1
|
Gérard R, Pauquet E, Ros B, Lehours P, Renesme L. Continuous Versus Intermittent Vancomycin Infusions for Coagulase-negative Staphylococcus Bacteremia in Neonates: A Propensity-matched Cohort Study. Pediatr Infect Dis J 2025; 44:131-135. [PMID: 39259856 DOI: 10.1097/inf.0000000000004538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
BACKGROUND Coagulase-negative staphylococci (CONS) are a major cause of late-onset neonatal sepsis, particularly in preterm infants, with high morbidity and mortality. While vancomycin is the first-line treatment for these infections, the optimal administration in neonates remains uncertain. OBJECTIVE We aim to compare the outcomes of neonates with CONS bacteremia treated with adjusted continuous infusion (CIV) versus standard intermittent infusion (IIV) of vancomycin. METHODS This retrospective study included 110 neonates, with 29 in the CIV group and 47 in the IIV group after propensity score matching. The primary outcome was treatment failure defined by the persistence of a positive blood culture for the same organism after at least 48 hours of vancomycin treatment. RESULTS After matching, the CIV group exhibited significantly lower treatment failure rates [5/29 (17%) vs. 26/47 (44%); P = 0.014] and a higher rate of achieving therapeutic vancomycin levels after 24 hours [20/29 (69%) vs. 26/47 (44%); P = 0.002] compared to the IIV group. No significant differences were observed in terms of acute kidney failure between the 2 groups. CONCLUSION Adjusted continuous vancomycin infusion in neonates with CONS bacteremia is associated with a lower treatment failure rate without an increase in renal toxicity compared to standard intermittent infusion. However, due to the observational design, larger prospective studies are needed to validate these results.
Collapse
Affiliation(s)
- Rémy Gérard
- From the Department of Pediatrics, Neonatal Intensive Care Unit
| | - Emilie Pauquet
- From the Department of Pediatrics, Neonatal Intensive Care Unit
| | - Barbara Ros
- Departement of Pediatrics, Neonatal and Pediatric Intensive Care Unit
| | - Philippe Lehours
- Microbiology Department, Bacteriology laboratory, University Hospital of Bordeaux
- Bordeaux Institute of Oncology, BRIC U1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Laurent Renesme
- Department of Pediatrics, Neonatal Intensive Care Unit, Children's Hospital of Eastern Ontario, Ottawa, Canada
| |
Collapse
|
2
|
Szemraj M, Glajzner P, Olszowiec K, Sienkiewicz M. The prevalence of multidrug resistance in Staphylococcus hominis isolated from clinical materials. Sci Rep 2025; 15:414. [PMID: 39747570 PMCID: PMC11696355 DOI: 10.1038/s41598-024-84500-2] [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: 07/26/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025] Open
Abstract
The treatment of infections caused by Staphylococcus hominis remains a challenge, mainly due to the increasing resistance of these bacteria to antibiotics. The aim of the study was to determine antibiotic resistance in 62 strains S. hominis isolated from clinical materials, and to identify the molecular basis of resistance to antibiotics. Forty-six strains were both methicillin-resistant and harbored the mecA gene. Twenty-three of these strains had mec complex A and ccr complex AB1. Such a combination of the mec and ccr complexes does not correspond to any cassettes that have been demonstrated so far. However, over 80% of the tested strains were multidrug-resistant, of which as many as 12 were resistant to at least seven antibiotics. More than a half of strains harbored the tetK, acc(6')-Ie aph(2''), and ant(4')-I genes. erm(C) was the most common resistant gene to antibiotics from the MLS group. Two strains had as many as five antibiotic resistance genes from the tested groups (erm(C), msr(A), msr(B), mph(C), lnu(A)). The presence of the vga gene encoding resistance to streptogramins A was detected in one strain. All of strains were sensitive to vancomycin. However, 11 of them had reduced sensitivity to this antibiotic and eight of them were characterized by a heterogeneous resistance profile to this antibiotic. Our results clearly shows increasing threat of S. hominis caused by their multi-resistance. Moreover, these bacteria can constitute a reservoir of resistance genes for more pathogenic bacteria.
Collapse
Affiliation(s)
- Magdalena Szemraj
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Łódź, Poland.
| | - Paulina Glajzner
- Department of Biopharmacy, Medical University of Lodz, Łódź, Poland
| | - Kamila Olszowiec
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Łódź, Poland
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Łódź, Poland
| |
Collapse
|
3
|
Silvadas JD, Pillai RS, Viswanadhan Girija R, Simi CK. Green Synthesis of Urethane-Linked Tamarind Seed Xyloglucan: Thermal Stability, Antibacterial Properties, and DFT Study. Biopolymers 2025; 116:e23648. [PMID: 39705111 DOI: 10.1002/bip.23648] [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: 06/28/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/22/2024]
Abstract
This study presents a feasible, one-pot synthesis approach for the preparation of a composite biopolymer material derived from tamarind seed xyloglucan (XG) by utilizing isocyanate chemistry. Through a facile reaction process, urethane bonds are formed in XG, resulting in the formation of a crosslinked network. FTIR spectra confirm the successful urethane link formation in XG via the OH-NCO reaction, and CHN analysis provides insights into the elemental composition. The synthesized XG-urethane composite (U-XG) exhibits enhanced thermal stability compared to native XG, with an enhanced degradation temperature (T5%) of 276°C (XG marked T5% at a lower temperature of 163°C). The optimized geometric structure, hydrogen bond types, and hydrogen bond strength of the synthesized U-XG are computationally studied by density functional theory (DFT) at the B3LYP/6-31G(d,p) level. This study also investigates the antibacterial efficacy of both XG and U-XG against a panel of pathogenic bacteria, including gram-positive bacteria such as S. aureus and S. epidermidis, as well as gram-negative E. coli. The U-XG demonstrates superior antibacterial activity against S. epidermidis compared to pristine XG. This research showcases the feasibility of a one-pot synthesis approach for preparing urethane-linked XG with enhanced thermal properties and superior antibacterial activity, offering promising prospects for biomedical and antimicrobial applications.
Collapse
Affiliation(s)
- Jesna Das Silvadas
- Post Graduate & Research Department of Chemistry, Mahatma Gandhi College, Thiruvananthapuram, India
| | | | - Resmi Viswanadhan Girija
- Post Graduate & Research Department of Chemistry, Mahatma Gandhi College, Thiruvananthapuram, India
| | - Chandroth Kalyad Simi
- Post Graduate & Research Department of Chemistry, Mahatma Gandhi College, Thiruvananthapuram, India
| |
Collapse
|
4
|
Kerkman PF, de Vor L, van der Vaart TW, ten Doesschate T, Muts RM, Depelteau JS, Scheepmaker L, Ruyken M, de Haas CJ, Aerts PC, Marijnissen R, Schuurman J, Beurskens FJ, Gorlani A, Bardoel B, Rooijakkers SH. Single-cell Sequencing of Circulating Human Plasmablasts during Staphylococcus aureus Bacteremia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1644-1655. [PMID: 39451041 PMCID: PMC7616744 DOI: 10.4049/jimmunol.2300858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Staphylococcus aureus is the major cause of healthcare-associated infections, including life-threatening conditions as bacteremia, endocarditis, and implant-associated infections. Despite adequate antibiotic treatment, the mortality of S. aureus bacteremia remains high. This calls for different strategies to treat this infection. In past years, sequencing of Ab repertoires from individuals previously exposed to a pathogen emerged as a successful method to discover novel therapeutic monoclonal Abs and understand circulating B cell diversity during infection. In this paper, we collected peripheral blood from 17 S. aureus bacteremia patients to study circulating plasmablast responses. Using single-cell transcriptome gene expression combined with sequencing of variable heavy and light Ig genes, we retrieved sequences from >400 plasmablasts revealing a high diversity with >300 unique variable heavy and light sequences. More than 200 variable sequences were synthesized to produce recombinant IgGs that were analyzed for binding to S. aureus whole bacterial cells. This revealed four novel monoclonal Abs that could specifically bind to the surface of S. aureus in the absence of Ig-binding surface SpA. Interestingly, three of four mAbs showed cross-reactivity with Staphylococcus epidermidis. Target identification revealed that the S. aureus-specific mAb BC153 targets wall teichoic acid, whereas cross-reactive mAbs BC019, BC020, and BC021 target lipoteichoic acid. All mAbs could induce Fc-dependent phagocytosis of staphylococci by human neutrophils. Altogether, we characterize the active B cell responses to S. aureus in infected patients and identify four functional mAbs against the S. aureus surface, of which three cross-react with S. epidermidis.
Collapse
Affiliation(s)
- Priscilla F. Kerkman
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lisanne de Vor
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Thomas W. van der Vaart
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Thijs ten Doesschate
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Internal Medicine, Jeroen Bosch Hospital, Den Bosch, The Netherlands
| | - Remy M. Muts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jamie S. Depelteau
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lisette Scheepmaker
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Maartje Ruyken
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Carla J.C. de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Piet C. Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | | | | | | | - Bart Bardoel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Suzan H.M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
5
|
[Recommendations for antibiotic use in hospitalized preterm infants with late-onset sepsis in the neonatal intensive care unit]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:1009-1018. [PMID: 39467668 PMCID: PMC11527416 DOI: 10.7499/j.issn.1008-8830.2408024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/14/2024] [Indexed: 10/30/2024]
Abstract
Late-onset sepsis (LOS) is commonly seen in neonates who are hospitalized for extended periods, particularly in very low birth weight infants (VLBWI) and extremely low birth weight infants (ELBWI). Currently, the management of LOS in preterm infants faces dual challenges of delayed diagnosis and treatment, as well as antibiotic overtreatment. To address these issues, the Hunan Neonatal Medical Quality Control Center and the Neonatology Group of Perinatal Medical Committee of Hunan Medical Association organized a group of neonatal experts from Hunan Province to formulate recommendations based on published literature and statistical data from the Hunan Neonatal Medical Quality Control Center, as well as real-world practices in most neonatal intensive care units in Hunan Province. The group of neonatal experts proposed 15 recommendations for the diagnosis and antibiotic treatment of LOS in hospitalized preterm infants in the neonatal intensive care unit.
Collapse
|
6
|
Thänert R, Schwartz DJ, Keen EC, Hall-Moore C, Wang B, Shaikh N, Ning J, Rouggly-Nickless LC, Thänert A, Ferreiro A, Fishbein SRS, Sullivan JE, Radmacher P, Escobedo M, Warner BB, Tarr PI, Dantas G. Clinical sequelae of gut microbiome development and disruption in hospitalized preterm infants. Cell Host Microbe 2024; 32:1822-1837.e5. [PMID: 39197454 PMCID: PMC11466706 DOI: 10.1016/j.chom.2024.07.027] [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: 05/10/2024] [Revised: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Aberrant preterm infant gut microbiota assembly predisposes to early-life disorders and persistent health problems. Here, we characterize gut microbiome dynamics over the first 3 months of life in 236 preterm infants hospitalized in three neonatal intensive care units using shotgun metagenomics of 2,512 stools and metatranscriptomics of 1,381 stools. Strain tracking, taxonomic and functional profiling, and comprehensive clinical metadata identify Enterobacteriaceae, enterococci, and staphylococci as primarily exploiting available niches to populate the gut microbiome. Clostridioides difficile lineages persist between individuals in single centers, and Staphylococcus epidermidis lineages persist within and, unexpectedly, between centers. Collectively, antibiotic and non-antibiotic medications influence gut microbiome composition to greater extents than maternal or baseline variables. Finally, we identify a persistent low-diversity gut microbiome in neonates who develop necrotizing enterocolitis after day of life 40. Overall, we comprehensively describe gut microbiome dynamics in response to medical interventions in preterm, hospitalized neonates.
Collapse
Affiliation(s)
- Robert Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Drew J Schwartz
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Women's Infectious Diseases Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric C Keen
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Anna Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aura Ferreiro
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Janice E Sullivan
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Paula Radmacher
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Marilyn Escobedo
- Department of Pediatrics, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
7
|
Mao R, Zhao Q, Lu H, Yang N, Li Y, Teng D, Hao Y, Gu X, Wang J. The Marine Antimicrobial Peptide AOD with Intact Disulfide Bonds Has Remarkable Antibacterial and Anti-Biofilm Activity. Mar Drugs 2024; 22:463. [PMID: 39452871 PMCID: PMC11509771 DOI: 10.3390/md22100463] [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: 08/30/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024] Open
Abstract
American Oyster Defensin (AOD) is a marine peptide that is derived from North American mussels. It has been demonstrated to exhibit potent antimicrobial activity and high safety in both in vitro and in vivo models. In this study, to facilitate synthesis, mutants of AOD with fewer disulfide bonds were designed and subjected to structural, antimicrobial, and anti-biofilm analysis. The antimicrobial activity of AOD-derived peptides decreased after reduction in the disulfide bond, and among its three derivatives, only AOD-1 inhibited very few bacteria with a MIC value of 64 μg/mL, whereas the others had no inhibitory effect on pathogenic bacteria. The findings demonstrated that full disulfide bonds are indispensable for bactericidal activity, with the α-helix playing a pivotal role in inhibiting bacterial membranes. Furthermore, the results of the ATP, ROS, membrane potential, and membrane fluidity assays demonstrated that intracellular ATP, reactive oxygen species, and membrane fluidity were all increased, while membrane potential was reduced. This indicated that AOD resulted in the impairment of membrane fluidity and induced metabolic disorders, ultimately leading to bacterial death. The inhibitory effect of AOD on the biofilm of S. epidermidis G-81 was determined through the crystal violet and confocal microscopy. The results demonstrated that AOD exhibited a notable inhibitory impact on the biofilm of S. epidermidis G-81. The minimum biofilm inhibitory concentration of AOD on S. epidermidis G-81 was 16 μg/mL, and the minimum biofilm scavenging concentration was 32 μg/mL, which exhibited superior efficacy compared to that of lincomycin. The inhibitory effect on the primary biofilm was 90.3%, and that on the mature biofilm was 82.85%, with a dose-dependent inhibition effect. Concurrently, AOD cleared intra-biofilm organisms and reduced the number of biofilm-holding bacteria by six orders of magnitude. These data indicate that disulfide bonds are essential to the structure and activity of AOD, and AOD may potentially become an effective dual-action antimicrobial and anti-biofilm agent.
Collapse
Affiliation(s)
- Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Qingyi Zhao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Haiqiang Lu
- Enzyme Engineering Laboratory, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Yuanyuan Li
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xinxi Gu
- Enzyme Engineering Laboratory, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
8
|
Koc F, Magner C, Murphy K, Kelleher ST, Tan MH, O'Toole M, Jenkins D, Boyle J, Lavelle M, Maguire N, Ross PR, Stanton C, McMahon CJ. Gut Microbiome in Children with Congenital Heart Disease After Cardiopulmonary Bypass Surgery (GuMiBear Study). Pediatr Cardiol 2024:10.1007/s00246-024-03634-2. [PMID: 39174731 DOI: 10.1007/s00246-024-03634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
The gut microbiome of infants with congenital heart disease (CHD) undergoing cardiopulmonary bypass surgery (CPB) is at risk of profound alteration. The aim of this study was to examine the gut microbiome pre- and post-bypass surgery to explore potential implications of altered gut biodiversity. A prospective cohort study involving infants with CHD who underwent CPB was performed. Faecal samples were collected from infants alongside the collection of demographic and clinical data in order to examine gut microbiome changes before and after surgery. 16S rRNA sequencing analysis was performed on DNA isolated from stool samples to determine changes in gut microbiome composition. Thirty-three patients were recruited, with samples from thirteen of these available for final analysis. Compared with healthy, matched controls, at a genus level, pre-operative samples for infants with CHD demonstrated a higher relative abundance of Escherichia-Shigella (31% vs 2-6%) and a lower relative abundance of Bifidobacterium (13% vs 40-60%). In post-operative samples, the relative abundance of Escherichia-Shigella (35%), Enterococcus (11%), Akkermansia (6%), and Staphylococcus (5%) were higher than pre-op samples. One infant developed post-operative necrotising-enterocolitis (NEC). They displayed a marked abundance of the Enterococcus (93%) genus pre-operatively. This study demonstrates that infants with CHD have an altered gut microbiome when compared with healthy controls and there might be a possible link between an abundance of virulent species and NEC.
Collapse
Affiliation(s)
- Fatma Koc
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Claire Magner
- School of Nursing, Midwifery and Health Systems, University College Dublin, Dublin, Ireland
| | - Kiera Murphy
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Ireland
| | - Sean T Kelleher
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Mong H Tan
- Paediatric Intensive Care Unit, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Molly O'Toole
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Dominic Jenkins
- Laboratory, Children's Health Ireland at Crumlin, Crumlin, Ireland
| | - Jordan Boyle
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Marie Lavelle
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Niamh Maguire
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Paul R Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Colin J McMahon
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland.
- School of Medicine, University College Dublin, Dublin, Ireland.
- School of Health Professions Education (SHE), Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
9
|
Jara J, Alba C, Del Campo R, Fernández L, Sáenz de Pipaón M, Rodríguez JM, Orgaz B. Linking preterm infant gut microbiota to nasograstric enteral feeding tubes: exploring potential interactions and microbial strain transmission. Front Pediatr 2024; 12:1397398. [PMID: 38952433 PMCID: PMC11215057 DOI: 10.3389/fped.2024.1397398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024] Open
Abstract
Introduction Preterm birth is a growing problem worldwide. Staying at a neonatal intensive care unit (NICU) after birth is critical for the survival of preterm infants whose feeding often requires the use of nasogastric enteral feeding tubes (NEFT). These can be colonized by hospital-associated pathobionts that can access the gut of the preterm infants through this route. Since the gut microbiota is the most impactful factor on maturation of the immune system, any disturbance in this may condition their health. Therefore, the aim of this study is to assess the impact of NEFT-associated microbial communities on the establishment of the gut microbiota in preterm infants. Material and methods A metataxonomic analysis of fecal and NEFT-related samples obtained during the first 2 weeks of life of preterm infants was performed. The potential sharing of strains isolated from the same set of samples of bacterial species involved in NICU's outbreaks, was assessed by Random Amplification of Polymorphic DNA (RAPD) genotyping. Results In the samples taken 48 h after birth (NEFT-1 and Me/F1), Staphylococcus spp. was the most abundant genera (62% and 14%, respectively) and it was latter displaced to 5.5% and 0.45%, respectively by Enterobacteriaceae. Significant differences in beta diversity were detected in NEFT and fecal samples taken at day 17 after birth (NEFT-3 and F3) (p = 0.003 and p = 0.024, respectively). Significant positive correlations were found between the most relevant genera detected in NEFT-3 and F3. 28% of the patients shared at least one RAPD-PCR profile in fecal and NEFT samples and 11% of the total profiles were found at least once simultaneously in NEFT and fecal samples from the same patient. Conclusion The results indicate a parallel bacterial colonization of the gut of preterm neonates and the NEFTs used for feeding, potentially involving strain sharing between these niches. Moreover, the same bacterial RAPD profiles were found in neonates hospitalized in different boxes, suggesting a microbial transference within the NICU environment. This study may assist clinical staff in implementing best practices to mitigate the spread of pathogens that could threaten the health of preterm infants.
Collapse
Affiliation(s)
- J. Jara
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - C. Alba
- Department of Nutrition and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - R. Del Campo
- Department of Microbiology, Ramón y Cajal University Hospital and Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - L. Fernández
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - M. Sáenz de Pipaón
- Department of Neonatology, La Paz University Hospital of Madrid, Madrid, Spain
- Department of Pediatrics, Autonoma University of Madrid, Madrid, Spain
| | - J. M. Rodríguez
- Department of Nutrition and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| | - B. Orgaz
- Department of Galenic Pharmacy and Food Science, School of Veterinary Sciences, University Complutense of Madrid (UCM), Madrid, Spain
| |
Collapse
|
10
|
Canale V, Skiba-Kurek I, Klesiewicz K, Papież M, Ropek M, Pomierny B, Piska K, Koczurkiewicz-Adamczyk P, Empel J, Karczewska E, Zajdel P. Improving Activity of New Arylurea Agents against Multidrug-Resistant and Biofilm-Producing Staphylococcus epidermidis. ACS Med Chem Lett 2024; 15:369-375. [PMID: 38505856 PMCID: PMC10945555 DOI: 10.1021/acsmedchemlett.3c00536] [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: 11/28/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 03/21/2024] Open
Abstract
Multidrug-resistant (MDR) strains of Staphylococcus epidermidis (S. epidermidis), prevalent in hospital environments, contribute to increased morbidity and mortality, especially among newborns, posing a critical concern for neonatal sepsis. In response to the pressing demand for novel antibacterial therapies, we present findings from synthetic chemistry and structure-activity relationship studies focused on arylsulfonamide/arylurea derivatives of aryloxy[1-(thien-2-yl)propyl]piperidines. Through bioisosteric replacement of the sulfonamide fragment with a urea moiety, compound 25 was identified, demonstrating potent bacteriostatic activity against clinical multidrug-resistant S. epidermidis strains (MIC50 and MIC90 = 1.6 and 3.125 μg/mL). Importantly, it showed activity against linezolid-resistant strains and exhibited selectivity over mammalian cells. Compound 25 displayed antibiofilm-forming properties against clinical S. epidermidis strains and demonstrated the capacity to eliminate existing biofilm layers. Additionally, it induced complete depolarization of the bacterial membrane in clinical S. epidermidis strains. In light of these findings, targeting bacterial cell membranes with compound 25 emerges as a promising strategy in the fight against multidrug-resistant S. epidermidis strains.
Collapse
Affiliation(s)
- Vittorio Canale
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Iwona Skiba-Kurek
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Karolina Klesiewicz
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Monika Papież
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Marlena Ropek
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Bartosz Pomierny
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Kamil Piska
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | | | - Joanna Empel
- Department
of Epidemiology and Clinical Microbiology, National Medicines Institute, 30/34 Chełmska Street, 00-725 Warsaw, Poland
| | - Elżbieta Karczewska
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Paweł Zajdel
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| |
Collapse
|
11
|
Papachatzi E, Gkentzi D, Tzifas S, Dassios T, Dimitriou G. Daptomycin Use for Persistent Coagulase-Negative Staphylococcal Bacteremia in a Neonatal Intensive Care Unit. Antibiotics (Basel) 2024; 13:254. [PMID: 38534689 DOI: 10.3390/antibiotics13030254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
During the last two decades, the incidence of late-onset sepsis (LOS) has increased due to improved survival of premature neonates. Persistent bacteremia (PB) in LOS is defined as more than two positive blood cultures obtained on different calendar days during the same infectious episode. Although rare, PB should be treated aggressively to prevent adverse outcomes. Daptomycin, a lipopeptide antibiotic, has been used in neonates with persistent coagulase-negative staphylococci (CoNS) bacteremia with promising results, but studies reporting on the efficacy and safety of the agent are scarce. The purpose of this study was to evaluate the efficacy and safety of daptomycin use for persistent CoNS bacteremia in a neonatal cohort. This is a retrospective, observational, single-center study of neonates treated with daptomycin during 2011-2022 in the Tertiary Neonatal Intensive Care Unit (NICU) of the University General Hospital of Patras, Greece. For the years 2011-2022, there were 3.413 admissions to the NICU. During the last 3 years (2020-2022)-the active epidemiological surveillance period-123 infants (out of 851 admissions, 14.4%) developed CoNS bacteremia (LOS). During the study period, twelve infants with PB were treated with daptomycin. They had a median gestational age of 32 weeks (IQR 31-34) and mean (SD) birth weight of 1.840 (867) grams. CoNS bacteremia isolates were s. epidermidis (50%), s. haemolyticus (20%), s. hominis (20%) and s. warneri (10%). The decision to start daptomycin (6 mg/kg/dose twice daily) was taken on median day 10 (ΙQR 7-15) of infection. None of the infants had focal complications or meningitis. Daptomycin therapy caused no renal, hepatic, muscular or gastrointestinal adverse events. One neonate developed seizures, and one death occurred due to multiple complications of prematurity. Most infants (11/12) were successfully treated and eventually had negative blood culture. Daptomycin monotherapy showed an adequate cure rate in premature neonates with persistent CoNS bacteremia in a tertiary NICU. In our study, daptomycin was effective and well tolerated; the safety profile, however, needs to be confirmed in larger studies and randomized controlled trials.
Collapse
Affiliation(s)
- Eleni Papachatzi
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Despoina Gkentzi
- Department of Pediatrics, University General Hospital of Patras, 26504 Patras, Greece
| | - Sotiris Tzifas
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Theodore Dassios
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Gabriel Dimitriou
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
- Department of Pediatrics, University General Hospital of Patras, 26504 Patras, Greece
| |
Collapse
|
12
|
Garg PM, Pittman I, Yi J, Shetty A, Taylor C, Reddy K, Inder TE, Varshney N, Hillegass WB, Garg PP. Clinical correlates of cerebellar injury in preterm infants with surgical necrotizing enterocolitis. J Neonatal Perinatal Med 2024; 17:705-716. [PMID: 39269856 PMCID: PMC11489020 DOI: 10.3233/npm-240022] [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] [Indexed: 09/15/2024]
Abstract
BACKGROUND The preterm infants are at risk of cerebellar injury and the risk factors for necrotizing enterocolitis (NEC) associated cerebellar injury are not fully understood. AIM Determine the risk factors of cerebellar injury in infants with surgical necrotizing enterocolitis (NEC). METHODS Retrospective study compared clinical/pathological information between surgical NEC infants with and those without cerebellar injury detected on brain MRI obtained at term equivalent age. Cerebellar Injury patterns that we identified on MRI brain were cerebellar hemorrhage, siderosis and/or cerebellar volume loss. RESULTS Cerebellar injury (21/65, 32.3%) in preterm infants with NEC was associated with patent ductus arteriosus (PDA) (18/21(85.7%) vs. 25/44(56.8%); p = 0.021), blood culture positive sepsis (13/21 (61.9%) vs. 11/44 (25%); p = 0.004) following NEC, predominantly grew gram positive bacteria (9/21(42.9%) vs. 4/44(9.1%); p = 0.001), greater red cell transfusion, higher rates of cholestasis following NEC and differences in intestinal histopathology (more hemorrhagic and reparative lesions) on univariate analysis. Those with cerebellar injury had higher grade white matter injury (14/21 (66.7%) vs. 4/44(9.1%) p = 0.0005) and higher-grade ROP (70.6% vs. 38.5%; p = 0.027) than those without cerebellar injury.On multilogistic regression, the positive blood culture sepsis (OR 3.9, CI 1.1-13.7, p = 0.03), PDA (OR 4.5, CI 1.0-19.9, p = 0.04) and severe intestinal pathological hemorrhage (grade 3-4) (OR 16.9, CI 2.1-135.5, p = 0.007) were independently associated with higher risk of cerebellar injury. CONCLUSION Preterm infants with surgical NEC with positive blood culture sepsis, PDA, and severe intestinal hemorrhagic lesions (grade 3-4) appear at greater risk for cerebellar injury.
Collapse
Affiliation(s)
- P M Garg
- Department of Pediatrics/Neonatology, Wake Forest University, Winston Salem, NC, USA
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - I Pittman
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - J Yi
- Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, NC, USA
| | - A Shetty
- Department of Pediatrics/Infectious Disease, Wake Forest University, Winston Salem, NC, USA
| | - C Taylor
- Department of Radiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - K Reddy
- Department of Radiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - T E Inder
- Children Hospital of Orange County, University of California, Irvine, Orange, CA, USA
| | - N Varshney
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - W B Hillegass
- Department of Data Science, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - P P Garg
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
13
|
Garg PM, Pittman I, Taylor C, Reddy K, Varshney N, Hillegass WB, Shetty A, Yi J, Inder T, Garg P. Clinical Correlates of Cerebellar Injury in Preterm Infants with Surgical Necrotizing Enterocolitis. RESEARCH SQUARE 2023:rs.3.rs-3720723. [PMID: 38168331 PMCID: PMC10760219 DOI: 10.21203/rs.3.rs-3720723/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Objective Determine the risk factors of cerebellar injury in infants with surgical necrotizing enterocolitis (NEC). Methods Retrospective study compared clinical/pathological information between surgical NEC infants with and those without cerebellar injury. Results Infants with cerebellar injury (21/65, 32.3%) had significantly more hemorrhagic and the reparative lesions on the intestinal histopathology, had patent ductus arteriosus (PDA) more often, received red cell transfusion frequently, had blood culture positive sepsis and grew gram positive organisms more often and had cholestasis frequently following NEC than those without cerebellar injury. On multilogistic regression, the positive blood culture sepsis (OR 3.9, CI 1.1-13.7, p = 0.03), PDA (OR 4.5, CI 1.0-19.9, p = 0.04) and severe hemorrhage (grade 3-4)(OR 16.9, CI 2.1-135.5, p = 0.007) were independently associated with higher risk of cerebellar injury. Conclusion The cerebellar injury was most likely associated with positive blood culture sepsis following NEC, PDA, and severe hemorrhage lesions (grade 3-4) in infants with surgical NEC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Joe Yi
- 3. Frank Porter Graham Child Development Institute, University of North Carolina
| | | | | |
Collapse
|
14
|
Huang YH, Yeh YR, Lien RI, Chiang MC, Huang YC. Molecular characteristics and clinical features of Staphylococcus epidermidis healthcare-associated late-onset bacteremia among infants hospitalized in neonatal intensive care units. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:1214-1225. [PMID: 37709633 DOI: 10.1016/j.jmii.2023.08.005] [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: 01/17/2023] [Revised: 08/16/2023] [Accepted: 08/27/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Though Staphylococcus epidermidis was the most common pathogen of late-onset sepsis (LOS) in neonatal intensive care units (NICUs), there haves been scanty reports on molecular epidemiology of S. epidermidis isolates from infants stayed in NICU and on correlation of molecular characteristics with clinical features in these infants. METHODS We collected and characterized S. epidermidis bloodstream isolates from infants hospitalized in NICU of a medical center in Taiwan between 2018 and 2020. Medical records of these infants were retrospectively reviewed. RESULTS A total of 107 isolates identified from 78 episodes of bacteremia in 75 infants were included for analysis. Of the 78 isolates (episodes), 24 pulsotypes, 11 sequence types (STs), and 5 types of staphylococcal chromosomal cassette (type I-V) were identified. ST59 and its single locus variant ST1124 (37.2%) comprised the most common strain, followed by ST35 (14.1%), ST2 (11.5%), and ST89 (10.3%). All but 5 isolates (73/78, 93.6%) belonged to clonal complex (CC) 2. Comparing infants infected with genetically different strains, the patients with underlying immune disease were significantly associated with ST2 infection (P = 0.021), while no statistically significant differences were found in terms of clinical and laboratory characteristics. Only 3.8% of the isolates were susceptible to oxacillin. CONCLUSIONS More than 90% of S. epidermidis bloodstream isolates from infants in NICU in Taiwan were resistant to oxacillin. Though diverse, more than 90% of the isolates (episodes) belonged to CC2. No statistically significant differences were found in terms of clinical characteristics among the infants infected with genetically different strains.
Collapse
Affiliation(s)
- Yi-Hsuan Huang
- Department of Medical Education, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan, Taiwan
| | - Yu-Rou Yeh
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Rey-In Lien
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan, Taiwan
| | - Ming-Chou Chiang
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan, Taiwan
| | - Yhu-Chering Huang
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|
15
|
Ghori NUH, Mullally CA, Nicol MP, Currie A, Hibbert J, Payne MS, Patole S, Strunk T. Skin-Microbiome Assembly in Preterm Infants during the First Three Weeks of Life and Impact of Topical Coconut Oil Application. Int J Mol Sci 2023; 24:16626. [PMID: 38068949 PMCID: PMC10706365 DOI: 10.3390/ijms242316626] [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: 10/31/2023] [Revised: 11/18/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
The structure and function of infant skin is not fully developed until 34 weeks of gestation, and this immaturity is associated with risk of late-onset sepsis (LOS). Topical coconut oil improves preterm-infant skin integrity and may reduce LOS. However, data on early-life skin-microbiome succession and potential effects of emollient skin care in preterm infants are scarce. We therefore collected skin-microbiome samples from the ear, axilla, and groin on days 1, 7, 14, and 21 from preterm infants born <30 weeks of gestation as part of a randomized clinical trial of standard skin care vs. topical coconut oil. We found that within-sample microbiome diversity was highest on day 1 after birth, with a subsequent decline and emergence of Staphylococcus genus dominance from day 7. Moreover, microbiome assembly was less diverse in infants receiving coconut oil vs. standard skin care. Our study provides novel data on preterm-infant skin-microbiome composition and highlights the modifying potential of emollient skin care.
Collapse
Affiliation(s)
- Noor-Ul-Huda Ghori
- Division of Infection and Immunity, School of Biomedical Sciences and The Marshall Centre, The University of Western Australia, Perth 6009, Australia (M.P.N.)
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth 6009, Australia
| | - Christopher A. Mullally
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth 6009, Australia
- Centre of Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Mark P. Nicol
- Division of Infection and Immunity, School of Biomedical Sciences and The Marshall Centre, The University of Western Australia, Perth 6009, Australia (M.P.N.)
| | - Andrew Currie
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth 6009, Australia
- Centre of Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Julie Hibbert
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth 6009, Australia
- Centre of Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Matthew S. Payne
- Division of Obstetrics and Gynecology, School of Medicine, The University of Western Australia, Perth 6009, Australia
| | - Sanjay Patole
- Neonatal Directorate, King Edward Memorial Hospital for Women, Child and Adolescent Health Service, Perth 6008, Australia
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth 6009, Australia
| | - Tobias Strunk
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth 6009, Australia
- Neonatal Directorate, King Edward Memorial Hospital for Women, Child and Adolescent Health Service, Perth 6008, Australia
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth 6009, Australia
| |
Collapse
|
16
|
Williams MR, Bagood MD, Enroth TJ, Bunch ZL, Jiang N, Liu E, Almoughrabie S, Khalil S, Li F, Brinton S, Cech NB, Horswill AR, Gallo RL. Staphylococcus epidermidis activates keratinocyte cytokine expression and promotes skin inflammation through the production of phenol-soluble modulins. Cell Rep 2023; 42:113024. [PMID: 37610872 PMCID: PMC10586132 DOI: 10.1016/j.celrep.2023.113024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/05/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023] Open
Abstract
Staphylococcus epidermidis is a common microbe on human skin and has beneficial functions in the skin microbiome. However, under conditions of allergic inflammation, the abundance of S. epidermidis increases, establishing potential danger to the epidermis. To understand how this commensal may injure the host, we investigate phenol-soluble modulin (PSM) peptides produced by S. epidermidis that are similar to peptides produced by Staphylococcus aureus. Synthetic S. epidermidis PSMs induce expression of host defense genes and are cytotoxic to human keratinocytes. Deletion mutants of S. epidermidis lacking these gene products support these observations and further show that PSMs require the action of the EcpA bacterial protease to induce inflammation when applied on mouse skin with an intact stratum corneum. The expression of PSMδ from S. epidermidis is also found to correlate with disease severity in patients with atopic dermatitis. These observations show how S. epidermidis PSMs can promote skin inflammation.
Collapse
Affiliation(s)
- Michael R Williams
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Michelle D Bagood
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Timothy J Enroth
- Department of Veterans Affairs Denver Health Care System, Denver, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Zoie L Bunch
- Department of Chemistry and Biochemistry, University of North Carolina Greensboro, Greensboro, NC 27402, USA
| | - Nina Jiang
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Edward Liu
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Samia Almoughrabie
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Shadi Khalil
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Samantha Brinton
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Nadja B Cech
- Department of Chemistry and Biochemistry, University of North Carolina Greensboro, Greensboro, NC 27402, USA
| | - Alexander R Horswill
- Department of Veterans Affairs Denver Health Care System, Denver, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
17
|
Szemraj M, Glajzner P, Sienkiewicz M. Decreased susceptibility to vancomycin and other mechanisms of resistance to antibiotics in Staphylococcus epidermidis as a therapeutic problem in hospital treatment. Sci Rep 2023; 13:13629. [PMID: 37604965 PMCID: PMC10442409 DOI: 10.1038/s41598-023-40866-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/17/2023] [Indexed: 08/23/2023] Open
Abstract
Multidrug-resistant coagulase-negative staphylococci represent a real therapeutic challenge. The aim of the study was to emphasize the importance of heteroresistance to vancomycin presence in methicillin-resistant strains of S. epidermidis. The research comprised 65 strains of S. epidermidis. Heteroresistance to vancomycin was detected with the use of the agar screening method with Brain Heart Infusion and a population profile analysis (PAP test). In addition, types of cassettes and genes responsible for resistance to antibiotics for 22 multidrug resistant strains were determined. Our investigations showed that 56 of 65 S. epidermidis strains were phenotypically resistant to methicillin. The tested strains were mostly resistant to erythromycin, gentamicin, clindamycin, and ciprofloxacin. Six strains showed decreased susceptibility to vancomycin and their heterogeneous resistance profiles were confirmed with the PAP test. All tested multi-resistant strains exhibited the mecA gene. More than half of them possessed type IV cassettes. ant(4')-Ia and aac(6')/aph(2''), ermC and tetK genes were most commonly found. The described phenomenon of heteroresistance to vancomycin in multidrug resistant bacteria of the Staphylococcus genus effectively inhibits a therapeutic effect of treatment with this antibiotic. That is why it is so important to search for markers that will enable to identify heteroresistance to vancomycin strains under laboratory conditions.
Collapse
Affiliation(s)
- Magdalena Szemraj
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Lodz, Poland.
| | - Paulina Glajzner
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
18
|
Licona G, Ballot D, Moon TD, Banerjee R, Amorim G, Agthe AG, Weitkamp JH. Multidrug-Resistant Bacterial Infections Among Very Low Birthweight Infants With Late-Onset Sepsis in Johannesburg, South Africa. Open Forum Infect Dis 2023; 10:ofad362. [PMID: 37564739 PMCID: PMC10411040 DOI: 10.1093/ofid/ofad362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Background An estimated 2.4 million babies died within the first 28 days of life in 2020. The third leading cause of neonatal death continues to be neonatal sepsis. Sepsis-causing bacterial pathogens vary temporally and geographically and, with a rise in multidrug-resistant organisms (MDROs), pose a threat to the neonatal population. Methods This was a single-center, retrospective study of very low birth weight (VLBW) infants with late-onset sepsis (LOS) admitted to a neonatal unit in South Africa. We aimed to calculate the prevalence of multidrug-resistant (MDR) infections in this population. The data collected included demographic and clinical characteristics, length of hospital stay, risk factors for MDRO and mortality, and microbiology results. Logistic regression was used to assess the association between prespecified risk factors with MDR infections and mortality. Results Of 2570 VLBW infants admitted, 34% had LOS, of which 33% was caused by MDROs. Infection with Acinetobacter spp., Pseudomonas spp., extended-spectrum beta-lactamase Klebsiella spp., or Escherichia coli was associated with the highest mortality in the LOS cohort. Infants with congenital infections (adjusted odds ratio [aOR], 5.13; 95% CI, 1.19-22.02; P = .028) or a history of necrotizing enterocolitis (aOR, 2.17; 95% CI, 1.05-4.49; P = .037) were at significantly higher risk for MDR infections. Conclusions More than one-third of LOS cases in VLBW infants were caused by MDROs in this study. MDR infections cause substantial neonatal mortality. Antimicrobial stewardship programs, infection control protocols, and ongoing surveillance are needed to prevent further emergence and spread of MDR infections worldwide.
Collapse
Affiliation(s)
- Genesis Licona
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Daynia Ballot
- Division of Neonatology, Department of Pediatrics, Charlotte Maxeke Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Troy D Moon
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ritu Banerjee
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gustavo Amorim
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alexander G Agthe
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jörn-Hendrik Weitkamp
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
19
|
Gravina G, Ardalan M, Chumak T, Nilsson AK, Ek JC, Danielsson H, Svedin P, Pekny M, Pekna M, Sävman K, Hellström A, Mallard C. Proteomics identifies lipocalin-2 in neonatal inflammation associated with cerebrovascular alteration in mice and preterm infants. iScience 2023; 26:107217. [PMID: 37496672 PMCID: PMC10366453 DOI: 10.1016/j.isci.2023.107217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/07/2023] [Accepted: 06/22/2023] [Indexed: 07/28/2023] Open
Abstract
Staphylococcus (S.) epidermidis is the most common nosocomial coagulase-negative staphylococci infection in preterm infants. Clinical signs of infection are often unspecific and novel markers to complement diagnosis are needed. We investigated proteomic alterations in mouse brain after S. epidermidis infection and in preterm infant blood. We identified lipocalin-2 (LCN2) as a crucial protein associated with cerebrovascular changes and astrocyte reactivity in mice. We further proved that LCN2 protein expression was associated with endothelial cells but not astrocyte reactivity. By combining network analysis and differential expression approaches, we identified LCN2 linked to blood C-reactive protein levels in preterm infants born <28 weeks of gestation. Blood LCN2 levels were associated with similar alterations of cytokines and chemokines in both infected mice and human preterm infants with increased levels of C-reactive protein. This experimental and clinical study suggests that LCN2 may be a marker of preterm infection/inflammation associated with cerebrovascular changes and neuroinflammation.
Collapse
Affiliation(s)
- Giacomo Gravina
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Translational Neuropsychiatric Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tetyana Chumak
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders K. Nilsson
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joakim C. Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hanna Danielsson
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Sach’s Children’s and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Pernilla Svedin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- University of Newcastle, Newcastle, NSW, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Marcela Pekna
- University of Newcastle, Newcastle, NSW, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Laboratory of Regenerative Neurobiology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Karin Sävman
- Department of Pediatrics, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden
- Region Västra Götaland, Department of Neonatology, The Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
20
|
Burke TL, Rupp ME, Fey PD. Staphylococcus epidermidis. Trends Microbiol 2023; 31:763-764. [PMID: 36682998 DOI: 10.1016/j.tim.2023.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/21/2023]
Affiliation(s)
- Taylor L Burke
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - M E Rupp
- Department of Internal Medicine, Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - P D Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
21
|
Jaloustre M, Cohen R, Biran V, Decobert F, Layese R, Audureau E, Le Saché N, Chevallier M, Boukhris MR, Bolot P, Caeymaex L, Tauzin M. Determinants of morbidity and mortality related to health care-associated primary bloodstream infections in neonatal intensive care units: a prospective cohort study from the SEPREVEN trial. Front Pediatr 2023; 11:1170863. [PMID: 37325351 PMCID: PMC10264575 DOI: 10.3389/fped.2023.1170863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Background Health care-associated primary bloodstream infections (BSIs), defined as not secondary to an infection at another body site, including central line-associated BSI, are a leading cause of morbidity and mortality in patients in neonatal intensive care units (NICUs). Our objective was to identify factors associated with severe morbidity and mortality after these infections in neonates in NICUs. Methods This ancillary study of the SEPREVEN trial included neonates hospitalized ≥2 days in one of 12 French NICUs and with ≥ 1 BSI during the 20-month study period. BSIs (all primary and health care-associated) were diagnosed in infants with symptoms suggestive of infection and classified prospectively as possible (one coagulase-negative staphylococci (CoNS)-growing blood culture) or proven (two same CoNS, or ≥1 recognized pathogen-growing blood culture). BSI consequences were collected prospectively as moderate morbidity (antibiotic treatment alone) or severe morbidity/mortality (life-saving procedure, permanent damage, prolonged hospitalization, and/or death). Results Of 557 BSIs identified in 494 patients, CoNS accounted for 378/557 (67.8%) and recognized bacterial or fungal pathogens for 179/557 (32.1%). Severe morbidity/mortality was reported in 148/557 (26.6%) BSIs. Independent factors associated with severe morbidity/mortality were corrected gestational age <28 weeks (CGA) at infection (P < .01), fetal growth restriction (FGR) (P = .04), and proven pathogen-related BSI vs. CoNS-related BSI (P < .01). There were no differences in severe morbidity and mortality between proven and possible CoNS BSIs. In possible BSI, S. epidermidis was associated with a lower risk of severe morbidity than other CoNS (P < .01), notably S. capitis and S. haemolyticus. Conclusions In BSIs in the NICU, severe morbidity/mortality was associated with low CGA at infection, FGR, and proven pathogen-related BSIs. When only one blood culture was positive, severe morbidity/mortality were less frequent if it grew with S. epidermidis compared to other CoNS. Further studies to help distinguish real CoNS BSIs from contaminations are needed. Study registration ClinicalTrials.gov (NCT02598609).
Collapse
Affiliation(s)
- Morgane Jaloustre
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Creteil, Creteil, France
| | - Robert Cohen
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Creteil, Creteil, France
- Faculty of Medicine, University Paris Est Creteil, Creteil, France
- Groupe de Pathologie Infectieuse Pédiatrique, Paris, France
| | - Valérie Biran
- Neonatal Intensive Care Unit, APHP, CHU Robert Debré, Paris, France
| | - Fabrice Decobert
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Creteil, Creteil, France
| | - Richard Layese
- Assistance Publique-Hôpitaux de Paris AP-HP, Hôpital Henri Mondor, Unité de Recherche Clinique (URC Mondor), Creteil, France
- University Paris Est Creteil, INSERM, IMRB, CEpiA Team, Creteil, France
| | - Etienne Audureau
- Assistance Publique-Hôpitaux de Paris AP-HP, Hôpital Henri Mondor, Unité de Recherche Clinique (URC Mondor), Creteil, France
- University Paris Est Creteil, INSERM, IMRB, CEpiA Team, Creteil, France
| | - Nolwenn Le Saché
- Pediatric Intensive Care and Neonatal Medicine, Bicêtre Hospital, AP-HP, Le Kremlin-Bicêtre, France
| | - Marie Chevallier
- Neonatal Intensive Care Unit, CHU Grenoble Alpes, Grenoble, France
| | | | - Pascal Bolot
- Neonatal Intensive Care Unit, Centre Hospitalier de Saint-Denis, Saint-Denis, France
| | - Laurence Caeymaex
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Creteil, Creteil, France
- Faculty of Medicine, University Paris Est Creteil, Creteil, France
| | - Manon Tauzin
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Creteil, Creteil, France
| | | |
Collapse
|
22
|
Martínez Chamás J, Isla MI, Zampini IC. Antibacterial and Antibiofilm Activity of Different Species of Fabiana sp. Extract Obtained via Maceration and Ultrasound-Assisted Extraction against Staphylococcus epidermidis. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12091830. [PMID: 37176887 PMCID: PMC10180551 DOI: 10.3390/plants12091830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/15/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Staphylococcus epidermidis is an opportunistic pathogen that, under certain conditions, can induce aggravated infectious processes, mainly in immunosuppressed patients. Moreover, S. epidermidis is one of the leading causes of medical device- and implant-associated infections and is also recognized as a canonical biofilm producer. Fabiana punensis, F. densa and F. patagonica are three medicinal plants that grow in arid environments in Argentina (Altoandina, Puna, Prepuna and Monte regions). In this work, we studied the antimicrobial activity of alcoholic extracts of these plant species obtained via maceration (M) and ultrasound-assisted extraction (UAE) against S. epidermidis. In addition, the antibiofilm activity of the F. densa extract was also evaluated. It was found that the extracts obtained via M did not present differences with those obtained via UAE regarding the chemical profile. F. densa showed the lowest minimum inhibitory concentration (MIC) value (75 µg GAE/mL). At concentrations higher than the MIC, the extract induced the release of cellular constituents. At the concentration of 1/8× MIC, the extract inhibited biofilm formation by 78%, reducing metabolic activity by 67%. On the other hand, it presented a low percentage of preformed biofilm removal. In all assays, gallic acid (GA) has been used as a reference antimicrobial compound. Finally, it was shown via microscopy visualization that the extract reduces adhesion to hydrophobic and hydrophilic surfaces. Thus, F. densa extracts could potentially be used for the antibiotic treatment of infections produced by S. epidermidis or as an inhibitor agent of production biofilm, avoiding infections caused by medical devices.
Collapse
Affiliation(s)
- José Martínez Chamás
- Laboratorio de Investigación de Productos Naturales (LIPRON), Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV-CONICET-UNT), Facultad de Ciencias Naturales e IML, Universidad Nacional de Tucumán, San Lorenzo 1469, San Miguel de Tucumán PC:4000, Tucumán, Argentina
| | - María Inés Isla
- Laboratorio de Investigación de Productos Naturales (LIPRON), Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV-CONICET-UNT), Facultad de Ciencias Naturales e IML, Universidad Nacional de Tucumán, San Lorenzo 1469, San Miguel de Tucumán PC:4000, Tucumán, Argentina
| | - Iris Catiana Zampini
- Laboratorio de Investigación de Productos Naturales (LIPRON), Instituto de Bioprospección y Fisiología Vegetal (INBIOFIV-CONICET-UNT), Facultad de Ciencias Naturales e IML, Universidad Nacional de Tucumán, San Lorenzo 1469, San Miguel de Tucumán PC:4000, Tucumán, Argentina
| |
Collapse
|
23
|
Hellmann KT, Challagundla L, Gray BM, Robinson DA. Improved Genomic Prediction of Staphylococcus epidermidis Isolation Sources with a Novel Polygenic Score. J Clin Microbiol 2023; 61:e0141222. [PMID: 36840569 PMCID: PMC10035303 DOI: 10.1128/jcm.01412-22] [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: 09/23/2022] [Accepted: 01/22/2023] [Indexed: 02/25/2023] Open
Abstract
Staphylococcus epidermidis infections can be challenging to diagnose due to the species frequent contamination of clinical specimens and indolent course of infection. Nevertheless, S. epidermidis is the major cause of late-onset sepsis among premature infants and of intravascular infection in all age groups. Prior work has shown that bacterial virulence factors, antimicrobial resistances, and strains have up to 80% in-sample accuracy to distinguish hospital from community sources, but are unable to distinguish true bacteremia from blood culture contamination. Here, a phylogeny-informed genome-wide association study of 88 isolates was used to estimate effect sizes of particular genomic variants for isolation sources. A "polygenic score" was calculated for each isolate as the summed effect sizes of its repertoire of genomic variants. Predictive models of isolation sources based on polygenic scores were tested with in-samples and out-samples from prior studies of different patient populations. Polygenic scores from accessory genes (AGs) distinguished hospital from community sources with the highest accuracy to date, up to 98% for in-samples and 65% to 91% for various out-samples, whereas scores from single nucleotide polymorphisms (SNPs) had lower accuracy. Scores from AGs and SNPs achieved the highest in-sample accuracy to date, up to 76%, in distinguishing infection from contaminant sources within a hospital. Model training and testing data sets with more similar population structures resulted in more accurate predictions. This study reports the first use of a polygenic score for predicting a complex bacterial phenotype and shows the potential of this approach for enhancing S. epidermidis diagnosis.
Collapse
Affiliation(s)
- K. Taylor Hellmann
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lavanya Challagundla
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Barry M. Gray
- Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| | - D. Ashley Robinson
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
24
|
França A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics (Basel) 2023; 12:antibiotics12030554. [PMID: 36978421 PMCID: PMC10044083 DOI: 10.3390/antibiotics12030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Infections are one of the most significant complications of neonates, especially those born preterm, with sepsis as one of the principal causes of mortality. Coagulase-negative staphylococci (CoNS), a group of staphylococcal species that naturally inhabit healthy human skin and mucosa, are the most common cause of late-onset sepsis, especially in preterms. One of the risk factors for the development of CoNS infections is the presence of implanted biomedical devices, which are frequently used for medications and/or nutrient delivery, as they serve as a scaffold for biofilm formation. The major concerns related to CoNS infections have to do with the increasing resistance to multiple antibiotics observed among this bacterial group and biofilm cells’ increased tolerance to antibiotics. As such, the treatment of CoNS biofilm-associated infections with antibiotics is increasingly challenging and considering that antibiotics remain the primary form of treatment, this issue will likely persist in upcoming years. For that reason, the development of innovative and efficient therapeutic measures is of utmost importance. This narrative review assesses the current challenges and emerging diagnostic tools and therapies for the treatment of CoNS biofilm-associated infections, with a special focus on late-onset sepsis.
Collapse
Affiliation(s)
- Angela França
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- LABBELS—Associate Laboratory in Biotechnology and Bioengineering and Microelectromechanical Systems, Braga and Guimarães, Portugal
| |
Collapse
|
25
|
Humberg A, Neuenburg L, Boeckel H, Fortmann MI, Härtel C, Herting E, Hinrichs H, Rademacher F, Harder J. Antimicrobial skin peptides in premature infants: Comparison with term infants and impact of perinatal factors. Front Immunol 2023; 14:1093340. [PMID: 36923410 PMCID: PMC10009099 DOI: 10.3389/fimmu.2023.1093340] [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: 12/15/2022] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
Introduction Preterm infants have an immature epidermis barrier function that may lead to an increased permeability to pathogens. On the surface of the human skin, antimicrobial peptides (AMPs) are important molecules of the innate immune system, have broad antimicrobial properties, and provide an essential role in integrity of the microbiome. Given the marked susceptibility of preterm infants to infection, we hypothesize a decreased expression of AMPs on the skin of preterm infants. Materials and methods In a prospective single-center study with 35 preterm and 20 term infants, we analyzed skin rinsing probes for the presence of the AMPs psoriasin (S100A7) and ribonuclease 7 (RNase 7) via enzyme-linked immunosorbent assay. Samples were taken from preterm infants < 34 0/7 weeks gestational age (mean ± SD gestational age, 28.8 ± 2.4 weeks) on days 0, 7, 14, and 28 after birth. Term infants (> 36 6/7 weeks) (controls) were washed on days 0 and 28. Results Psoriasin and RNase 7 were both expressed on skin of preterm and term infants and increased in concentration significantly over time. RNase 7 was more expressed in term infants on day 0 [preterm = 1.1 (0.7-2.9) vs. term = 2.0 (1.1-3.4) ng/ml, p = 0.017]. On day 28, premature infants showed higher values of psoriasin [preterm = 10.9 (5.6-14.2) vs. term = 6.3 (3.4-9.0) ng/ml, p < 0.001]. Notably, preterm infants with infectious or inflammatory context driven by histological proof of chorioamnionitis and early-onset or late-onset sepsis had higher concentrations of psoriasin as compared with non-affected preterm infants. After exclusion of infants with inflammatory hit, median concentrations of RNase 7 and psoriasin did not differ between preterm and full-term infants on days 0 and 28. Discussion Psoriasin and RNase 7 concentrations increase over time on the skin of newborn infants and seem to play a role in the first defense against infection. This is of particularly interest as the role of AMPs on a maturing skin microbiome and its possible new prevention strategies is unclear and needs to be determined.
Collapse
Affiliation(s)
- Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Muenster, Muenster, Germany
- Institute of Medical Biometry and Statistics, University of Luebeck, Luebeck, Germany
| | - Lisa Neuenburg
- Department of Pediatrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Hannah Boeckel
- Department of Pediatrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital, Wuerzburg, Germany
| | - Egbert Herting
- Department of Pediatrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Heilwig Hinrichs
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Franziska Rademacher
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, Kiel, Germany
| |
Collapse
|
26
|
Harvey EJ, Ashiru-Oredope D, Hill LF, Demirjian A. Need for standardized vancomycin dosing for coagulase-negative staphylococci in hospitalized infants. Clin Microbiol Infect 2023; 29:10-12. [PMID: 36195185 DOI: 10.1016/j.cmi.2022.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/01/2022] [Accepted: 09/19/2022] [Indexed: 01/26/2023]
Affiliation(s)
- Eleanor J Harvey
- Healthcare-Associated Infection, Fungal, Antimicrobial Resistance, Antimicrobial Use & Sepsis Division, United Kingdom Health Security Agency, London, United Kingdom
| | - Diane Ashiru-Oredope
- Healthcare-Associated Infection, Fungal, Antimicrobial Resistance, Antimicrobial Use & Sepsis Division, United Kingdom Health Security Agency, London, United Kingdom; National Institute for Health and Care Research Health Protection Unit in Healthcare-Associated Infections and Antimicrobial Resistance, Imperial College, London, United Kingdom
| | - Louise F Hill
- Centre for Neonatal and Paediatric Infection, Institute for Infection and Immunity, St George's, University of London, United Kingdom
| | - Alicia Demirjian
- Healthcare-Associated Infection, Fungal, Antimicrobial Resistance, Antimicrobial Use & Sepsis Division, United Kingdom Health Security Agency, London, United Kingdom; National Institute for Health and Care Research Health Protection Unit in Healthcare-Associated Infections and Antimicrobial Resistance, Imperial College, London, United Kingdom; Department of Paediatric Infectious Diseases & Immunology, Evelina London Children's Hospital, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom; Faculty of Life Sciences & Medicine, King's College London, United Kingdom.
| |
Collapse
|
27
|
Kennedy KM, de Goffau MC, Perez-Muñoz ME, Arrieta MC, Bäckhed F, Bork P, Braun T, Bushman FD, Dore J, de Vos WM, Earl AM, Eisen JA, Elovitz MA, Ganal-Vonarburg SC, Gänzle MG, Garrett WS, Hall LJ, Hornef MW, Huttenhower C, Konnikova L, Lebeer S, Macpherson AJ, Massey RC, McHardy AC, Koren O, Lawley TD, Ley RE, O'Mahony L, O'Toole PW, Pamer EG, Parkhill J, Raes J, Rattei T, Salonen A, Segal E, Segata N, Shanahan F, Sloboda DM, Smith GCS, Sokol H, Spector TD, Surette MG, Tannock GW, Walker AW, Yassour M, Walter J. Questioning the fetal microbiome illustrates pitfalls of low-biomass microbial studies. Nature 2023; 613:639-649. [PMID: 36697862 PMCID: PMC11333990 DOI: 10.1038/s41586-022-05546-8] [Citation(s) in RCA: 171] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/09/2022] [Indexed: 01/26/2023]
Abstract
Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.
Collapse
Affiliation(s)
- Katherine M Kennedy
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Marcus C de Goffau
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Wellcome Sanger Institute, Cambridge, UK
| | - Maria Elisa Perez-Muñoz
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marie-Claire Arrieta
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, South Korea
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Thorsten Braun
- Department of Obstetrics and Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederic D Bushman
- Department of Microbiology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel Dore
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jonathan A Eisen
- Department of Evolution and Ecology, University of California, Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephanie C Ganal-Vonarburg
- Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michael G Gänzle
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
- Department of Medicine and Division of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Chair of Intestinal Microbiome, ZIEL-Institute for Food and Health, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Curtis Huttenhower
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liza Konnikova
- Departments of Pediatrics and Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Andrew J Macpherson
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Ruth C Massey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Alice Carolyn McHardy
- Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), Hannover Braunschweig site, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Trevor D Lawley
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jeroen Raes
- VIB Center for Microbiology, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thomas Rattei
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eran Segal
- Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Harry Sokol
- Gastroenterology Department, AP-HP, Saint Antoine Hospital, Centre de Recherche Saint-Antoine, CRSA, INSERM and Sorbonne Université, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gerald W Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alan W Walker
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Moran Yassour
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
- Department of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
28
|
Sethi G, Varghese RP, Krishna R. Identification and design of a multi-epitope subunit vaccine against the opportunistic pathogen Staphylococcus epidermidis: An immunoinformatics approach. J Biomol Struct Dyn 2022; 40:13859-13871. [PMID: 34726118 DOI: 10.1080/07391102.2021.1997819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Staphylococcus epidermidis is one of the major causes of nosocomial infections around the globe that leads to a high rate of mortality and morbidity in both immunocompromised patients and preterm infants. Despite the alarming increase in multi-drug resistance, no promising vaccines are readily available against this pathogen. Thus, the present study is focused on designing a multi-epitope subunit vaccine using five antigenic proteins of S. epidermidis through an immunoinformatics approach. The final vaccine comprised B-cell, HTL, and CTL binding epitopes followed by Lipoprotein LprA adjuvant added at N-terminal to augment the immunogenicity. Physicochemical assessment of the vaccine reveals the antigenic and non-allergic nature. The vaccine structure was designed, refined, validated, and disulfide engineered to obtain the best model. Molecular docking and dynamics simulation of the proposed vaccine with toll-like receptors (TLR-2 and TLR-4) showed strong and stable interactions. MM-PBSA analysis was implemented as an efficient tool to determine the intermolecular binding free energies of the system. The vaccine was subjected to immune simulation to predict its immunogenic profile. In silico cloning suggested that the proposed vaccine can be expressed efficiently in E.coli. Furthermore, in vivo animal experiment is needed to determine the effectiveness of the in silico designed vaccine.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Guneswar Sethi
- Centre for Bioinformatics, Pondicherry University, Puducherry, India
| | | | - Ramadas Krishna
- Centre for Bioinformatics, Pondicherry University, Puducherry, India
| |
Collapse
|
29
|
Abstract
This narrative review seeks to examine the relationships between bacterial microbiomes and infectious disease. This is achieved by detailing how different human host microbiomes develop and function, from the earliest infant acquisitions of maternal and environmental species through to the full development of microbiomes by adulthood. Communication between bacterial species or communities of species within and outside of the microbiome is a factor in both maintenance of homeostasis and management of threats from the external environment. Dysbiosis of this homeostasis is key to understanding the development of disease states. Several microbiomes and the microbiota within are used as prime examples of how changes in species composition, particularly at the phylum level, leads to such diverse conditions as inflammatory bowel disease (IBD), type 2 diabetes, psoriasis, Parkinson's disease, reflux oesophagitis and others. The review examines spatial relationships between microbiomes to understand how dysbiosis in the gut microbiome in particular can influence diseases in distant host sites via routes such as the gut-lung, gut-skin and gut-brain axes. Microbiome interaction with host processes such as adaptive immunity is increasingly identified as critical to developing the capacity of the immune system to react to pathogens. Dysbiosis of essential bacteria involved in modification of host substrates such as bile acid components can result in development of Crohn's disease, small intestine bacterial overgrowth, hepatic cancer and obesity. Interactions between microbiomes in distantly located sites are being increasingly being identified, resulting in a 'whole of body' effect by the combined host microbiome.
Collapse
Affiliation(s)
- Jim Manos
- Infection, Immunity and InflammationSchool of Medical SciencesFaculty of Medicine and HealthThe Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| |
Collapse
|
30
|
Garg PM, Paschal JL, Ansari MAY, Block D, Inagaki K, Weitkamp JH. Clinical impact of NEC-associated sepsis on outcomes in preterm infants. Pediatr Res 2022; 92:1705-1715. [PMID: 35352003 DOI: 10.1038/s41390-022-02034-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To determine risk factors and outcomes of necrotizing enterocolitis (NEC)-associated sepsis in infants with NEC. METHODS A retrospective review comparing demographic and clinical information in infants with and without NEC-associated sepsis (defined as positive blood culture at the time of NEC onset). RESULTS A total of 209 infants with medical (n = 98) and surgical NEC (n = 111) had a median gestational age of 27 weeks (IQR 25; 30.5) and a median birth weight of 910 g [IQR 655; 1138]. Fifty of 209 (23.9%) infants had NEC-associated sepsis. Infants with NEC-associated sepsis had lower median GA (26.4 vs. 27.4 weeks; p = 0.01), lower birth weight (745 vs. 930 g; p = 0.009), were more likely mechanically ventilated [p < 0.001], received dopamine [p < 0.001], had more evidence of acute kidney injury [60% vs. 38.4%, p = 0.01], longer postoperative ileus (16 [13.0; 22.0] vs. 12 [8; 16] days; p = 0.006), higher levels of C-reactive protein, lower platelet counts, longer hospitalization compared to infants without NEC-associated sepsis. On multivariate regression, cholestasis was an independent risk factor for NEC-associated sepsis (OR 2.94; 95% CI 1.1-8.8, p = 0.038). CONCLUSION NEC-associated sepsis was associated with greater hemodynamic support, acute kidney injury, longer postoperative ileus, and hospitalization on bivariate analysis, and cholestasis was associated with higher odds of sepsis on multi regression analysis. IMPACT NEC-associated sepsis was present in 24% of infants with NEC. Gram-positive bacteria, Gram-negative bacteria, and Candida were found in 15.3%, 10.5%, and 2.8% of cases, respectively. Infants with NEC-associated sepsis had a greater inflammatory response (CRP levels), received more blood transfusion before NEC onset, frequently needed assisted ventilation ionotropic support, and had acute kidney injury after NEC onset. NEC infants with Gram-negative sepsis had higher portal venous gas, received more platelet transfusions before NEC onset, and had higher CRP levels and lower median lymphocyte counts at 24 h after NEC onset than those with Gram-positive sepsis.
Collapse
Affiliation(s)
- Parvesh Mohan Garg
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Jaslyn L Paschal
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Md Abu Yusuf Ansari
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Danielle Block
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kengo Inagaki
- Department of Pediatrics/Infectious Disease, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jörn-Hendrik Weitkamp
- Department of Pediatrics/Neonatology, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN, USA
| |
Collapse
|
31
|
Abstract
Neonatal late-onset sepsis (LOS) continues to threaten morbidity and mortality in the NICU and poses ongoing diagnostic and therapeutic challenges. Early recognition of clinical signs, rapid evaluation, and prompt initiation of treatment are critical to prevent life-threatening deterioration. Preterm infants-born at ever-decreasing gestational ages-are at particularly high risk for life-long morbidities and death. This changing NICU population necessitates continual reassessments of diagnostic and preventive measures and evidence-based treatment for LOS. The clinical presentation of LOS is varied and nonspecific. Despite ongoing research, reliable, specific laboratory biomarkers facilitating early diagnosis are lacking. These limitations drive an ongoing practice of liberal initiation of empiric antibiotics among infants with suspected LOS. Subsequent promotion of multidrug-resistant microorganisms threatens the future of antimicrobial therapy and puts preterm and chronically ill infants at even higher risk of nosocomial infection. Efforts to identify adjunctive therapies counteracting sepsis-driven hyperinflammation and sepsis-related functional immunosuppression are ongoing. However, most approaches have either failed to improve LOS prognosis or are not yet ready for clinical application. This article provides an overview of the epidemiology, risk factors, diagnostic tools, and treatment options of LOS in the context of increasing numbers of extremely preterm infants. It addresses the question of whether LOS could be identified earlier and more precisely to allow for earlier and more targeted therapy and discusses rational approaches to antibiotic therapy to avoid overuse. Finally, this review elucidates the necessity of long-term follow-up of infants with a history of LOS.
Collapse
Affiliation(s)
- Sarah A. Coggins
- Division of Neonatology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kirsten Glaser
- Division of Neonatology, Department of Women’s and Children’s Health, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
32
|
Kolesnichenko SI, Kadyrova IA, Lavrinenko AV, Zhumadilova ZA, Avdienko OV, Vinogradskaya YV, Fominykh YA, Panibratec LG, Akhmaltdinova LL. Mortality Risk Factors of Early Neonatal Sepsis During COVID-19 Pandemic. Infect Drug Resist 2022; 15:6307-6316. [PMID: 36337929 PMCID: PMC9635391 DOI: 10.2147/idr.s390723] [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: 09/21/2022] [Accepted: 10/22/2022] [Indexed: 11/05/2022] Open
Abstract
Purpose This study aimed to determine predisposing factors for negative outcome in infants with early neonatal sepsis during COVID-19. Patients and Methods A prospective cohort study of 172 newborns up to 4 days diagnosed with neonatal sepsis was carried out in Karaganda (Kazakhstan). The microbiological examination was used to identify a causative agent of bloodstream infection. ELISA was performed to determine the total anti-SARS-CoV-2 antibodies. Gestational age, mode of delivery, birth weight, C-reactive protein and procalcitonin levels, comorbidities, type of pathogen, duration of hospitalization and mother’s infection diseases were used for statistical analysis. Results Mortality in infants with neonatal sepsis was 22% (38/172). Anti-SARS-CoV-2 antibodies were detected in 68.3% of the newborns. Culture-negative ELBW infants have a 5.3-fold higher risk of death (p<0.001). Low gestational age and a shorter period of hospitalization were statistically associated with fatality. CRP is generally higher in deceased children (p=0.002). Necrotizing enterocolitis (p<0.001), pneumonia (p=0.009) and anemia (p=0.016) were significantly associated with negative outcome. And, 31.4% of the infants with sepsis had positive blood cultures. The leading cause of sepsis in newborns was CoNS – 57%. Conclusion During COVID-19 pandemic neonatal sepsis mortality was associated with low birth weight, gestational age, and comorbidities as in non-pandemic time. The relationship between COVID-19 in the mother and neonatal mortality was not found. However, anti-SARS-CoV-2 antibodies were detected in more than half of newborns.
Collapse
Affiliation(s)
- Svetlana I Kolesnichenko
- Shared Resource Laboratory, Research Centre, Karaganda Medical University, Karaganda, Kazakhstan,Correspondence: Svetlana I Kolesnichenko, Shared Resource Laboratory, Research Centre, Karaganda Medical University, 40 Gogol St, Karaganda, 100008, Kazakhstan, Tel +7 702 599 0225, Email
| | - Irina A Kadyrova
- Shared Resource Laboratory, Research Centre, Karaganda Medical University, Karaganda, Kazakhstan
| | - Alyona V Lavrinenko
- Shared Resource Laboratory, Research Centre, Karaganda Medical University, Karaganda, Kazakhstan
| | - Zhibek A Zhumadilova
- Shared Resource Laboratory, Research Centre, Karaganda Medical University, Karaganda, Kazakhstan
| | - Olga V Avdienko
- Shared Resource Laboratory, Research Centre, Karaganda Medical University, Karaganda, Kazakhstan
| | | | - Yevgeniy A Fominykh
- Regional Clinical Hospital of Karaganda, Perinatal Center No. 2, Karaganda, Kazakhstan
| | - Lyudmila G Panibratec
- Regional Clinical Hospital of Karaganda, Perinatal Center No. 2, Karaganda, Kazakhstan
| | - Lyudmila L Akhmaltdinova
- Shared Resource Laboratory, Research Centre, Karaganda Medical University, Karaganda, Kazakhstan,National Scientific Cardiac Surgery Center, Nur-Sultan, Kazakhstan
| |
Collapse
|
33
|
Skovdal SM, Jørgensen NP, Meyer RL. JMM Profile: Staphylococcus epidermidis. J Med Microbiol 2022; 71. [DOI: 10.1099/jmm.0.001597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus epidermidis
is the most abundant commensal bacterium of human skin. Despite protecting us from foreign invaders,
S. epidermidis
itself exploits human vulnerability when given the opportunity. Such opportunities arise when patients are immunocompromised or when biomedical implants present an opportunity to colonize the surface and form biofilms.
S. epidermidis
is one of the most frequently isolated organisms from implanted devices and from bloodstream infections. However,
S. epidermidis
infections are often recalcitrant to antibiotics because of biofilm-associated antibiotic tolerance. Furthermore, the emergence and spread of nearly pan-resistant strains is a considerable health concern. Symptoms can be subclinical, making diagnosis challenging, and treatment with antibiotics is inefficient. For now, infection prevention remains the best strategy available.
Collapse
Affiliation(s)
- Sandra Maria Skovdal
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | | | - Rikke Louise Meyer
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Aarhus, Denmark
- Department of Biology, Faculty of Natural Sciences, Aarhus University, Aarhus, Denmark
| |
Collapse
|
34
|
de Vor L, Beudeker CR, Flier A, Scheepmaker LM, Aerts PC, Vijlbrief DC, Bekker MN, Beurskens FJ, van Kessel KPM, de Haas CJC, Rooijakkers SHM, van der Flier M. Monoclonal antibodies effectively potentiate complement activation and phagocytosis of Staphylococcus epidermidis in neonatal human plasma. Front Immunol 2022; 13:933251. [PMID: 35967335 PMCID: PMC9372458 DOI: 10.3389/fimmu.2022.933251] [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: 04/30/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
Central line associated bloodstream infections (CLABSI) with Staphylococcus epidermidis are a major cause of morbidity in neonates, who have an increased risk of infection because of their immature immune system. As especially preterm neonates suffer from antibody deficiency, clinical studies into preventive therapies have thus far focused on antibody supplementation with pooled intravenous immunoglobulins from healthy donors (IVIG) but with little success. Here we study the potential of monoclonal antibodies (mAbs) against S. epidermidis to induce phagocytic killing by human neutrophils. Nine different mAbs recognizing Staphylococcal surface components were cloned and expressed as human IgG1s. In binding assays, clones rF1, CR5133 and CR6453 showed the strongest binding to S. epidermidis ATCC14990 and CR5133 and CR6453 bound the majority of clinical isolates from neonatal sepsis (19 out of 20). To study the immune-activating potential of rF1, CR5133 and CR6453, bacteria were opsonized with mAbs in the presence or absence of complement. We observed that activation of the complement system is essential to induce efficient phagocytosis of S. epidermidis. Complement activation and phagocytic killing could be enhanced by Fc-mutations that improve IgG1 hexamerization on cellular surfaces. Finally, we studied the ability of the mAbs to activate complement in r-Hirudin neonatal plasma conditions. We show that classical pathway complement activity in plasma isolated from neonatal cord blood is comparable to adult levels. Furthermore, mAbs could greatly enhance phagocytosis of S. epidermidis in neonatal plasma. Altogether, our findings provide insights that are crucial for optimizing anti-S. epidermidis mAbs as prophylactic agents for neonatal CLABSI.
Collapse
Affiliation(s)
- Lisanne de Vor
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Coco R. Beudeker
- Department of Paediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Anne Flier
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lisette M. Scheepmaker
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Piet C. Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daniel C. Vijlbrief
- Department of Neonatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mireille N. Bekker
- Department of Obstetrics, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Kok P. M. van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Carla J. C. de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Michiel van der Flier
- Department of Paediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
35
|
Abstract
Our skin is the interface through which we mediate lifelong interactions with our surrounding environment. Initial development of the skin's epidermis, adnexal structures, and barrier function is necessary for normal cutaneous microbial colonization, immune development, and prevention of disease. Early life microbial exposures can have unique and long-lasting impacts on skin health. The identity of neonatal skin microbes and the context in which they are first encountered, i.e., through a compromised skin barrier or in conjunction with cutaneous inflammation, can have additional short- and long-term health consequences. Here, we discuss key attributes of infant skin and endogenous and exogenous factors that shape its relationship to the early life cutaneous microbiome, with a focus on their clinical implications.
Collapse
Affiliation(s)
- Laura R Dwyer
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
36
|
Muk T, Brunse A, Henriksen NL, Aasmul-Olsen K, Nguyen DN. Glucose supply and glycolysis inhibition shape the clinical fate of Staphylococcus epidermidis-infected preterm newborns. JCI Insight 2022; 7:157234. [PMID: 35503431 DOI: 10.1172/jci.insight.157234] [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] [Received: 12/03/2021] [Accepted: 04/29/2022] [Indexed: 11/17/2022] Open
Abstract
Preterm infants are susceptible to bloodstream infection by coagulase-negative staphylococci (CONS) that can lead to sepsis. High parenteral glucose supplement is commonly used to support their growth and energy expenditure, but may exceed endogenous regulation during infection, causing dysregulated immune response and clinical deterioration. Using a preterm piglet model of neonatal CONS sepsis induced by Staphylococcus epidermidis infection, we demonstrate the delicate interplay between immunity and glucose metabolism to regulate the host infection response. Circulating glucose levels, glycolysis and inflammatory response to infection are closely connected across the states of tolerance, resistance and immunoparalysis. Further, high parenteral glucose provision during infection induces hyperglycemia, elevated glycolysis and inflammation, leading to metabolic acidosis and sepsis, whereas glucose restricted individuals are clinically unaffected with increased gluconeogenesis to maintain moderate hypoglycemia. Finally, standard glucose supply maintaining normoglycemia or pharmacological glycolysis inhibition enhances bacterial clearance and dampens inflammation but fails to prevent sepsis. Our results uncover how blood glucose and glycolysis controls circulating immune responses, in turn determining the clinical fate of CONS infected preterm individuals. This questions the current practice of parenteral glucose supply for preterm infants during infection.
Collapse
Affiliation(s)
- Tik Muk
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Anders Brunse
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Nicole L Henriksen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Karoline Aasmul-Olsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Duc Ninh Nguyen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
37
|
Towards Understanding the Lymph Node Response to Skin Infection with Saprophytic Staphylococcus epidermidis. Biomedicines 2022; 10:biomedicines10051021. [PMID: 35625758 PMCID: PMC9138836 DOI: 10.3390/biomedicines10051021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/21/2022] [Accepted: 04/28/2022] [Indexed: 11/20/2022] Open
Abstract
In individuals with lymphedema, diabetic foot, or other diseases, infections with saprophytes are common. The response of major cell subpopulations in the draining lymph nodes to skin infection with Staphylococcus epidermidis was assessed using the rat model. After massive subepidermal infection, a cytometric evaluation showed an increase in cytotoxic and helper T lymphocytes and major subpopulations of the innate immune response. Three weeks later, signs of inflammation reduction with an increase in the content of memory T helper lymphocytes and effector memory T cytotoxic lymphocytes were observed. After skin re-infection, a rapid response of cytotoxic, helper, and memory T lymphocytes, memory B lymphocytes and plasmablasts, and macrophages was detected. In addition, a reduction in the number of naïve B lymphocytes, activated MHC class II+ cells, and some cells of the innate immune system was observed. T regulatory lymphocyte response after the initial and secondary S. epidermidis skin infection was not detected. The morphometric evaluation showed significant changes in the main cell subpopulations in each functional zone of the node and then confirmed the efficient elimination of the administered antigen, as evidenced by the observations on day 28. Notably, after re-infection, the cellular response did not exceed the level after the initial infection and was reduced in many cell subpopulations. Understanding how the lymph nodes eliminate S. epidermidis can provide valuable insights into creating immunological therapies against infections with saprophytes.
Collapse
|
38
|
Implementation of a Vancomycin Dose-Optimization Protocol in Neonates: Impact on Vancomycin Exposure, Biological Parameters, and Clinical Outcomes. Antimicrob Agents Chemother 2022; 66:e0219121. [PMID: 35465728 DOI: 10.1128/aac.02191-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Vancomycin dosing used in neonates results frequently in insufficient concentrations. A vancomycin dose-optimization protocol consisting of an individualization of loading and maintenance doses (administered during continuous infusion) through a previously validated pharmacokinetic model was implemented in our center. This monocenter retrospective study aimed to compare vancomycin average concentration (Cavg) in the therapeutic range (15 to 25 mg/L) and biological and clinical parameters before and after implementation of this protocol. A total of 60 and 59 courses of vancomycin treatment in 45 and 49 patients were analyzed in groups before and after implementation, respectively. Initial vancomycin Cavg were more frequently in the therapeutic range in the group after implementation (74.6% versus 28.3%, P < 0.001), with 1.6-fold higher Cavg (20.3 [17.0-22.2] mg/L versus 12.9 [11.3-17.0] mg/L, P < 0.001). Considering all Cavg during longitudinal therapeutic drug monitoring (TDM), the frequency of therapeutic Cavg was higher in the group after implementation (74.8% [n = 103] versus 31% [n = 116], P < 0.001). The dose optimization protocol was also associated with a reduced time to obtain a negative blood culture (P < 0.001) and fewer antibiotic switches (P = 0.025), without increasing the frequency of nephrotoxicity. Clinical outcomes also appeared to be improved, with less periventricular leukomalacia (P = 0.021), trended toward less respiratory instability (P = 0.15) and a shorter duration of vasoactive drug use (P = 0.18) for neonates receiving personalized doses of vancomycin. This personalized vancomycin dose protocol improves vancomycin exposure in neonates, with good safety, and suggests an improvement in biological and clinical outcomes.
Collapse
|
39
|
Doualeh M, Payne M, Litton E, Raby E, Currie A. Molecular Methodologies for Improved Polymicrobial Sepsis Diagnosis. Int J Mol Sci 2022; 23:ijms23094484. [PMID: 35562877 PMCID: PMC9104822 DOI: 10.3390/ijms23094484] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 12/19/2022] Open
Abstract
Polymicrobial sepsis is associated with worse patient outcomes than monomicrobial sepsis. Routinely used culture-dependent microbiological diagnostic techniques have low sensitivity, often leading to missed identification of all causative organisms. To overcome these limitations, culture-independent methods incorporating advanced molecular technologies have recently been explored. However, contamination, assay inhibition and interference from host DNA are issues that must be addressed before these methods can be relied on for routine clinical use. While the host component of the complex sepsis host–pathogen interplay is well described, less is known about the pathogen’s role, including pathogen–pathogen interactions in polymicrobial sepsis. This review highlights the clinical significance of polymicrobial sepsis and addresses how promising alternative molecular microbiology methods can be improved to detect polymicrobial infections. It also discusses how the application of shotgun metagenomics can be used to uncover pathogen/pathogen interactions in polymicrobial sepsis cases and their potential role in the clinical course of this condition.
Collapse
Affiliation(s)
- Mariam Doualeh
- Centre for Molecular Medicine & Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia;
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA 6009, Australia
- Women and Infants Research Foundation, Perth, WA 6008, Australia;
| | - Matthew Payne
- Women and Infants Research Foundation, Perth, WA 6008, Australia;
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, WA 6008, Australia
| | - Edward Litton
- Intensive Care Unit, Fiona Stanley Hospital, Murdoch, WA 6150, Australia;
- Intensive Care Unit, St. John of God Hospital, Subiaco, WA 6009, Australia
| | - Edward Raby
- State Burns Unit, Fiona Stanley Hospital, Murdoch, WA 6150, Australia;
- Microbiology Department, Path West Laboratory Medicine, Murdoch, WA 6150, Australia
| | - Andrew Currie
- Centre for Molecular Medicine & Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia;
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA 6009, Australia
- Women and Infants Research Foundation, Perth, WA 6008, Australia;
- Correspondence: ; Tel.: +61-(08)-9360-7426
| |
Collapse
|
40
|
Jovandaric MZ, Stefanovic S, Babic S, Milenkovic SJ, Babovic IR. Importance of Staphylococcus epidermidis findings in the blood and cerebrospinal fluid of a full-term newborn: a case report. J Int Med Res 2022; 50:3000605221093216. [PMID: 35435035 PMCID: PMC9019340 DOI: 10.1177/03000605221093216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Methicillin-resistant Staphylococcus epidermidis (MRS) predominantly colonizes the skin and mucous membranes of humans and other animals. We describe the case of a male newborn of gestational age 39 weeks whose primary and repeated blood cultures and cerebrospinal fluid samples isolated MRS. The choice and duration of antibiotic therapy were determined by the clinical presentation, infection parameters, and results of bacteriological analyses of blood and cerebrospinal fluid samples obtained from the newborn on the day 5 of life. After 28 days of antibiotic therapy for sepsis accompanied by meningitis, the newborn was discharged home without sequelae.
Collapse
Affiliation(s)
- Miljana Z Jovandaric
- Clinic for Gynecology and Obstetrics, Department of Neonatology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Slobodanka Stefanovic
- Department of Medical Microbiology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Sandra Babic
- Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, Belgrade, Serbia
| | - Svetlana J Milenkovic
- Clinic for Gynecology and Obstetrics, Department of Neonatology, University Clinical Center of Serbia, Belgrade, Serbia
| | | |
Collapse
|
41
|
Sethi G, Sethi S, Krishna R. Multi-epitope based vaccine design against Staphylococcus epidermidis: A subtractive proteomics and immunoinformatics approach. Microb Pathog 2022; 165:105484. [PMID: 35301068 DOI: 10.1016/j.micpath.2022.105484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/06/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Staphylococcus epidermidis has emerged as a major contributor of nosocomial infections across the world. With the increased rate of emerging resistant and previously undefined infectious diseases, there is a growing need to develop a novel vaccine possessing required immunogenic properties. The adopted reverse vaccinology approach identified "IMPNQILTI" of LysM domain protein, "YSYTYTIDA" of staphylococcal secretory antigen SsaA, and "YNYDANTGQ" neutral metalloproteinaseas potential peptides for vaccine design. The 9-mer epitope of target proteins is antigenic, virulent, surface-exposed, non-allergenic, and conserved across various strains of S. epidermidis. Protein-protein interactions study indicated the involvement of target proteins in major biological pathways for S. epidermidis pathogenesis. Protein-peptide docking was performed, and population coverage analysis showed significant interactions of T-cell epitopes with the HLA-binding molecules while covering 90.58% of the world's population. Further, a multi-epitope vaccine of 177 amino acids long was constructed. Docking with Toll-like receptor (TLR-2) molecule confirmed the effective interaction of the vaccine with the receptor. The vaccine efficiency in generating an effective immune response in the host was evaluated by immune simulation. Finally, in silico cloning confirmed that the constructed vaccine can be efficiently expressed in E. coli. However, the designed vaccine needs experimental validation to determine the effectiveness and immunogenicity profile, which will ensure an active immunity against S. epidermidis.
Collapse
Affiliation(s)
- Guneswar Sethi
- Department of Bioinformatics, Pondicherry University, Puducherry, 605014, India.
| | - Satyanarayan Sethi
- Central Institute of Freshwater Aquaculture, Research Institute, Bhubaneswar, 751002, India.
| | - Ramadas Krishna
- Department of Bioinformatics, Pondicherry University, Puducherry, 605014, India.
| |
Collapse
|
42
|
Lemme-Dumit JM, Song Y, Lwin HW, Hernandez-Chavez C, Sundararajan S, Viscardi RM, Ravel J, Pasetti MF, Ma B. Altered Gut Microbiome and Fecal Immune Phenotype in Early Preterm Infants With Leaky Gut. Front Immunol 2022; 13:815046. [PMID: 35280991 PMCID: PMC8905226 DOI: 10.3389/fimmu.2022.815046] [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: 11/14/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal barrier immaturity, or "leaky gut", is the proximate cause of susceptibility to necrotizing enterocolitis in preterm neonates. Exacerbated intestinal immune responses, gut microbiota dysbiosis, and heightened barrier injury are considered primary triggers of aberrant intestinal maturation in early life. Inordinate host immunity contributes to this process, but the precise elements remain largely uncharacterized, leaving a significant knowledge gap in the biological underpinnings of gut maturation. In this study, we investigated the fecal cytokine profile and gut microbiota in a cohort of 40 early preterm infants <33-weeks-gestation to identify immune markers of intestinal barrier maturation. Three distinct microbiota types were demonstrated to be differentially associated with intestinal permeability (IP), maternal breast milk feeding, and immunological profiles. The Staphylococcus epidermidis- and Enterobacteriaceae-predominant microbiota types were associated with an elevated IP, reduced breast milk feeding, and less defined fecal cytokine profile. On the other hand, a lower IP was associated with increased levels of fecal IL-1α/β and a microbiota type that included a wide array of anaerobes with expanded fermentative capacity. Our study demonstrated the critical role of both immunological and microbiological factors in the early development of intestinal barrier that collectively shape the intestinal microenvironment influencing gut homeostasis and postnatal intestinal maturation in early preterm newborns.
Collapse
Affiliation(s)
- Jose M Lemme-Dumit
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Hnin Wai Lwin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Claudia Hernandez-Chavez
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sripriya Sundararajan
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rose M Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcela F Pasetti
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
43
|
Molecular Diagnosis, Antimicrobial Resistance Profiles and Disease Patterns of Gram-Positive Pathogens Recovered from Clinical Infections in Major Ha’il Hospitals. MICROBIOLOGY RESEARCH 2022. [DOI: 10.3390/microbiolres13010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Nosocomial resistance in staphylococci and enterococci is challenging. The aim of this work was to conduct a multipoint study using molecular detections, antimicrobial resistances profiles, patient demographics and disease patterns for objective assessments of Staphilococcus aureus and other Gram-positive pathogens recovered from clinical infections in the Ha’il region. We have surveyed 188 non-duplicate Gram-positives against 22 antimicrobials for molecular-differentiation, resistance, patient demographics, and disease patterns from January–April 2021. According to definitions for acquired resistance, Staphylococcus aureus was the most frequent with multidrug resistant (65.4%), where MRSA was 60% (n = 72 out of 121). In age-identified patients, 43% were seniors ≥50 years, 38% 21–49 years, and 19% 0–20 years. In gender-identified patients, 63% were males, and 37% were females. While 25% of specimens were from the ICU, the majority (60%) of specimens were from surgical infection in other wards. Staphylococcus epidermidis was the second (15.4%) species of infection identified with 81% from bloodstream infections at the ICU and other wards. The majority of S. epidermidis patients (69%) were seniors ≥50 years, while other age groups 0–20 and 21–49 each had 14% isolates. Although S. epidermidis was multidrug-resistant, it was susceptible to many drugs. Enterococcus faecalis (13%) ranked third with two major infections; bloodstream (64%) and urinary-tract infections (36%) in mainly seniors (86%). Its isolates were fully resistant to oxacillin, penicillin, cefoxitin, and cefotaxime but nearly 100% susceptible to seven others. Other Gram-positive bacteria (6%) were susceptible to many antibiotics. The use of combinations of objective criteria is a well thought out approach in infection control. While the low-frequency of Gram-positives is an impressive achievement, future large-scale investigations should include all private hospitals, clinics and other cities over a longer sampling time to gain more insights. Although geriatric susceptibility can be justified by age and comorbidities, the staphylococcal infections in young adults and children is a global concern and warrants more vertical studies.
Collapse
|
44
|
Look Who's Talking: Host and Pathogen Drivers of Staphylococcus epidermidis Virulence in Neonatal Sepsis. Int J Mol Sci 2022; 23:ijms23020860. [PMID: 35055041 PMCID: PMC8775791 DOI: 10.3390/ijms23020860] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
Preterm infants are at increased risk for invasive neonatal bacterial infections. S. epidermidis, a ubiquitous skin commensal, is a major cause of late-onset neonatal sepsis, particularly in high-resource settings. The vulnerability of preterm infants to serious bacterial infections is commonly attributed to their distinct and developing immune system. While developmentally immature immune defences play a large role in facilitating bacterial invasion, this fails to explain why only a subset of infants develop infections with low-virulence organisms when exposed to similar risk factors in the neonatal ICU. Experimental research has explored potential virulence mechanisms contributing to the pathogenic shift of commensal S. epidermidis strains. Furthermore, comparative genomics studies have yielded insights into the emergence and spread of nosocomial S. epidermidis strains, and their genetic and functional characteristics implicated in invasive disease in neonates. These studies have highlighted the multifactorial nature of S. epidermidis traits relating to pathogenicity and commensalism. In this review, we discuss the known host and pathogen drivers of S. epidermidis virulence in neonatal sepsis and provide future perspectives to close the gap in our understanding of S. epidermidis as a cause of neonatal morbidity and mortality.
Collapse
|
45
|
Rampelotto RF, Coelho SS, Franco LN, Mota ADD, Calegari LF, Jacobi LF, Hörner R. Coagulase-negative staphylococci isolates from blood cultures of newborns in a tertiary hospital in southern Brazil. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | - Laísa Nunes Franco
- University Federal of Santa Maria, Brazil; University Federal of Santa Maria, Brazil
| | - Augusto Dias da Mota
- University Federal of Santa Maria, Brazil; University Federal of Santa Maria, Brazil
| | | | | | - Rosmari Hörner
- University Federal of Santa Maria, Brazil; University Federal of Santa Maria, Brazil
| |
Collapse
|
46
|
Viability-Resolved Metagenomics Reveals Antagonistic Colonization Dynamics of Staphylococcus epidermidis Strains on Preterm Infant Skin. mSphere 2021; 6:e0053821. [PMID: 34523979 PMCID: PMC8550141 DOI: 10.1128/msphere.00538-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Preterm infants are at increased risk of infections caused by coagulase-negative staphylococci (CoNS) that colonize skin. Technical barriers in sequencing low-microbial-biomass skin swabs from preterm infants hinder attempts to gain a strain-level understanding of CoNS colonization dynamics within their developing skin microbiome. Here, the microbiome of five skin sites and available stool was studied from four preterm infants hospitalized over their first 2 months of life. We used propidium monoazide treatment of samples to enrich for the viable microbiome and metagenomic shotgun sequencing to resolve species and strains. The microbiome of different skin sites overlapped with each other, was dominated by the CoNS species Staphylococcus epidermidis and Staphylococcus capitis, and was distinct from stool. Species diversity on skin increased over time despite antibiotic exposure. Evidence of antagonism between the most common S. epidermidis strains, ST2 and ST59, included negative relationships for species correlation networks and in situ replication rates and that ST2 colonized skin earlier but was often replaced by ST59 over time. Experiments done with reference isolates showed that ST2 produced more biofilm than ST59 on plastic surfaces, which was reduced in mixed culture. We also discovered that a rare S. epidermidis strain, ST5, grew rapidly in stool in association with Stenotrophomonas maltophilia from a suspected episode of infection. Viability treatment of samples and moderate throughput shotgun sequencing provides strain-level information about CoNS colonization dynamics of preterm infant skin that ultimately might be exploited to prevent infections. IMPORTANCE The skin is a habitat for microbes that commonly infect preterm infants, but the use of sequencing for fine-scale study of the microbial communities of skin that develop in these infants has been limited by technical barriers. We treated skin swabs of preterm infants with a photoreactive dye that eliminates DNA from nonviable microbes and then sequenced the remaining DNA. We found that two strains of the most common species, Staphylococcus epidermidis, showed an antagonistic relationship on skin by cooccurring with different species, replicating fastest in different samples, and dominating skin sites at different times. Representatives of these strains also differed in their ability to stick to plastic surfaces—an important pathogenicity trait of this species. Our study shows the feasibility of gaining detailed information about strain colonization dynamics from this difficult-to-sequence body site of preterm infants, which might be used to guide novel approaches to prevent infections.
Collapse
|
47
|
Skiba-Kurek I, Nowak P, Empel J, Tomczak M, Klepacka J, Sowa-Sierant I, Żak I, Pomierny B, Karczewska E. Evaluation of Biofilm Formation and Prevalence of Multidrug-Resistant Strains of Staphylococcus epidermidis Isolated from Neonates with Sepsis in Southern Poland. Pathogens 2021; 10:pathogens10070877. [PMID: 34358027 PMCID: PMC8308537 DOI: 10.3390/pathogens10070877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 01/11/2023] Open
Abstract
Staphylococcus epidermidis strains play an important role in nosocomial infections, especially in the ones associated with biofilm formation on medical devices. The paper was aimed at analyzing the mechanisms of antibiotic resistance and confirming the biofilm-forming ability among S. epidermidis strains isolated from the blood of hospitalized newborns. Genetic analysis of resistance mechanism determinants included multiplex PCR detection of mecA, ermA, ermB, ermC, msrA, and mef genes. Biofilm analysis comprised phenotypic and genotypic methods including Christensen and Freeman methods and PCR detection of the icaADB gene complex. Among the tested S. epidermidis strains, 89% of the isolates were resistant to methicillin, 67%—to erythromycin, 53%—to clindamycin, 63%—to gentamicin, and 23%—to teicoplanin, while all the strains were susceptible to vancomycin and linezolid. The mecA gene was detected in 89% of the isolates, the ermC gene was the most common and present among 56% of the strains, while the msrA gene was observed in 11% isolates. Eighty-five percent of the strains were described as biofilm-positive by phenotypic methods and carried the icaADB gene cluster. Multidrug resistance and the biofilm-forming ability in most of the strains tested may contribute to antimicrobial therapy failure (p < 0.05).
Collapse
Affiliation(s)
- Iwona Skiba-Kurek
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Krakow, Poland; (I.S.-K.); (P.N.)
| | - Paweł Nowak
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Krakow, Poland; (I.S.-K.); (P.N.)
| | - Joanna Empel
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, Chełmska 30/34 Street, 00-725 Warsaw, Poland; (J.E.); (M.T.)
| | - Magdalena Tomczak
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, Chełmska 30/34 Street, 00-725 Warsaw, Poland; (J.E.); (M.T.)
| | - Joanna Klepacka
- Department of Clinical Microbiology, University Children’s Hospital of Krakow, Wielicka 256 Street, 30-663 Krakow, Poland; (J.K.); (I.S.-S.); (I.Ż.)
| | - Iwona Sowa-Sierant
- Department of Clinical Microbiology, University Children’s Hospital of Krakow, Wielicka 256 Street, 30-663 Krakow, Poland; (J.K.); (I.S.-S.); (I.Ż.)
| | - Iwona Żak
- Department of Clinical Microbiology, University Children’s Hospital of Krakow, Wielicka 256 Street, 30-663 Krakow, Poland; (J.K.); (I.S.-S.); (I.Ż.)
| | - Bartosz Pomierny
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland;
| | - Elżbieta Karczewska
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Krakow, Poland; (I.S.-K.); (P.N.)
- Correspondence: ; Tel.: +481-2620-5750; Fax: +481-2620-5758
| |
Collapse
|
48
|
Tajouri L, Campos M, Olsen M, Lohning A, Jones P, Moloney S, Grimwood K, Ugail H, Mahboub B, Alawar H, McKirdy S, Alghafri R. The role of mobile phones as a possible pathway for pathogen movement, a cross-sectional microbial analysis. Travel Med Infect Dis 2021; 43:102095. [PMID: 34116242 DOI: 10.1016/j.tmaid.2021.102095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Mobile phones are used the world over, including in healthcare settings. This study aimed to investigate the viable microbial colonisation of mobile phones used by healthcare personnel. METHODS Swabs collected on the same day from 30 mobile phones belonging to healthcare workers from three separate paediatric wards of an Australian hospital were cultured on five types of agar plate, then colonies from each phone were pooled, extracted and sequenced by shotgun metagenomics. Questionnaires completed by staff whose phones were sampled assisted in the analysis and interpretation of results. RESULTS AND DISCUSSION All phones sampled cultured viable bacteria. Overall, 399 bacterial operational taxonomic units were identified from 30 phones, with 1432 cumulative hits. Among these were 58 recognised human pathogenic and commensal bacteria (37 Gram-negative, 21 Gram-positive). The total number of virulence factor genes detected was 347, with 1258 cumulative hits. Antibiotic resistance genes (ARGs) were detected on all sampled phones and overall, 133 ARGs were detected with 520 cumulative hits. The most important classes of ARGs detected encoded resistance to beta-lactam, aminoglycoside and macrolide antibiotics and efflux pump mediated resistance mechanisms. CONCLUSION Mobile phones carry viable bacterial pathogens and may act as fomites by contaminating the hands of their users and indirectly providing a transmission pathway for hospital-acquired infections and dissemination of antibiotic resistance. Further research is needed, but meanwhile adding touching mobile phones to the five moments of hand hygiene is a simple infection control strategy worth considering in hospital and community settings. Additionally, the implementation of practical and effective guidelines to decontaminate mobile phone devices would likely be beneficial to the hospital population and community at large.
Collapse
Affiliation(s)
- Lotti Tajouri
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, Australia; Harry Butler Institute, Murdoch University, Murdoch, WA, 6150, Australia; Dubai Future Council on Community Security, Dubai, United Arab Emirates; Dubai Police Scientists Council, Dubai Police, Dubai, United Arab Emirates.
| | - Mariana Campos
- Harry Butler Institute, Murdoch University, Murdoch, WA, 6150, Australia; CSIRO Health & Biosecurity, CSIRO Land & Water, Australia
| | - Matthew Olsen
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, Australia
| | - Anna Lohning
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, Australia
| | - Peter Jones
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, Australia
| | - Susan Moloney
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, Australia; Department of Paediatrics, Gold Coast University Hospital, Southport, Australia
| | - Keith Grimwood
- Griffith University and Gold Coast Health, Southport, QLD, Australia; Department of Paediatrics, Gold Coast University Hospital, Southport, Australia
| | - Hassan Ugail
- Centre for Visual Computing, University of Bradford, Bradford, United Kingdom
| | | | - Hamad Alawar
- General Department of Forensic Science and Criminology, Dubai Police, Dubai, United Arab Emirates
| | - Simon McKirdy
- Harry Butler Institute, Murdoch University, Murdoch, WA, 6150, Australia
| | - Rashed Alghafri
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, Australia; Harry Butler Institute, Murdoch University, Murdoch, WA, 6150, Australia; General Department of Forensic Science and Criminology, Dubai Police, Dubai, United Arab Emirates; Dubai Future Council on Community Security, Dubai, United Arab Emirates; Dubai Police Scientists Council, Dubai Police, Dubai, United Arab Emirates
| |
Collapse
|
49
|
Ron-Doitch S, Frušić-Zlotkin M, Soroka Y, Duanis-Assaf D, Amar D, Kohen R, Steinberg D. eDNA-Mediated Cutaneous Protection Against UVB Damage Conferred by Staphylococcal Epidermal Colonization. Microorganisms 2021; 9:microorganisms9040788. [PMID: 33918948 PMCID: PMC8068790 DOI: 10.3390/microorganisms9040788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/10/2023] Open
Abstract
The human skin is a lush microbial habitat which is occupied by a wide array of microorganisms. Among the most common inhabitants are Staphylococcus spp., namely Staphylococcus epidermidis and, in ≈20% of healthy individuals, Staphylococcus aureus. Both bacteria have been associated with cutaneous maladies, where they mostly arrange in a biofilm, thus achieving improved surface adhesion and stability. Moreover, our skin is constantly exposed to numerous oxidative environmental stressors, such as UV-irradiation. Thus, skin cells are equipped with an important antioxidant defense mechanism, the Nrf2–Keap1 pathway. In this work, we aimed to explore the morphology of S. aureus and S. epidermidis as they adhered to healthy human skin and characterize their matrix composition. Furthermore, we hypothesized that the localization of both types of bacteria on a healthy skin surface may provide protective effects against oxidative stressors, such as UV-irradiation. Our results indicate for the first time that S. aureus and S. epidermidis assume a biofilm-like morphology as they adhere to ex vivo healthy human skin and that the cultures’ extracellular matrix (ECM) is composed of extracellular polysaccharides (EPS) and extracellular DNA (eDNA). Both bacterial cultures, as well as isolated S. aureus biofilm eDNA, conferred cutaneous protection against UVB-induced apoptosis. This work emphasized the importance of skin microbiota representatives in the maintenance of a healthy cutaneous redox balance by activating the skin’s natural defense mechanism.
Collapse
Affiliation(s)
- Sapir Ron-Doitch
- Biofilm Research Laboratory, Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University-Hadassah, Jerusalem 91120, Israel; (S.R.-D.); (D.D.-A.)
- The Myers Skin Research Laboratory, Faculty of Medicine, Institute for Drug Research, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem 9103401, Israel; (M.F.-Z.); (Y.S.); (R.K.)
| | - Marina Frušić-Zlotkin
- The Myers Skin Research Laboratory, Faculty of Medicine, Institute for Drug Research, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem 9103401, Israel; (M.F.-Z.); (Y.S.); (R.K.)
| | - Yoram Soroka
- The Myers Skin Research Laboratory, Faculty of Medicine, Institute for Drug Research, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem 9103401, Israel; (M.F.-Z.); (Y.S.); (R.K.)
| | - Danielle Duanis-Assaf
- Biofilm Research Laboratory, Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University-Hadassah, Jerusalem 91120, Israel; (S.R.-D.); (D.D.-A.)
| | - Dalit Amar
- Department of Plastic, Reconstructive and Aesthetic Surgery, Faculty of Medicine, Hebrew University of Jerusalem, Israel, Hadassah Medical Center, Jerusalem 9103401, Israel;
| | - Ron Kohen
- The Myers Skin Research Laboratory, Faculty of Medicine, Institute for Drug Research, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem 9103401, Israel; (M.F.-Z.); (Y.S.); (R.K.)
| | - Doron Steinberg
- Biofilm Research Laboratory, Faculty of Dental Medicine, Institute of Dental Sciences, Hebrew University-Hadassah, Jerusalem 91120, Israel; (S.R.-D.); (D.D.-A.)
- Correspondence:
| |
Collapse
|
50
|
Williams MR, Cau L, Wang Y, Kaul D, Sanford JA, Zaramela LS, Khalil S, Butcher AM, Zengler K, Horswill AR, Dupont CL, Hovnanian A, Gallo RL. Interplay of Staphylococcal and Host Proteases Promotes Skin Barrier Disruption in Netherton Syndrome. Cell Rep 2021; 30:2923-2933.e7. [PMID: 32130897 PMCID: PMC7183042 DOI: 10.1016/j.celrep.2020.02.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/04/2019] [Accepted: 02/05/2020] [Indexed: 01/09/2023] Open
Abstract
Netherton syndrome (NS) is a monogenic skin disease resulting from loss of function of lymphoepithelial Kazal-type-related protease inhibitor (LEKTI-1). In this study we examine if bacteria residing on the skin are influenced by the loss of LEKTI-1 and if interaction between this human gene and resident bacteria contributes to skin disease. Shotgun sequencing of the skin microbiome demonstrates that lesional skin of NS subjects is dominated by Staphylococcus aureus (S. aureus) and Staphylococcus epidermidis (S. epidermidis). Isolates of either species from NS subjects are able to induce skin inflammation and barrier damage on mice. These microbes promote skin inflammation in the setting of LEKTI-1 deficiency due to excess proteolytic activity promoted by S. aureus phenol-soluble modulin α as well as increased bacterial proteases staphopain A and B from S. aureus or EcpA from S. epidermidis. These findings demonstrate the critical need for maintaining homeostasis of host and microbial proteases to prevent a human skin disease.
Collapse
Affiliation(s)
- Michael R Williams
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA.
| | - Laura Cau
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA; SILAB, R&D Department, Brive, France.
| | - Yichen Wang
- INSERM, UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Drishti Kaul
- J. Craig Venter Institute, La Jolla, CA 92093, USA
| | - James A Sanford
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Livia S Zaramela
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Shadi Khalil
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA; University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Anna M Butcher
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Karsten Zengler
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, San Diego, CA 92093, USA; Department of Bioengineering, University of California, San Diego, CA 92093, USA
| | - Alexander R Horswill
- Department of Veterans Affairs Denver Health Care System, Denver, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | | | - Alain Hovnanian
- INSERM, UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|