread, and lethality by preventing acidification of bacteria-containing macrophage phagosomes, thereby reducing effective killing of both S. aureus and Gram-negative bacteria. Prophylaxis or early treatment with a single α toxin neutralizing monoclonal antibody prevented proliferation of co-infecting Gram-negative pathogens and lethality while also promoting S. aureus clearance. These studies suggest that some pathogen-specific, antibody-based approaches may also work to reduce infection risk in patients colonized or co-infected with S. aureus and disparate drug-resistant Gram-negative bacterial opportunists.
Collapse
Affiliation(s)
- Taylor S Cohen
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Jamese J Hilliard
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Omari Jones-Nelson
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Ashley E Keller
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Terrence O'Day
- Department of Translational Science, MedImmune, Gaithersburg, MD 20878, USA
| | - Christine Tkaczyk
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | | | - Melissa Hamilton
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Mark Pelletier
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Qun Wang
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Binh An Diep
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA. Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Vien T M Le
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Lily Cheng
- Department of Translational Science, MedImmune, Gaithersburg, MD 20878, USA
| | - JoAnn Suzich
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - C Kendall Stover
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Bret R Sellman
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA.
| |
Collapse
|
41
|
Cohen TS, Jones-Nelson O, Hotz M, Cheng L, Miller LS, Suzich J, Stover CK, Sellman BR. S. aureus blocks efferocytosis of neutrophils by macrophages through the activity of its virulence factor alpha toxin. Sci Rep 2016; 6:35466. [PMID: 27739519 PMCID: PMC5064327 DOI: 10.1038/srep35466] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022] Open
Abstract
Bacterial pneumonia, such as those caused by Staphylococcus aureus, is associated with an influx of inflammatory neutrophils into the lung tissue and airways. Regulation and clearance of recruited neutrophils is essential for preventing tissue damage by “friendly fire”, a responsibility of macrophages in a process called efferocytosis. We hypothesized that S. aureus impairs efferocytosis by alveolar macrophages (AMs) through the activity of the secreted virulence factor alpha toxin (AT), which has been implicated in altering the antimicrobial function of AMs. Infection of mice lacking AMs resulted in significantly increased numbers of neutrophils in the lung, while clearance of neutrophils delivered intranasally into uninfected mice was reduced in AM depleted animals. In vitro, sublytic levels of AT impaired uptake of apoptotic neutrophils by purified AMs. In vivo, the presence of AT reduced uptake of neutrophils by AMs. Differential uptake of neutrophils was not due to changes in either the CD47/CD172 axis or CD36 levels. AT significantly reduced lung expression of CCN1 and altered AM surface localization of DD1α, two proteins known to influence efferocytosis. We conclude that AT may contribute to tissue damage during S. aureus pneumonia by inhibiting the ability of AM to clear neutrophils at the site of infection.
Collapse
Affiliation(s)
- Taylor S Cohen
- Department of Infectious Disease, Medimmune, LLC One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Omari Jones-Nelson
- Department of Infectious Disease, Medimmune, LLC One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Meghan Hotz
- Department of Infectious Disease, Medimmune, LLC One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Lily Cheng
- Department of Translational Science, Medimmune, LLC One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Lloyd S Miller
- Department of Dermatology, John Hopkins University School of Medicine, Baltimore MD 21231, USA
| | - JoAnn Suzich
- Department of Infectious Disease, Medimmune, LLC One MedImmune Way, Gaithersburg, MD 20878, USA
| | - C Kendall Stover
- Department of Infectious Disease, Medimmune, LLC One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Bret R Sellman
- Department of Infectious Disease, Medimmune, LLC One MedImmune Way, Gaithersburg, MD 20878, USA
| |
Collapse
|
42
|
Craven DE, Hudcova J, Lei Y, Craven KA, Waqas A. Pre-emptive antibiotic therapy to reduce ventilator-associated pneumonia: "thinking outside the box". Crit Care 2016; 20:300. [PMID: 27680980 PMCID: PMC5041322 DOI: 10.1186/s13054-016-1472-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Mechanically ventilated, intubated patients are at increased risk for tracheal colonization with bacterial pathogens that may progress to heavy bacterial colonization, ventilator-associated tracheobronchitis (VAT), and/or ventilator-associated pneumonia (VAP). Previous studies report that 10 to 30 % of patients with VAT progress to VAP, resulting in increased morbidity and significant acute and chronic healthcare costs. Several natural history studies, randomized, controlled trials, and a meta-analysis have reported antibiotic treatment for VAT can reduce VAP, ventilator days, length of intensive care unit (ICU) stay, and patient morbidity and mortality. We discuss early diagnostic criteria, etiologic agents, and benefits of initiating, early, appropriate intravenous or aerosolized antibiotic(s) to treat VAT and reduce VAP, to improve patient outcomes by reducing lung damage, length of ICU stay, and healthcare costs.
Collapse
Affiliation(s)
- Donald E Craven
- Center for Infectious Diseases & Prevention, Lahey Hospital and Medical Center, 31 Mall Rd, Burlington, MA, 01805, USA. .,Tufts University School of Medicine, Boston, MA, USA.
| | - Jana Hudcova
- Surgical Critical Care, Lahey Hospital and Medical Center, Burlington, MA, USA.,Tufts University School of Medicine, Boston, MA, USA
| | - Yuxiu Lei
- Pulmonary and Critical Care Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Kathleen A Craven
- New England Independent Review Board for Human Research, Wellesley, MA, USA
| | - Ahsan Waqas
- Brookdale University Medical Center, Brooklyn, NY, USA
| |
Collapse
|
43
|
Improved Protection in a Rabbit Model of Community-Associated Methicillin-Resistant Staphylococcus aureus Necrotizing Pneumonia upon Neutralization of Leukocidins in Addition to Alpha-Hemolysin. Antimicrob Agents Chemother 2016; 60:6333-40. [PMID: 27527081 DOI: 10.1128/aac.01213-16] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/06/2016] [Indexed: 12/31/2022] Open
Abstract
Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA), especially the USA300 pulsotype, is a frequent cause of skin and soft tissue infections and severe pneumonia. Despite appropriate antibiotic treatment, complications are common and pneumonia is associated with high mortality. S. aureus strains express multiple cytotoxins, including alpha-hemolysin (Hla) and up to five bicomponent leukocidins that specifically target phagocytic cells for lysis. CA-MRSA USA300 strains carry the genes for all six cytotoxins. Species specificity of the leukocidins greatly contributes to the ambiguity regarding their role in S. aureus pathogenesis. We performed a comparative analysis of the leukocidin susceptibility of human, rabbit, and mouse polymorphonuclear leukocytes (PMNs) to assess the translational value of mouse and rabbit S. aureus models. We found that mouse PMNs were largely resistant to LukSF-PV, HlgAB, and HlgCB and susceptible only to LukED, whereas rabbit and human PMNs were highly sensitive to all these cytotoxins. In the rabbit pneumonia model with a USA300 CA-MRSA strain, passive immunization with a previously identified human monoclonal antibody (MAb), Hla-F#5, which cross-neutralizes Hla, LukSF-PV, HlgAB, HlgCB, and LukED, provided full protection, whereas an Hla-specific MAb was only partially protective. In the mouse USA300 CA-MRSA pneumonia model, both types of antibodies demonstrated full protection, suggesting that Hla, but not leukocidin(s), is the principal virulence determinant in mice. As the rabbit recapitulates the high susceptibility to leukocidins characteristic of humans, this species represents a valuable model for assessing novel, cytotoxin-targeting anti-S. aureus therapeutic approaches.
Collapse
|
44
|
Giulieri SG, Holmes NE, Stinear TP, Howden BP. Use of bacterial whole-genome sequencing to understand and improve the management of invasive Staphylococcus aureus infections. Expert Rev Anti Infect Ther 2016; 14:1023-1036. [PMID: 27626511 DOI: 10.1080/14787210.2016.1233815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Management of invasive Staphylococcus aureus infections is complex. Dramatic improvements in bacterial whole genome sequencing (WGS) offer new opportunities for personalising the treatment of S. aureus infections. Areas covered: We address recent achievements in S. aureus genomics, describe genetic determinants of antibiotic resistance and summarise studies that have defined molecular characteristics associated with risk and outcome of S. aureus invasive infections. Potential clinical use of WGS for resistance prediction, infection outcome stratification and management of persistent /relapsing infections is critically discussed. Expert commentary: WGS is not only providing invaluable information to track the emergence and spread of important S. aureus clones, but also allows rapid determination of resistance genotypes in the clinical environment. An evolving opportunity is to infer clinically important outcomes and optimal therapeutic approaches from widely available S. aureus genome data, with the goal of individualizing management of invasive S. aureus infections.
Collapse
Affiliation(s)
- Stefano G Giulieri
- a Microbiological Diagnostic Unit Public Health Laboratory , Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne , Melbourne , Australia.,b Infectious Diseases Service , Department of Medicine, Lausanne University Hospital , Lausanne , Switzerland
| | - Natasha E Holmes
- c Infectious Diseases Department , Austin Health , Heidelberg , Australia
| | - Timothy P Stinear
- d Doherty Applied Microbial Genomics , Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne , Melbourne , Australia.,e Department of Microbiology and Immunology , The University of Melbourne at the Peter Doherty Institute for Infection and Immunity , Melbourne , Australia
| | - Benjamin P Howden
- a Microbiological Diagnostic Unit Public Health Laboratory , Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne , Melbourne , Australia.,c Infectious Diseases Department , Austin Health , Heidelberg , Australia.,e Department of Microbiology and Immunology , The University of Melbourne at the Peter Doherty Institute for Infection and Immunity , Melbourne , Australia
| |
Collapse
|
45
|
Staphylococcus aureus Alpha-Toxin Is Conserved among Diverse Hospital Respiratory Isolates Collected from a Global Surveillance Study and Is Neutralized by Monoclonal Antibody MEDI4893. Antimicrob Agents Chemother 2016; 60:5312-21. [PMID: 27324766 PMCID: PMC4997823 DOI: 10.1128/aac.00357-16] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus infections lead to an array of illnesses ranging from mild skin infections to serious diseases, such endocarditis, osteomyelitis, and pneumonia. Alpha-toxin (Hla) is a pore-forming toxin, encoded by the hla gene, that is thought to play a key role in S. aureus pathogenesis. A monoclonal antibody targeting Hla, MEDI4893, is in clinical development for the prevention of S. aureus ventilator-associated pneumonia (VAP). The presence of the hla gene and Hla protein in 994 respiratory isolates collected from patients in 34 countries in Asia, Europe, the United States, Latin America, the Middle East, Africa, and Australia was determined. Hla levels were correlated with the geographic location, age of the subject, and length of stay in the hospital. hla gene sequence analysis was performed, and mutations were mapped to the Hla crystal structure. S. aureus supernatants containing Hla variants were tested for susceptibility or resistance to MEDI4893. The hla gene was present and Hla was expressed in 99.0% and 83.2% of the isolates, respectively, regardless of geographic region, hospital locale, or age of the subject. More methicillin-susceptible than methicillin-resistant isolates expressed Hla (86.9% versus 78.8%; P = 0.0007), and S. aureus isolates from pediatric patients expressed the largest amounts of Hla. Fifty-seven different Hla subtypes were identified, and 91% of the isolates encoded an Hla subtype that was neutralized by MED4893. This study demonstrates that Hla is conserved in diverse S. aureus isolates from around the world and is an attractive target for prophylactic monoclonal antibody (MAb) or vaccine development.
Collapse
|
46
|
Lei Y, Hudcova J, Rashid J, Sarwar A, Gillespie W, Finn C, Goggin M, Omran MB, Boroda E, Craven DE. Natural History, Outcomes and Antibiotic Treatment for Ventilator-Associated Tracheobronchitis in Critical Ill Patients. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/mri.2016.51001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
47
|
Antibiotic therapy for ventilator-associated tracheobronchitis: a standard of care to reduce pneumonia, morbidity and costs? Curr Opin Pulm Med 2015; 21:250-9. [PMID: 25784245 DOI: 10.1097/mcp.0000000000000158] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW The present review draws our attention to ventilator-associated tracheobronchitis (VAT) as a distinct clinical entity that has been associated with progression to ventilator-associated pneumonia (VAP) and worse patient outcomes. In contrast to VAP, which has been extensively investigated for over the past 30 years, most VAT studies have been conducted in the past decade. There are ample data which demonstrate that VAT may progress to VAP, have more ventilator days, and have longer ICU stay that may translate into higher healthcare costs. RECENT FINDINGS The article focuses on the diagnostic criteria for VAT, causative agents, and studies analyzing associations between VAT and patient outcomes in relation to early, appropriate intravenous, and/or aerosolized antibiotic therapy. Aerosolized antibiotic treatment delivered by improved device technology is a novel approach that has proved to be effective for the treatment and eradication of multidrug-resistant bacterial pathogens. Aerosolized antibiotics are effective in decreasing the use of systemic antibiotics, reducing bacterial resistance, and may also facilitate clinical resolution of infection. SUMMARY Evidence presented in this review supports treatment of VAT with early and appropriate antibiotic therapy as a standard of care to reduce VAP, ventilator days, and duration of ICU stay in high-risk patient population.
Collapse
|
48
|
Arroliga AC, Velazco JF, Midturi JK, Ghamande SA. Back to the future: α-hemolysin activity on blood agar to predict ventilator-associated pneumonia caused by Staphylococcus aureus. Am J Respir Crit Care Med 2015; 190:1086-8. [PMID: 25398106 DOI: 10.1164/rccm.201410-1886ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
49
|
Koymans KJ, Vrieling M, Gorham RD, van Strijp JAG. Staphylococcal Immune Evasion Proteins: Structure, Function, and Host Adaptation. Curr Top Microbiol Immunol 2015; 409:441-489. [PMID: 26919864 DOI: 10.1007/82_2015_5017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Staphylococcus aureus is a successful human and animal pathogen. Its pathogenicity is linked to its ability to secrete a large amount of virulence factors. These secreted proteins interfere with many critical components of the immune system, both innate and adaptive, and hamper proper immune functioning. In recent years, numerous studies have been conducted in order to understand the molecular mechanism underlying the interaction of evasion molecules with the host immune system. Structural studies have fundamentally contributed to our understanding of the mechanisms of action of the individual factors. Furthermore, such studies revealed one of the most striking characteristics of the secreted immune evasion molecules: their conserved structure. Despite high-sequence variability, most immune evasion molecules belong to a small number of structural categories. Another remarkable characteristic is that S. aureus carries most of these virulence factors on mobile genetic elements (MGE) or ex-MGE in its accessory genome. Coevolution of pathogen and host has resulted in immune evasion molecules with a highly host-specific function and prevalence. In this review, we explore how these shared structures and genomic locations relate to function and host specificity. This is discussed in the context of therapeutic options for these immune evasion molecules in infectious as well as in inflammatory diseases.
Collapse
Affiliation(s)
- Kirsten J Koymans
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - Manouk Vrieling
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ronald D Gorham
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, G04-614, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|