1
|
Nazli A, Tao W, You H, He X, He Y. Treatment of MRSA Infection: Where are We? Curr Med Chem 2024; 31:4425-4460. [PMID: 38310393 DOI: 10.2174/0109298673249381231130111352] [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: 02/19/2023] [Revised: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 02/05/2024]
Abstract
Staphylococcus aureus is a leading cause of septicemia, endocarditis, pneumonia, skin and soft tissue infections, bone and joint infections, and hospital-acquired infections. In particular, methicillin-resistant Staphylococcus aureus (MRSA) is associated with high morbidity and mortality, and continues to be a major public health problem. The emergence of multidrug-resistant MRSA strains along with the wide consumption of antibiotics has made anti-MRSA treatment a huge challenge. Novel treatment strategies (e.g., novel antimicrobials and new administrations) against MRSA are urgently needed. In the past decade, pharmaceutical companies have invested more in the research and development (R&D) of new antimicrobials and strategies, spurred by favorable policies. All research articles were collected from authentic online databases, including Google Scholar, PubMed, Scopus, and Web of Science, by using different combinations of keywords, including 'anti-MRSA', 'antibiotic', 'antimicrobial', 'clinical trial', 'clinical phase', clinical studies', and 'pipeline'. The information extracted from articles was compared to information provided on the drug manufacturer's website and Clinical Trials.gov (https://clinicaltrials.gov/) to confirm the latest development phase of anti-MRSA agents. The present review focuses on the current development status of new anti-MRSA strategies concerning chemistry, pharmacological target(s), indications, route of administration, efficacy and safety, pharmacokinetics, and pharmacodynamics, and aims to discuss the challenges and opportunities in developing drugs for anti-MRSA infections.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Wenlan Tao
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Hengyao You
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaoli He
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| |
Collapse
|
2
|
Prabha S, Chauhan P, Warkare S, Pandey KM. A computational investigation of potential plant-based bioactive compounds against drug-resistant Staphylococcus aureus of multiple target proteins. J Biomol Struct Dyn 2023:1-19. [PMID: 38133950 DOI: 10.1080/07391102.2023.2297009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023]
Abstract
Drug-resistant Staphylococcus aureus (DRSA) poses a significant global health threat, like bacteremia, endocarditis, skin, soft tissue, bone, and joint infections. Nowadays, the resistance against conventional drugs has been a prompt and focused medical concern. The present study aimed to explore the inhibitory potential of plant-based bioactive compounds (PBBCs) against effective target proteins using a computational approach. We retrieved and verified 22 target proteins associated with DRSA and conducted a screening process that involved testing 87 PBBCs. Molecular docking was performed between screened PBBCs and reference drugs with selected target proteins via AutoDock. Subsequently, we filtered the target proteins and top PBBCs based on their binding affinity scores. Furthermore, molecular dynamic simulation was carried out through GROMACS for a duration of 100 ns, and the binding free energy was calculated using the gmx_MMPBSA. The result showed consistent hydrogen bonding interactions among the amino acid residues Ser 149, Arg 151, Thr 165, Thr 216, Glu 239, Ser 240, Ile 14, as well as Asn 18, Gln 19, Lys 45, Thr 46, Tyr 109, with their respective target proteins of the penicillin-binding protein and dihydrofolate reductase complex. Additionally, we assessed the pharmacokinetic properties of screened PBBCs via SwissADME and AdmetSAR. The findings suggest that β-amyrin, oleanolic acid, kaempferol, quercetin, and friedelin have the potential to inhibit the selected target proteins. In future research, both in vitro and in vivo, experiments will be needed to establish these PBBCs as potent antimicrobial drugs for DRSA.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sarit Prabha
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | | | - Sudeesh Warkare
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | - Khushhali M Pandey
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| |
Collapse
|
3
|
He J, Yang X, Yang K, Xu H, Chen C, Wang J, Zeng J. TPST2-mediated receptor tyrosine sulfation enhances leukocidin cytotoxicity and S. aureus infection. Front Immunol 2023; 14:1242330. [PMID: 37671153 PMCID: PMC10476081 DOI: 10.3389/fimmu.2023.1242330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Background An essential fact underlying the severity of Staphylococcus aureus (S. aureus) infection is the bicomponent leukocidins released by the pathogen to target and lyse host phagocytes through specific binding cell membrane receptors. However, little is known about the impact of post-transcriptional modification of receptors on the leukocidin binding. Method In this study, we used small interfering RNA library (Horizon/Dharmacon) to screen potential genes that affect leukocidin binding on receptors. The cell permeability was investigated through flow cytometry measuring the internalization of 4',6-diamidino-2-phenylindole. Expression of C5a anaphylatoxin chemotactic receptor 1 (C5aR1), sulfated C5aR1 in, and binding of 6x-His-tagged Hemolysin C (HlgC) and Panton-Valentine leukocidin (PVL) slow-component to THP-1 cell lines was detected and analyzed via flow cytometry. Bacterial burden and Survival analysis experiment was conducted in WT and myeloid TPST-cko C57BL/6N mice. Results After short hairpin RNA (shRNA) knockdown of TPST2 gene in THP-1, HL-60, and RAW264.7, the cytotoxicity of HlgAB, HlgCB, and Panton-Valentine leukocidin on THP-1 or HL-60 cells was decreased significantly, and the cytotoxicity of HlgAB on RAW264.7 cells was also decreased significantly. Knockdown of TPST2 did not affect the C5aR1 expression but downregulated cell surface C5aR1 tyrosine sulfation on THP-1. In addition, we found that the binding of HlgC and LukS-PV on cell surface receptor C5aR1 was impaired in C5aR1+TPST2- and C5aR1-TPST2- cells. Phagocyte knockout of TPST2 protects mice from S. aureus infection and improves the survival of mice infected with S. aureus. Conclusion These results indicate that phagocyte TPST2 mediates the bicomponent leukocidin cytotoxicity by promoting cell membrane receptor sulfation modification that facilitates its binding to leukocidin S component.
Collapse
Affiliation(s)
- Jie He
- Division of Pulmonary and Critical Care Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xianggui Yang
- Department of Laboratory Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Kai Yang
- Division of Pulmonary and Critical Care Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | | | | | | | - Jun Zeng
- Division of Pulmonary and Critical Care Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
4
|
Omobolanle Adesanya E, Daniel Ogunlakin A. Potential Use of African Botanicals and Other Compounds in the Treatment of Methicillin-Resistant Staphylococcus aureus Infections. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.108351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Infections caused by the group of Staphylococcus bacteria are commonly called Staph infections, and over 30 types of Staphylococcal bacteria exist with Staphylococcus aureus causing about 90% of the infections from the genus. Staphylococcus aureus (S. aureus) is a major cause of both hospital- and community-acquired infections with major concern arising from its strain of species that is resistant to many antibiotics. One of such strain is the Methicillin-resistant Staphylococcus aureus (MRSA) that has been described to be a resistance to methicillin drugs. Another is glycopeptides-resistant emerging from the increased use of glycopeptides drugs. This continuous emergence and spread of new resistant strains of S. aureus is a major challenge which makes the search for novel anti-resistant agents imperative. The development of vaccines from natural and synthetic products is some of the measures being proposed for the protection against the infections. Also, the development of monoclonal or polyclonal antibodies for passive immunization is sought for, and attentions with regard to arriving at successful trials have been directed back to medicinal plant research as an alternative. This review discusses the treatment strategies of MRSA, the antibacterial property of various medicinal plants, and the influence of their active compounds on methicillin-resistant S. aureus (MRSA), as well as to recommend the path to future research in this area.
Collapse
|
5
|
Duan L, Zhang J, Chen Z, Gou Q, Xiong Q, Yuan Y, Jing H, Zhu J, Ni L, Zheng Y, Liu Z, Zhang X, Zeng H, Zou Q, Zhao Z. Antibiotic Combined with Epitope-Specific Monoclonal Antibody Cocktail Protects Mice Against Bacteremia and Acute Pneumonia from Methicillin-Resistant Staphylococcus aureus Infection. J Inflamm Res 2021; 14:4267-4282. [PMID: 34511967 PMCID: PMC8415768 DOI: 10.2147/jir.s325286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose We previously reported that monoclonal antibody (mAb) cocktail improves survival in Staphylococcus aureus infection. In this study, we used acute pneumonia model and lethal sepsis model to investigate the efficacy of antibiotic combined with epitope-specific mAb cocktail in treating MRSA252 infection. Methods MRSA252 was challenged by tail vein injection or tracheal intubation to establish sepsis model or pneumonia model. One hour after infection, the mice received a single intravenous injection of normal saline, vancomycin, and vancomycin combined monoclonal antibody, linezolid alone or linezolid combined monoclonal antibody. Daily record survival rate (total 7 days), bacterial load, histology, cytokine analysis of serum and alveolar lavage fluid, and in vitro determination of the neutralizing ability of antibodies to SEB toxin and Hla toxin explained the mechanism of antibody action. Results The mAb cocktail combined with low doses of vancomycin or linezolid improved survival rates in acute pneumonia model (70%, 80%) and lethal sepsis model (80%, 80%). Epitope-specific monoclonal antibodies reduced bacterial colonization in the kidneys and lungs of mice and inhibited the biological functions of the toxins Hla and SEB in vitro. Compared to the antibiotic alone or PBS groups, the combination group had higher levels of IL-1α, IL-1β and IFN-γ and lower levels of IL-6, IL-10, TNF-α. Further, the combination of antibiotic and mAb cocktail improved infection survival against the clinical MRSA isolates in a lethal sepsis model. Conclusion This study demonstrates a novel method to treat people with low immunity against drug-resistant S. aureus infections.
Collapse
Affiliation(s)
- LianLi Duan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Qiang Gou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Qingshan Xiong
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Yue Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Jiang Zhu
- Department of Pathology, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Li Ni
- Obstetrics and Gynecology, The First People's Hospital of Jiulongpo District, Chongqing, 400050, People's Republic of China
| | - Yuling Zheng
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Zhiyong Liu
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Xiaokai Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| |
Collapse
|
6
|
Bekeredjian-Ding I. Challenges for Clinical Development of Vaccines for Prevention of Hospital-Acquired Bacterial Infections. Front Immunol 2020; 11:1755. [PMID: 32849627 PMCID: PMC7419648 DOI: 10.3389/fimmu.2020.01755] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
Increasing antibiotic resistance in bacteria causing endogenous infections has entailed a need for innovative approaches to therapy and prophylaxis of these infections and raised a new interest in vaccines for prevention of colonization and infection by typically antibiotic resistant pathogens. Nevertheless, there has been a long history of failures in late stage clinical development of this type of vaccines, which remains not fully understood. This article provides an overview on present and past vaccine developments targeting nosocomial bacterial pathogens; it further highlights the specific challenges associated with demonstrating clinical efficacy of these vaccines and the facts to be considered in future study designs. Notably, these vaccines are mainly applied to subjects with preexistent immunity to the target pathogen, transient or chronic immunosuppression and ill-defined microbiome status. Unpredictable attack rates and changing epidemiology as well as highly variable genetic and immunological strain characteristics complicate the development. In views of the clinical need, re-thinking of the study designs and expectations seems warranted: first of all, vaccine development needs to be footed on a clear rationale for choosing the immunological mechanism of action and the optimal time point for vaccination, e.g., (1) prevention (or reduction) of colonization vs. prevention of infection and (2) boosting of a preexistent immune response vs. altering the quality of the immune response. Furthermore, there are different, probably redundant, immunological and microbiological defense mechanisms that provide protection from infection. Their interplay is not well-understood but as a consequence their effect might superimpose vaccine-mediated resolution of infection and lead to failure to demonstrate efficacy. This implies that improved characterization of patient subpopulations within the trial population should be obtained by pro- and retrospective analyses of trial data on subject level. Statistical and systems biology approaches could help to define immune and microbiological biomarkers that discern populations that benefit from vaccination from those where vaccines might not be effective.
Collapse
Affiliation(s)
- Isabelle Bekeredjian-Ding
- Division of Microbiology, Langen, Germany.,Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
7
|
Liu Y, Song Z, Ge S, Zhang J, Xu L, Yang F, Lu D, Luo P, Gu J, Zou Q, Zeng H. Determining the immunological characteristics of a novel human monoclonal antibody developed against staphylococcal enterotoxin B. Hum Vaccin Immunother 2020; 16:1708-1718. [PMID: 32275466 DOI: 10.1080/21645515.2020.1744362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Staphylococci are the main cause of nosocomial infections globally. The exotoxin staphylococcal enterotoxin B (SEB) produced by methicillin-resistant Staphylococcus aureus is a major cause of pathology after a staphylococcal infection. We previously isolated an anti-SEB human monoclonal antibody designated as M0313. Here we further characterize this antibody in vitro and in vivo. Immunoblotting analysis and ELISA results indicated that M0313 accurately recognized and bound to SEB. Its binding affinity to native SEB was measured at the low nM level. M0313 effectively inhibited SEB from inducing mouse splenic lymphocyte and human peripheral blood mononuclear cell proliferation and cytokine release in cell culture. M0313 also neutralized SEB toxicity in BALB/c female mice. Most importantly, M0313 promoted the survival of mice treated with SEB-expressing bacteria. In-vivo imaging revealed that M0313 treatment significantly reduced the replication of SEB-expressing bacteria in mice. The neutralization capacity of M0313 correlated with its ability to block SEB from binding to major histocompatibility complex II and T-cell receptor by binding to the SEB residues 85-102 and 90-92. Thus, the monoclonal antibody M0313 may be developed into a therapeutic agent.
Collapse
Affiliation(s)
- Yuanyuan Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Zhen Song
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China.,Clinical Laboratory Department, Army 954th Hospital, General Hospital of Tibet Military Region , Tibet, PR China
| | - Shuang Ge
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Limin Xu
- Research and Development Department, Chengdu Olymvax Biotechnology Co., Ltd ., Chengdu, Sichuan, PR China
| | - Feng Yang
- Research and Development Department, Chengdu Olymvax Biotechnology Co., Ltd ., Chengdu, Sichuan, PR China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Jiang Gu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| |
Collapse
|
8
|
Chan R, Buckley PT, O'Malley A, Sause WE, Alonzo F, Lubkin A, Boguslawski KM, Payne A, Fernandez J, Strohl WR, Whitaker B, Lynch AS, Torres VJ. Identification of biologic agents to neutralize the bicomponent leukocidins of Staphylococcus aureus. Sci Transl Med 2020; 11:11/475/eaat0882. [PMID: 30651319 DOI: 10.1126/scitranslmed.aat0882] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 01/22/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022]
Abstract
A key aspect underlying the severity of infections caused by Staphylococcus aureus is the abundance of virulence factors that the pathogen uses to thwart critical components of the human immune response. One such mechanism involves the destruction of host immune cells by cytolytic toxins secreted by S. aureus, including five bicomponent leukocidins: PVL, HlgAB, HlgCB, LukED, and LukAB. Purified leukocidins can lyse immune cells ex vivo, and systemic injections of purified LukED or HlgAB can acutely kill mice. Here, we describe the generation and characterization of centyrins that bind S. aureus leukocidins with high affinity and protect primary human immune cells from toxin-mediated cytolysis. Centyrins are small protein scaffolds derived from the fibronectin type III-binding domain of the human protein tenascin-C. Although centyrins are potent in tissue culture assays, their short serum half-lives limit their efficacies in vivo. By extending the serum half-lives of centyrins through their fusion to an albumin-binding consensus domain, we demonstrate the in vivo efficacy of these biologics in a murine intoxication model and in models of both prophylactic and therapeutic treatment of live S. aureus systemic infections. These biologics that target S. aureus virulence factors have potential for treating and preventing serious staphylococcal infections.
Collapse
Affiliation(s)
- Rita Chan
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Peter T Buckley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - William E Sause
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Francis Alonzo
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Ashira Lubkin
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Angela Payne
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Jeffrey Fernandez
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - William R Strohl
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Brian Whitaker
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Anthony Simon Lynch
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA.
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA.
| |
Collapse
|
9
|
Staphylococcus aureus Toxins: From Their Pathogenic Roles to Anti-virulence Therapy Using Natural Products. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0059-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
10
|
Manara S, Pasolli E, Dolce D, Ravenni N, Campana S, Armanini F, Asnicar F, Mengoni A, Galli L, Montagnani C, Venturini E, Rota-Stabelli O, Grandi G, Taccetti G, Segata N. Whole-genome epidemiology, characterisation, and phylogenetic reconstruction of Staphylococcus aureus strains in a paediatric hospital. Genome Med 2018; 10:82. [PMID: 30424799 PMCID: PMC6234625 DOI: 10.1186/s13073-018-0593-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/29/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is an opportunistic pathogen and a leading cause of nosocomial infections. It can acquire resistance to all the antibiotics that entered the clinics to date, and the World Health Organization defined it as a high-priority pathogen for research and development of new antibiotics. A deeper understanding of the genetic variability of S. aureus in clinical settings would lead to a better comprehension of its pathogenic potential and improved strategies to contrast its virulence and resistance. However, the number of comprehensive studies addressing clinical cohorts of S. aureus infections by simultaneously looking at the epidemiology, phylogenetic reconstruction, genomic characterisation, and transmission pathways of infective clones is currently low, thus limiting global surveillance and epidemiological monitoring. METHODS We applied whole-genome shotgun sequencing (WGS) to 184 S. aureus isolates from 135 patients treated in different operative units of an Italian paediatric hospital over a timespan of 3 years, including both methicillin-resistant S. aureus (MRSA) and methicillin-sensitive S. aureus (MSSA) from different infection types. We typed known and unknown clones from their genomes by multilocus sequence typing (MLST), Staphylococcal Cassette Chromosome mec (SCCmec), Staphylococcal protein A gene (spa), and Panton-Valentine Leukocidin (PVL), and we inferred their whole-genome phylogeny. We explored the prevalence of virulence and antibiotic resistance genes in our cohort, and the conservation of genes encoding vaccine candidates. We also performed a timed phylogenetic investigation for a potential outbreak of a newly emerging nosocomial clone. RESULTS The phylogeny of the 135 single-patient S. aureus isolates showed a high level of diversity, including 80 different lineages, and co-presence of local, global, livestock-associated, and hypervirulent clones. Five of these clones do not have representative genomes in public databases. Variability in the epidemiology is mirrored by variability in the SCCmec cassettes, with some novel variants of the type IV cassette carrying extra antibiotic resistances. Virulence and resistance genes were unevenly distributed across different clones and infection types, with highly resistant and lowly virulent clones showing strong association with chronic diseases, and highly virulent strains only reported in acute infections. Antigens included in vaccine formulations undergoing clinical trials were conserved at different levels in our cohort, with only a few highly prevalent genes fully conserved, potentially explaining the difficulty of developing a vaccine against S. aureus. We also found a recently diverged ST1-SCCmecIV-t127 PVL- clone suspected to be hospital-specific, but time-resolved integrative phylogenetic analysis refuted this hypothesis and suggested that this quickly emerging lineage was acquired independently by patients. CONCLUSIONS Whole genome sequencing allowed us to study the epidemiology and genomic repertoire of S. aureus in a clinical setting and provided evidence of its often underestimated complexity. Some virulence factors and clones are specific of disease types, but the variability and dispensability of many antigens considered for vaccine development together with the quickly changing epidemiology of S. aureus makes it very challenging to develop full-coverage therapies and vaccines. Expanding WGS-based surveillance of S. aureus to many more hospitals would allow the identification of specific strains representing the main burden of infection and therefore reassessing the efforts for the discovery of new treatments and clinical practices.
Collapse
Affiliation(s)
- Serena Manara
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Edoardo Pasolli
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Daniela Dolce
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Novella Ravenni
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Silvia Campana
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | | | | | - Alessio Mengoni
- Department of Biology, University of Florence, Florence, Italy
| | - Luisa Galli
- Department of Health Sciences, University of Florence, Florence, Italy
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Carlotta Montagnani
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Elisabetta Venturini
- Infectious Diseases Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Omar Rota-Stabelli
- Department of Sustainable Agro-Ecosystems and Bioresources, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Guido Grandi
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Giovanni Taccetti
- Cystic Fibrosis Center, Interdisciplinary Specialist Department, Anna Meyer Children's University Hospital, Florence, Italy
| | - Nicola Segata
- Centre for Integrative Biology, University of Trento, Trento, Italy.
| |
Collapse
|
11
|
Liu Q, Mazhar M, Miller LS. Immune and Inflammatory Reponses to Staphylococcus aureus Skin Infections. CURRENT DERMATOLOGY REPORTS 2018; 7:338-349. [PMID: 30989002 DOI: 10.1007/s13671-018-0235-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Purpose of Review There have been recent advances in our understanding of cutaneous immune responses to the important human skin pathogen, Staphylococcus aureus (S. aureus). This review will highlight these insights into innate and adaptive immune mechanisms in host defense and cutaneous inflammation in response to S. aureus skin infections. Recent Findings Antimicrobial peptides, pattern recognition receptors and inflammasome activation function in innate immunity as well as T cells and their effector cytokines play a key role in adaptive immunity against S. aureus skin infections. In addition, certain mechanisms by which S. aureus contributes to aberrant cutaneous inflammation, such as in flares of the inflammatory skin disease atopic dermatitis have also been identified. Summary These cutaneous immune mechanisms could provide new targets for future vaccines and immune-based therapies to combat skin infections and cutaneous inflammation caused by S. aureus.
Collapse
Affiliation(s)
- Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Momina Mazhar
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
12
|
Gudiol C, Cuervo G, Shaw E, Pujol M, Carratalà J. Pharmacotherapeutic options for treating Staphylococcus aureus bacteremia. Expert Opin Pharmacother 2017; 18:1947-1963. [DOI: 10.1080/14656566.2017.1403585] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Carlota Gudiol
- Infectious Diseases Department, Bellvitge University Hospital, IDIBELL. L’Hospitalet de Llobregat, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- REIPI (Spanish Network for Research in Infectious Disease), Instituto de Salud Carlos III, Madrid, Spain
| | - Guillermo Cuervo
- Infectious Diseases Department, Bellvitge University Hospital, IDIBELL. L’Hospitalet de Llobregat, Barcelona, Spain
- REIPI (Spanish Network for Research in Infectious Disease), Instituto de Salud Carlos III, Madrid, Spain
| | - Evelyn Shaw
- Infectious Diseases Department, Bellvitge University Hospital, IDIBELL. L’Hospitalet de Llobregat, Barcelona, Spain
- REIPI (Spanish Network for Research in Infectious Disease), Instituto de Salud Carlos III, Madrid, Spain
| | - Miquel Pujol
- Infectious Diseases Department, Bellvitge University Hospital, IDIBELL. L’Hospitalet de Llobregat, Barcelona, Spain
- REIPI (Spanish Network for Research in Infectious Disease), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Carratalà
- Infectious Diseases Department, Bellvitge University Hospital, IDIBELL. L’Hospitalet de Llobregat, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- REIPI (Spanish Network for Research in Infectious Disease), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|