1
|
Parolini C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167761. [PMID: 40044061 DOI: 10.1016/j.bbadis.2025.167761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
In 2020, sepsis has been defined a worldwide health major issue (World Health Organization). Lung, urinary tract and abdominal cavity are the preferred sites of sepsis-linked infection. Research has highlighted that the advancement of sepsis is not only related to the presence of inflammation or microbial or host pattern recognition. Clinicians and researchers now recognized that a severe immunosuppression is also a common feature found in patients with sepsis, increasing the susceptibility to secondary infections. Lipopolysaccharides (LPS) are expressed on the cell surface of Gram-negative, whereas Gram-positive bacteria express peptidoglycan (PGN) and lipoteichoic acid (LTA). The main mechanism by which LPS trigger host innate immune responses is binding to TLR4-MD2 (toll-like receptor4-myeloid differentiation factor 2), whereas, PGN and LTA are exogenous ligands of TLR2. Nucleotide-binding oligomerization domain (NOD)-like receptors are the most well-characterized cytosolic pattern recognition receptors, which bind microbial molecules, endogenous by-products and environmental triggers. It has been demonstrated that high-density lipoproteins (HDL), besides their major role in promoting cholesterol efflux, possess diverse pleiotropic properties, ranging from a modulation of the immune system to anti-inflammatory, anti-apoptotic, and anti-oxidant functions. In addition, HDL are able at i) binding LPS, preventing the activating of TLR4, and ii) inducing the expression of ATF3 (Activating transcription factor 3), a negative regulator of the TLR signalling pathways, contributing at justifying their capacity to hamper infection-based illnesses. Therefore, reconstituted HDL (rHDL), constituted by apolipoprotein A-I/apolipoprotein A-IMilano complexed with phospholipids, may be considered as a new therapeutic tool for the management of sepsis.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", via Balzaretti 9 - Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
2
|
Lu CH, Ma J, Lin MC, Wu CJ, Kuo CY, Chiang-Ni C, Kuo ML. Streptococcal pyrogenic exotoxin a induces regulatory T cells via TNF-α-TNFR2 signaling. Med Microbiol Immunol 2025; 214:25. [PMID: 40402252 DOI: 10.1007/s00430-025-00835-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025]
Abstract
Bacterial superantigens are potent immune activators that trigger T cell proliferation and intensive release of cytokines, leading to toxic shock syndrome. Also, they impair host immune responses, increasing bacterial carriage and transmission. Several studies proposed that superantigens can induce regulatory T (Treg) cells, which may suppress immune responses against bacterial infection. However, the mechanism of Treg cell induction by superantigens is still elusive. We here demonstrated that streptococcal pyrogenic exotoxin A (SPEA) promoted human CD4+CD25+Foxp3+ T cell induction in a dose- and time-dependent manner and the induction required antigen-presenting cells (APCs). SPEA-induced CD4+CD25+ T cells could suppress allogeneic T cell proliferation and IL-2 secretion. Flow cytometric analyses demonstrated high expression of TNFR2 on SPEA-induced CD4+CD25+Foxp3+ T cells. Blocking the interaction between TNF-⍺ and TNFR2 reduced SPEA-induced CD25+Foxp3+ Treg cells. Our present study suggests a mechanism that the TNF-⍺ and TNFR2 axis is required for the induction of human CD4+CD25+Foxp3+ Treg cells by SPEA, which implicates a potential strategy to enhance the clearance of Group A streptococcus infection through reducing Treg cell induction by the inhibition of TNFR2 signaling.
Collapse
Affiliation(s)
- Chun-Hao Lu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jason Ma
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
| | - Ming-Chieh Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Cheng-Jang Wu
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, USA
| | - Chieh-Ying Kuo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
| | - Chuan Chiang-Ni
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan.
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan.
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
3
|
Dufresne K, Tuffs SW, Walton NR, Kasper KJ, Mohorovic I, Hasan F, Bentall T, Heinrichs DE, Delport J, Mele TS, McCormick JK. Evidence that staphylococcal superantigens promote within-patient bacterial persistence following post-operative surgical site infection. Infect Immun 2025; 93:e0040724. [PMID: 39878522 PMCID: PMC11895439 DOI: 10.1128/iai.00407-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Staphylococcus aureus is a predominant cause of post-operative surgical site infections and persistent bacteremia. Here, we describe a patient who experienced three episodes of S. aureus infection over a period of 4 months following a total knee arthroplasty. The initial bloodstream isolate (SAB-0429) was a clonal complex 5 (CC5) and methicillin-resistant S. aureus (MRSA), whereas two subsequent isolates (SAB-0485 and SAB-0495) were CC5 isolates but methicillin-sensitive S. aureus. The two latter isolates harbored a plasmid encoding three superantigen genes that were not present in the primary MRSA isolate. SAB-0485 and SAB-0495 both expressed the plasmid-encoded staphylococcal enterotoxin R exotoxin and demonstrated increased superantigen activity compared with SAB-0429. Compared to SAB-0429, the latter isolates also demonstrated an increased bacterial burden in a mouse bacteremia model that was dependent on increased interferon-γ production. Curing of the plasmid from SAB-0485 reduced this virulence phenotype. These findings suggest that the superantigen exotoxins may provide a selective advantage in chronic post-surgical infections.IMPORTANCEIn this study, we investigated bacterial isolates from a patient who experienced three recurrent S. aureus infections over a 4 month period following total knee arthroplasty. Genomic and phenotypic characterization of these isolates revealed that they all belonged to clonal complex 5, yet the latter two strains contained an additional plasmid encoding superantigen exotoxins. Subsequent experimental infection experiments in mice demonstrated that the plasmid-encoded superantigens exacerbated bacteremia by promoting liver abscess formation. These experiments suggest that despite appropriate antibiotic therapy, bacterial superantigens may be able to promote persistent infection following post-surgery.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Stephen W. Tuffs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Nicholas R. Walton
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Katherine J. Kasper
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Ivor Mohorovic
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Farah Hasan
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Tracey Bentall
- Division of Critical Care Medicine, Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - David E. Heinrichs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Johan Delport
- Department of Pathology, University of Western Ontario, London, Ontario, Canada
| | - Tina S. Mele
- Division of Critical Care Medicine, Department of Medicine, University of Western Ontario, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, London Health Sciences Centre, University Hospital, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
4
|
Rahimi-Jamnani F, Moradi HR, Fateh A, Azizi M, Nazari F, Soezi M, Siadat SD. The Therapeutic Efficacy of MS473, a Fully Human Single-Chain Variable Fragment Targeting Staphylococcus aureus Toxic Shock Syndrome Toxin-1, in a D-Galactosamine-Sensitized Mouse Model of Lethal Shock. BMC Microbiol 2025; 25:95. [PMID: 40011833 PMCID: PMC11863401 DOI: 10.1186/s12866-025-03825-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/13/2025] [Indexed: 02/28/2025] Open
Abstract
Toxic shock syndrome toxin-1 (TSST-1), produced by Staphylococcus aureus, is one of the most potent superantigens involved in causing life-threatening TSS and contributes to the onset of some autoimmune diseases. To this end, we have previously identified a fully human single-chain variable fragment antibody (scFv), MS473, exhibiting high binding affinity and specificity for TSST-1 and demonstrating in vitro neutralization activity. In the present study, the therapeutic activity of MS473 was assessed in a D-galactosamine-sensitized mouse model of lethal shock. D-galactosamine-sensitized mice were injected with TSST-1 and then received a single dose of MS473 intraperitoneally (15 mg per kg of mouse body weight) after five minutes or intravenously (3 mg per kg of mouse body weight) after 10 min. The survival rate was examined for seven days. Furthermore, blood samples from different groups of mice were subjected to biochemical assessment, and their kidneys and livers were analyzed histopathologically 24 h after the toxin injection. The findings demonstrated a 100% survival rate with no significant damage to kidney and liver function in the treated groups, receiving MS473 through two different administration routes compared to the control groups, including the toxin-injected mice receiving normal saline or an unrelated scFv. Targeting disseminated TSST-1 with the scFv, which has appropriate permeability and distribution throughout the body, may be an effective way to alleviate the malfunctioning of the immune system caused by TSST-1.
Collapse
Affiliation(s)
- Fatemeh Rahimi-Jamnani
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Hamid Reza Moradi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Masoumeh Azizi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Farzaneh Nazari
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdieh Soezi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
5
|
Dufresne K, Al KF, Craig HC, Coleman CEM, Kasper KJ, Burton JP, McCormick JK. TSST-1 promotes colonization of Staphylococcus aureus within the vaginal tract by activation of CD8 + T cells. Infect Immun 2025; 93:e0043924. [PMID: 39840991 PMCID: PMC11834441 DOI: 10.1128/iai.00439-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/14/2024] [Indexed: 01/23/2025] Open
Abstract
Toxic shock syndrome toxin-1 (TSST-1) is a superantigen produced by Staphylococcus aureus and is the determinant of menstrual toxic shock syndrome (mTSS); however, the impact of TSST-1 on the vaginal environment beyond mTSS is not understood. Herein, we assessed how TSST-1 affects vaginal colonization by S. aureus, host inflammatory responses, and changes in microbial communities within the murine vagina. We demonstrated that TSST-1 induced a CD8+ T-cell-dependent inflammatory response in 24 h that correlated with S. aureus persistence within the vaginal tract. This increase was due to superantigen-dependent T-cell activation that triggered a change in microbial composition within the vaginal tract. Altogether, this study demonstrates that within the vaginal tract, TSST-1 modulates the vaginal microbiota to favor the survival of S. aureus in the absence of mTSS.IMPORTANCEToxic shock syndrome toxin-1 (TSST-1) is a superantigen toxin produced from Staphylococcus aureus that causes the menstrual form of toxic shock syndrome. This research demonstrates that TSST-1 also has a wider function within the vaginal tract than previously expected. We show that TSST-1, by activating CD8+ T cells, induces an inflammatory environment that modifies the vaginal microbiota to favor colonization by S. aureus. These are important findings as S. aureus can colonize the human vaginal tract efficiently and subsequently trigger dysbiosis within the microbial communities leading to several adverse outcomes such as decreased fertility, increased risks for sexually transmitted diseases, and issues related to pregnancy and birth.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Kait F. Al
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Heather C. Craig
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Charlotte E. M. Coleman
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Katherine J. Kasper
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Jeremy P. Burton
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
6
|
Thacharodi A, Hassan S, Vithlani A, Ahmed T, Kavish S, Geli Blacknell NM, Alqahtani A, Pugazhendhi A. The burden of group A Streptococcus (GAS) infections: The challenge continues in the twenty-first century. iScience 2025; 28:111677. [PMID: 39877071 PMCID: PMC11773489 DOI: 10.1016/j.isci.2024.111677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
Streptococcus pyogenes is a Gram-positive bacterium, also known as Group A Streptococcus (GAS), that has become a significant threat to the healthcare system, infecting more than 18 million people and resulting in more than 500,000 deaths annually worldwide. GAS infection rates decreased gradually during the 20th century in Western countries, largely due to improved living conditions and access to antibiotics. However, post-COVID-19, the situation has led to a steep increase in GAS infection rates in Europe, the United States, Australia, and New Zealand, which triggers a global concern. GAS infections are normally moderate, with symptoms of fever, pharyngitis, and pyoderma; nevertheless, if left untreated or with continued exposure to GAS or with recurring infections it can result in fatal outcomes. GAS produces a variety of virulence factors and exotoxins that can lead to deadly infections such as necrotizing fasciitis, impetigo, cellulitis, pneumonia, empyema, streptococcal toxic shock syndrome, bacteremia, and puerperal sepsis. In addition, post-immune mediated disorders such as post-streptococcal glomerulonephritis, acute rheumatic fever, and rheumatic heart disease contribute to extremely high death rates in developing nations. Despite substantial research on GAS infections, it is still unclear what molecular pathways are responsible for their emergence and how to best manage them. This review thus provides insights into the most recent research on the pathogenesis, virulence, resistance, and host interaction mechanisms of GAS, as well as novel management options to assist scientific communities in combating GAS infections.
Collapse
Affiliation(s)
- Aswin Thacharodi
- Dr. Thacharodi’s Laboratories, Department of Research and Development, Puducherry 605005, India
| | - Saqib Hassan
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu 600119, India
- Future Leaders Mentoring Fellow, American Society for Microbiology, Washington 20036, USA
| | - Avadh Vithlani
- Senior Resident, Department of Pulmonary Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Tawfeeq Ahmed
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu 600119, India
| | - Sanjana Kavish
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu 600119, India
| | | | - Ali Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Arivalagan Pugazhendhi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Engineering & Technology, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
7
|
Zhang H, Monk IR, Braverman J, Jones CM, Brooks AG, Stinear TP, Wakim LM. Staphylococcal superantigens evoke temporary and reversible T cell anergy, but fail to block the development of a bacterium specific cellular immune response. Nat Commun 2024; 15:9872. [PMID: 39543088 PMCID: PMC11564628 DOI: 10.1038/s41467-024-54074-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Superantigens (sAgs) are bacterial virulence factors that induce a state of immune hyperactivation by forming a bridge between certain subsets of T cell receptor (TCR) β chains on T lymphocytes, and class II major histocompatibility complex (MHC-II) molecules; this cross-linking leads to indiscriminate T cell activation, cytokine storm and toxic shock. Here we show that sAg exposure drives the preferential expansion of naive and central memory T cell subsets, but not effector or resident memory T cells, which instead, hyper release pro-inflammatory cytokines. A targeted therapeutic approach to minimise cytokine release by effector memory T cells attenuated sAg-induced cytokine release. Irrespective of antigen experience, sAg activation does not render mature T cells permanently dysfunctional, and full restoration of effector function is observed following a transient and reversible anergy. Moreover, we show that in the face of sAg induced immune hyperactivation, an intact bacterium-specific CD4+ T cell response can be mounted.
Collapse
Affiliation(s)
- Heran Zhang
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Ian R Monk
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Jessica Braverman
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Claerwen M Jones
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
8
|
Piechowicz L, Kosznik-Kwaśnicka K, Jarzembowski T, Daca A, Necel A, Bonawenturczak A, Werbowy O, Stasiłojć M, Pałubicka A. Staphylococcus aureus Co-Infection in COVID-19 Patients: Virulence Genes and Their Influence on Respiratory Epithelial Cells in Light of Risk of Severe Secondary Infection. Int J Mol Sci 2024; 25:10050. [PMID: 39337536 PMCID: PMC11431965 DOI: 10.3390/ijms251810050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Pandemics from viral respiratory tract infections in the 20th and early 21st centuries were associated with high mortality, which was not always caused by a primary viral infection. It has been observed that severe course of infection, complications and mortality were often the result of co-infection with other pathogens, especially Staphylococcus aureus. During the COVID-19 pandemic, it was also noticed that patients infected with S. aureus had a significantly higher mortality rate (61.7%) compared to patients infected with SARS-CoV-2 alone. Our previous studies have shown that S. aureus strains isolated from patients with COVID-19 had a different protein profile than the strains in non-COVID-19 patients. Therefore, this study aims to analyze S. aureus strains isolated from COVID-19 patients in terms of their pathogenicity by analyzing their virulence genes, adhesion, cytotoxicity and penetration to the human pulmonary epithelial cell line A549. We have observed that half of the tested S. aureus strains isolated from patients with COVID-19 had a necrotizing effect on the A549 cells. The strains also showed greater variability in terms of their adhesion to the human cells than their non-COVID-19 counterparts.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Tomasz Jarzembowski
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Agnieszka Daca
- Department of Physiopathology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Ada Bonawenturczak
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland
| | - Anna Pałubicka
- Specialist Hospital in Koscierzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Koscierzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland
| |
Collapse
|
9
|
Inada M, Iwamoto N, Nomoto H, Tsuzuki S, Takemoto N, Fuwa N, Moriya A, Ohmagari N. Characteristics of Streptococcal Toxic Shock Syndrome Caused by Different Beta-hemolytic Streptococci Species: A Single-center Retrospective Study. Open Forum Infect Dis 2024; 11:ofae486. [PMID: 39296344 PMCID: PMC11409875 DOI: 10.1093/ofid/ofae486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Background Streptococcal toxic shock syndrome (STSS) is a life-threatening condition caused by beta-hemolytic streptococci (BHS). Streptococcus pyogenes is the main causative agent of this disease; other BHS such as Streptococcus agalactiae or Streptococcus dysgalactiae could also cause STSS. However, the clinical characteristics of STSS caused by other types of BHS remain poorly understood. In this study, we evaluated the likelihood of STSS development in various streptococcal species. Methods We conducted a retrospective observational study using adult medical records of patients with invasive BHS in a tertiary care institution from 2002 to 2022 and classified them into STSS or non-STSS groups. Multivariable analysis of bacterial species adjusted for age and diabetes mellitus was conducted. S pyogenes cases were propensity-matched (1:4) to non-pyogenes BHS cases. Results A total of 43 STSS and 285 non-STSS cases were identified. S pyogenes, S agalactiae, and S dysgalactiae accounted for 17, 13, and 13 STSS cases, respectively. The crude mortality of STSS was approximately 35% in all groups. A multivariable analysis suggested that STSS was less frequent in S agalactiae and S dysgalactiae cases with odds ratio 0.24 (95% confidence interval [CI], 0.10-0.54; P < .001) and 0.23 (95% CI, .10-.55; P < .001), respectively. Propensity score matching showed that S pyogenes caused STSS more frequently than other BHS cases with an odds ratio of 3.28 (95% CI 1.21-8.77; P = .010). Conclusions This study described and compared the clinical characteristics of STSS caused by different BHS. We demonstrated that S pyogenes caused STSS more often than other BHS.
Collapse
Affiliation(s)
- Makoto Inada
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Noriko Iwamoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hidetoshi Nomoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinya Tsuzuki
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiko Takemoto
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Noriko Fuwa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ataru Moriya
- Laboratory Testing Department, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
10
|
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
|
11
|
Auger N, Carrier FM, Waechter J, Brousseau É, Maniraho A, Ayoub A, Bégin P. Long-term outcomes of patients with toxic shock syndrome: A matched cohort study. J Infect 2024; 89:106213. [PMID: 38944286 DOI: 10.1016/j.jinf.2024.106213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/12/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024]
Abstract
OBJECTIVES We examined long-term outcomes of toxic shock syndrome. METHODS We conducted a matched cohort study of 630 patients with toxic shock syndrome and 5009 healthy controls between 2006 and 2021 in Quebec, Canada. Outcomes included hospitalization for renal, cardiovascular, hepatic, and other morbidity during 15 years of follow-up. We estimated adjusted hazard ratios (HR) and 95% confidence intervals (CI) for the risk of these outcomes over time, comparing patients with toxic shock syndrome relative to matched controls. RESULTS Compared with healthy controls, rehospitalization rates at 15 years were higher for men with toxic shock syndrome (52.0 vs 30.0 per 100) but not for women (38.7 vs 45.6 per 100). In men, toxic shock syndrome was associated with an elevated risk of renal (HR 17.43, 95% CI 6.35-47.82), cardiovascular (HR 2.57; 95% CI 1.52-4.34), and hepatic hospitalization (HR 19.83, 95% CI 4.72-83.34). In women, toxic shock syndrome was associated with renal hospitalization (HR 4.71, 95% CI 1.94-11.45). Streptococcal toxic shock was associated with a greater risk of rehospitalization than staphylococcal toxic shock. CONCLUSIONS Toxic shock syndrome is associated with rehospitalization up to 15 years later, especially in men. These patients may benefit from continued follow-up to prevent long-term morbidity.
Collapse
Affiliation(s)
- Nathalie Auger
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Institut national de santé publique du Québec, Montreal, Quebec, Canada; Department of Social and Preventive Medicine, School of Public Health, University of Montreal, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada.
| | - François M Carrier
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Departments of Anesthesiology and Medicine, Critical Care Division, University of Montreal Hospital Centre, Montreal, Quebec, Canada
| | - Jason Waechter
- Department of Critical Care Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Émilie Brousseau
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Institut national de santé publique du Québec, Montreal, Quebec, Canada
| | - Amanda Maniraho
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Institut national de santé publique du Québec, Montreal, Quebec, Canada
| | - Aimina Ayoub
- University of Montreal Hospital Research Centre, Montreal, Quebec, Canada; Institut national de santé publique du Québec, Montreal, Quebec, Canada
| | - Philippe Bégin
- Sainte-Justine Hospital Research Centre, Montreal, Quebec, Canada; Department of Clinical Immunology, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Dufresne K, DiMaggio DA, Maduta CS, Brinsmade SR, McCormick JK. Discovery of an antivirulence compound that targets the Staphylococcus aureus SaeRS two-component system to inhibit toxic shock syndrome toxin-1 production. J Biol Chem 2024; 300:107455. [PMID: 38852884 PMCID: PMC11328871 DOI: 10.1016/j.jbc.2024.107455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/11/2024] Open
Abstract
Menstrual toxic shock syndrome (mTSS) is a rare but severe disorder associated with the use of menstrual products such as high-absorbency tampons and is caused by Staphylococcus aureus strains that produce the toxic shock syndrome toxin-1 (TSST-1) superantigen. Herein, we screened a library of 3920 small bioactive molecules for the ability to inhibit transcription of the TSST-1 gene without inhibiting the growth of S. aureus. The dominant positive regulator of TSST-1 is the SaeRS two-component system (TCS), and we identified phenazopyridine hydrochloride (PP-HCl) that repressed the production of TSST-1 by inhibiting the kinase function of SaeS. PP-HCl competed with ATP for binding of the kinase SaeS leading to decreased phosphorylation of SaeR and reduced expression of TSST-1 as well as several other secreted virulence factors known to be regulated by SaeRS. PP-HCl targets the virulence of S. aureus, and it also decreases the impact of TSST-1 on human lymphocytes without affecting the healthy vaginal microbiota. Our findings demonstrate the promising potential of PP-HCl as a therapeutic strategy against mTSS.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Dennis A DiMaggio
- Department of Biology, Georgetown University, Washington, District of Columbia, USA
| | - Carla S Maduta
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Shaun R Brinsmade
- Department of Biology, Georgetown University, Washington, District of Columbia, USA
| | - John K McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
13
|
Bak YS, Park JY, Kim JB, Cho SH. Molecular characterization and antibiotic resistance of Staphylococcus aureus strains isolated from patients with diarrhea in Korea between the years 2007 and 2022. Food Sci Biotechnol 2024; 33:1965-1974. [PMID: 38752118 PMCID: PMC11091020 DOI: 10.1007/s10068-023-01478-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 05/18/2024] Open
Abstract
To investigate the molecular characteristics and antibiotic resistance of Staphylococcus aureus isolates from patients with diarrhea in Korea, 327 S. aureus strains were collected between 2007 and 2022. The presence of staphylococcal enterotoxin (SE) and toxic shock syndrome toxin-1 (TSST-1) genes in S. aureus isolates was determined by PCR. The highest expression of the TSST-1 gene was found in the GIMNO type (43.1% of GIMNO type). GIMNO type (Type I) refers to each staphylococcal enterotoxin (SE) gene gene (initials of genes): G = seg; I = sei; M = selm; N = seln; O = selo. Moreover, Type I isolates showed a significantly higher resistance to most antibiotics. A total of 195 GIMNO-type S. aureus strains were analyzed using multilocus sequence typing (MLST), and 18 unique sequence types (STs) were identified. The most frequent sequence type was ST72 (36.9%), followed by ST5 (22.1%) and ST30 (16.9%). Interestingly, ST72 strains showed a higher prevalence of MRSA than the other STs. In conclusion, our results were the first reported for S. aureus strains in Korea, which significantly expanded S. aureus genotype information for the surveillance of pathogenic S. aureus and may provide important epidemiological information to resolve several infectious diseases caused by S. aureus. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01478-9.
Collapse
Affiliation(s)
- Young-Seok Bak
- Department of Emergency Medical Services, Sun Moon University, Asan-si, Chungcheongnam-do 31460 Korea
| | - Jun-Young Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jung-Beom Kim
- Department of Food Science and Technology, Suncheon National University, Suncheon, 57922 Korea
| | - Seung-Hak Cho
- Department of Emergency Medical Services, Sun Moon University, Asan-si, Chungcheongnam-do 31460 Korea
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
- Division of Bacterial Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Heungdeok-Gu, Cheongju, 363-951 Republic of Korea
| |
Collapse
|
14
|
Tuffs SW, Dufresne K, Rishi A, Walton NR, McCormick JK. Novel insights into the immune response to bacterial T cell superantigens. Nat Rev Immunol 2024; 24:417-434. [PMID: 38225276 DOI: 10.1038/s41577-023-00979-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Bacterial T cell superantigens (SAgs) are a family of microbial exotoxins that function to activate large numbers of T cells simultaneously. SAgs activate T cells by direct binding and crosslinking of the lateral regions of MHC class II molecules on antigen-presenting cells with T cell receptors (TCRs) on T cells; these interactions alter the normal TCR-peptide-MHC class II architecture to activate T cells in a manner that is independent of the antigen specificity of the TCR. SAgs have well-recognized, central roles in human diseases such as toxic shock syndrome and scarlet fever through their quantitative effects on the T cell response; in addition, numerous other consequences of SAg-driven T cell activation are now being recognized, including direct roles in the pathogenesis of endocarditis, bloodstream infections, skin disease and pharyngitis. In this Review, we summarize the expanding family of bacterial SAgs and how these toxins can engage highly diverse adaptive immune receptors. We highlight recent findings regarding how SAg-driven manipulation of the adaptive immune response may operate in multiple human diseases, as well as contributing to the biology and life cycle of SAg-producing bacterial pathogens.
Collapse
Affiliation(s)
- Stephen W Tuffs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Aanchal Rishi
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Nicholas R Walton
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - John K McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
15
|
Zhang Z, Kean IRL, Dratva LM, Clark JA, Syrimi E, Khan N, Daubney E, White D, O'Neill L, Chisholm C, Payne C, Benkenstein S, Kupiec K, Galassini R, Wright V, Winmill H, Robbins C, Brown K, Ramnarayan P, Scholefield B, Peters M, Klein N, Montgomery H, Meyer KB, Teichmann SA, Bryant C, Taylor G, Pathan N. Enhanced CD95 and interleukin 18 signalling accompany T cell receptor Vβ21.3+ activation in multi-inflammatory syndrome in children. Nat Commun 2024; 15:4227. [PMID: 38762592 PMCID: PMC11102542 DOI: 10.1038/s41467-024-48699-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Multisystem inflammatory syndrome in children is a post-infectious presentation SARS-CoV-2 associated with expansion of the T cell receptor Vβ21.3+ T-cell subgroup. Here we apply muti-single cell omics to compare the inflammatory process in children with acute respiratory COVID-19 and those presenting with non SARS-CoV-2 infections in children. Here we show that in Multi-Inflammatory Syndrome in Children (MIS-C), the natural killer cell and monocyte population demonstrate heightened CD95 (Fas) and Interleuking 18 receptor expression. Additionally, TCR Vβ21.3+ CD4+ T-cells exhibit skewed differentiation towards T helper 1, 17 and regulatory T cells, with increased expression of the co-stimulation receptors ICOS, CD28 and interleukin 18 receptor. We observe no functional evidence for NLRP3 inflammasome pathway overactivation, though MIS-C monocytes show elevated active caspase 8. This, coupled with raised IL18 mRNA expression in CD16- NK cells on single cell RNA sequencing analysis, suggests interleukin 18 and CD95 signalling may trigger activation of TCR Vβ21.3+ T-cells in MIS-C, driven by increased IL-18 production from activated monocytes and CD16- Natural Killer cells.
Collapse
MESH Headings
- Humans
- Interleukin-18/metabolism
- Child
- Signal Transduction
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- fas Receptor/metabolism
- fas Receptor/genetics
- Monocytes/immunology
- Monocytes/metabolism
- Systemic Inflammatory Response Syndrome/immunology
- Systemic Inflammatory Response Syndrome/metabolism
- COVID-19/immunology
- COVID-19/virology
- COVID-19/metabolism
- COVID-19/complications
- Inflammasomes/metabolism
- Inflammasomes/immunology
- SARS-CoV-2/immunology
- Adolescent
- Male
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Female
- Child, Preschool
- Single-Cell Analysis
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD28 Antigens/metabolism
- Lymphocyte Activation/immunology
- Receptors, Interleukin-18/metabolism
- Receptors, Interleukin-18/genetics
- Receptors, Interleukin-18/immunology
Collapse
Affiliation(s)
- Zhenguang Zhang
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Iain R L Kean
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Lisa M Dratva
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - John A Clark
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Eleni Syrimi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Naeem Khan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Esther Daubney
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK
| | - Deborah White
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK
| | - Lauran O'Neill
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Catherine Chisholm
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Caroline Payne
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Sarah Benkenstein
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Klaudia Kupiec
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | | | - Victoria Wright
- Department of Paediatrics, Imperial College London, London, UK
| | - Helen Winmill
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
| | - Ceri Robbins
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
| | - Katherine Brown
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | | | - Barnaby Scholefield
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Mark Peters
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
- Departments of Paediatrics, University College London, London, UK
| | - Nigel Klein
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
- Departments of Paediatrics, University College London, London, UK
| | | | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Theory of Condensed Matter, Cavendish Laboratory, Department of Physics University of Cambridge, Cambridge, UK
| | - Clare Bryant
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Graham Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Nazima Pathan
- Departments of Paediatrics, University of Cambridge, Cambridge, UK.
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK.
| |
Collapse
|
16
|
Sarkar R, Yadav A, Maheshwari A. Fever with Rash in a Child: Revisited. Indian J Dermatol 2024; 69:282. [PMID: 39119300 PMCID: PMC11305486 DOI: 10.4103/ijd.ijd_913_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Rash and fever are some of the most common chief complaints present in paediatric dermatology emergencies. The spectrum of differential diagnosis is broad, including many different infectious and some non-infectious agents. A systematic approach involving detailed history taking, careful clinical examination along with particular attention to epidemiological features are the most important factors to make a diagnosis. This article reviews the morphological patterns of various causes of fever with rash in children, including infectious as well as non-infectious causes, with special emphasis on the Indian scenario. We intend to highlight the clinical characteristics of each cause, which will not only help make a clinical diagnosis but also distinguish benign versus life-threatening causes of skin rash in febrile paediatric patients and provide early medical intervention.
Collapse
Affiliation(s)
- Rashmi Sarkar
- From the Department of Dermatology, Lady Hardinge Medical College, New Delhi, India
| | - Anukriti Yadav
- From the Department of Dermatology, Lady Hardinge Medical College, New Delhi, India
| | - Apoorva Maheshwari
- From the Department of Dermatology, Lady Hardinge Medical College, New Delhi, India
| |
Collapse
|
17
|
Sun YK, Wang C, Lin PQ, Hu L, Ye J, Gao ZG, Lin R, Li HM, Shu Q, Huang LS, Tan LH. Severe pediatric COVID-19: a review from the clinical and immunopathophysiological perspectives. World J Pediatr 2024; 20:307-324. [PMID: 38321331 PMCID: PMC11052880 DOI: 10.1007/s12519-023-00790-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/14/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) tends to have mild presentations in children. However, severe and critical cases do arise in the pediatric population with debilitating systemic impacts and can be fatal at times, meriting further attention from clinicians. Meanwhile, the intricate interactions between the pathogen virulence factors and host defense mechanisms are believed to play indispensable roles in severe COVID-19 pathophysiology but remain incompletely understood. DATA SOURCES A comprehensive literature review was conducted for pertinent publications by reviewers independently using the PubMed, Embase, and Wanfang databases. Searched keywords included "COVID-19 in children", "severe pediatric COVID-19", and "critical illness in children with COVID-19". RESULTS Risks of developing severe COVID-19 in children escalate with increasing numbers of co-morbidities and an unvaccinated status. Acute respiratory distress stress and necrotizing pneumonia are prominent pulmonary manifestations, while various forms of cardiovascular and neurological involvement may also be seen. Multiple immunological processes are implicated in the host response to COVID-19 including the type I interferon and inflammasome pathways, whose dysregulation in severe and critical diseases translates into adverse clinical manifestations. Multisystem inflammatory syndrome in children (MIS-C), a potentially life-threatening immune-mediated condition chronologically associated with COVID-19 exposure, denotes another scientific and clinical conundrum that exemplifies the complexity of pediatric immunity. Despite the considerable dissimilarities between the pediatric and adult immune systems, clinical trials dedicated to children are lacking and current management recommendations are largely adapted from adult guidelines. CONCLUSIONS Severe pediatric COVID-19 can affect multiple organ systems. The dysregulated immune pathways in severe COVID-19 shape the disease course, epitomize the vast functional diversity of the pediatric immune system and highlight the immunophenotypical differences between children and adults. Consequently, further research may be warranted to adequately address them in pediatric-specific clinical practice guidelines.
Collapse
Affiliation(s)
- Yi-Kan Sun
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310030, China
| | - Can Wang
- Surgical Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Pei-Quan Lin
- Surgical Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Lei Hu
- Surgical Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Jing Ye
- Surgical Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Zhi-Gang Gao
- Department of General Surgery, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Ru Lin
- Department of Cardiopulmonary and Extracorporeal Life Support, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Hao-Min Li
- Clinical Data Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Qiang Shu
- Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
- National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Li-Su Huang
- National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
- Department of Infectious Diseases, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| | - Lin-Hua Tan
- Surgical Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
18
|
Maduta CS, McCormick JK, Dufresne K. Vaginal community state types (CSTs) alter environmental cues and production of the Staphylococcus aureus toxic shock syndrome toxin-1 (TSST-1). J Bacteriol 2024; 206:e0044723. [PMID: 38334326 PMCID: PMC10955855 DOI: 10.1128/jb.00447-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/20/2024] [Indexed: 02/10/2024] Open
Abstract
Menstrual toxic shock syndrome (mTSS) is a rare but life-threatening disease associated with the use of high-absorbency tampons. The production of the Staphylococcus aureus toxic shock syndrome toxin-1 (TSST-1) superantigen is involved in nearly all cases of mTSS and is tightly controlled by regulators responding to the environment. In the prototypic mTSS strain S. aureus MN8, the major repressor of TSST-1 is the carbon catabolite protein A (CcpA), which responds to glucose concentrations in the vaginal tract. Healthy vaginal Lactobacillus species also depend on glucose for both growth and acidification of the vaginal environment through lactic acid production. We hypothesized that interactions between the vaginal microbiota [herein referred to as community state types (CSTs)] and S. aureus MN8 depend on environmental cues and that these interactions subsequently affect TSST-1 production. Using S. aureus MN8 ΔccpA growing in various glucose concentrations, we demonstrate that the supernatants from different CSTs grown in vaginally defined medium (VDM) could significantly decrease tst expression. When co-culturing CST species with MN8 ∆ccpA, we show that Lactobacillus jensenii completely inhibits TSST-1 production in conditions mimicking healthy menstruation or mTSS. Finally, we show that growing S. aureus in "unhealthy" or "transitional" CST supernatants results in higher interleukin 2 (IL-2) production from T cells. These findings suggest that dysbiotic CSTs may encourage TSST-1 production in the vaginal tract and further indicate that the CSTs are likely important for the protection from mTSS.IMPORTANCEIn this study, we investigate the impact of the vaginal microbiota against Staphylococcus aureus in conditions mimicking the vaginal environment at various stages of the menstrual cycle. We demonstrate that Lactobacillus jensenii can inhibit toxic shock syndrome toxin-1 (TSST-1) production, suggesting the potential for probiotic activity in treating and preventing menstrual toxic shock syndrome (mTSS). On the other side of the spectrum, "unhealthy" or "transient" bacteria such as Gardnerella vaginalis and Lactobacillus iners support more TSST-1 production by S. aureus, suggesting that community state types are important in the development of mTSS. This study sets forward a model for examining contact-independent interactions between pathogenic bacteria and the vaginal microbiota. It also demonstrates the necessity of replicating the environment when studying one as dynamic as the vagina.
Collapse
Affiliation(s)
- Carla S. Maduta
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
19
|
Ohashi A, Murayama MA, Miyabe Y, Yudoh K, Miyabe C. Streptococcal infection and autoimmune diseases. Front Immunol 2024; 15:1361123. [PMID: 38464518 PMCID: PMC10920276 DOI: 10.3389/fimmu.2024.1361123] [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: 12/25/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Excessive activation of immune cells by environmental factors, such as infection or individual genetic risk, causes various autoimmune diseases. Streptococcus species are gram-positive bacteria that colonize the nasopharynx, respiratory tract, gastrointestinal tract, genitourinary tract, and skin. Group A Streptococcus (GAS) species cause various symptoms, ranging from mild infections, such as tonsillitis and pharyngitis, to serious infections, such as necrotizing fasciitis and streptococcal toxic shock syndrome. The contribution of GAS infections to several autoimmune diseases, including acute rheumatic fever, vasculitis, and neuropsychiatric disorders, has been studied. In this review, we focus on the association between streptococcal infections and autoimmune diseases, and discuss current research on the mechanisms underlying the initiation and progression of autoimmune diseases.
Collapse
Affiliation(s)
- Ayaka Ohashi
- Department of Immunology and Parasitology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Masanori A. Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Yoshishige Miyabe
- Department of Immunology and Parasitology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kazuo Yudoh
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Chie Miyabe
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
20
|
Wang P, Fredj Z, Zhang H, Rong G, Bian S, Sawan M. Blocking Superantigen-Mediated Diseases: Challenges and Future Trends. J Immunol Res 2024; 2024:2313062. [PMID: 38268531 PMCID: PMC10807946 DOI: 10.1155/2024/2313062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/15/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024] Open
Abstract
Superantigens are virulence factors secreted by microorganisms that can cause various immune diseases, such as overactivating the immune system, resulting in cytokine storms, rheumatoid arthritis, and multiple sclerosis. Some studies have demonstrated that superantigens do not require intracellular processing and instated bind as intact proteins to the antigen-binding groove of major histocompatibility complex II on antigen-presenting cells, resulting in the activation of T cells with different T-cell receptor Vβ and subsequent overstimulation. To combat superantigen-mediated diseases, researchers have employed different approaches, such as antibodies and simulated peptides. However, due to the complex nature of superantigens, these approaches have not been entirely successful in achieving optimal therapeutic outcomes. CD28 interacts with members of the B7 molecule family to activate T cells. Its mimicking peptide has been suggested as a potential candidate to block superantigens, but it can lead to reduced T-cell activity while increasing the host's infection risk. Thus, this review focuses on the use of drug delivery methods to accurately target and block superantigens, while reducing the adverse effects associated with CD28 mimic peptides. We believe that this method has the potential to provide an effective and safe therapeutic strategy for superantigen-mediated diseases.
Collapse
Affiliation(s)
- Pengbo Wang
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Zina Fredj
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Hongyong Zhang
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Guoguang Rong
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Sumin Bian
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Mohamad Sawan
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| |
Collapse
|
21
|
Cascais I, Losa A, Correia C, Pinto DH, Meireles D, Sarmento A. Toxic Shock Syndrome: Eighteen Years of Experience in a Pediatric Intensive Care Unit. Cureus 2024; 16:e52898. [PMID: 38406070 PMCID: PMC10891456 DOI: 10.7759/cureus.52898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Toxic shock syndrome (TSS) is a life-threatening disease usually caused by a Staphylococcus aureus or group Aβ-hemolytic Streptococcus infection. Methods In this retrospective study, we included patients with TSS admitted to a tertiary hospital's pediatric intensive care unit (PICU) over the last 18 years. We compared the staphylococcal TSS (Staph-TSS) and streptococcal TSS (Strep-TSS) groups. Results We included 17 patients (64.7% male), with a median age of 6.1 years (3.0 years for streptococcal TSS versus 13.3 years for staphylococcal TSS, p = 0.040), a median of 3.0 days from symptom onset to diagnosis, and a median of 6.0 days of hospitalization. Ten patients met the Centers for Disease Control and Prevention (CDC) criteria for staphylococcal TSS (one menstrual-related) and seven met the criteria for streptococcal TSS (four of them occurring since the COVID-19 pandemic was declared). Fifteen patients had identified risk factors, primarily cutaneous lesions (29.4%). In 15 patients, at least three organs or systems were affected, with fever, rash, and hypotension as universal findings. Mucous membrane hyperemia was present in 16 patients, gastrointestinal symptoms in 14 patients, and desquamation in nine. Muscular involvement was present in seven patients, all with staphylococcal TSS (p = 0.010). All patients received two or more antibiotics, including a protein synthesis inhibitor (except for one), and required fluid resuscitation and vasoactive amines (median three days). Six patients needed invasive mechanical ventilation (median seven days). Albumin infusion was necessary in six patients, significantly more frequently in patients with streptococcal TSS (p = 0.035). Two patients with staphylococcal TSS died, while the seven patients with streptococcal TSS survived hospital discharge. There were no recurrent cases. Conclusions Our study revealed TSS severity and multiorgan involvement, emphasizing the importance of early diagnosis and intervention. Risk factors were prevalent, and we noted an increased frequency of group A streptococcal (GAS) TSS post-COVID-19 pandemic.
Collapse
Affiliation(s)
- Inês Cascais
- Department of Pediatrics, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário de Santo António, Porto, PRT
| | - Ana Losa
- Department of Pediatrics, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário de Santo António, Porto, PRT
| | - Cláudia Correia
- Department of Pediatrics, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário de Santo António, Porto, PRT
| | - Diana H Pinto
- Department of Pediatrics, Centro Hospitalar Entre Douro e Vouga, Santa Maria da Feira, PRT
| | - Daniel Meireles
- Department of Pediatric Intensive Care, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário de Santo António, Porto, PRT
| | - Alzira Sarmento
- Department of Pediatric Intensive Care, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário de Santo António, Porto, PRT
| |
Collapse
|
22
|
Tapp K, Henson A. "For men, by men": Menstrual victimization and the weaponization of period products in carceral settings. WOMEN'S HEALTH (LONDON, ENGLAND) 2024; 20:17455057241240931. [PMID: 38554068 PMCID: PMC10981853 DOI: 10.1177/17455057241240931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/20/2024] [Accepted: 03/01/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND The literature on menstruation defines period poverty as the inability to access sufficient period products, education, and sanitary facilities needed to manage menstruation healthily and effectively. While research has identified shortcomings of healthcare in the carceral setting, period poverty behind bars has remained largely absent from criminal legal discourse. OBJECTIVES The current study examines the interplay of period poverty and carceral control to introduce the novel concept of menstrual victimization, defined as the physical, emotional, and financial victimization that results from period poverty perpetuated through carceral control. METHODS The study uses qualitative content analysis to systematically gather and code journalistic accounts pertaining to the menstrual experiences of incarcerated and previously incarcerated females, criminal justice practitioners, and journalists. The analysis uses literary pieces (n = 99), which were coded deductively and guided by concepts related to structural violence and radical feminist criminology. RESULTS The findings shed light on the unique structural harms incarcerated menstruators face and reveal the dearth of needed empirical research on period poverty in carceral spaces. The narratives in the sample revealed how manufactured scarcity of period products within carceral spaces is used as a means of oppression by institutional agents. The emergent themes highlight how the intersection of period poverty and carceral control led to menstrual victimization characterized through shame, humiliation, control, and coercion. CONCLUSION Potential outcomes associated with understanding menstrual victimization in the carceral setting are discussed, including reducing menstrual stigma, disseminating health education, minimizing health disparities, and ultimately, shifting modes of holding accountability away from oppressive, retributive, and controlling tactics.
Collapse
Affiliation(s)
- Kathryn Tapp
- School of Criminology and Criminal Justice, Arizona State University, Phoenix, AZ, USA
| | - Abigail Henson
- School of Criminology and Criminal Justice, Arizona State University, Phoenix, AZ, USA
| |
Collapse
|
23
|
Nakajima I, Fukuda K, Ishida W, Kishimoto T, Kuwana A, Suzuki T, Kaito C, Yamashiro K. Staphylococcus aureus-derived virulent phenol-soluble modulin α triggers alarmin release to drive IL-36-dependent corneal inflammation. Microbes Infect 2024; 26:105237. [PMID: 37805122 DOI: 10.1016/j.micinf.2023.105237] [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: 01/10/2023] [Revised: 08/22/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) isolated from patients with keratitis produces substantial amounts of phenol-soluble modulin α (PSMα). However, the role of PSMα in S. aureus keratitis remains unclear. We observed that PSMα-producing and PSMα-deficient strains could infect the cornea in our experimental mouse keratitis model; however, only the PSMα-producing strain delayed epithelial wound healing and induced stromal inflammation. PSMα induced damage to the epithelium, the release of alarmins IL-1α and IL-36α, and the expression of inflammatory chemokines by resident corneal cells in the mouse corneal organ culture. The IL-36 (but not IL-1) receptor antagonist attenuated mouse keratitis induced by PSMα-containing bacterial culture supernatants, as well as by infection with PSMα-producing S. aureus, suggesting that the corneal inflammations were dependent on IL-36. Recombinant PSMα elicited IL-36-dependent corneal inflammation in mice. Thus, PSMα and the subsequently released IL-36 are critical factors triggering inflammation during S. aureus keratitis.
Collapse
Affiliation(s)
- Isana Nakajima
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Kochi, Japan
| | - Ken Fukuda
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Kochi, Japan.
| | - Waka Ishida
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Kochi, Japan
| | - Tatsuma Kishimoto
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Kochi, Japan
| | - Aozora Kuwana
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Kochi, Japan
| | - Takashi Suzuki
- Department of Ophthalmology, Toho University, Tokyo, Japan
| | - Chikara Kaito
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Kochi, Japan
| |
Collapse
|
24
|
Zhu Z, Hu Z, Li S, Fang R, Ono HK, Hu DL. Molecular Characteristics and Pathogenicity of Staphylococcus aureus Exotoxins. Int J Mol Sci 2023; 25:395. [PMID: 38203566 PMCID: PMC10778951 DOI: 10.3390/ijms25010395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/24/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Staphylococcus aureus stands as one of the most pervasive pathogens given its morbidity and mortality worldwide due to its roles as an infectious agent that causes a wide variety of diseases ranging from moderately severe skin infections to fatal pneumonia and sepsis. S. aureus produces a variety of exotoxins that serve as important virulence factors in S. aureus-related infectious diseases and food poisoning in both humans and animals. For example, staphylococcal enterotoxins (SEs) produced by S. aureus induce staphylococcal foodborne poisoning; toxic shock syndrome toxin-1 (TSST-1), as a typical superantigen, induces toxic shock syndrome; hemolysins induce cell damage in erythrocytes and leukocytes; and exfoliative toxin induces staphylococcal skin scalded syndrome. Recently, Panton-Valentine leucocidin, a cytotoxin produced by community-associated methicillin-resistant S. aureus (CA-MRSA), has been reported, and new types of SEs and staphylococcal enterotoxin-like toxins (SEls) were discovered and reported successively. This review addresses the progress of and novel insights into the molecular structure, biological activities, and pathogenicity of both the classic and the newly identified exotoxins produced by S. aureus.
Collapse
Affiliation(s)
- Zhihao Zhu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Zuo Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Shaowen Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Rendong Fang
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China;
| | - Hisaya K. Ono
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Dong-Liang Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| |
Collapse
|
25
|
Schlievert PM, Nelson JD, Kilgore SH, Radoshevich L, Klingelhutz AJ, Leung DYM. Purification, characterization, and cloning of a novel pro-inflammatory secreted protein from Staphylococcus aureus. Microbiol Spectr 2023; 11:e0289823. [PMID: 37937984 PMCID: PMC10715055 DOI: 10.1128/spectrum.02898-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
IMPORTANCE Staphylococcus aureus causes a myriad of human diseases, ranging from relatively mild soft tissue infections to highly fatal pneumonia, sepsis, and toxic shock syndrome. The organisms primarily cause diseases across mucosal and skin barriers. In order to facilitate penetration of barriers, S. aureus causes harmful inflammation by inducing chemokines from epithelial cells. We report the cloning and characterization of a novel secreted S. aureus protein that induces chemokine production from epithelial cells as its major demonstrable function. This secreted protein possibly helps S. aureus and its secreted proteins to penetrate host barriers.
Collapse
Affiliation(s)
- Patrick M. Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jacob D. Nelson
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Samuel H. Kilgore
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Aloysius J. Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
26
|
Wang Y, Yang L, Xu G. New-Onset Acute Interstitial Nephritis Post-SARS-CoV-2 Infection and COVID-19 Vaccination: A Panoramic Review. J Epidemiol Glob Health 2023; 13:615-636. [PMID: 37870719 PMCID: PMC10686899 DOI: 10.1007/s44197-023-00159-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
The 2019 coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has posed a considerable challenge to global healthcare. Acute interstitial nephritis (AIN) post SARS-CoV-2 infection and vaccination has been reported, but its clinical features and pathogenesis remained unclear. We reviewed so far the largest 22 cases of AIN post SARS-CoV-2 infection and 36 cases of AIN following COVID-19 vaccination. The onset of AIN was mainly related to messenger RNA vaccines (52.8%). Apart from fever, proteinuria (45.5%) was the main manifestation of AIN post SARS-CoV-2 infection, left acute kidney injury (AKI, 63.9%) in patients post COVID-19 vaccination. The potential mechanism of vaccination induced AIN was conjugating vaccines with proteins to form a hapten, which activated dendritic cells and promoted a cascade immunological reaction leading to AIN.
Collapse
Affiliation(s)
- Yu Wang
- Department of Nephrology, Donghu District, the Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi, China
| | - Ling Yang
- Department of Health Management Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, Donghu District, the Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
27
|
Chakma V, Barman DN, Das SC, Hossain A, Momin MB, Tasneem M, Gupta SD. In silico analysis of a novel hypothetical protein (YP_498675.1) from Staphylococcus aureus unravels the protein of tryptophan synthase beta superfamily (Try-synth-beta_ II). J Genet Eng Biotechnol 2023; 21:135. [PMID: 37995054 PMCID: PMC10667181 DOI: 10.1186/s43141-023-00613-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Staphylococcus aureus is a gram-positive spherical bacteria and the most common cause of nosocomial infections in the world. Given its clinical significance, the genome sequence of S. aureus has been elucidated to enhance our comprehension of its lifestyle and pathogenicity. The research aimed to summarize a potential hypothetical protein that may play an important role in S. aureus virulence and pathogenicity, covering its anticipated structure, probable biological functions, and importance in this context. RESULTS A hypothetical protein, YP_498675.1 with 281 amino acid residues of S. aureus, was chosen for analysis and modeling by several bioinformatics tools and databases in this work. According to primary and secondary structure analyses, YP_498675.1 is a stable hydrophilic protein with a significant proportion of α-helices. Subcellular localization predictions by CELLO, PSORTb, and SOSUI server indicate that it is a cytoplasmic protein. NCBI-CDD, Pfam, and InterProScan functional genomics research revealed that the hypothetical protein may include the pyridoxal phosphate (PLP)-dependent 2, 3-diaminopropionate biosynthesis protein SbnA domain. In the homology modeling method, the HHpred server was employed to create its 3D structure using the template structure of a Staphyloferrin B precursor biosynthetic enzyme SbnA bound to PLP (PDB ID: 5D84_A), an X-ray diffraction model having 100% sequence identity with the hypothetical protein. After energy minimization, several quality assessments and validation factors determined that the generated protein model was reliable and of reasonable quality. CONCLUSION The present study has characterized and functionally annotated the hypothetical protein YP_498675.1 of S. aureus. Further experimental validation would aid in determining the actual function of YP_498675.1 as well as confirm the protein's value as a therapeutic target.
Collapse
Affiliation(s)
- Vivian Chakma
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Dhirendra Nath Barman
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Shuvo Chandra Das
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Anwar Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Monira Binte Momin
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Maisha Tasneem
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Shipan Das Gupta
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh.
| |
Collapse
|
28
|
Moran MC, Brewer MG, Schlievert PM, Beck LA. S. aureus virulence factors decrease epithelial barrier function and increase susceptibility to viral infection. Microbiol Spectr 2023; 11:e0168423. [PMID: 37737609 PMCID: PMC10581065 DOI: 10.1128/spectrum.01684-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/03/2023] [Indexed: 09/23/2023] Open
Abstract
Individuals with atopic dermatitis (AD) are highly colonized by Staphylococcus aureus and are more susceptible to severe viral complications. We hypothesized that S. aureus secreted virulence factors may alter keratinocyte biology to enhance viral susceptibility through disruption of the skin barrier, impaired keratinocyte differentiation, and/or inflammation. To address this hypothesis, human keratinocytes were exposed to conditioned media from multiple S. aureus strains that vary in virulence factor production (USA300, HG003, and RN4220) or select purified virulence factors. We have identified the S. aureus enterotoxin-like superantigen SElQ, as a virulence factor of interest, since it is highly produced by USA300 and was detected on the skin of 53% of AD subjects (n = 72) in a study conducted by our group. Treatment with USA300 conditioned media or purified SElQ resulted in a significant increase in keratinocyte susceptibility to infection with vaccinia virus, and also significantly decreased barrier function. Importantly, we have previously demonstrated that keratinocyte differentiation influences susceptibility to viral infection, and our qPCR observations indicated that USA300 S. aureus and SElQ alter differentiation in keratinocytes. CRISPR/Cas9 was used to knock out CD40, a potential enterotoxin receptor on epithelial cells. We found that CD40 expression on keratinocytes was not completely necessary for SElQ-mediated responses, as measured by proinflammatory cytokine expression and barrier function. Together, these findings support that select S. aureus virulence factors, particularly SElQ, enhance the susceptibility of epidermal cells to viral infection, which may contribute to the increased cutaneous infections observed in individuals with AD. IMPORTANCE Staphylococcus aureus skin colonization and infection are frequently observed in individuals with atopic dermatitis. Many S. aureus strains belong to the clonal group USA300, and these strains produce superantigens including the staphylococcal enterotoxin-like Q (SElQ). Our studies highlight that SElQ may play a key role by altering keratinocyte differentiation and reducing barrier function; collectively, this may explain the AD-specific enhanced infection risk to cutaneous viruses. It is unclear what receptor mediates SElQ's effects on keratinocytes. We have shown that one putative surface receptor, CD40, was not critical for its effects on proinflammatory cytokine production or barrier function.
Collapse
Affiliation(s)
- Mary C. Moran
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Matthew G. Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
29
|
Weiss S, Holtfreter S, Meyer TC, Schmiedeke F, Cammann C, Dörr M, Felix SB, Grabe HJ, Homuth G, Kohler C, Mahncke C, Michalik S, Nauck M, Friedrich N, Samietz S, Völzke H, Völker U, Bröker BM. Toxin exposure and HLA alleles determine serum antibody binding to toxic shock syndrome toxin 1 (TSST-1) of Staphylococcus aureus. Front Immunol 2023; 14:1229562. [PMID: 37731490 PMCID: PMC10507260 DOI: 10.3389/fimmu.2023.1229562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Life-threatening toxic shock syndrome is often caused by the superantigen toxic shock syndrome toxin-1 (TSST-1) produced by Staphylococcus aureus. A well-known risk factor is the lack of neutralizing antibodies. To identify determinants of the anti-TSST-1 antibody response, we examined 976 participants of the German population-based epidemiological Study of Health in Pomerania (SHIP-TREND-0). We measured anti-TSST-1 antibody levels, analyzed the colonization with TSST-1-encoding S. aureus strains, and performed a genome-wide association analysis of genetic risk factors. TSST-1-specific serum IgG levels varied over a range of 4.2 logs and were elevated by a factor of 12.3 upon nasal colonization with TSST-1-encoding S. aureus. Moreover, the anti-TSST-1 antibody levels were strongly associated with HLA class II gene loci. HLA-DRB1*03:01 and HLA-DQB1*02:01 were positively, and HLA-DRB1*01:01 as well as HLA-DQB1*05:01 negatively associated with the anti-TSST-1 antibody levels. Thus, both toxin exposure and HLA alleles affect the human antibody response to TSST-1.
Collapse
Affiliation(s)
- Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Tanja C. Meyer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Frieder Schmiedeke
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Clemens Cammann
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Marcus Dörr
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B. Felix
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Hans J. Grabe
- Department of Psychatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Christian Kohler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Cedric Mahncke
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Nele Friedrich
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Stefanie Samietz
- Department of Prosthetic Dentistry, Gerodontology and Biomaterials, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
30
|
Schlievert PM, Gaitán AV, Kilgore SH, Roe AL, Maukonen J, Lehtoranta L, Leung DYM, Marsman DS. Inhibition of Toxic Shock Syndrome-Associated Staphylococcus aureus by Probiotic Lactobacilli. Microbiol Spectr 2023; 11:e0173523. [PMID: 37404182 PMCID: PMC10434015 DOI: 10.1128/spectrum.01735-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
Staphylococcus aureus is a human pathogen with many infections originating on mucosal surfaces. One common group of S. aureus is the USA200 (CC30) clonal group, which produces toxic shock syndrome toxin-1 (TSST-1). Many USA200 infections occur on mucosal surfaces, particularly in the vagina and gastrointestinal tract. This allows these organisms to cause cases of menstrual TSS and enterocolitis. The current study examined the ability of two lactobacilli, Lactobacillus acidophilus strain LA-14 and Lacticaseibacillus rhamnosus strain HN001, for their ability to inhibit the growth of TSST-1 positive S. aureus, the production of TSST-1, and the ability of TSST-1 to induce pro-inflammatory chemokines from human vaginal epithelial cells (HVECs). In competition growth experiments, L. rhamnosus did not affect the growth of TSS S. aureus but did inhibit the production of TSST-1; this effect was partially due to acidification of the growth medium. L. acidophilus was both bactericidal and prevented the production of TSST-1 by S. aureus. This effect appeared to be partially due to acidification of the growth medium, production of H2O2, and production of other antibacterial molecules. When both organisms were incubated with S. aureus, the effect of L. acidophilus LA-14 dominated. In in vitro experiments with HVECs, neither lactobacillus induced significant production of the chemokine interleukin-8, whereas TSST-1 did induce production of the chemokine. When the lactobacilli were incubated with HVECs in the presence of TSST-1, the lactobacilli reduced chemokine production. These data suggest that these two bacteria in probiotics could reduce the incidence of menstrual and enterocolitis-associated TSS. IMPORTANCE Toxic shock syndrome (TSS) Staphylococcus aureus commonly colonize mucosal surfaces, giving them the ability to cause TSS through the action of TSS toxin-1 (TSST-1). This study examined the ability of two probiotic lactobacilli to inhibit S. aureus growth and TSST-1 production, and the reduction of pro-inflammatory chemokine production by TSST-1. Lacticaseibacillus rhamnosus strain HN001 inhibited TSST-1 production due to acid production but did not affect S. aureus growth. Lactobacillus acidophilus strain LA-14 was bactericidal against S. aureus, partially due to acid and H2O2 production, and consequently also inhibited TSST-1 production. Neither lactobacillus induced the production of pro-inflammatory chemokines by human vaginal epithelial cells, and both inhibited chemokine production by TSST-1. These data suggest that the two probiotics could reduce the incidence of mucosa-associated TSS, including menstrual TSS and cases originating as enterocolitis.
Collapse
Affiliation(s)
- Patrick M. Schlievert
- Department of Microbiology and Immunology, University of Iowa; Carver College of Medicine, Iowa City, Iowa, USA
| | | | - Samuel H. Kilgore
- Department of Microbiology and Immunology, University of Iowa; Carver College of Medicine, Iowa City, Iowa, USA
| | - Amy L. Roe
- The Procter & Gamble Company, Cincinnati, Ohio, USA
| | | | | | | | | |
Collapse
|
31
|
Truong VK, Hayles A, Bright R, Luu TQ, Dickey MD, Kalantar-Zadeh K, Vasilev K. Gallium Liquid Metal: Nanotoolbox for Antimicrobial Applications. ACS NANO 2023; 17:14406-14423. [PMID: 37506260 DOI: 10.1021/acsnano.3c06486] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
The proliferation of drug resistance in microbial pathogens poses a significant threat to human health. Hence, treatment measures are essential to surmount this growing problem. In this context, liquid metal nanoparticles are promising. Gallium, a post-transition metal notable for being a liquid at physiological temperature, has drawn attention for its distinctive properties, high antimicrobial efficacy, and low toxicity. Moreover, gallium nanoparticles demonstrate anti-inflammatory properties in immune cells. Gallium can alloy with other metals and be prepared in various composites to modify and tailor its characteristics and functionality. More importantly, the bactericidal mechanism of gallium liquid metal could sidestep the threat of emerging drug resistance mechanisms. Building on this rationale, gallium-based liquid metal nanoparticles can enable impactful and innovative strategic pathways in the battle against antimicrobial resistance. This review outlines the characteristics of gallium-based liquid metals at the nanoscale and their corresponding antimicrobial mechanisms to provide a comprehensive yet succinct overview of their current antimicrobial applications. In addition, challenges and opportunities that require further research efforts have been identified and discussed.
Collapse
Affiliation(s)
- Vi Khanh Truong
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Andrew Hayles
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Richard Bright
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Trong Quan Luu
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Michael D Dickey
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Kourosh Kalantar-Zadeh
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Krasimir Vasilev
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| |
Collapse
|
32
|
Hsieh RC, Liu R, Burgin DJ, Otto M. Understanding mechanisms of virulence in MRSA: implications for antivirulence treatment strategies. Expert Rev Anti Infect Ther 2023; 21:911-928. [PMID: 37501364 DOI: 10.1080/14787210.2023.2242585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) is a widespread pathogen, often causing recurrent and deadly infections in the hospital and community. Many S. aureus virulence factors have been suggested as potential targets for antivirulence therapy to decrease the threat of diminishing antibiotic availability. Antivirulence methods hold promise due to their adjunctive and prophylactic potential and decreased risk for selective pressure. AREAS COVERED This review describes the dominant virulence mechanisms exerted by MRSA and antivirulence therapeutics that are currently undergoing testing in clinical or preclinical stages. We also discuss the advantages and downsides of several investigational antivirulence approaches, including the targeting of bacterial transporters, host-directed therapy, and quorum-sensing inhibitors. For this review, a systematic search of literature on PubMed, Google Scholar, and Web of Science for relevant search terms was performed in April and May 2023. EXPERT OPINION Vaccine and antibody strategies have failed in clinical trials and could benefit from more basic science-informed approaches. Antivirulence-targeting approaches need to be set up better to meet the requirements of drug development, rather than only providing limited results to provide 'proof-of-principle' translational value of pathogenesis research. Nevertheless, there is great potential of such strategies and potential particular promise for novel probiotic approaches.
Collapse
Affiliation(s)
- Roger C Hsieh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Dylan J Burgin
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
33
|
Brouwer S, Rivera-Hernandez T, Curren BF, Harbison-Price N, De Oliveira DMP, Jespersen MG, Davies MR, Walker MJ. Pathogenesis, epidemiology and control of Group A Streptococcus infection. Nat Rev Microbiol 2023; 21:431-447. [PMID: 36894668 PMCID: PMC9998027 DOI: 10.1038/s41579-023-00865-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/11/2023]
Abstract
Streptococcus pyogenes (Group A Streptococcus; GAS) is exquisitely adapted to the human host, resulting in asymptomatic infection, pharyngitis, pyoderma, scarlet fever or invasive diseases, with potential for triggering post-infection immune sequelae. GAS deploys a range of virulence determinants to allow colonization, dissemination within the host and transmission, disrupting both innate and adaptive immune responses to infection. Fluctuating global GAS epidemiology is characterized by the emergence of new GAS clones, often associated with the acquisition of new virulence or antimicrobial determinants that are better adapted to the infection niche or averting host immunity. The recent identification of clinical GAS isolates with reduced penicillin sensitivity and increasing macrolide resistance threatens both frontline and penicillin-adjunctive antibiotic treatment. The World Health Organization (WHO) has developed a GAS research and technology road map and has outlined preferred vaccine characteristics, stimulating renewed interest in the development of safe and effective GAS vaccines.
Collapse
Affiliation(s)
- Stephan Brouwer
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Bodie F Curren
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Nichaela Harbison-Price
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David M P De Oliveira
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Magnus G Jespersen
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Mark R Davies
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
34
|
Schlievert P, Kilgore S, Leung D. Agr Regulation of Streptococcal Pyrogenic Exotoxin A in Staphylococcus aureus. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000795. [PMID: 37179971 PMCID: PMC10167548 DOI: 10.17912/micropub.biology.000795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/22/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023]
Abstract
Group A streptococcal pyrogenic exotoxins (SPEs A, B, and C) are superantigens. SPE A shares high sequence similarity with Staphylococcus aureus enterotoxins (SEs) B and C. Since SPE A is bacteriophage-encoded, we hypothesized that its gene ( speA ) was acquired from S. aureus . speA , when cloned into S. aureus , was stably expressed, its protein resistant to proteases, and the gene under accessory gene regulator control. speA was acquired by streptococci from cross-species transduction. speB was not expressed in S. aureus. SPE C was degraded by staphylococcal proteases. The genes speB and speC were not recently acquired from S. aureus.
Collapse
|
35
|
Filippatos F, Tatsi EB, Michos A. Immunology of Multisystem Inflammatory Syndrome after COVID-19 in Children: A Review of the Current Evidence. Int J Mol Sci 2023; 24:ijms24065711. [PMID: 36982783 PMCID: PMC10057510 DOI: 10.3390/ijms24065711] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Immune responses following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in children are still under investigation. Even though coronavirus disease 2019 (COVID-19) is usually mild in the pediatric population, some children exhibit severe clinical manifestations, require hospitalization, or develop the most severe condition: a multisystem inflammatory syndrome in children (MIS-C) associated with SARS-CoV-2 infection. The activated innate, humoral and T-cell-mediated immunological pathways that lead certain pediatric populations to present with MIS-C or remain asymptomatic after SARS-CoV-2 infection are yet to be established. This review focuses on the immunological aspects of MIS-C with respect to innate, humoral, and cellular immunity. In addition, presents the role of the SARS-CoV-2 Spike protein as a superantigen in the pathophysiological mechanisms, discusses the great heterogeneity among the immunological studies in the pediatric population, and highlights possible reasons why some children with a certain genetic background present with MIS-C.
Collapse
|
36
|
Linz MS, Mattappallil A, Finkel D, Parker D. Clinical Impact of Staphylococcus aureus Skin and Soft Tissue Infections. Antibiotics (Basel) 2023; 12:557. [PMID: 36978425 PMCID: PMC10044708 DOI: 10.3390/antibiotics12030557] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The pathogenic bacterium Staphylococcus aureus is the most common pathogen isolated in skin-and-soft-tissue infections (SSTIs) in the United States. Most S. aureus SSTIs are caused by the epidemic clone USA300 in the USA. These infections can be serious; in 2019, SSTIs with S. aureus were associated with an all-cause, age-standardized mortality rate of 0.5 globally. Clinical presentations of S. aureus SSTIs vary from superficial infections with local symptoms to monomicrobial necrotizing fasciitis, which can cause systemic manifestations and may lead to serious complications or death. In order to cause skin infections, S. aureus employs a host of virulence factors including cytolytic proteins, superantigenic factors, cell wall-anchored proteins, and molecules used for immune evasion. The immune response to S. aureus SSTIs involves initial responders such as keratinocytes and neutrophils, which are supported by dendritic cells and T-lymphocytes later during infection. Treatment for S. aureus SSTIs is usually oral therapy, with parenteral therapy reserved for severe presentations; it ranges from cephalosporins and penicillin agents such as oxacillin, which is generally used for methicillin-sensitive S. aureus (MSSA), to vancomycin for methicillin-resistant S. aureus (MRSA). Treatment challenges include adverse effects, risk for Clostridioides difficile infection, and potential for antibiotic resistance.
Collapse
Affiliation(s)
- Matthew S. Linz
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Arun Mattappallil
- Department of Pharmaceutical Services, University Hospital, Newark, NJ 07103, USA
| | - Diana Finkel
- Division of Infectious Diseases, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
37
|
Pomputius WF, Kilgore SH, Schlievert PM. Probable enterotoxin-associated toxic shock syndrome caused by Staphylococcus epidermidis. BMC Pediatr 2023; 23:108. [PMID: 36882717 PMCID: PMC9989563 DOI: 10.1186/s12887-023-03914-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND We describe a case of a toxic shock-like syndrome in a child, which was associated with Staphylococcus epidermidis instead of Staphylococcus aureus or Streptococcus pyogenes, the usual causes of toxic shock syndrome. CASE PRESENTATION The patient was an 8-year-old boy who developed a toxic shock syndrome-like illness, including fever, hypotension, and rash. The Staphylococcus epidermidis isolate was cultured from urine, but this organism was unavailable for toxin testing. Multiple blood cultures were negative. Instead, a highly novel assay was used on acute plasma from the patient which demonstrated the presence of the genes for superantigens, staphylococcal enterotoxins A, C, D, and E. Superantigens are the known causes of toxic shock syndrome. CONCLUSIONS Our study suggests strongly that Staphylococcus epidermidis was causing the TSS symptoms through the known Staphylococcus aureus superantigens. It is unknown how many other such patients exist; this should be explored. Of great importance is that PCR performed directly on blood plasma in the absence of microbial isolation could be used to demonstrate superantigen genes.
Collapse
Affiliation(s)
- William F Pomputius
- Division of Infectious Disease, Children's Minnesota, Minneapolis, MN, 55455, USA
| | - Samuel H Kilgore
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, Iowa, 52242, USA
| | - Patrick M Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, Iowa, 52242, USA. .,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
38
|
Schlievert PM. Staphylococcal Enterotoxin B and C Mutants and Vaccine Toxoids. Microbiol Spectr 2023; 11:e0444622. [PMID: 36815779 PMCID: PMC10101070 DOI: 10.1128/spectrum.04446-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/26/2023] [Indexed: 02/24/2023] Open
Abstract
Three mutants individually of both staphylococcal enterotoxins B and C were prepared by site-specific mutagenesis of enterotoxin amino acids that contact host T lymphocyte immune cell receptor sites (N23A, Q210A, and N23A/Q210A); these amino acids are shared between the two enterotoxins, and mutations reduce the interaction with the variable part of the β-chain of the T lymphocyte receptor. The mutant proteins, as expressed in Staphylococcus aureus RN4220, lacked biological toxicity as measured by the loss of (i) stimulation of rabbit splenocyte proliferation, (ii) pyrogenicity, and (iii) the ability to enhance the lethality of endotoxin shock, compared to wild-type enterotoxins. In addition, the mutants were able to vaccinate rabbits against pyrogenicity, the enhancement of endotoxin shock, and lethality in a pneumonia model when animals were challenged with methicillin-resistant S. aureus. Three vaccine injections (one primary and two boosters) protected rabbits for at least 3.5 months postvaccination when challenged with wild-type enterotoxins (last time point tested). These mutant proteins have the potential to function as toxoid vaccines against these two causes of nonmenstrual toxic shock syndrome (TSS). IMPORTANCE Toxic shock syndrome toxin 1 (TSST-1) and staphylococcal enterotoxins B and C cause the majority of cases of staphylococcal toxic shock syndrome. Previously, vaccine toxoids of TSST-1 have been prepared. In this study, vaccine toxoids of enterotoxins B and C were prepared. The toxoids lost biological toxicity but were able to vaccinate rabbits against lethal TSS.
Collapse
Affiliation(s)
- Patrick M. Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
39
|
Schlievert PM, Kilgore SH, Beck LA, Yoshida T, Klingelhutz AJ, Leung DYM. Host Cationic Antimicrobial Molecules Inhibit S. aureus Exotoxin Production. mSphere 2023; 8:e0057622. [PMID: 36598227 PMCID: PMC9942567 DOI: 10.1128/msphere.00576-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/02/2022] [Indexed: 01/05/2023] Open
Abstract
Innate immune molecules, including antimicrobial peptides (for example, defensins) and lysozyme, function to delay or prevent bacterial infections. These molecules are commonly found on mucosal and skin surfaces. Staphylococcus aureus is a common pathogen and causes millions of infections annually. It is well known that innate immune molecules, such as defensins and lysozyme, either poorly inhibit or do not inhibit the growth of S. aureus. Our current studies show that the α-defensin human neutrophil α-defensin-1 (HNP-1) and lysozyme inhibit exotoxin production, both hemolysins and superantigens, which are required for S. aureus infection. HNP-1 inhibited exotoxin production at concentrations as low as 0.001 μg/mL. Lysozyme inhibited exotoxin production at 0.05 to 0.5 μg/mL. Both HNP-1 and lysozyme functioned through at least one two-component system (SrrA/B). The β-defensin human β-defensin 1 (HBD-1) inhibited hemolysin but not superantigen production. The cation chelator S100A8/A9 (calprotectin), compared to EDTA, was tested for the ability to inhibit exotoxin production. EDTA at high concentrations inhibited exotoxin production; these were the same concentrations that interfered with staphylococcal growth. S100A8/A9 at the highest concentration tested (10 μg/mL) had no effect on S. aureus growth but enhanced exotoxin production. Lower concentrations had no effect on growth or exotoxin production. Lysostaphin is regularly used to lyse S. aureus. The lytic concentrations of lysostaphin were the only concentrations that also inhibited growth and exotoxin production. Our studies demonstrate that a major activity of innate defensin peptides and lysozyme is inhibition of staphylococcal exotoxin production but not inhibition of growth. IMPORTANCE Staphylococcus aureus causes large numbers of both relatively benign and serious human infections, which are mediated in large part by the organisms' secreted exotoxins. Since 1921, it has been known that lysozyme and, as shown later in the 1900s, other innate immune peptides, including human neutrophil α-defensin-1 (HNP-1) and human β-defensin 1 (HBD-1), are either not antistaphylococcal or are only weakly inhibitory to growth. Our study confirms those findings but, importantly, shows that at subgrowth inhibitory concentrations, these positively charged innate immune peptides inhibit exotoxin production, including both hemolysins and the superantigen toxic shock syndrome toxin-1. The data show that the principal activity of innate immune peptides in the host is likely to be inhibition of exotoxin production required for staphylococcal mucosal or skin colonization rather than growth inhibition.
Collapse
Affiliation(s)
- Patrick M. Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Samuel H. Kilgore
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Takeshi Yoshida
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Aloysius J. Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
40
|
Rahman S, Sarkar K, Das AK. Exploring staphylococcal superantigens to design a potential multi-epitope vaccine against Staphylococcus aureus: an in-silico reverse vaccinology approach. J Biomol Struct Dyn 2023; 41:13098-13112. [PMID: 36729064 DOI: 10.1080/07391102.2023.2171138] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/13/2023] [Indexed: 02/03/2023]
Abstract
Staphylococcus aureus is a horrifying bacteria capable of causing millions of deaths yearly across the globe. A major contribution to the success of S. aureus as an ESKAPE pathogen is the abundance of virulence factors that can manipulate the innate and adaptive immune system of the individual. Currently, no vaccine is available to treat S. aureus-mediated infections. In this study, we present in-silico approaches to design a stable, safe and immunogenic vaccine that could help to control the infections associated with the bacteria. Three vital pathogenic secreted toxins of S. aureus, such as staphylococcal enterotoxin A (SEA), staphylococcal enterotoxin B (SEB), Toxic-shock syndrome toxin (TSST-1), were selected using the reverse vaccinology approach to design the multi-epitope vaccine (MEV). Linear B-lymphocyte, cytotoxic T-lymphocyte (CTL) and helper T-lymphocyte (HTL) epitopes were predicted from these selected proteins. For designing the multi-epitope vaccine (MEV), B-cell epitopes were joined with the KK linker, CTL epitopes were joined with the AAY linker, and HTL epitopes were joined with the GPGPG linker. Finally, to increase the immune response to the vaccine, a human β-defensin-3 (hBD-3) adjuvant was added to the N-terminus of the MEV construct. The final MEV was found to be antigenic and non-allergen in nature. In-silico immune simulation and cloning analysis predicted the immune-stimulating potential of the designed MEV construct along with the cloning feasibility in the pET28a(+) vector with the E. coli expression system. This immunoinformatics study provides a platform for designing a suitable, safe and effective vaccine against S. aureus.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shakilur Rahman
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Kasturi Sarkar
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Amit Kumar Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
41
|
Giryes S, McGonagle D. Immune and non-immune mechanisms that determine vasculitis and coronary artery aneurysm topography in Kawasaki disease and MIS-C. Clin Exp Rheumatol 2023; 22:103240. [PMID: 36496111 DOI: 10.1016/j.autrev.2022.103240] [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: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
The overlap between multisystem inflammatory syndrome in children (MIS-C) and Kawasaki disease (KD) including coronary artery aneurysms (CAA) and broadly shared gastrointestinal and mucocutaneous disease is poorly defined. In this perspective, we highlight common age-related extravascular epicardial microanatomical and immunological factors that might culminate in CAA expression in both MIS-C and KD. Specifically, the coronary vasa vasorum originates outside the major coronary arteries. Widespread inflammation in the epicardial interstitial compartment in shared between KD and MIS-C. Age-related changes in the neonatal and immature coronary vasculature including the impact of coronary artery biomechanical factors including coronary vessel calibre, age-related vessel distensibility, flow, and vessel neurovascular innervation may explain the decreasing CAA frequency from neonates to older children and the virtual absence of CAA in young adults with the MIS-C phenotype. Other KD and MIS-C features including mucocutaneous disease with keratinocyte-related immunopathology corroborate that disease phenotypes are centrally influenced by inflammation originating outside vessel walls but a potential role for primary coronary artery vascular wall inflammation cannot be excluded. Hence, common extravascular originating tissue-specific responses to aetiologically diverse triggers including superantigens may lead to widespread interstitial tissue inflammation characteristically manifesting as CAA development, especially in younger subjects. Given that CAA is virtually absent in adults, further studies are needed to ascertain whether epicardial interstitial inflammation may impact on both coronary artery physiology and cardiac conduction tissue and contribute to cardiovascular disease- a hitherto unappreciated consideration.
Collapse
Affiliation(s)
- Sami Giryes
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom; National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, United Kingdom.
| |
Collapse
|
42
|
Obanda BA, Cook EAJ, Fèvre EM, Bebora L, Ogara W, Wang SH, Gebreyes W, Ngetich R, Wandede D, Muyodi J, Blane B, Coll F, Harrison EM, Peacock SJ, Gitao GC. Characteristics of Staphylococcus aureus Isolated from Patients in Busia County Referral Hospital, Kenya. Pathogens 2022; 11:1504. [PMID: 36558838 PMCID: PMC9781741 DOI: 10.3390/pathogens11121504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus is an important pathogen associated with hospital, community, and livestock-acquired infections, with the ability to develop resistance to antibiotics. Nasal carriage by hospital inpatients is a risk for opportunistic infections. Antibiotic susceptibility patterns, virulence genes and genetic population structure of S. aureus nasal isolates, from inpatients at Busia County Referral Hospital (BCRH) were analyzed. A total of 263 inpatients were randomly sampled, from May to July 2015. The majority of inpatients (85.9%) were treated empirically with antimicrobials, including ceftriaxone (65.8%) and metronidazole (49.8%). Thirty S. aureus isolates were cultured from 29 inpatients with a prevalence of 11% (10.3% methicillin-susceptible S. aureus (MSSA), 0.8% methicillin resistant S. aureus (MRSA)). Phenotypic and genotypic resistance was highest to penicillin-G (96.8%), trimethoprim (73.3%), and tetracycline (13.3%) with 20% of isolates classified as multidrug resistant. Virulence genes, Panton-Valentine leukocidin (pvl), toxic shock syndrome toxin-1 (tsst-1), and sasX gene were detected in 16.7%, 23.3% and 3.3% of isolates. Phylogenetic analysis showed 4 predominant clonal complexes CC152, CC8, CC80, and CC508. This study has identified that inpatients of BCRH were carriers of S. aureus harbouring virulence genes and resistance to a range of antibiotics. This may indicate a public health risk to other patients and the community.
Collapse
Affiliation(s)
- Benear Apollo Obanda
- Department of Veterinary Pathology, Microbiology and Parasitology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
- Global One Health Initiative, Office of International Affairs, The Ohio State University, Columbus, OH 43210, USA
- Centre for Microbiology Research Nairobi, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | | | - Eric M. Fèvre
- International Livestock Research Institute, Nairobi P.O. Box 30709-00100, Kenya
- Institute of Infection, Veterinary & Ecological Sciences, Leahurst Campus, University of Liverpool, Chester High Road, Neston CH64 7TE, UK
| | - Lilly Bebora
- Department of Veterinary Pathology, Microbiology and Parasitology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - William Ogara
- Department of Public Health, Pharmacology and Toxicology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Shu-Hua Wang
- Global One Health Initiative, Office of International Affairs, The Ohio State University, Columbus, OH 43210, USA
- Division of Infectious Disease, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Wondwossen Gebreyes
- Global One Health Initiative, Office of International Affairs, The Ohio State University, Columbus, OH 43210, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ronald Ngetich
- Centre for Microbiology Research Nairobi, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | - Dolphine Wandede
- Centre for Microbiology Research Nairobi, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | - Johnstone Muyodi
- The Centre for Infectious and Parasitic Diseases Control Research, Busia P.O. Box 3-50400, Kenya
| | - Beth Blane
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK
| | - Francesc Coll
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, University of London, London WC1E 7HT, UK
| | - Ewan M. Harrison
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK
- Wellcome Sanger Institute, Hinxton CB10 1SA, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Sharon J. Peacock
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK
| | - George C. Gitao
- Department of Veterinary Pathology, Microbiology and Parasitology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| |
Collapse
|
43
|
Obanda BA, Gibbons CL, Fèvre EM, Bebora L, Gitao G, Ogara W, Wang SH, Gebreyes W, Ngetich R, Blane B, Coll F, Harrison EM, Kariuki S, Peacock SJ, Cook EAJ. Multi-Drug Resistant Staphylococcus aureus Carriage in Abattoir Workers in Busia, Kenya. Antibiotics (Basel) 2022; 11:1726. [PMID: 36551383 PMCID: PMC9774130 DOI: 10.3390/antibiotics11121726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Abattoir workers have been identified as high-risk for livestock-associated Staphylococcus aureus carriage. This study investigated S. aureus carriage in abattoir workers in Western Kenya. Nasal swabs were collected once from participants between February-November 2012. S. aureus was isolated using bacterial culture and antibiotic susceptibility testing performed using the VITEK 2 instrument and disc diffusion methods. Isolates underwent whole genome sequencing and Multi Locus Sequence Types were derived from these data. S. aureus (n = 126) was isolated from 118/737 (16.0%) participants. Carriage was higher in HIV-positive (24/89, 27.0%) than HIV−negative participants (94/648, 14.5%; p = 0.003). There were 23 sequence types (STs) identified, and half of the isolates were ST152 (34.1%) or ST8 (15.1%). Many isolates carried the Panton-Valentine leucocidin toxin gene (42.9%). Only three isolates were methicillin resistant S. aureus (MRSA) (3/126, 2.4%) and the prevalence of MRSA carriage was 0.4% (3/737). All MRSA were ST88. Isolates from HIV-positive participants (37.0%) were more frequently resistant to sulfamethoxazole/trimethoprim compared to isolates from HIV-negative participants (6.1%; p < 0.001). Similarly, trimethoprim resistance genes were more frequently detected in isolates from HIV-positive (81.5%) compared to HIV-negative participants (60.6%; p = 0.044). S. aureus in abattoir workers were representative of major sequence types in Africa, with a high proportion being toxigenic isolates. HIV-positive individuals were more frequently colonized by antimicrobial resistant S. aureus which may be explained by prophylactic antimicrobial use.
Collapse
Affiliation(s)
- Benear Apollo Obanda
- Department of Veterinary Pathology, Microbiology and Parasitology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
- Global One Health Initiative, The Ohio State University, Columbus, OH 43210, USA
- Centre for Microbiology Research Nairobi, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | | | - Eric M. Fèvre
- Institute of Infection, Veterinary & Ecological Sciences, Leahurst Campus, University of Liverpool, Chester High Road, Neston CH64 7TE, UK
- International Livestock Research Institute, Nairobi P.O. Box 30709-00100, Kenya
| | - Lilly Bebora
- Department of Veterinary Pathology, Microbiology and Parasitology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - George Gitao
- Department of Veterinary Pathology, Microbiology and Parasitology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - William Ogara
- Department of Public Health Pharmacology and Toxicology, University of Nairobi, Nairobi P.O. Box 29053-00625, Kenya
| | - Shu-Hua Wang
- Global One Health Initiative, The Ohio State University, Columbus, OH 43210, USA
- Division of Infectious Disease, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Wondwossen Gebreyes
- Global One Health Initiative, The Ohio State University, Columbus, OH 43210, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ronald Ngetich
- Centre for Microbiology Research Nairobi, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | - Beth Blane
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Francesc Coll
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Ewan M. Harrison
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Wellcome Sanger Institute, Hinxton CB10 1SA, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Samuel Kariuki
- Centre for Microbiology Research Nairobi, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya
| | - Sharon J. Peacock
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | | |
Collapse
|
44
|
Noli Truant S, Redolfi DM, Sarratea MB, Malchiodi EL, Fernández MM. Superantigens, a Paradox of the Immune Response. Toxins (Basel) 2022; 14:toxins14110800. [PMID: 36422975 PMCID: PMC9692936 DOI: 10.3390/toxins14110800] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Staphylococcal enterotoxins are a wide family of bacterial exotoxins with the capacity to activate as much as 20% of the host T cells, which is why they were called superantigens. Superantigens (SAgs) can cause multiple diseases in humans and cattle, ranging from mild to life-threatening infections. Almost all S. aureus isolates encode at least one of these toxins, though there is no complete knowledge about how their production is triggered. One of the main problems with the available evidence for these toxins is that most studies have been conducted with a few superantigens; however, the resulting characteristics are attributed to the whole group. Although these toxins share homology and a two-domain structure organization, the similarity ratio varies from 20 to 89% among different SAgs, implying wide heterogeneity. Furthermore, every attempt to structurally classify these proteins has failed to answer differential biological functionalities. Taking these concerns into account, it might not be appropriate to extrapolate all the information that is currently available to every staphylococcal SAg. Here, we aimed to gather the available information about all staphylococcal SAgs, considering their functions and pathogenicity, their ability to interact with the immune system as well as their capacity to be used as immunotherapeutic agents, resembling the two faces of Dr. Jekyll and Mr. Hyde.
Collapse
|
45
|
Glucose Mediates Niche-Specific Repression of Staphylococcus aureus Toxic Shock Syndrome Toxin-1 through the Activity of CcpA in the Vaginal Environment. J Bacteriol 2022; 204:e0026922. [PMID: 36106854 PMCID: PMC9578429 DOI: 10.1128/jb.00269-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus chronically colonizes up to 30% of the human population on the skin or mucous membranes, including the nasal tract or vaginal canal. While colonization is often benign, this bacterium also has the capability to cause serious infections. Menstrual toxic shock syndrome (mTSS) is a serious toxinosis associated with improper use of tampons, which can induce an environment that is favorable to the production of the superantigen known as toxic shock syndrome toxin-1 (TSST-1). To better understand environmental signaling that influences TSST-1 production, we analyzed expression in the prototype mTSS strain S. aureus MN8. Using transcriptional and protein-based analysis in two niche-related media, we observed that TSST-1 expression was significantly higher in synthetic nasal medium (SNM) than in vaginally defined medium (VDM). One major divergence in medium composition was high glucose concentration in VDM. The glucose-dependent virulence regulator gene ccpA was deleted in MN8, and, compared with wild-type MN8, we observed increased TSST-1 expression in the ΔccpA mutant when grown in VDM, suggesting that TSST-1 is repressed by catabolite control protein A (CcpA) in the vaginal environment. We were able to relieve CcpA-mediated repression by modifying the glucose level in vaginal conditions, confirming that changes in nutritional conditions contribute to the overexpression of TSST-1 that can lead to mTSS. We also compared CcpA-mediated repression to other key regulators of tst, finding that CcpA regulation is dominant compared to other characterized regulatory mechanisms. This study underlines the importance of environmental signaling for S. aureus pathogenesis in the context of mTSS. IMPORTANCE Menstrual toxic shock syndrome (mTSS) is caused by strains of Staphylococcus aureus that overproduce a toxin known as toxic shock syndrome toxin-1 (TSST-1). This work studied how glucose levels in a model vaginal environment could influence the amount of TSST-1 that is produced by S. aureus. We found that high levels of glucose repress TSST-1 production, and this is done by a regulatory protein called catabolite control protein A (CcpA). The research also demonstrated that, compared with other regulatory proteins, the CcpA regulator appears to be the most important for maintaining low levels of TSST-1 in the vaginal environment, and this information helps to understand how changes in the vaginal environmental can lead to mTSS.
Collapse
|
46
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
47
|
Molecular Evolution and Genomic Insights into Community-Acquired Methicillin-Resistant Staphylococcus aureus Sequence Type 88. Microbiol Spectr 2022; 10:e0034222. [PMID: 35730953 PMCID: PMC9430171 DOI: 10.1128/spectrum.00342-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sequence type 88 (ST88) methicillin-resistant Staphylococcus aureus (MRSA) has been recognized as an important pathogen that causes infections in humans, especially when it has strong biofilm production and multidrug resistance (MDR). However, knowledge of the determinants of resistance or virulence and genomic characteristics of ST88 MRSA from China is still limited. In this study, we employed the antimicrobial resistance (AMR), biofilm formation, and genomic characteristics of ST88 MRSA collected from various foods in China and estimated the worldwide divergence of ST88 MRSA with publicly available ST88 genomes. All ST88 isolates studied were identified as having resistance genes, while 50% (41/82) harbored MDR genes. All isolates carried core virulence genes related to immune modulation, adherence, secreted enzymes, and hemolysin. In addition, all 20 Chinese ST88 isolates showed biofilm production capacity, three strongly so. Bayesian phylogenetic analysis showed that Chinese ST88 clones formed an independent MRSA lineage, with two subclades associated with acquisition of type IVc staphylococcal cassette chromosome mec (SCCmec) elements. In contrast, all African ST88 strains were subtyped as SCCmecIVa, where the African clades were mixed with a few European and American isolates, suggesting potential transmission from Africa to these regions. In summary, our results revealed the evolution of ST88 MRSA in humans, animals, and foods in Africa and Asia. The food-associated ST88 genomes in this study will remedy the lack of food-associated ST88 isolates, and the study in general will extend the discussion of the potential exchanges of ST88 between humans and foods or food animals. IMPORTANCE ST88 MRSA has frequently been detected in humans, animals, and foods mainly in Africa and Asia. It can colonize and cause mild to severe infections in humans, especially children. Several studies from African countries have described its genotypic characteristics but, limited information is available on the evolution and characterization of ST88 MRSA in Asia, especially China. Meanwhile, the molecular history of its global spread remains largely unclear. In this study, we analyzed the genomic evolution of global ST88 MRSA strains in detail and identified key genetic changes associated with specific hosts or regions. Our results suggested geographical differentiation between ST88 MRSA’s evolution in Africa and its evolution in Asia, with a more recent clonal evolution in China. The introduction of ST88 MRSA in China was aligned with the acquisition of SCCmecIVc elements, specific virulent prophages, and AMR genes.
Collapse
|
48
|
Schlievert PM, Kilgore SH, Benavides A, Klingelhutz AJ. Pathogen Stimulation of Interleukin-8 from Human Vaginal Epithelial Cells through CD40. Microbiol Spectr 2022; 10:e0010622. [PMID: 35297656 PMCID: PMC9045207 DOI: 10.1128/spectrum.00106-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/08/2022] [Indexed: 01/08/2023] Open
Abstract
Many bacterial and fungal pathogens cause disease across mucosal surfaces, and to a lesser extent through skin surfaces. Pathogens that potentially cause disease vaginally across epithelial cells include Staphylococcus aureus, group A and B streptococci, Escherichia coli, Neisseria gonorrhoeae, and Candida albicans. We have previously shown that staphylococcal and streptococcal superantigens induce inflammatory chemokines from vaginal epithelial cells through the immune costimulatory molecule CD40 through use of a CRISPR cas9 knockout mutant and complemented epithelial cell line. In this study, we show that the potential vaginal pathogens S. aureus, group A and B streptococci, E. coli, an Enterococcus faecalis strain, and C. albicans in part use CD40 to stimulate interleukin-8 (IL-8) production from human vaginal epithelial cells. In contrast, N. gonorrhoeae does not appear to use CD40 to signal IL-8 production. Normal flora Lactobacillus crispatus and an Enterococcus faecalis strain that produces reutericyclin do not induce IL-8. These data indicate that many potential pathogens, but no normal commensals, induce IL-8 to help disrupt the human vaginal epithelial barrier through CD40, thus providing a potential therapeutic target for drug development. IMPORTANCE Most bacterial and fungal pathogens cause disease across mucosal, and to a lesser extent, skin barriers with the help of induced chemokines from epithelial cells. In this study, we showed that potential vaginal pathogens Staphylococcus aureus, group A and B streptococci, some Enterococcus faecalis strains, Escherichia coli, and Candida albicans use the immune costimulatory molecule CD40 to induce the chemokine interleukin-8 production. In contrast, Neisseria gonorrhoeae does not use CD40 to stimulate interleukin-8. Normal flora lactobacilli and at least one E. faecalis strain do not induce interleukin-8.
Collapse
Affiliation(s)
- Patrick M. Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Samuel H. Kilgore
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Andrea Benavides
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Aloysius J. Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
49
|
Ababneh Q, Jaradat Z, Khanfar M, Alnohoud R, Alzu'bi M, Makahleh S, Abulaila S. Methicillin‐resistant
Staphylococcus aureus
contamination of high‐touched surfaces in a university campus. J Appl Microbiol 2022; 132:4486-4500. [DOI: 10.1111/jam.15526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/23/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Qutaiba Ababneh
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts Jordan University of Science and Technology P. O. Box 3030 Irbid 22110 Jordan
| | - Ziad Jaradat
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts Jordan University of Science and Technology P. O. Box 3030 Irbid 22110 Jordan
| | - Malak Khanfar
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts Jordan University of Science and Technology P. O. Box 3030 Irbid 22110 Jordan
| | - Rahaf Alnohoud
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts Jordan University of Science and Technology P. O. Box 3030 Irbid 22110 Jordan
| | - Mallak Alzu'bi
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts Jordan University of Science and Technology P. O. Box 3030 Irbid 22110 Jordan
| | - Sara Makahleh
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts Jordan University of Science and Technology P. O. Box 3030 Irbid 22110 Jordan
| | - Sally Abulaila
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts Jordan University of Science and Technology P. O. Box 3030 Irbid 22110 Jordan
| |
Collapse
|
50
|
Bergbower EAS, Odonkor P. CON: Acute Normovolemic Hemodilution (ANH) Should Not Be Used as a Transfusion Strategy in Patients With Infective Endocarditis. J Cardiothorac Vasc Anesth 2022; 36:2815-2818. [DOI: 10.1053/j.jvca.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 11/11/2022]
|