1
|
Napolitano M, Esposito M, Fargnoli MC, Girolomoni G, Romita P, Nicoli E, Matruglio P, Foti C. Infections in Patients with Atopic Dermatitis and the Influence of Treatment. Am J Clin Dermatol 2025; 26:183-197. [PMID: 39915363 PMCID: PMC11850493 DOI: 10.1007/s40257-025-00917-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 02/25/2025]
Abstract
Atopic dermatitis (AD) is a T helper 2-mediated chronic inflammatory skin disease that affects children and adults. Patients with AD are prone to recurrent infections of the skin and other organs, which can severely worsen the disease course. This review summarises the current evidence on the aetiology, pathogenesis, treatment and prevention of infections in patients with AD. PubMed was searched for English-language research articles, systematic reviews, meta-analyses and guidelines published until February 2023 using the key term "atopic dermatitis" and terms relevant to infections. Patients with AD have an increased risk of bacterial, viral and fungal infections of the skin, mainly due to impaired barrier function, altered immune response and frequent scratching. The most common pathogens are Staphylococcus aureus and herpes simplex virus, which can cause impetigo, folliculitis, abscesses, eczema herpeticum and other complications. They also appear to increase susceptibility to systemic infections, including respiratory and urinary tract infections and sepsis. Certain systemic treatments for AD, such as mycophenolate mofetil and Janus kinase inhibitors, increase the risk of viral infections. Prevention and treatment of recurrent infections in patients with AD require a multifaceted approach that includes topical and systemic antimicrobials, skin care and effective control of AD symptoms (to break the itch-scratch cycle). Preventing and limiting the development of infections are important considerations in choosing an AD treatment.
Collapse
Affiliation(s)
- Maddalena Napolitano
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Esposito
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
- UOSD General and Oncological Dermatology, Ospedale San Salvatore, L'Aquila, Italy
| | | | - Giampiero Girolomoni
- Department of Medicine, Section of Dermatology, University of Verona, Verona, Italy
| | - Paolo Romita
- Department of Precision Medicine and Regeneration and Ionian Area, Unit of Dermatology, University of Bari Aldo Moro, Bari, Italy
| | | | | | - Caterina Foti
- Department of Precision Medicine and Regeneration and Ionian Area, Unit of Dermatology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
2
|
Tartor YH, Enany ME, Ismail NI, El-Demerdash AS, Eidaroos NH, Algendy RM, Mahmmod Y, Elsohaby I. Vancomycin-resistant Staphylococcus aureus endangers Egyptian dairy herds. Sci Rep 2024; 14:30606. [PMID: 39715776 DOI: 10.1038/s41598-024-81516-6] [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: 08/20/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
The emergence of pandrug-resistant (PDR) and extensive drug-resistant (XDR) methicillin-resistant and vancomycin-resistant Staphylococcus aureus (MRSA and VRSA) isolates from bovine milk samples along with biofilm formation ability and harboring various virulence genes complicates the treatment of bovine mastitis and highlights the serious threat to public health. This study investigated for the first time the frequency, antimicrobial resistance profiles, biofilm-forming ability, virulence factors, spa and staphylococcal cassette chromosome mec (SCCmec) types of MRSA and VRSA isolated from clinical and subclinical bovine mastitis in Egypt. A total of 808 milk samples were collected from each quarter of 202 dairy animals, including 31 buffaloes and 171 cattle. The frequency of mastitis in the collected milk samples was 48.4% (60/124) in buffaloes and 29.2% (200/684) in cattle. A total of 65 Staphylococcus species isolates were recovered, including 27 coagulase-positive S. aureus (CoPS) isolates and 38 coagulase-negative staphylococci (CoNS). The CoNS included 27 mammaliicocci (20 Mammaliicoccus lentus and 7 M. sciuri) and 11 Non-aureus staphylococci (S. lugdunensis) isolates. All the CoPS isolates were mecA positive and resistant to 20-33 tested antimicrobials with multiple antibiotic resistance index ranging from 0.61 to 1. Three isolates were PDR, four were XDR, and 20 were multidrug resistant isolates. VRSA was detected in 85.2% of CoPS isolates with minimal inhibitory concentration (MIC) ranging from 64 to 1024 µg/mL. The vanA gene was found in 60.8%, vanB in 73.9%, and both genes in 43.5% of VRSA isolates. All the CoPS isolates exhibited biofilm formation ability, with 55.6% being strong, and 44.4% moderate biofilm producers, and harbored icaA (74.1%) and icaD (74.1%) biofilm-forming genes. All S. aureus isolates harbored both beta-haemolysin (hlb) and leucotoxin (lukMF) genes, while 44.4% were positive for toxic shock syndrome toxin (tsst) gene. Enterotoxin genes sea, seb, sec, sed, and see were found in 59.3%, 40.7%, 18.5%, 33.3%, and 14.8% of isolates, respectively. Additionally, 70.4% of the isolates had spa X-region gene, and exhibited eight different MRSA spa types (t127, t267, t037, t011, t843, t1081, t2663, and t1575), with spa t127 being the most common. Three SCCmec types (I, II and III) were identified, with SCCmec I being predominant, and were further classified into subtypes 1.1.1, 1.1.2, 1.n.1, and 4.1.1. The ability of MRSA and VRSA isolates to produce biofilms and resist antimicrobials highlights the serious threat these pathogens pose to bovine milk safety, animal welfare, and public health. Therefore, strict hygiene practices and antimicrobial surveillance are crucial to reduce the risk of MRSA and VRSA colonization and dissemination.
Collapse
Affiliation(s)
- Yasmine H Tartor
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Mohamed E Enany
- Department of Bacteriology, Immunology, and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | | | - Azza S El-Demerdash
- Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig, 44516, Egypt
| | - Nada H Eidaroos
- Department of Bacteriology, Immunology, and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Reem M Algendy
- Department of Food Hygiene, Safety and Technology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Yasser Mahmmod
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, NY, 11548, USA
| | - Ibrahim Elsohaby
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, SAR, China
- Centre for Applied One Health Research and Policy Advice (OHRP), City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
3
|
Liu C, Tian T, Shi Y, Li M, Hong L, Zhou J, Liu J, Zhong Y, Wang X, Wang Z, Bai X, Wang L, Li C, Wu Z. Enhancing antibacterial photodynamic therapy with NIR‐activated gold nanoclusters: Atomic‐precision size effect on reducing bacterial biofilm formation and virulence. AGGREGATE 2024. [DOI: 10.1002/agt2.666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
AbstractPersistent biofilm infections pose a critical health threat with their relentless presence and amplified antibiotic resistance. Traditional antibacterial photodynamic therapy can inhibit bacteria extracellularly but struggles to control biofilm formation and virulence. Thus, there is an urgent need to develop photosensitizers, such as ultra‐small gold nanoclusters (AuNCs), that can penetrate biofilms and internalize into bacteria. However, AuNCs still face the challenge of insufficient reactive oxygen species (ROS) production and limited near‐infrared light absorption. This study develops a model of indocyanine green (ICG)‐sensitized AuNCs with atomic‐precision size effect. This approach achieved near‐infrared light absorption while inhibiting radiation transitions, thereby regulating the generation of ROS. Notably, different‐sized AuNCs (Au10NCs, Au15NCs, Au25NCs) yielded varied ROS types, resulting from different energy level distributions and electron transfer rates. ICG‐Au15NCs achieved a treatment efficacy of 99.94% against Staphylococcus aureus infections in vitro and significantly accelerated wound healing in vivo. Moreover, this study highlights the unique role of ICG‐AuNCs in suppressing quorum sensing, virulence, and ABC transporters compared to their larger counterparts. This strategy demonstrates that atomic‐precision size effect of AuNCs paves the way for innovative approaches in antibacterial photodynamic therapy for infection control.
Collapse
Affiliation(s)
- Chengyu Liu
- Department of Prosthodontics Jilin Provincial Key Laboratory of Tooth Development and Remodeling School and Hospital of Stomatology Jilin University Changchun China
| | - Tenghui Tian
- Hospital of Affiliated Changchun University of Chinese Medicine Branch of National Clinical Research Center for Chinese Medicine Cardiology Changchun China
| | - Yujia Shi
- Department of Oral Implantology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering Jilin University Changchun China
| | - Meiqi Li
- Department of Oral Implantology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering Jilin University Changchun China
| | - Le Hong
- Hospital of Affiliated Changchun University of Chinese Medicine Branch of National Clinical Research Center for Chinese Medicine Cardiology Changchun China
| | - Jing Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering Jilin University Changchun China
| | - Jia Liu
- Department of Oral Implantology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering Jilin University Changchun China
| | - Yuan Zhong
- State Key Laboratory of Integrated Optoelectronics and College of Electronic Science and Engineering Jilin university Changchun China
| | - Xue Wang
- State Key Laboratory of Integrated Optoelectronics and College of Electronic Science and Engineering Jilin university Changchun China
| | - Zhenyu Wang
- State Key Laboratory of Integrated Optoelectronics and College of Electronic Science and Engineering Jilin university Changchun China
| | - Xue Bai
- State Key Laboratory of Integrated Optoelectronics and College of Electronic Science and Engineering Jilin university Changchun China
| | - Lin Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering Jilin University Changchun China
| | - Chunyan Li
- Department of Prosthodontics Jilin Provincial Key Laboratory of Tooth Development and Remodeling School and Hospital of Stomatology Jilin University Changchun China
| | - Zhennan Wu
- State Key Laboratory of Integrated Optoelectronics and College of Electronic Science and Engineering Jilin university Changchun China
| |
Collapse
|
4
|
Cieza MYR, Bonsaglia ECR, Rall VLM, dos Santos MV, Silva NCC. Staphylococcal Enterotoxins: Description and Importance in Food. Pathogens 2024; 13:676. [PMID: 39204276 PMCID: PMC11357529 DOI: 10.3390/pathogens13080676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Staphylococcus aureus stands out as one of the most virulent pathogens in the genus Staphylococcus. This characteristic is due to its ability to produce a wide variety of staphylococcal enterotoxins (SEs) and exotoxins, which in turn can cause staphylococcal food poisoning (SFP), clinical syndromes such as skin infections, inflammation, pneumonia, and sepsis, in addition to being associated with the development of inflammation in the mammary glands of dairy cattle, which results in chronic mastitis and cell necrosis. SEs are small globular proteins that combine superantigenic and emetic activities; they are resistant to heat, low temperatures, and proteolytic enzymes and are tolerant to a wide pH range. More than 24 SE genes have been well described (SEA-SEE, SEG, SEH, SEI, SEJ, SElK, SElL, SElM, SElN, SElO, SElP, SElQ, SElR, SElS, SElT, SElU, SElV, SElW, SElX, SElY, and SElZ), being a part of different SFP outbreaks, clinical cases, and isolated animal strains. In recent years, new genes (sel26, sel27, sel28, sel31, sel32, and sel33) from SEs have been described, as well as two variants (seh-2p and ses-3p) resulting in a total of thirty-three genes from Ses, including the nine variants that are still in the process of genetic and molecular structure evaluation. SEs are encoded by genes that are located in mobile genetic elements, such as plasmids, prophages, pathogenicity islands, and the enterotoxin gene cluster (egc), and housed in the genomic island of S. aureus. Both classical SEs and SE-like toxins (SEls) share phylogenetic relationships, structure, function, and sequence homology, which are characteristics for the production of new SEs through recombination processes. Due to the epidemiological importance of SEs, their rapid assessment and detection have been crucial for food security and public health; for this reason, different methods of identification of SEs have been developed, such as liquid chromatography coupled with high-resolution mass spectrometry (LC-HRMS), molecular methods, and whole-genome sequencing; providing the diagnosis of SEs and a better understanding of the occurrence, spread, and eradication of SEs. This review provides scientific information on the enterotoxins produced by S. aureus, such as structural characteristics, genetic organization, regulatory mechanisms, superantigen activity, mechanisms of action used by SEs at the time of interaction with the immune system, methods of detection of SEs, and recent biocontrol techniques used in food.
Collapse
Affiliation(s)
- Mirian Yuliza Rubio Cieza
- Department of Food Science and Nutrition, School of Food Engineering, University of Campinas (UNICAMP), Campinas 13083-862, Brazil;
| | - Erika Carolina Romão Bonsaglia
- Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Sciences, University of São Paulo (USP), Pirassununga 13635-900, Brazil; (E.C.R.B.); (M.V.d.S.)
| | - Vera Lucia Mores Rall
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu 18618-691, Brazil;
| | - Marcos Veiga dos Santos
- Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Sciences, University of São Paulo (USP), Pirassununga 13635-900, Brazil; (E.C.R.B.); (M.V.d.S.)
| | - Nathália Cristina Cirone Silva
- Department of Food Science and Nutrition, School of Food Engineering, University of Campinas (UNICAMP), Campinas 13083-862, Brazil;
| |
Collapse
|
5
|
Patel H, Rawat S. A genetic regulatory see-saw of biofilm and virulence in MRSA pathogenesis. Front Microbiol 2023; 14:1204428. [PMID: 37434702 PMCID: PMC10332168 DOI: 10.3389/fmicb.2023.1204428] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Staphylococcus aureus is one of the most common opportunistic human pathogens causing several infectious diseases. Ever since the emergence of the first methicillin-resistant Staphylococcus aureus (MRSA) strain decades back, the organism has been a major cause of hospital-acquired infections (HA-MRSA). The spread of this pathogen across the community led to the emergence of a more virulent subtype of the strain, i.e., Community acquired Methicillin resistant Staphylococcus aureus (CA-MRSA). Hence, WHO has declared Staphylococcus aureus as a high-priority pathogen. MRSA pathogenesis is remarkable because of the ability of this "superbug" to form robust biofilm both in vivo and in vitro by the formation of polysaccharide intercellular adhesin (PIA), extracellular DNA (eDNA), wall teichoic acids (WTAs), and capsule (CP), which are major components that impart stability to a biofilm. On the other hand, secretion of a diverse array of virulence factors such as hemolysins, leukotoxins, enterotoxins, and Protein A regulated by agr and sae two-component systems (TCS) aids in combating host immune response. The up- and downregulation of adhesion genes involved in biofilm formation and genes responsible for synthesizing virulence factors during different stages of infection act as a genetic regulatory see-saw in the pathogenesis of MRSA. This review provides insight into the evolution and pathogenesis of MRSA infections with a focus on genetic regulation of biofilm formation and virulence factors secretion.
Collapse
Affiliation(s)
| | - Seema Rawat
- Microbiology Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| |
Collapse
|
6
|
Huang J, Xu Y. Autoimmunity: A New Focus on Nasal Polyps. Int J Mol Sci 2023; 24:ijms24098444. [PMID: 37176151 PMCID: PMC10179643 DOI: 10.3390/ijms24098444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) has long been considered a benign, chronic inflammatory, and hyperplastic disease. Recent studies have shown that autoimmune-related mechanisms are involved in the pathology of nasal polyps. Activated plasma cells, eosinophils, basophils, innate type 2 lymphocytes, mast cells, and proinflammatory cytokine in polyp tissue indicate the mobilization of innate and adaptive immune pathways during polyp formation. The discovery of a series of autoantibodies further supports the autoimmune nature of nasal polyps. Local homeostasis dysregulation, infection, and chronic inflammation may trigger autoimmunity through several mechanisms, including autoantigens overproduction, microbial translocation, molecular mimicry, superantigens, activation or inhibition of receptors, bystander activation, dysregulation of Toll-Like Receptors (TLRs), epitope spreading, autoantigens complementarity. In this paper, we elaborated on the microbiome-mediated mechanism, abnormal host immunity, and genetic changes to update the role of autoimmunity in the pathogenesis of chronic rhinosinusitis with nasal polyps.
Collapse
Affiliation(s)
- Jingyu Huang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
7
|
Johnson WL, Sohn M, Woeller CF, Wozniak RAF. Staphylococcal Enterotoxins Promote Virulence in Bacterial Keratitis. Invest Ophthalmol Vis Sci 2023; 64:5. [PMID: 37133835 PMCID: PMC10166116 DOI: 10.1167/iovs.64.5.5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
Purpose Staphylococcus aureus is an important cause of corneal infections (keratitis). To better understand the virulence mechanisms mediating keratitis, a recent comparative genomics study revealed that a set of secreted enterotoxins were found with higher prevalence among ocular versus non-ocular S. aureus clinical infection isolates, suggesting a key role for these toxins in keratitis. Although well known to cause toxic shock syndrome and S. aureus food poisoning, enterotoxins have not yet been shown to mediate virulence in keratitis. Methods A set of clinical isolate test strains, including a keratitis isolate that encodes five enterotoxins (sed, sej, sek, seq, ser), its corresponding enterotoxin deletion mutant and complementation strain, a keratitis isolate devoid of enterotoxins, and the non-ocular S. aureus strain USA300 along with its corresponding enterotoxin deletion and complementation strains, were evaluated for cellular adhesion, invasion and cytotoxicity in a primary corneal epithelial model as well as with microscopy. Additionally, strains were evaluated in an in vivo model of keratitis to quantify enterotoxin gene expression and measure disease severity. Results We demonstrate that, although enterotoxins do not impact bacterial adhesion or invasion, they do elicit direct cytotoxicity in vitro toward corneal epithelial cells. In an in vivo model, sed, sej, sek, seq, ser were found to have variable gene expression across 72 hours of infection and test strains encoding enterotoxins resulted in increased bacterial burden as well as a reduced host cytokine response. Conclusions Our results support a novel role for staphylococcal enterotoxins in promoting virulence in S. aureus keratitis.
Collapse
Affiliation(s)
- William L Johnson
- Department of Ophthalmology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - Michael Sohn
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - Collynn F Woeller
- Department of Ophthalmology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - Rachel A F Wozniak
- Department of Ophthalmology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| |
Collapse
|
8
|
Jiang L, Ma Y, Chen Y, Cai M, Wu Z, Xiong Y, Duan X, Liao X, Wang J. Multi-target antibacterial mechanism of ruthenium polypyridine complexes with anthraquinone groups against Staphylococcus aureus. RSC Med Chem 2023; 14:700-709. [PMID: 37122548 PMCID: PMC10131643 DOI: 10.1039/d2md00430e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
Three new Ru(ii) complexes, [Ru(dtb)2PPAD](PF6)2 (Ru-1), [Ru(dmob)2PPAD](PF6)2 (Ru-2) and [Ru(bpy)2PPAD](PF6)2 (Ru-3) (dtb = 4,4'-di-tert-butyl-2,2'-bipyridine, dmob = 4,4'-dimethyl-2,2'-bipyridine, bpy = 2,2'-bipyridine and PPAD = 2-(pyridine-3-yl)-1H-imidazo[4,5f][1.10]phenanthracene-9,10-dione), were synthesized and characterized by 1H NMR and 13C NMR spectroscopy, HRMS and HPLC. Among them, Ru-1 showed excellent antimicrobial activity against Gram-positive bacteria Staphylococcus aureus (minimum inhibitory concentration (MIC) = 1 μg mL-1) and low hemolytic and cytotoxic activity. In addition, Ru-1 showed obviously rapid bactericidal activity, low resistance rate, bacterial biofilm destroying activity and high biosafety in vivo. Moreover, skin infection models and a mouse model of sepsis indicated that the anti-infective efficacy of Ru-1 was comparable to that of vancomycin. Mechanism exploration results showed that the antibacterial behavior is probably related with targeting of the bacterial cell membrane and inhibiting topoisomerase I.
Collapse
Affiliation(s)
- Li Jiang
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Yuanyuan Ma
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Yiman Chen
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Mengcheng Cai
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Zhixing Wu
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Yanshi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Xuemin Duan
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Xiangwen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Jintao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| |
Collapse
|
9
|
Francis D, Bhairaddy A, Joy A, Hari GV, Francis A. Secretory proteins in the orchestration of microbial virulence: The curious case of Staphylococcus aureus. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:271-350. [PMID: 36707204 DOI: 10.1016/bs.apcsb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | | | - Ashik Francis
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| |
Collapse
|
10
|
Hatanaka D, Nakamura T, Kusakari M, Takahashi H, Hasegawa H. A case of necrotizing tracheobronchitis successfully treated with immunoglobulin. Pediatr Int 2021; 63:1538-1540. [PMID: 34477264 DOI: 10.1111/ped.14670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/07/2021] [Accepted: 02/08/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Daisuke Hatanaka
- Department of Neonatology, Japanese Red Cross Musashino Hospital, Tokyo, Japan
| | - Toshihiko Nakamura
- Department of Neonatology, Japanese Red Cross Musashino Hospital, Tokyo, Japan
| | - Michiko Kusakari
- Department of Neonatology, Japanese Red Cross Musashino Hospital, Tokyo, Japan
| | - Hidehiro Takahashi
- Department of Neonatology, Japanese Red Cross Musashino Hospital, Tokyo, Japan
| | - Hisaya Hasegawa
- Division of Neonatal Intensive Care, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| |
Collapse
|
11
|
Ahmad-Mansour N, Loubet P, Pouget C, Dunyach-Remy C, Sotto A, Lavigne JP, Molle V. Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments. Toxins (Basel) 2021; 13:677. [PMID: 34678970 PMCID: PMC8540901 DOI: 10.3390/toxins13100677] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus aureus is a clinically important pathogen that causes a wide range of human infections, from minor skin infections to severe tissue infection and sepsis. S. aureus has a high level of antibiotic resistance and is a common cause of infections in hospitals and the community. The rising prevalence of community-acquired methicillin-resistant S. aureus (CA-MRSA), combined with the important severity of S. aureus infections in general, has resulted in the frequent use of anti-staphylococcal antibiotics, leading to increasing resistance rates. Antibiotic-resistant S. aureus continues to be a major health concern, necessitating the development of novel therapeutic strategies. S. aureus uses a wide range of virulence factors, such as toxins, to develop an infection in the host. Recently, anti-virulence treatments that directly or indirectly neutralize S. aureus toxins have showed promise. In this review, we provide an update on toxin pathogenic characteristics, as well as anti-toxin therapeutical strategies.
Collapse
Affiliation(s)
- Nour Ahmad-Mansour
- Laboratory of Pathogen Host Interactions, CNRS UMR5235, Université de Montpellier, 34000 Montpellier, France;
| | - Paul Loubet
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, 30908 Nîmes, France; (P.L.); (A.S.)
| | - Cassandra Pouget
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, 30908 Nîmes, France;
| | - Catherine Dunyach-Remy
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Microbiology and Hospital Hygiene, Université de Montpellier, 30908 Nîmes, France; (C.D.-R.); (J.-P.L.)
| | - Albert Sotto
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, 30908 Nîmes, France; (P.L.); (A.S.)
| | - Jean-Philippe Lavigne
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Microbiology and Hospital Hygiene, Université de Montpellier, 30908 Nîmes, France; (C.D.-R.); (J.-P.L.)
| | - Virginie Molle
- Laboratory of Pathogen Host Interactions, CNRS UMR5235, Université de Montpellier, 34000 Montpellier, France;
| |
Collapse
|
12
|
Alenius H, Sinkko H, Moitinho-Silva L, Rodriguez E, Broderick C, Alexander H, Reiger M, Hjort Hjelmsø M, Fyhrquist N, Olah P, Bryce P, Smith C, Koning F, Eyerich K, Greco D, van den Bogaard EH, Neumann AU, Traidl-Hoffmann C, Homey B, Flohr C, Bønnelykke K, Stokholm J, Weidinger S. The power and potential of BIOMAP to elucidate host-microbiome interplay in skin inflammatory diseases. Exp Dermatol 2021; 30:1517-1531. [PMID: 34387406 DOI: 10.1111/exd.14446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/02/2021] [Accepted: 08/02/2021] [Indexed: 11/27/2022]
Abstract
The two most common chronic inflammatory skin diseases are atopic dermatitis (AD) and psoriasis. The underpinnings of the remarkable degree of clinical heterogeneity of AD and psoriasis are poorly understood and, as a consequence, disease onset and progression are unpredictable and the optimal type and time-point for intervention are as yet unknown. The BIOMAP project is the first IMI (Innovative Medicines Initiative) project dedicated to investigating the causes and mechanisms of AD and psoriasis and to identify potential biomarkers responsible for the variation in disease outcome. The consortium includes 7 large pharmaceutical companies and 25 non-industry partners including academia. Since there is mounting evidence supporting an important role for microbial exposures and our microbiota as factors mediating immune polarization and AD and psoriasis pathogenesis, an entire work package is dedicated to the investigation of skin and gut microbiome linked to AD or psoriasis. The large collaborative BIOMAP project will enable the integration of patient cohorts, data and knowledge in unprecedented proportions. The project has a unique opportunity with a potential to bridge and fill the gaps between current problems and solutions. This review highlights the power and potential of BIOMAP project in the investigation of microbe-host interplay in AD and psoriasis.
Collapse
Affiliation(s)
- Harri Alenius
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden.,Human Microbiome Research Program (HUMI), Faculty of Medicine, University of Helsinki, Finland
| | - Hanna Sinkko
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden.,Human Microbiome Research Program (HUMI), Faculty of Medicine, University of Helsinki, Finland
| | - Lucas Moitinho-Silva
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Elke Rodriguez
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Conor Broderick
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Helen Alexander
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Matthias Reiger
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany
| | - Mathis Hjort Hjelmsø
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nanna Fyhrquist
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden
| | - Peter Olah
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Department of Dermatology, Venereology and Oncodermatology, Medical Faculty, University of Pécs, Hungary
| | - Paul Bryce
- Type 2 Inflammation & Fibrosis Cluster, Immunology & Inflammation Therapeutic Area, Sanofi US, Cambridge, MA, United States of America
| | - Catherine Smith
- St John's Institute of Dermatology, Kings College London, and Guys and St Thomas' NHS Foundation Trust, 9th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Frits Koning
- Department of Immunology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Kilian Eyerich
- Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Dario Greco
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Avidan U Neumann
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Claudia Traidl-Hoffmann
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,CK CARE, Christine Kühne Center for Allergy Research and Education, Davos, Switzerland.,ZIEL - Institute for Food & Health, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Carsten Flohr
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.,Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
13
|
Cavalcante FS, Saintive S, Carvalho Ferreira D, Rocha Silva AB, Guimarães LC, Braga BS, Dios Abad ED, Ribeiro M, Netto Dos Santos KR. Methicillin-resistant Staphylococcus aureus from infected skin lesions present several virulence genes and are associated with the CC30 in Brazilian children with atopic dermatitis. Virulence 2021; 12:260-269. [PMID: 33356835 PMCID: PMC7808431 DOI: 10.1080/21505594.2020.1869484] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease and colonization by Staphylococcus aureus may affect up to 100% of these patients. Virulent and resistant isolates can worsen AD patient clinical condition and jeopardize the treatment. We aimed to detect virulence genes and to evaluate the biofilm production of S. aureus isolates from infected skin lesions of children with AD. Methicillin resistance was detected by phenotypic and molecular tests and the virulence genes were detected by PCR. Biofilm formation was assessed by bacterial growing on microtiter plates and later stained with safranin. Genotyping was performed by Pulsed-Field Gel Electrophoresis and Multilocus Sequence Typing. Among 106 AD patients, 55 (51.8%) had developed S. aureus cutaneous infections and 23 (41.6%) were methicillin-resistant (MRSA). All 55 isolates carried the fnbA, hla, icaA, sasG, and seu genes, and more than 70% presented cna, eap, ebpS, hlg, and pvl genes. Clonal complex (CC) 30 was the main lineage found (34.5%), especially among MRSA isolates (52.2%). The egc cluster and the bbp gene were significantly the most frequent in MRSA isolates and in USA1100/ST30/CC30 lineage. Most of the isolates (74.5%) were non-biofilm producers and many of them only started to produce it in the presence of fibrinogen. There was no significant association between S. aureus isolates features and the AD severity. This study demonstrated a high frequency of CC30 MRSA isolates presenting several virulence genes in infected skin lesions of AD children in Brazil, that may influence the severity of the disease and the treatments required.
Collapse
Affiliation(s)
| | - Simone Saintive
- Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro , Rio de Janeiro, Brazil
| | - Dennis Carvalho Ferreira
- Faculdade de Odontologia, Universidade Veiga de Almeida , Rio de Janeiro, Brazil.,Faculdade de Odontologia, Universidade Estácio de Sá , Rio de Janeiro, Brazil
| | - Adriana Barbosa Rocha Silva
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro , Rio de Janeiro, Brazil
| | - Lorrayne Cardoso Guimarães
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro , Rio de Janeiro, Brazil
| | | | - Eliane de Dios Abad
- Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro , Rio de Janeiro, Brazil
| | - Marcia Ribeiro
- Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro , Rio de Janeiro, Brazil
| | | |
Collapse
|
14
|
Miller LS, Fowler VG, Shukla SK, Rose WE, Proctor RA. Development of a vaccine against Staphylococcus aureus invasive infections: Evidence based on human immunity, genetics and bacterial evasion mechanisms. FEMS Microbiol Rev 2020; 44:123-153. [PMID: 31841134 PMCID: PMC7053580 DOI: 10.1093/femsre/fuz030] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Invasive Staphylococcus aureus infections are a leading cause of morbidity and mortality in both hospital and community settings, especially with the widespread emergence of virulent and multi-drug resistant methicillin-resistant S. aureus strains. There is an urgent and unmet clinical need for non-antibiotic immune-based approaches to treat these infections as the increasing antibiotic resistance is creating a serious threat to public health. However, all vaccination attempts aimed at preventing S. aureus invasive infections have failed in human trials, especially all vaccines aimed at generating high titers of opsonic antibodies against S. aureus surface antigens to facilitate antibody-mediated bacterial clearance. In this review, we summarize the data from humans regarding the immune responses that protect against invasive S. aureus infections as well as host genetic factors and bacterial evasion mechanisms, which are important to consider for the future development of effective and successful vaccines and immunotherapies against invasive S. aureus infections in humans. The evidence presented form the basis for a hypothesis that staphylococcal toxins (including superantigens and pore-forming toxins) are important virulence factors, and targeting the neutralization of these toxins are more likely to provide a therapeutic benefit in contrast to prior vaccine attempts to generate antibodies to facilitate opsonophagocytosis.
Collapse
Affiliation(s)
- Lloyd S Miller
- Immunology, Janssen Research and Development, 1400 McKean Road, Spring House, PA, 19477, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Cancer Research Building 2, Suite 209, Baltimore, MD, 21231, USA.,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, 1830 East Monument Street, Baltimore, MD, 21287, USA.,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, 601 North Caroline Street, Baltimore, MD, 21287, USA.,Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Vance G Fowler
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, 315 Trent Drive, Hanes House, Durham, NC, 27710, USA.,Duke Clinical Research Institute, Duke University Medical Center, 40 Duke Medicine Circle, Durham, NC, 27710, USA
| | - Sanjay K Shukla
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, 1000 North Oak Avenue, Marshfield, WI, 54449, USA.,Computation and Informatics in Biology and Medicine, University of Wisconsin, 425 Henry Mall, Room 3445, Madison, WI, 53706, USA
| | - Warren E Rose
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA.,Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, 4123 Rennebohm Hall, Madison, WI, 53705 USA
| | - Richard A Proctor
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA.,Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, 1550 Linden Drive, Microbial Sciences Building, Room 1334, Madison, WI, 53705, USA
| |
Collapse
|
15
|
Hnasko R, Lin AV, McGarvey JA. Rapid Detection of Staphylococcal Enterotoxin-B by Lateral Flow Assay. Monoclon Antib Immunodiagn Immunother 2020; 38:209-212. [PMID: 31603743 PMCID: PMC6791480 DOI: 10.1089/mab.2019.0028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A cohort of monoclonal antibodies (mAbs) were generated against Staphylococcal enterotoxin-B (SEB) and selected by double sandwich enzyme-linked immunosorbent assay (ELISA) for solution capture of the toxin. Clonal hybridoma cell lines were established and a pair of anti-SEB mAbs selected for the development of a sandwich ELISA. Immobilized 3D6 mAb (IgG1, kappa) when paired with 4C9 mAb (IgG1, kappa) conjugated to horseradish peroxidase generates a typical dose–response curve with an EC50 of 24.8 ng/mL for purified SEB using chemiluminescent detection. These mAbs bind SEB by Western blot and ELISA binding to classical enterotoxin serotypes show that the 3D6 mAb binds both SEB and the SEC1 serotypes, whereas 4C9 binds only SEB. These mAbs effectively port onto lateral flow test strips with a visual detection sensitivity for SEB of 5 ng/mL in <10 minutes using a 4C9 conjugated to a 40 nm gold reporter.
Collapse
Affiliation(s)
- Robert Hnasko
- USDA-ARS Produce Safety and Microbiology Research Unit (PSM), Albany, California
| | - Alice V Lin
- USDA-ARS Produce Safety and Microbiology Research Unit (PSM), Albany, California
| | - Jeffery A McGarvey
- USDA-ARS Foodborne Toxin Detection and Prevention Research Unit (FTDP), Albany, California
| |
Collapse
|
16
|
Meilleur CE, Wardell CM, Mele TS, Dikeakos JD, Bennink JR, Mu HH, McCormick JK, Haeryfar SMM. Bacterial Superantigens Expand and Activate, Rather than Delete or Incapacitate, Preexisting Antigen-Specific Memory CD8+ T Cells. J Infect Dis 2020; 219:1307-1317. [PMID: 30418594 DOI: 10.1093/infdis/jiy647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/07/2018] [Indexed: 11/13/2022] Open
Abstract
Superantigens (SAgs) released by common Gram-positive bacterial pathogens have been reported to delete, anergize, or activate mouse T cells. However, little is known about their effects on preexisting memory CD8+ T cell (TCD8) pools. Furthermore, whether SAgs manipulate human memory TCD8 responses to cognate antigens is unknown. We used a human peripheral blood mononuclear cell culture system and a nontransgenic mouse model in which the impact of stimulation by two fundamentally distinct SAgs, staphylococcal enterotoxin B and Mycoplasma arthritidis mitogen, on influenza virus- and/or cytomegalovirus-specific memory TCD8 could be monitored. Bacterial SAgs surprisingly expanded antiviral memory TCD8 generated naturally through infection or artificially through vaccination. Mechanistically, this was a T cell-intrinsic and T cell receptor β-chain variable-dependent phenomenon. Importantly, SAg-expanded TCD8 displayed an effector memory phenotype and were capable of producing interferon-γ and destroying target cells ex vivo or in vivo. These findings have clear implications for antimicrobial defense and rational vaccine design.
Collapse
Affiliation(s)
- Courtney E Meilleur
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Christine M Wardell
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Tina S Mele
- Division of General Surgery, Department of Surgery, Western University, London, Canada.,Division of Critical Care Medicine, Western University, London, Canada
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Jack R Bennink
- Viral Immunology Section, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Hong-Hua Mu
- Division of Rheumatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City
| | - John K McCormick
- Department of Microbiology and Immunology, Western University, London, Canada.,Centre for Human Immunology, Western University, London, Canada.,Lawson Health Research Institute, London, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Canada.,Division of General Surgery, Department of Surgery, Western University, London, Canada.,Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Canada.,Centre for Human Immunology, Western University, London, Canada.,Lawson Health Research Institute, London, Canada
| |
Collapse
|
17
|
Leonard A, Wang J, Yu L, Liu H, Estrada Y, Greenlees L, McPhee R, Ruzin A, Guttman-Yassky E, Howell MD. Atopic Dermatitis Endotypes Based on Allergen Sensitization, Reactivity to Staphylococcus aureus Antigens, and Underlying Systemic Inflammation. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 8:236-247.e3. [DOI: 10.1016/j.jaip.2019.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 10/26/2022]
|
18
|
Alexander H, Paller AS, Traidl-Hoffmann C, Beck LA, De Benedetto A, Dhar S, Girolomoni G, Irvine AD, Spuls P, Su J, Thyssen JP, Vestergaard C, Werfel T, Wollenberg A, Deleuran M, Flohr C. The role of bacterial skin infections in atopic dermatitis: expert statement and review from the International Eczema Council Skin Infection Group. Br J Dermatol 2019; 182:1331-1342. [PMID: 31677162 PMCID: PMC7317931 DOI: 10.1111/bjd.18643] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/27/2022]
Abstract
Patients with atopic dermatitis (AD) have an increased risk of bacterial skin infections, which cause significant morbidity and, if untreated, may become systemic. Staphylococcus aureus colonizes the skin of most patients with AD and is the most common organism to cause infections. Overt bacterial infection is easily recognized by the appearance of weeping lesions, honey‐coloured crusts and pustules. However, the wide variability in clinical presentation of bacterial infection in AD and the inherent features of AD – cutaneous erythema and warmth, oozing associated with oedema, and regional lymphadenopathy – overlap with those of infection, making clinical diagnosis challenging. Furthermore, some features may be masked because of anatomical site‐ and skin‐type‐specific features, and the high frequency of S. aureus colonization in AD makes positive skin swab culture of suspected infection unreliable as a diagnostic tool. The host mechanisms and microbial virulence factors that underlie S. aureus colonization and infection in AD are incompletely understood. The aim of this article is to present the latest evidence from animal and human studies, including recent microbiome research, to define the clinical features of bacterial infections in AD, and to summarize our current understanding of the host and bacterial factors that influence microbial colonization and virulence.
Collapse
Affiliation(s)
- H Alexander
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, SE1 7EH, U.K
| | - A S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, U.S.A
| | - C Traidl-Hoffmann
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany.,CK-CARE, Christine Kühne Center for Allergy Research and Education, Davos, Switzerland
| | - L A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, U.S.A
| | - A De Benedetto
- Department of Dermatology, College of Medicine, University of Florida, Gainesville, FL, U.S.A
| | - S Dhar
- Department of Pediatric Dermatology, Institute of Child Health, Kolkata, India
| | - G Girolomoni
- Department of Medicine, Section of Dermatology and Venereology, University of Verona, Verona, Italy
| | - A D Irvine
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland.,Dermatology, Children's Health Ireland, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland
| | - P Spuls
- Department of Dermatology, Amsterdam Public Health, Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - J Su
- Departments of Dermatology and Paediatrics, Murdoch Children's Research Institute, University of Melbourne and Monash University, Eastern Health, Melbourne, VIC, Australia
| | - J P Thyssen
- Department of Dermatology and Allergy, Herlev-Gentofte Hospital, Hellerup, Denmark
| | - C Vestergaard
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - T Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - A Wollenberg
- Department of Dermatology and Allergology, Ludwig Maximilian University, Munich, Germany
| | - M Deleuran
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - C Flohr
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, SE1 7EH, U.K
| |
Collapse
|
19
|
Zeng C, Liu Z, Han Z. Structure of Staphylococcal Enterotoxin N: Implications for Binding Properties to Its Cellular Proteins. Int J Mol Sci 2019; 20:ijms20235921. [PMID: 31775346 PMCID: PMC6928602 DOI: 10.3390/ijms20235921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 01/26/2023] Open
Abstract
Staphylococcus aureus strains produce a unique family of immunostimulatory exotoxins termed as bacterial superantigens (SAgs), which cross-link major histocompatibility complex class II (MHC II) molecule and T-cell receptor (TCR) to stimulate large numbers of T cells at extremely low concentrations. SAgs are associated with food poisoning and toxic shock syndrome. To date, 26 genetically distinct staphylococcal SAgs have been reported. This study reports the first X-ray structure of newly characterized staphylococcal enterotoxin N (SEN). SEN possesses the classical two domain architecture that includes an N-terminal oligonucleotide-binding fold and a C-terminal β-grasp domain. Amino acid and structure alignments revealed that several critical amino acids that are proposed to be responsible for MHC II and TCR molecule engagements are variable in SEN, suggesting that SEN may adopt a different binding mode to its cellular receptors. This work helps better understand the mechanisms of action of SAgs.
Collapse
Affiliation(s)
- Chi Zeng
- College of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (C.Z.); (Z.L.)
- Hubei Province Fresh Food Engineering Research Center, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Zhaoxin Liu
- College of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (C.Z.); (Z.L.)
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhenggang Han
- College of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (C.Z.); (Z.L.)
- Correspondence:
| |
Collapse
|
20
|
Barie PS, Narayan M, Sawyer RG. Immunization Against Staphylococcus aureus Infections. Surg Infect (Larchmt) 2019; 19:750-756. [PMID: 31033407 DOI: 10.1089/sur.2018.263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: Infections caused by Staphylococcus aureus continue to plague surgical patients, whether as surgical site infections or other nosocomial infections that complicate surgical care. The only meaningful methods available to decrease the risk of developing such infections are topical skin antisepsis (pre-operative skin preparation) and peri-operative antibiotic prophylaxis, neither of which offer a panacea. Alternatives to the latter are sought so as to minimize antibiotic selection pressure as a factor in the increasing problem of antimicrobial drug resistance. This review considers the possibility that immunization against S. aureus may offer a viable alternative for prophylaxis. Methods: Review and synthesis of pertinent English-language medical literature. Results: Vaccination against viral pathogens has been in successful clinical use for more than two centuries and was instrumental in the eradication of smallpox and the near-elimination of diseases such as poliomyelitis. Vaccinations against a limited number of bacterial pathogens (e.g., Bordetella pertussis, Clostridium tetanii, Corynebacterium diphtheriae, Haemophilus influenzae type b, Neisseria meningiditis, Streptococcus pneumoniae) have also been introduced with success, whereas others against bacteria are in development (C. difficile, Pseudomonas aeruginosa, S. aureus). Vaccination against S. aureus infection is in current veterinary use (e.g., to prevent mastitis among dairy cattle) but has not been successful to date in human beings despite multiple attempts, although development continues. Conclusions: Because of its complex microbiology, including multiple virulence factors and the ability to evade host immune surveillance, S. aureus presents numerous antigenic targets for vaccine development. Failure of two prior single-antigen vaccines in clinical trials has led to the consensus that future vaccine candidates must be directed against multiple antigens. Two distinct four-antigen vaccines are in clinical trials, but efficacy is yet to be determined.
Collapse
Affiliation(s)
- Philip S Barie
- 1 Department of Surgery, Weill Cornell Medicine , New York, New York
- 2 Department of Medicine, Weill Cornell Medicine , New York, New York
| | - Mayur Narayan
- 1 Department of Surgery, Weill Cornell Medicine , New York, New York
| | - Robert G Sawyer
- 3 Department of Surgery, Western Michigan University , Kalamazoo, Michigan
| |
Collapse
|
21
|
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: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
22
|
Proctor RA. Immunity to Staphylococcus aureus: Implications for Vaccine Development. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0037-2018. [PMID: 31298209 PMCID: PMC10957185 DOI: 10.1128/microbiolspec.gpp3-0037-2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Indexed: 12/19/2022] Open
Abstract
Cell-mediated immunity seems to be critical for prevention and resolution of invasive S. aureus infections, but an imbalance in this immunity may also produce SIRS and death or an inadequate protective response with prolonged bacteremia and death. This dysregulation is likely at the heart of mortality and severe disease in humans. Anti-toxin antibodies may also come into play in reducing the severity of S. aureus infections, but these antibodies might also address superantigen-induced immune dysregulation. Thus, while changing intrinsic T cell responses may be therapeutically difficult, monoclonal antibodies against superantigens may have utility in addressing dysfunctional immune responses to S. aureus. The models above are hypotheses for examining, and potentially dramatically improving immune response to and safety of S. aureus vaccines.
Collapse
Affiliation(s)
- Richard A Proctor
- University of Wisconsin, Medical Microbiology/Immunology, Madison, WI 53705
| |
Collapse
|
23
|
Radcliff FJ, Waldvogel-Thurlow S, Clow F, Mahadevan M, Johnston J, Li G, Proft T, Douglas RG, Fraser JD. Impact of Superantigen-Producing Bacteria on T Cells from Tonsillar Hyperplasia. Pathogens 2019; 8:pathogens8030090. [PMID: 31252586 PMCID: PMC6789895 DOI: 10.3390/pathogens8030090] [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: 05/08/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus and Group A Streptococcus (GAS) are common occupants of the tonsils and many strains produce potent exotoxins (mitogens) that directly target T cells, which could be a driver for tonsillar hyperplasia. Tonsil tissues from 41 patients were tested for these bacteria in conjunction with profiling of B and T cells by flow cytometry. S. aureus and GAS were detected in tonsil tissue from 44% and 7%, respectively, of patients by bacteriological culture; immuno-histology showed bacteria in close proximity to both B and T lymphocytes. The presence of tonsillar S. aureus did not alter B or T cell populations, whereas peripheral blood mucosal-associated invariant T (MAIT) cells were significantly increased in S. aureus culture positive individuals (p < 0.006). Alterations of tonsil CD4+ TCR Vβ family members relative to peripheral blood were evident in 29 patients. Three patients had strong TCR Vβ skewing indicative of recent exposure to superantigens, their tonsils contained mitogenic bacteria, and supernatants from these bacteria were used to partially recapitulate the skewing profile in vitro, supporting the notion that superantigens can target tonsillar T cells in situ. Tonsils are a reservoir for superantigen-producing bacteria with the capacity to alter the composition and function of key immune cells.
Collapse
Affiliation(s)
- Fiona J Radcliff
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand.
| | | | - Fiona Clow
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Murali Mahadevan
- Department of Surgery, University of Auckland, Auckland 1023, New Zealand
| | - James Johnston
- Department of Surgery, University of Auckland, Auckland 1023, New Zealand
| | - Gen Li
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Thomas Proft
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Richard G Douglas
- Department of Surgery, University of Auckland, Auckland 1023, New Zealand
| | - John D Fraser
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
24
|
Berger S, Kunerl A, Wasmuth S, Tierno P, Wagner K, Brügger J. Menstrual toxic shock syndrome: case report and systematic review of the literature. THE LANCET. INFECTIOUS DISEASES 2019; 19:e313-e321. [PMID: 31151811 DOI: 10.1016/s1473-3099(19)30041-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/20/2019] [Accepted: 01/25/2019] [Indexed: 01/20/2023]
Abstract
Menstrual toxic shock syndrome (mTSS) is a life-threatening disease caused by superantigen-producing Staphylococcus aureus. Incidence ranges from 0·03 to 0·50 cases per 100 000 people, with overall mortality around 8%. In this Grand Round, we present the case of a previously healthy 23-year-old menstruating woman who was diagnosed with mTSS after she presented at our hospital with a septic condition for the second time. The diagnosis was confirmed by fulfilment of the clinical criteria outlined by the US Centers for Disease Control and Prevention (CDC; fever, rash, desquamation, hypotension, and multi-system involvement) as well as a nasal swab positive for the S aureus strain and presence of the gene encoding for toxic shock syndrome toxin 1 (TSST-1). In the early 1980s, when mTSS was first described, use of tampons was considered the main risk factor. Today, the complex interplay between pathogenic factors of S aureus, immunological mechanisms of the host, and changes in the vaginal ecosystem during menstruation has broadened current understanding of the disease, and the CDC criteria have appreciable limitations in everyday clinical practice.
Collapse
Affiliation(s)
- Selina Berger
- Department of Internal Medicine, Sonnenhofspital, Bern, Switzerland
| | - Anika Kunerl
- Department of Internal Medicine, Sonnenhofspital, Bern, Switzerland
| | - Stefan Wasmuth
- Department of Internal Medicine, Sonnenhofspital, Bern, Switzerland
| | - Philip Tierno
- Department of Pathology, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Karoline Wagner
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Jan Brügger
- Department of Internal Medicine, Sonnenhofspital, Bern, Switzerland; University of Zurich, Zurich, Switzerland.
| |
Collapse
|
25
|
Botelho AMN, Cerqueira e Costa MO, Moustafa AM, Beltrame CO, Ferreira FA, Côrtes MF, Costa BSS, Silva DNS, Bandeira PT, Lima NCB, Souza RC, de Almeida LGP, Vasconcelos ATR, Narechania A, Ryan C, O’Brien K, Kolokotronis SO, Planet PJ, Nicolás MF, Figueiredo AMS. Local Diversification of Methicillin- Resistant Staphylococcus aureus ST239 in South America After Its Rapid Worldwide Dissemination. Front Microbiol 2019; 10:82. [PMID: 30873127 PMCID: PMC6400870 DOI: 10.3389/fmicb.2019.00082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
The global spread of specific clones of methicillin-resistant Staphylococcus aureus (MRSA) has become a major public health problem, and understanding the dynamics of geographical spread requires worldwide surveillance. Over the past 20 years, the ST239 lineage of MRSA has been recognized as an emerging clone across the globe, with detailed studies focusing on isolates from Europe and Asia. Less is known about this lineage in South America, and, particularly, Brazil where it was the predominant lineage of MRSA in the early 1990s to 2000s. To gain a better understanding about the introduction and spread of ST239 MRSA in Brazil we undertook a comparative phylogenomic analysis of ST239 genomes, adding seven completed, closed Brazilian genomes. Brazilian ST239 isolates grouped in a subtree with those from South American, and Western, romance-language-speaking, European countries, here designated the South American clade. After an initial worldwide radiation in the 1960s and 1970s, we estimate that ST239 began to spread in South America and Brazil in approximately 1988. This clone demonstrates specific genomic changes that are suggestive of local divergence and adaptational change including agrC single-nucleotide polymorphisms variants, and a distinct pattern of virulence-associated genes (mainly the presence of the chp and the absence of sea and sasX). A survey of a geographically and chronologically diverse set of 100 Brazilian ST239 isolates identified this virulence genotype as the predominant pattern in Brazil, and uncovered an unexpectedly high prevalence of agr-dysfunction (30%). ST239 isolates from Brazil also appear to have undergone transposon (IS256) insertions in or near global regulatory genes (agr and mgr) that likely led to rapid reprogramming of bacterial traits. In general, the overall pattern observed in phylogenomic analyses of ST239 is of a rapid initial global radiation, with subsequent local spread and adaptation in multiple different geographic locations. Most ST239 isolates harbor the ardA gene, which we show here to have in vivo anti-restriction activity. We hypothesize that this gene may have improved the ability of this lineage to acquire multiple resistance genes and distinct virulence-associated genes in each local context. The allopatric divergence pattern of ST239 also may suggest strong selective pressures for specific traits in different geographical locations.
Collapse
Affiliation(s)
- Ana Maria Nunes Botelho
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ahmed M. Moustafa
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, United States
| | - Cristiana Ossaille Beltrame
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabienne Antunes Ferreira
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marina Farrel Côrtes
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Souza Scramignon Costa
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Deborah Nascimento Santos Silva
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Terra Bandeira
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Rangel Celso Souza
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | | | | | - Apurva Narechania
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, United States
| | - Chanelle Ryan
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, United States
| | - Kelsey O’Brien
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, United States
| | - Sergios-Orestis Kolokotronis
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, United States
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Paul J. Planet
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, United States
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, United States
| | | | - Agnes Marie Sá Figueiredo
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Kumar R, Feltrup TM, Kukreja RV, Patel KB, Cai S, Singh BR. Evolutionary Features in the Structure and Function of Bacterial Toxins. Toxins (Basel) 2019; 11:toxins11010015. [PMID: 30609803 PMCID: PMC6356308 DOI: 10.3390/toxins11010015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/24/2018] [Accepted: 12/25/2018] [Indexed: 12/21/2022] Open
Abstract
Toxins can function both as a harmful and therapeutic molecule, depending on their concentrations. The diversity in their function allows us to ask some very pertinent questions related to their origin and roles: (a) What makes them such effective molecules? (b) Are there evolutionary features encoded within the structures of the toxins for their function? (c) Is structural hierarchy in the toxins important for maintaining their structure and function? (d) Do protein dynamics play a role in the function of toxins? and (e) Do the evolutionary connections to these unique features and functions provide the fundamental points in driving evolution? In light of the growing evidence in structural biology, it would be appropriate to suggest that protein dynamics and flexibility play a much bigger role in the function of the toxin than the structure itself. Discovery of IDPs (intrinsically disorder proteins), multifunctionality, and the concept of native aggregation are shaking the paradigm of the requirement of a fixed three-dimensional structure for the protein’s function. Growing evidence supporting the above concepts allow us to redesign the structure-function aspects of the protein molecules. An evolutionary model is necessary and needs to be developed to study these important aspects. The criteria for a well-defined model would be: (a) diversity in structure and function, (b) unique functionality, and (c) must belong to a family to define the evolutionary relationships. All these characteristics are largely fulfilled by bacterial toxins. Bacterial toxins are diverse and widely distributed in all three forms of life (Bacteria, Archaea and Eukaryotes). Some of the unique characteristics include structural folding, sequence and functional combination of domains, targeting a cellular process to execute their function, and most importantly their flexibility and dynamics. In this work, we summarize certain unique aspects of bacterial toxins, including role of structure in defining toxin function, uniqueness in their enzymatic function, and interaction with their substrates and other proteins. Finally, we have discussed the evolutionary aspects of toxins in detail, which will help us rethink the current evolutionary theories. A careful study, and appropriate interpretations, will provide answers to several questions related to the structure-function relationship of proteins, in general. Additionally, this will also allow us to refine the current evolution theories.
Collapse
Affiliation(s)
- Raj Kumar
- Botulinum Research Center, Institute of Advanced Sciences, Dartmouth, MA 02747, USA.
| | - Thomas M Feltrup
- Botulinum Research Center, Institute of Advanced Sciences, Dartmouth, MA 02747, USA.
| | - Roshan V Kukreja
- Botulinum Research Center, Institute of Advanced Sciences, Dartmouth, MA 02747, USA.
| | - Kruti B Patel
- Botulinum Research Center, Institute of Advanced Sciences, Dartmouth, MA 02747, USA.
| | - Shuowei Cai
- Department of Chemistry and Biochemistry, University of Massachusetts, Dartmouth, MA 02747, USA.
| | - Bal Ram Singh
- Botulinum Research Center, Institute of Advanced Sciences, Dartmouth, MA 02747, USA.
| |
Collapse
|
27
|
Wu X, Yang M, Fang X, Zhen S, Zhang J, Yang X, Qiao L, Yang Y, Zhang C. Expression and regulation of phenol-soluble modulins and enterotoxins in foodborne Staphylococcus aureus. AMB Express 2018; 8:187. [PMID: 30467730 PMCID: PMC6250609 DOI: 10.1186/s13568-018-0717-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/17/2018] [Indexed: 12/15/2022] Open
Abstract
Although high levels of staphylococcal phenol-soluble modulins (PSMs) in clinical methicillin-resistant Staphylococcus aureus (MRSA) has been shown to correlate with bacterial virulence, the PSMs expression in foodborne Staphylococcus aureus (S. aureus), as well as its association with staphylococcal food poisoning (SFP) was not yet clear. We collected a panel of 350 foodborne and 127 clinic-derived S. aureus strains and compared their PSMs expression. Overall, foodborne strains exhibited higher PSMs than clinical isolates, indicating a potential pathological significance of PSMs in staphylococcal food contamination. Furthermore, PSMs expression and staphylococcal enterotoxins (SEs) levels in relation to antibiotic sensitive and resistant strains were analysed. While the co-expression of PSMs and SEs was confirmed, one typical foodborne strain simultaneously yielding PSMs, SEB and SED was selected. By comparing this wildtype strain to a series of gene-deficient mutants, we concluded that PSMs and SEs expressions both relied on staphylococcal accessory regulator A initiation in the early stage of accessory gene regulator control, yet their succedent regulations differentiated to RNAIII-dependent and independent, respectively. These data provided preliminary insight into PSMs and SEs expression in foodborne S. aureus, and may guide the further studies on PSMs effects in SFP.
Collapse
|
28
|
Bovine Staphylococcus aureus Superantigens Stimulate the Entire T Cell Repertoire of Cattle. Infect Immun 2018; 86:IAI.00505-18. [PMID: 30201699 PMCID: PMC6204692 DOI: 10.1128/iai.00505-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/29/2018] [Indexed: 11/20/2022] Open
Abstract
Superantigens (SAgs) represent a diverse family of bacterial toxins that induce Vβ-specific T cell proliferation associated with an array of important diseases in humans and animals, including mastitis of dairy cows. However, an understanding of the diversity and distribution of SAg genes among bovine Staphylococcus aureus strains and their role in the pathogenesis of mastitis is lacking. Superantigens (SAgs) represent a diverse family of bacterial toxins that induce Vβ-specific T cell proliferation associated with an array of important diseases in humans and animals, including mastitis of dairy cows. However, an understanding of the diversity and distribution of SAg genes among bovine Staphylococcus aureus strains and their role in the pathogenesis of mastitis is lacking. Population genomic analysis of 195 bovine S. aureus isolates representing 57 unique sequence types revealed that strains encode 2 to 13 distinct SAgs and that the majority of isolates contain 5 or more SAg genes. A genome-scale analysis of bovine reference strain RF122 revealed a complement of 11 predicted SAg genes, which were all expressed in vitro. Detection of specific antibodies in convalescent cows suggests expression of 7 of 11 SAgs during natural S. aureus infection. We determined the Vβ T cell activation profile for all functional SAgs encoded by RF122, revealing evidence for bovine host-specific activity among the recently identified RF122-encoded SAgs SElY and SElZ. Remarkably, we discovered that some strains have evolved the capacity to stimulate the entire T cell repertoire of cattle through an array of diverse SAgs, suggesting a key role in bovine immune evasion.
Collapse
|
29
|
Noviello S, Huang DB, Corey GR. Iclaprim: a differentiated option for the treatment of skin and skin structure infections. Expert Rev Anti Infect Ther 2018; 16:793-803. [PMID: 30317894 DOI: 10.1080/14787210.2018.1536545] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Iclaprim is a selective bacterial dihydrofolate reductase (DHFR) inhibitor. Although there are alternative options for the treatment of acute bacterial skin and skin structure infections (ABSSSI), iclaprim is differentiated from other available antibiotics. Areas covered: Iclaprim is under clinical development for ABSSSI. This review summarizes the mechanism of action, pharmacokinetics, microbiology, clinical development program, and the differentiation of iclaprim from other antibiotics. Expert commentary: Iclaprim has a different mechanism of action (DHFR inhibitor) compared to most other antibiotics, is active and rapidly bactericidal against Gram-positive pathogens including antibiotic-resistant pathogens, and suppresses bacterial exotoxins (alpha hemolysin, Panton Valentine leukocidin, and toxic shock syndrome toxin-1). Compared to trimethoprim, iclaprim has lower MIC90s, can be given without a sulfonamide, overcomes select trimethoprim resistance, and does not cause hyperkalemia. Iclaprim is administered as a fixed dose, does not require dose adjustment in renally-impaired or obese patients, and was not associated with nephrotoxicity in the Phase 3 pivotal REVIVE studies. Iclaprim represents a novel, alternative option for the treatment of severe skin and skin structure infections due to Gram-positive bacteria, particularly in patients at risk of acute kidney injury.
Collapse
Affiliation(s)
| | - David B Huang
- a Motif BioSciences , Princeton , NJ , USA.,b Department of Internal Medicine, Division of Infectious Diseases , Rutgers New Jersey Medical School , Trenton , NJ , USA
| | - G Ralph Corey
- c Department of Medicine , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
30
|
He C, Xu S, Zhao H, Hu F, Xu X, Jin S, Yang H, Gong F, Liu Q. Leukotoxin and pyrogenic toxin Superantigen gene backgrounds in bloodstream and wound Staphylococcus aureus isolates from eastern region of China. BMC Infect Dis 2018; 18:395. [PMID: 30103694 PMCID: PMC6090790 DOI: 10.1186/s12879-018-3297-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 08/01/2018] [Indexed: 11/10/2022] Open
Abstract
Background The bicomponent leukotoxins and the pyrogenic toxin superantigens (PTSAgs) are important virulence factors of Staphylococcus aureus. It is necessary to survey the prevalence and expression of these toxin-encoding genes for understanding the possible pathogenic capacity of S. aureus to cause disease. Methods Five leukotoxin genes and thirteen PTSAg determinants were detected for 177 S. aureus isolates from blood (n = 88) and wound (n = 89) infections by Polymerase Chain Reaction (PCR). The expression of leukotoxin ED (lukED) was determined by quantitative real-time PCR (qRT-PCR). The genetic backgrounds of isolates were analyzed by Staphylococcal Cassette Chromosome mec (SCCmec) typing (for methicillin-resistant S. aureus isolates), Pulsed-Field Gel Electrophoresis (PFGE), accessory gene regulator (agr) typing and Multilocus Sequence Typing (MLST, for representative isolates based on PFGE type) methods. Results 99.4% (176/177) isolates contained at least one of leukotoxin genes. Among them, 94.9% (168/177), 81.4% (144/177) and 67.8% (120/177) isolates harbored hlgBC, lukED and lukAB, respectively. Compared to leukotoxin genes, there was a relatively lower overall prevalence of PTSAg genes [99.4% versus 72.9% (129/177), P < 0.001], and they were organized in 59 patterns, with the most common combination of the egc cluster with or without other PTSAg genes. Genetic analysis showed the distributions of certain toxin genes were associated with the genetic backgrounds of isolates. The egc cluster was a common feature of CC5 isolates, among which ST5 and ST764 isolates harbored more PTSAg genes. The lukED was not present in ST398 isolates, and its expression was quite different among isolates. No significant correlations were observed between the lukED expression levels of strains and the ST or agr types. Conclusions The present study elucidated the distribution of leukotoxin and PTSAg genes and the expression of lukED in blood and wound isolates, and analyzed the relationship between them with genetic characteristics of isolates. These data improve the current understanding of the possible pathogenicity of S. aureus. Electronic supplementary material The online version of this article (10.1186/s12879-018-3297-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chunyan He
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Su Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200080, China
| | - Huanqiang Zhao
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Fupin Hu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200080, China
| | - Xiaogang Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200080, China
| | - Shu Jin
- Experimental Research Center, Shanghai People's Hospital of Putuo District, Shanghai, 200080, China
| | - Han Yang
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Fang Gong
- Department of Clinical Laboratory, the Third Hospital Affiliated to Nantong University, Wuxi, 226000, China
| | - Qingzhong Liu
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
31
|
Contribution of toxic shock syndrome toxin-1 to systemic inflammation investigated by a mouse model of cervicovaginal infection with Staphylococcus aureus. Med Microbiol Immunol 2018; 207:297-306. [DOI: 10.1007/s00430-018-0551-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022]
|
32
|
Woetmann A, Alhede M, Dabelsteen S, Bjarnsholt T, Rybtke M, Nastasi C, Krejsgaard T, Andersen MH, Bonefeld CM, Geisler C, Givskov M, Odum N. Interleukin-26 (IL-26) is a novel anti-microbial peptide produced by T cells in response to staphylococcal enterotoxin. Oncotarget 2018; 9:19481-19489. [PMID: 29731960 PMCID: PMC5929403 DOI: 10.18632/oncotarget.24603] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/24/2018] [Indexed: 01/08/2023] Open
Abstract
Anti-microbial peptides are produced at outer and inner surfaces by epithelia and innate immune cells in response to bacterial infection. Staphylococcus aureus is an enterotoxin producing, Gram-positive pathogen, which is a major cause of soft tissue infections and life-threatening bacteremia and sepsis. Here we show that (i) skin T cells in chronic wounds infected with S. aureus express interleukin-26 (IL-26) in situ, (ii) staphylococcal enterotoxins (SE) trigger IL-26 expression in T cell lines and primary skin T cells, and (iii) IL-26 triggers death and inhibits biofilm formation and growth of S. aureus. Thus, we provide novel evidence that IL-26 is an anti-microbial peptide produced by T cells in response to SE. Accordingly, we propose that IL-26 producing T cells take part in the innate immune response to SE producing S. aureus and thus play a novel role in the primary innate immune defense in addition to their classical role in adaptive immunity.
Collapse
Affiliation(s)
- Anders Woetmann
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Morten Alhede
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Sally Dabelsteen
- Department of Odontology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Morten Rybtke
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Claudia Nastasi
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Thorbjørn Krejsgaard
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Charlotte M Bonefeld
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Geisler
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Givskov
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark.,Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark.,Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore
| | - Niels Odum
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Benkerroum N. Staphylococcal enterotoxins and enterotoxin-like toxins with special reference to dairy products: An overview. Crit Rev Food Sci Nutr 2017; 58:1943-1970. [DOI: 10.1080/10408398.2017.1289149] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Noreddine Benkerroum
- Department of Food Science and Agricultural Chemistry, Macdonald-Stewart Building, McGill University, Macdonald Campus, Sainte-Anne-de-Bellevue, Canada
| |
Collapse
|
34
|
Chen Y, Huang Y, Liang B, Dong H, Yao S, Xie Y, Long Y, Zhong H, Yang Y, Zhu B, Gong S, Zhou Z. Inverse relationship between toxic shock syndrome toxin-1 antibodies and interferon-γ and interleukin-6 in peripheral blood mononuclear cells from patients with pediatric tonsillitis caused by Staphylococcus aureus. Int J Pediatr Otorhinolaryngol 2017; 97:211-217. [PMID: 28483238 DOI: 10.1016/j.ijporl.2017.04.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Pediatric tonsillitis is frequently caused by Staphylococcus aureus, which is the most common pathogen that causes serious pyogenic infections in humans and endangers human health. S. aureus produces numerous potent virulence factors that play a critical role in the pathogenesis of the infection caused by this bacterium, and one of the most important toxins produced by S. aureus is toxic shock syndrome toxin-1 (TSST-1). The aim of this study is to investigate the first time the levels of IFN-γ and interleukin IL-6 in TSST-1-stimulated PBMCs from pediatric tonsillitis patients and the correlation of these cytokine levels with TSST-1-specific IgG in serum. METHODS TSST-1 gene of S. aureus was cloned and expressed in a prokaryotic expression system, and purified recombinant TSST-1 protein was used for measuring TSST-1-specific antibodies in the serum of patients with pediatric tonsillitis caused by S. aureus. Moreover, the levels of interferon (IFN)-γ and interleukin (IL)-6 in TSST-1-stimulated peripheral blood mononuclear cells (PBMCs) from pediatric tonsillitis patients were investigated. RESULTS In patients with pediatric tonsillitis caused by S. aureus, significantly higher levels of serum TSST-1-specific IgG (P < 0.05) and IgG1 (P < 0.05) were detected than in healthy children. Moreover, PBMCs from the patients exhibited higher IFN-γ (P < 0.05) production in response to TSST-1 than did PBMCs from healthy children. In patients with pediatric tonsillitis caused by S. aureus, the positive rate of TSST-1-specific IgG was 70%, and the patients who tested negative for TSST-1-specific IgG exhibited significantly higher levels of IFN-γ (P < 0.05) and IL-6 (P < 0.05) than did the IgG-positive patients, in accord, the levels of TSST-1-specific IgG correlated inversely with the levels of IFN-γ and IL-6 in patients PBMCs stimulated with TSST-1. CONCLUSIONS TSST-1 induces humoral and cellular immunity in pediatric tonsillitis caused by S. aureus, which suggests that TSST-1 may play an important role in the pathogenesis of pediatric tonsillitis.
Collapse
Affiliation(s)
- Yinshuang Chen
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China; The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Yanmei Huang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Bingshao Liang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Hui Dong
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Shuwen Yao
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Yongqiang Xie
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Yan Long
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Huamin Zhong
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Yiyu Yang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Bing Zhu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Sitang Gong
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| | - Zhenwen Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.318 Renminzhong Road, Yuexiu, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
35
|
Martin AR, Pollack RA, Capoferri A, Ambinder RF, Durand CM, Siliciano RF. Rapamycin-mediated mTOR inhibition uncouples HIV-1 latency reversal from cytokine-associated toxicity. J Clin Invest 2017; 127:651-656. [PMID: 28094770 DOI: 10.1172/jci89552] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/30/2016] [Indexed: 12/13/2022] Open
Abstract
Current strategies for HIV-1 eradication require the reactivation of latent HIV-1 in resting CD4+ T cells (rCD4s). Global T cell activation is a well-characterized means of inducing HIV-1 transcription, but is considered too toxic for clinical applications. Here, we have explored a strategy that involves a combination of immune activation and the immunosuppressive mTOR inhibitor rapamycin. In purified rCD4s from HIV-1-infected individuals on antiretroviral therapy, rapamycin treatment downregulated markers of toxicity, including proinflammatory cytokine release and cellular proliferation that were induced after potent T cell activation using αCD3/αCD28 antibodies. Using an ex vivo assay for HIV-1 mRNA, we demonstrated that despite this immunomodulatory effect, rapamycin did not affect HIV-1 gene expression induced by T cell activation in these rCD4s. In contrast, treating activated rCD4s with the immunosuppressant cyclosporin, a calcineurin inhibitor, robustly inhibited HIV-1 reactivation. Importantly, rapamycin treatment did not impair cytotoxic T lymphocyte (CTL) recognition and killing of infected cells. These findings raise the possibility of using rapamycin in conjunction with T cell-activating agents in HIV-1 cure strategies.
Collapse
|
36
|
Kametani Y, Ohshima S, Miyamoto A, Shigenari A, Takasu M, Imaeda N, Matsubara T, Tanaka M, Shiina T, Kamiguchi H, Suzuki R, Kitagawa H, Kulski JK, Hirayama N, Inoko H, Ando A. Production of a Locus- and Allele-Specific Monoclonal Antibody for the Characterization of SLA-1*0401 mRNA and Protein Expression Levels in MHC-Defined Microminipigs. PLoS One 2016; 11:e0164995. [PMID: 27760184 PMCID: PMC5070868 DOI: 10.1371/journal.pone.0164995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/04/2016] [Indexed: 12/17/2022] Open
Abstract
The class I major histocompatibility complex (MHC) presents self-developed peptides to specific T cells to induce cytotoxity against infection. The MHC proteins are encoded by multiple loci that express numerous alleles to preserve the variability of the antigen-presenting ability in each species. The mechanism regulating MHC mRNA and protein expression at each locus is difficult to analyze because of the structural and sequence similarities between alleles. In this study, we examined the correlation between the mRNA and surface protein expression of swine leukocyte antigen (SLA)-1*0401 after the stimulation of peripheral blood mononuclear cells (PBMCs) by Staphylococcus aureus superantigen toxic shock syndrome toxin-1 (TSST-1). We prepared a monoclonal antibody (mAb) against a domain composed of Y102, L103 and L109 in the α2 domain. The Hp-16.0 haplotype swine possess only SLA-1*0401, which has the mAb epitope, while other haplotypes possess 0 to 3 SLA classical class I loci with the mAb epitopes. When PBMCs from SLA-1*0401 homozygous pigs were stimulated, the SLA-1*0401 mRNA expression level increased until 24 hrs and decreased at 48 hrs. The kinetics of the interferon regulatory transcription factor-1 (IRF-1) mRNA level were similar to those of the SLA-1*0401 mRNA. However, the surface protein expression level continued to increase until 72 hrs. Similar results were observed in the Hp-10.0 pigs with three mAb epitopes. These results suggest that TSST-1 stimulation induced both mRNA and surface protein expression of class I SLA in the swine PBMCs differentially and that the surface protein level was sustained independently of mRNA regulation.
Collapse
Affiliation(s)
- Yoshie Kametani
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Institute of Advanced Biosciences, Tokai University, Kanagawa, Japan
- * E-mail:
| | - Shino Ohshima
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Asuka Miyamoto
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Atsuko Shigenari
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Masaki Takasu
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, Gifu, Japan
| | - Noriaki Imaeda
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, Gifu, Japan
| | - Tatsuya Matsubara
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, Gifu, Japan
| | - Masafumi Tanaka
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Takashi Shiina
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiroshi Kamiguchi
- Teaching and Research Support Center, Tokai University School of Medicine, Isehara, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Sagamihara, Kanagawa, Japan
| | - Hitoshi Kitagawa
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, Gifu, Japan
| | - Jerzy K. Kulski
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Crawley WA, Australia
| | - Noriaki Hirayama
- Institute of Glycoscience, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Hidetoshi Inoko
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Asako Ando
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
37
|
Li L, Pian Y, Chen S, Hao H, Zheng Y, Zhu L, Xu B, Liu K, Li M, Jiang H, Jiang Y. Phenol-soluble modulin α4 mediates Staphylococcus aureus-associated vascular leakage by stimulating heparin-binding protein release from neutrophils. Sci Rep 2016; 6:29373. [PMID: 27383625 PMCID: PMC4935938 DOI: 10.1038/srep29373] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/16/2016] [Indexed: 11/09/2022] Open
Abstract
Vascular leakage frequently occurs in patients with severe Staphylococcus aureus infection. However, the mechanism underlying S. aureus infection-induced vascular leakage remains unclear. Here, we identified the S. aureus virulence factor phenol-soluble modulin (PSM)α4 from the culture supernatant of strain USA300 as a stimulator of heparin-binding protein (HBP) release from polymorphonuclear neutrophils (PMNs) and demonstrated that PSMα4-induced HBP release from PMNs leads to vascular leakage. PSMα4 appeared less cytolytic than PSMα1-3 and was insensitive to lipoproteins; it significantly increased myeloperoxidase and elastase release from PMNs and cell surface CD63 expression in PMNs. PSMα4-induced HBP release required formyl peptide receptor 2 (FPR2) and phosphoinositide 3-kinase (PI3K) and depended on Ca(2+) influx and cytoskeleton rearrangement. Thus, PSMα4 may stimulate HBP release by activating FPR2 and PI3K to initiate PMN degranulation. PSMα4-induced HBP release from PMNs increased endothelial cell monolayer permeability in vitro and induced vascular leakage in mice. This novel function of PSMα4 may contribute to the pathogenesis of S. aureus and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Lin Li
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yaya Pian
- Key Laboratory of infection and immunity, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Shaolong Chen
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Huaijie Hao
- Institution of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Bin Xu
- National Center of Biomedical Analysis, Beijing, China
| | - Keke Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Min Li
- Department of laboratory medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
38
|
Whole-Genome Sequencing for Routine Pathogen Surveillance in Public Health: a Population Snapshot of Invasive Staphylococcus aureus in Europe. mBio 2016; 7:mBio.00444-16. [PMID: 27150362 PMCID: PMC4959656 DOI: 10.1128/mbio.00444-16] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The implementation of routine whole-genome sequencing (WGS) promises to transform our ability to monitor the emergence and spread of bacterial pathogens. Here we combined WGS data from 308 invasive Staphylococcus aureus isolates corresponding to a pan-European population snapshot, with epidemiological and resistance data. Geospatial visualization of the data is made possible by a generic software tool designed for public health purposes that is available at the project URL (http://www.microreact.org/project/EkUvg9uY?tt=rc). Our analysis demonstrates that high-risk clones can be identified on the basis of population level properties such as clonal relatedness, abundance, and spatial structuring and by inferring virulence and resistance properties on the basis of gene content. We also show that in silico predictions of antibiotic resistance profiles are at least as reliable as phenotypic testing. We argue that this work provides a comprehensive road map illustrating the three vital components for future molecular epidemiological surveillance: (i) large-scale structured surveys, (ii) WGS, and (iii) community-oriented database infrastructure and analysis tools. The spread of antibiotic-resistant bacteria is a public health emergency of global concern, threatening medical intervention at every level of health care delivery. Several recent studies have demonstrated the promise of routine whole-genome sequencing (WGS) of bacterial pathogens for epidemiological surveillance, outbreak detection, and infection control. However, as this technology becomes more widely adopted, the key challenges of generating representative national and international data sets and the development of bioinformatic tools to manage and interpret the data become increasingly pertinent. This study provides a road map for the integration of WGS data into routine pathogen surveillance. We emphasize the importance of large-scale routine surveys to provide the population context for more targeted or localized investigation and the development of open-access bioinformatic tools to provide the means to combine and compare independently generated data with publicly available data sets.
Collapse
|
39
|
Kong C, Neoh HM, Nathan S. Targeting Staphylococcus aureus Toxins: A Potential form of Anti-Virulence Therapy. Toxins (Basel) 2016; 8:toxins8030072. [PMID: 26999200 PMCID: PMC4810217 DOI: 10.3390/toxins8030072] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/03/2016] [Accepted: 03/10/2016] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen and the leading cause of a wide range of severe clinical infections. The range of diseases reflects the diversity of virulence factors produced by this pathogen. To establish an infection in the host, S. aureus expresses an inclusive set of virulence factors such as toxins, enzymes, adhesins, and other surface proteins that allow the pathogen to survive under extreme conditions and are essential for the bacteria’s ability to spread through tissues. Expression and secretion of this array of toxins and enzymes are tightly controlled by a number of regulatory systems. S. aureus is also notorious for its ability to resist the arsenal of currently available antibiotics and dissemination of various multidrug-resistant S. aureus clones limits therapeutic options for a S. aureus infection. Recently, the development of anti-virulence therapeutics that neutralize S. aureus toxins or block the pathways that regulate toxin production has shown potential in thwarting the bacteria’s acquisition of antibiotic resistance. In this review, we provide insights into the regulation of S. aureus toxin production and potential anti-virulence strategies that target S. aureus toxins.
Collapse
Affiliation(s)
- Cin Kong
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor Darul Ehsan, Malaysia.
| | - Hui-min Neoh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Sheila Nathan
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
40
|
Ataee RA, Kashefi R, Alishiri GH, Esmaieli D. Assay of Blood and Synovial Fluid of Patients With Rheumatoid Arthritis for Staphylococcus aureus Enterotoxin D: Absence of Bacteria But Presence of Its Toxin. Jundishapur J Microbiol 2015; 8:e28395. [PMID: 26870313 PMCID: PMC4746793 DOI: 10.5812/jjm.28395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/10/2015] [Accepted: 05/21/2015] [Indexed: 01/04/2023] Open
Abstract
Background: Rheumatoid arthritis (RA) is the most common chronic inflammatory disease. The staphylococcal superantigens are considered as the causative agent of RA disease. Objectives: This study aimed to assess the presence of staphylococcal enterotoxin D in synovial fluid and blood of patients with RA. Patients and Methods: A total of 120 blood and SF samples of patients with RA were studied. Bacterial culture, primer pairs design, polymerase chain reaction (PCR), and enzyme-linked immunosorbent assay (ELISA) methods have been used to assess of the staphylococcal enterotoxin D. The data were analyzed through descriptive statistics. Results: During this study and after sequential subcultures, only 5 bacterial strains were isolated. The results of PCR showed the presence of staphylococcal enterotoxin D gene in almost 50% of SF and also in 48.4% of blood samples of patients with RA. Similarly, the ELISA method detected staphylococcal enterotoxin D in 36.16% of SF and in 33.33% of blood of patients with RA. Conclusions: The result of this study showed that a high percentage of patients with RA have shown staphylococcal enterotoxin D (superantigen D) or entD gene in SF and in blood. However, the origin of this superantigen was not clarified and no Staphylococcus aureus enterotoxin D producer was isolated. This finding indicates other role of this superantigen besides its intoxication. Therefore, staphylococcal enterotoxin D as a biomarker may provide a good model for the diagnosis and treatment of patients with RA.
Collapse
Affiliation(s)
- Ramezan Ali Ataee
- Department of Medical Microbiology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Reyhane Kashefi
- Department of Microbial Biotechnology, Faculty of Science, Payame Noor University, Tehran, IR Iran
- Corresponding author: Reyhane Kashefi, Department of Microbial Biotechnology, Faculty of Science, Payame Noor University, Tehran, IR Iran. Tel: +98-989122190418, Fax: +98-2126127258, E-mail:
| | - Gholam Hossein Alishiri
- Department of Rheumatology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Davoud Esmaieli
- Department of Medical Microbiology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| |
Collapse
|
41
|
Vuong C, Yeh AJ, Cheung GYC, Otto M. Investigational drugs to treat methicillin-resistant Staphylococcus aureus. Expert Opin Investig Drugs 2015; 25:73-93. [PMID: 26536498 DOI: 10.1517/13543784.2016.1109077] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Staphylococcus aureus remains one of the leading causes of morbidity and mortality worldwide. This is to a large extent due to antibiotic-resistant strains, in particular methicillin-resistant S. aureus (MRSA). While the toll of invasive MRSA infections appears to decrease in U.S. hospitals, the rate of community-associated MRSA infections remains constant and there is a surge of MRSA in many other countries, a situation that calls for continuing if not increased efforts to find novel strategies to combat MRSA infections. AREAS COVERED This review provides an overview of current investigational drugs and therapeutic antibodies against S. aureus in early clinical development (up to phase II clinical development). It includes a short description of the mechanism of action and a presentation of microbiological and clinical data. EXPERT OPINION Increased recent antibiotic development efforts and results from pathogenesis research have led to several new antibiotics and therapies, such as anti-virulence drugs, as well as a more informed selection of targets for vaccination efforts against MRSA. This developing portfolio of novel anti-staphylococcal drugs will hopefully provide us with additional and more efficient ways to combat MRSA infections in the near future and prevent us from running out of treatment options, even if new resistances arise.
Collapse
Affiliation(s)
- Cuong Vuong
- a Principal Scientist/Laboratory Head, Bacteriology , AiCuris GmbH & Co. KG, Friedrich-Ebert-Str. 475/Geb. 302, 42117 Wuppertal , Germany
| | - Anthony J Yeh
- b Post-baccalaureate IRTA, Laboratory of Bacteriology , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| | - Gordon Y C Cheung
- c Staff Scientist, National Institute of Allergy and Infectious Diseases , National Institutes of Health, Laboratory of Bacteriology , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| | - Michael Otto
- d Senior Investigator, National Institute of Allergy and Infectious Diseases , National Institutes of Health, Laboratory of Bacteriology , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| |
Collapse
|
42
|
Thammavongsa V, Kim HK, Missiakas D, Schneewind O. Staphylococcal manipulation of host immune responses. Nat Rev Microbiol 2015; 13:529-43. [PMID: 26272408 DOI: 10.1038/nrmicro3521] [Citation(s) in RCA: 418] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Staphylococcus aureus, a bacterial commensal of the human nares and skin, is a frequent cause of soft tissue and bloodstream infections. A hallmark of staphylococcal infections is their frequent recurrence, even when treated with antibiotics and surgical intervention, which demonstrates the bacterium's ability to manipulate innate and adaptive immune responses. In this Review, we highlight how S. aureus virulence factors inhibit complement activation, block and destroy phagocytic cells and modify host B cell and T cell responses, and we discuss how these insights might be useful for the development of novel therapies against infections with antibiotic resistant strains such as methicillin-resistant S. aureus.
Collapse
Affiliation(s)
- Vilasack Thammavongsa
- 1] Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, Illinois 60637, USA. [2] Regeneron Pharmaceuticals, 755 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Hwan Keun Kim
- Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, Illinois 60637, USA
| | - Dominique Missiakas
- Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, Illinois 60637, USA
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, Illinois 60637, USA
| |
Collapse
|
43
|
Breitwieser FP, Pardo CA, Salzberg SL. Re-analysis of metagenomic sequences from acute flaccid myelitis patients reveals alternatives to enterovirus D68 infection. F1000Res 2015; 4:180. [PMID: 26309730 PMCID: PMC4536609 DOI: 10.12688/f1000research.6743.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2015] [Indexed: 02/01/2023] Open
Abstract
Metagenomic sequence data can be used to detect the presence of infectious viruses and bacteria, but normal microbial flora make this process challenging. We re-analyzed metagenomic RNA sequence data collected during a recent outbreak of acute flaccid myelitis (AFM), caused in some cases by infection with enterovirus D68. We found that among the patients whose symptoms were previously attributed to enterovirus D68, one patient had clear evidence of infection with
Haemophilus influenzae, and a second patient had a severe
Staphylococcus aureus infection caused by a methicillin-resistant strain. Neither of these bacteria were identified in the original study. These observations may have relevance in cases that present with flaccid paralysis because bacterial infections, co-infections or post-infection immune responses may trigger pathogenic processes that may present as poliomyelitis-like syndromes and may mimic AFM. A separate finding was that large numbers of human sequences were present in each of the publicly released samples, although the original study reported that human sequences had been removed before deposition.
Collapse
Affiliation(s)
- Florian P Breitwieser
- Center for Computational Biology, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins Hospital, Baltimore, MD, 21205, USA
| | - Steven L Salzberg
- Center for Computational Biology, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA ; Departments of Biomedical Engineering, Computer Science, and Biostatistics, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
44
|
Healthcare- and Community-Associated Methicillin-Resistant Staphylococcus aureus (MRSA) and Fatal Pneumonia with Pediatric Deaths in Krasnoyarsk, Siberian Russia: Unique MRSA's Multiple Virulence Factors, Genome, and Stepwise Evolution. PLoS One 2015; 10:e0128017. [PMID: 26047024 PMCID: PMC4457420 DOI: 10.1371/journal.pone.0128017] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 04/21/2015] [Indexed: 12/25/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a common multidrug-resistant (MDR) pathogen. We herein discussed MRSA and its infections in Krasnoyarsk, Siberian Russia between 2007 and 2011. The incidence of MRSA in 3,662 subjects was 22.0% and 2.9% for healthcare- and community-associated MRSA (HA- and CA-MRSA), respectively. The 15-day mortality rates for MRSA hospital- and community-acquired pneumonia (HAP and CAP) were 6.5% and 50%, respectively. MRSA CAP cases included pediatric deaths; of the MRSA pneumonia episodes available, ≥27.3% were associated with bacteremia. Most cases of HA-MRSA examined exhibited ST239/spa3(t037)/SCCmecIII.1.1.2 (designated as ST239Kras), while all CA-MRSA cases examined were ST8/spa1(t008)/SCCmecIV.3.1.1(IVc) (designated as ST8Kras). ST239Kras and ST8Kras strongly expressed cytolytic peptide (phenol-soluble modulin α, PSMα; and δ-hemolysin, Hld) genes, similar to CA-MRSA. ST239Kras pneumonia may have been attributed to a unique set of multiple virulence factors (MVFs): toxic shock syndrome toxin-1 (TSST-1), elevated PSMα/Hld expression, α-hemolysin, the staphylococcal enterotoxin SEK/SEQ, the immune evasion factor SCIN/SAK, and collagen adhesin. Regarding ST8Kras, SEA was included in MVFs, some of which were common to ST239Kras. The ST239Kras (strain OC3) genome contained: a completely unique phage, φSa7-like (W), with no att repetition; S. aureus pathogenicity island SaPI2R, the first TSST-1 gene-positive (tst+) SaPI in the ST239 lineage; and a super copy of IS256 (≥22 copies/genome). ST239Kras carried the Brazilian SCCmecIII.1.1.2 and United Kingdom-type tst. ST239Kras and ST8Kras were MDR, with the same levofloxacin resistance mutations; small, but transmissible chloramphenicol resistance plasmids spread widely enough to not be ignored. These results suggest that novel MDR and MVF+ HA- and CA-MRSA (ST239Kras and ST8Kras) emerged in Siberian Russia (Krasnoyarsk) associated with fatal pneumonia, and also with ST239Kras, a new (Siberian Russian) clade of the ST239 lineage, which was created through stepwise evolution during its potential transmission route of Brazil-Europe-Russia/Krasnoyarsk, thereby selective advantages from unique MVFs and the MDR.
Collapse
|
45
|
Lee KY. A common immunopathogenesis mechanism for infectious diseases: the protein-homeostasis-system hypothesis. Infect Chemother 2015; 47:12-26. [PMID: 25844259 PMCID: PMC4384454 DOI: 10.3947/ic.2015.47.1.12] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Indexed: 01/09/2023] Open
Abstract
It was once believed that host cell injury in various infectious diseases is caused solely by pathogens themselves; however, it is now known that host immune reactions to the substances from the infectious agents and/or from the injured host cells by infectious insults are also involved. All biological phenomena in living organisms, including biochemical, physiological and pathological processes, are performed by the proteins that have various sizes and shapes, which in turn are controlled by an interacting network within the living organisms. The author proposes that this network is controlled by the protein homeostasis system (PHS), and that the immune system is one part of the PHS of the host. Each immune cell in the host may recognize and respond to substances, including pathogenic proteins (PPs) that are toxic to target cells of the host, in ways that depend on the size and property of the PPs. Every infectious disease has its own set of toxic substances, including PPs, associated with disease onset, and the PPs and the corresponding immune cells may be responsible for the inflammatory processes that develop in those infectious diseases.
Collapse
Affiliation(s)
- Kyung-Yil Lee
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea. ; Department of Pediatrics, The Catholic University of Korea, Daejeon St. Mary's Hospital, Daejeon, Korea
| |
Collapse
|