51
|
Linehan K, Dempsey EM, Ryan CA, Ross RP, Stanton C. First encounters of the microbial kind: perinatal factors direct infant gut microbiome establishment. MICROBIOME RESEARCH REPORTS 2022; 1:10. [PMID: 38045649 PMCID: PMC10688792 DOI: 10.20517/mrr.2021.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 12/05/2023]
Abstract
The human gut microbiome harbors a diverse range of microbes that play a fundamental role in the health and well-being of their host. The early-life microbiome has a major influence on human development and long-term health. Perinatal factors such as maternal nutrition, antibiotic use, gestational age and mode of delivery influence the initial colonization, development, and function of the neonatal gut microbiome. The perturbed early-life gut microbiome predisposes infants to diseases in early and later life. Understanding how perinatal factors guide and shape the composition of the early-life microbiome is essential to improving infant health. The following review provides a synopsis of perinatal factors with the most decisive influences on initial microbial colonization of the infant gut.
Collapse
Affiliation(s)
- Kevin Linehan
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Eugene M. Dempsey
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- Department of Paediatrics & Child Health and INFANT Centre, University College Cork, Cork T12 YN60, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- Department of Paediatrics & Child Health and INFANT Centre, University College Cork, Cork T12 YN60, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
| |
Collapse
|
52
|
Xu X, Marffy ALL, Keightley A, McCarthy AJ, Geisbrecht BV. Group B Streptococcus Surface Protein β: Structural Characterization of a Complement Factor H-Binding Motif and Its Contribution to Immune Evasion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1232-1247. [PMID: 35110419 PMCID: PMC8881398 DOI: 10.4049/jimmunol.2101078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022]
Abstract
The β protein from group B Streptococcus (GBS) is a ∼132-kDa, cell-surface exposed molecule that binds to multiple host-derived ligands, including complement factor H (FH). Many details regarding this interaction and its significance to immune evasion by GBS remain unclear. In this study, we identified a three-helix bundle domain within the C-terminal half of the B75KN region of β as the major FH-binding determinant and determined its crystal structure at 2.5 Å resolution. Analysis of this structure suggested a role in FH binding for a loop region connecting helices α1 and α2, which we confirmed by mutagenesis and direct binding studies. Using a combination of protein cross-linking and mass spectrometry, we observed that B75KN bound to complement control protein (CCP)3 and CCP4 domains of FH. Although this binding site lies within a complement regulatory region of FH, we determined that FH bound by β retained its decay acceleration and cofactor activities. Heterologous expression of β by Lactococcus lactis resulted in recruitment of FH to the bacterial surface and a significant reduction of C3b deposition following exposure to human serum. Surprisingly, we found that FH binding by β was not required for bacterial resistance to phagocytosis by neutrophils or killing of bacteria by whole human blood. However, loss of the B75KN region significantly diminished bacterial survival in both assays. Although our results show that FH recruited to the bacterial surface through a high-affinity interaction maintains key complement-regulatory functions, they raise questions about the importance of FH binding to immune evasion by GBS as a whole.
Collapse
Affiliation(s)
- Xin Xu
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS U.S.A
| | - Alexander L. Lewis Marffy
- Department of Infectious Diseases, Section of Molecular Microbiology, MRC Centre for Molecular Bacteriology & Infection, Imperial College London; London, U.K
| | - Andrew Keightley
- Department of Opthamology, School of Medicine, University of Missouri-Kansas City; Kansas City, MO U.S.A
| | - Alex J. McCarthy
- Department of Infectious Diseases, Section of Molecular Microbiology, MRC Centre for Molecular Bacteriology & Infection, Imperial College London; London, U.K
| | - Brian V. Geisbrecht
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS U.S.A.,To whom correspondence should be addressed: Brian V. Geisbrecht, Ph.D., Kansas State University, 141 Chalmers Hall, 1711 Claflin Road, Manhattan, KS 66506, PH: 785.532.3154,
| |
Collapse
|
53
|
Chacko A, Delbaz A, Choudhury IN, Eindorf T, Shah M, Godfrey C, Sullivan MJ, St John JA, Ulett GC, Ekberg JAK. Streptococcus agalactiae Infects Glial Cells and Invades the Central Nervous System via the Olfactory and Trigeminal Nerves. Front Cell Infect Microbiol 2022; 12:793416. [PMID: 35281448 PMCID: PMC8907725 DOI: 10.3389/fcimb.2022.793416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus agalactiae causes neonatal meningitis and can also infect the adult central nervous system (CNS). S. agalactiae can cross the blood-brain barrier but may also reach the CNS via other paths. Several species of bacteria can directly invade the CNS via the olfactory and trigeminal nerves, which extend between the nasal cavity and brain and injury to the nasal epithelium can increase the risk/severity of infection. Preterm birth is associated with increased risk of S. agalactiae infection and with nasogastric tube feeding. The tubes, also used in adults, can cause nasal injuries and may be contaminated with bacteria, including S. agalactiae. We here investigated whether S. agalactiae could invade the CNS after intranasal inoculation in mice. S. agalactiae rapidly infected the olfactory nerve and brain. Methimazole-mediated model of nasal epithelial injury led to increased bacterial load in these tissues, as well as trigeminal nerve infection. S. agalactiae infected and survived intracellularly in cultured olfactory/trigeminal nerve- and brain-derived glia, resulting in cytokine production, with some differences between glial types. Furthermore, a non-capsulated S. agalactiae was used to understand the role of capsule on glial cells interaction. Interestingly, we found that the S. agalactiae capsule significantly altered cytokine and chemokine responses and affected intracellular survival in trigeminal glia. In summary, this study shows that S. agalactiae can infect the CNS via the nose-to-brain path with increased load after epithelial injury, and that the bacteria can survive in glia.
Collapse
Affiliation(s)
- Anu Chacko
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Ali Delbaz
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Indra N. Choudhury
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Tanja Eindorf
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Megha Shah
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Christopher Godfrey
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Matthew J. Sullivan
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | - James A. St John
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Glen C. Ulett
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | - Jenny A. K. Ekberg
- Menzies Health Institute Queensland and School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
- *Correspondence: Jenny A. K. Ekberg,
| |
Collapse
|
54
|
Shahi I, Llaneras CN, Perelman SS, Torres VJ, Ratner AJ. Genome-Wide CRISPR-Cas9 Screen Does Not Identify Host Factors Modulating Streptococcus agalactiae β-Hemolysin/Cytolysin-Induced Cell Death. Microbiol Spectr 2022; 10:e0218621. [PMID: 35196804 PMCID: PMC8865549 DOI: 10.1128/spectrum.02186-21] [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: 11/08/2021] [Accepted: 01/07/2022] [Indexed: 11/20/2022] Open
Abstract
Pore-forming toxins (PFTs) are commonly produced by pathogenic bacteria, and understanding them is key to the development of virulence-targeted therapies. Streptococcus agalactiae, or group B Streptococcus (GBS), produces several factors that enhance its pathogenicity, including the PFT β-hemolysin/cytolysin (βhc). Little is understood about the cellular factors involved in βhc pore formation. We conducted a whole-genome CRISPR-Cas9 forward genetic screen to identify host genes that might contribute to βhc pore formation and cell death. While the screen identified the established receptor, CD59, in control experiments using the toxin intermedilysin (ILY), no clear candidate genes were identified that were required for βhc-mediated lethality. Of the top targets from the screen, two genes involved in membrane remodeling and repair represented candidates that might modulate the kinetics of βhc-induced cell death. Upon attempted validation of the results using monoclonal cell lines with targeted disruption of these genes, no effect on βhc-mediated cell lysis was observed. The CRISPR-Cas9 screen results are consistent with the hypothesis that βhc does not require a single nonessential host factor to mediate target cell death. IMPORTANCE CRISPR-Cas9 forward genetic screens have been used to identify host cell targets required by bacterial toxins. They have been used successfully to both verify known targets and elucidate novel host factors required by toxins. Here, we show that this approach fails to identify host factors required for cell death due to βhc, a toxin required for GBS virulence. These data suggest that βhc may not require a host cell receptor for toxin function or may require a host receptor that is an essential gene and would not be identified using this screening strategy.
Collapse
Affiliation(s)
- Ifrah Shahi
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Cristina N. Llaneras
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sofya S. Perelman
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Adam J. Ratner
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
55
|
Rajaei A, Rowe HM, Neely MN. The LCP Family Protein, Psr, Is Required for Cell Wall Integrity and Virulence in Streptococcus agalactiae. Microorganisms 2022; 10:microorganisms10020217. [PMID: 35208672 PMCID: PMC8875755 DOI: 10.3390/microorganisms10020217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023] Open
Abstract
A robust cell envelope is the first line of protection for an infecting pathogen when encountering the immune defense of its host. In Gram-positive organisms, LytR-CpsA-Psr (LCP) family proteins play a major role in the synthesis and assembly of the cell envelope. While these proteins could be considered for potential new drug targets, not enough is known about how they function to support the integrity of the cell wall. Streptococcus agalactiae (group B streptococcus or GBS) is known to encode at least three LCP family proteins, including CpsA, LytR (BrpA) and Psr. Using strains of GBS that have mutations in two of the three LCP proteins, we were able to determine a role for these proteins in GBS cell wall integrity. The results presented here demonstrate that the absence of Psr results in a decreased growth rate, decreased viability over time, inconsistent cocci morphology and diminished cell wall integrity, as well as an increased penicillin susceptibility, decreased capsule levels and attenuation in virulence in a zebrafish model of infectious disease. A strain that is missing two of the LCP family proteins, CpsA and Psr, exhibits an increase in these defective phenotypes, indicating that CpsA and Psr are partially redundant in function.
Collapse
Affiliation(s)
- Atefeh Rajaei
- Molecular and Biomedical Sciences Department, University of Maine, Orono, ME 04469, USA;
| | - Hannah M. Rowe
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA;
| | - Melody N. Neely
- Molecular and Biomedical Sciences Department, University of Maine, Orono, ME 04469, USA;
- Correspondence:
| |
Collapse
|
56
|
Chandna J, Liu WH, Dangor Z, Leahy S, Sridhar S, John HB, Mucasse H, Bassat Q, Bardaji A, Abubakar A, Nasambu C, Newton CR, Sánchez Yanotti C, Libster R, Milner K, Paul P, Lawn JE. Emotional and Behavioral Outcomes in Childhood for Survivors of Invasive Group B Streptococcus Disease in Infancy: Findings From 5 Low- and Middle-Income Countries. Clin Infect Dis 2022; 74:S35-S43. [PMID: 34725686 PMCID: PMC8776308 DOI: 10.1093/cid/ciab821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Survivors of invasive group B Streptococcus (iGBS) disease, notably meningitis, are at increased risk of neurodevelopmental impairment. However, the limited studies to date have a median follow-up to 18 months and have mainly focused on moderate or severe neurodevelopmental impairment, with no previous studies on emotional-behavioral problems among iGBS survivors. METHODS In this multicountry, matched cohort study, we included children aged 18 months to 17 years with infant iGBS sepsis and meningitis from health demographic surveillance systems, or hospital records in Argentina, India, Kenya, Mozambique, and South Africa. Children without an iGBS history were matched to iGBS survivors for sex and age. Our primary outcomes were emotional-behavioral problems and psychopathological conditions as measured with the Child Behavior Checklist (CBCL). The CBCL was completed by the child's primary caregiver. RESULTS Between October 2019 and April 2021, 573 children (mean age, 7.18 years) were assessed, including 156 iGBS survivors and 417 non-iGBS comparison children. On average, we observed more total problems and more anxiety, attention, and conduct problems for school-aged iGBS survivors compared with the non-iGBS group. No differences were found in the proportion of clinically significant psychopathological conditions defined by the Diagnostic and Statistical Manual of Mental Disorders (Fifth Edition). CONCLUSIONS Our findings suggested that school-aged iGBS survivors experienced increased mild emotional behavioral problems that may affect children and families. At-risk neonates including iGBS survivors need long-term follow-up with integrated emotional-behavioral assessments and appropriate care. Scale-up will require simplified assessments that are free and culturally adapted.
Collapse
Affiliation(s)
- Jaya Chandna
- Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Wan-Hsin Liu
- Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Division of General Paediatrics, Department of Paediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ziyaad Dangor
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shannon Leahy
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Hima B John
- Neonatology Department, Christian Medical College, Vellore, India
| | | | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- Pediatrics Department, Hospital Sant Joan de Déu (University of Barcelona), Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, Madrid, Spain
| | - Azucena Bardaji
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Amina Abubakar
- Neuroscience Research Group, Department of Clinical Sciences, KEMRI Wellcome Trust, Kilifi, Kenya
- Institute of Human Development, Aga Khan University, Nairobi, Kenya
| | - Carophine Nasambu
- Neuroscience Research Group, Department of Clinical Sciences, KEMRI Wellcome Trust, Kilifi, Kenya
| | - Charles R Newton
- Neuroscience Research Group, Department of Clinical Sciences, KEMRI Wellcome Trust, Kilifi, Kenya
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | | | | | - Kate Milner
- Neurodisability & Rehabilitation Research Group, Murdoch Children’s Research Institute 2, Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Proma Paul
- Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Joy E Lawn
- Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
57
|
Miselli F, Frabboni I, Di Martino M, Zinani I, Buttera M, Insalaco A, Stefanelli F, Lugli L, Berardi A. Transmission of Group B Streptococcus in late-onset neonatal disease: a narrative review of current evidence. Ther Adv Infect Dis 2022; 9:20499361221142732. [PMID: 36569815 PMCID: PMC9780763 DOI: 10.1177/20499361221142732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Group B streptococcus (GBS) late-onset disease (LOD, occurring from 7 through 89 days of life) is an important cause of sepsis and meningitis in infants. The pathogenesis and modes of transmission of LOD to neonates are yet to be elucidated. Established risk factors for the incidence of LOD include maternal GBS colonisation, young maternal age, preterm birth, HIV exposure and African ethnicity. The mucosal colonisation by GBS may be acquired perinatally or in the postpartum period from maternal or other sources. Growing evidence has demonstrated the predominant role of maternal sources in the transmission of LOD. Intrapartum antibiotic prophylaxis (IAP) to prevent early-onset disease reduces neonatal GBS colonisation during delivery; however, a significant proportion of IAP-exposed neonates born to GBS-carrier mothers acquire the pathogen at mucosal sites in the first weeks of life. GBS-infected breast milk, with or without presence of mastitis, is considered a potential vehicle for transmitting GBS. Furthermore, horizontal transmission is possible from nosocomial and other community sources. Although unfrequently reported, nosocomial transmission of GBS in the neonatal intensive care unit is probably less rare than is usually believed. GBS disease can sometime recur and is usually caused by the same GBS serotype that caused the primary infection. This review aims to discuss the dynamics of transmission of GBS in the neonatal LOD.
Collapse
Affiliation(s)
- Francesca Miselli
- Neonatal Intensive Care Unit, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Ilaria Frabboni
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Marianna Di Martino
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Isotta Zinani
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Martina Buttera
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Anna Insalaco
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Francesca Stefanelli
- Pediatric Post-Graduate School, University of Modena e Reggio Emilia, Modena, Italy
| | - Licia Lugli
- Neonatal Intensive Care Unit, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Alberto Berardi
- Neonatal Intensive Care Unit, Policlinico University Hospital, 41124 Modena, Italy
| |
Collapse
|
58
|
Fister P, Peček J, Jeverica S, Primec ZR, Paro-Panjan D. Neonatal Group B Streptococcal Meningitis: Predictors for Poor Neurologic Outcome at 18 Months. J Child Neurol 2022; 37:64-72. [PMID: 34816748 DOI: 10.1177/08830738211053128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AIM To find early predictors for poor neurodevelopmental outcome after neonatal group B streptococcal meningitis. METHODS We retrospectively analyzed clinical characteristics of 23 patients with neonatal group B streptococcal meningitis and their neurodevelopmental outcome at 18 months. Available group B Streptococcus strains were serotyped and their genomes characterized. RESULTS We found several differences between patients with early- (n = 5) and late-onset (n = 18) disease. Nine children had neurologic abnormalities at 18 months and 4 had epilepsy, all of them after late-onset disease. Most important risk factors for poor outcome were impaired consciousness at admission, hemodynamic instability, seizures, or abnormal electroencephalogram during the acute illness and abnormal neurologic and ophthalmologic examination at the end of treatment, whereas abnormalities in laboratory and imaging studies were not predictive. Hypervirulent serotype III, multilocus sequence type 17 group B Streptococcus was the predominant pathogen. CONCLUSIONS Neurodevelopmental impairment after neonatal group B streptococcal meningitis is likelier in those with clinical and neurophysiological features indicating worse disease severity.
Collapse
Affiliation(s)
- Petja Fister
- Department of Neonatology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia.,37664Faculty of Medicine, University of Ljubljana, Slovenia
| | - Jerneja Peček
- Department of Neonatology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia.,37664Faculty of Medicine, University of Ljubljana, Slovenia
| | - Samo Jeverica
- 68924National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Zvonka Rener Primec
- 37664Faculty of Medicine, University of Ljubljana, Slovenia.,Department of Neurology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Darja Paro-Panjan
- Department of Neonatology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia.,37664Faculty of Medicine, University of Ljubljana, Slovenia
| |
Collapse
|
59
|
Shabayek S, Ferrieri P, Spellerberg B. Group B Streptococcal Colonization in African Countries: Prevalence, Capsular Serotypes, and Molecular Sequence Types. Pathogens 2021; 10:pathogens10121606. [PMID: 34959562 PMCID: PMC8706430 DOI: 10.3390/pathogens10121606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 11/20/2022] Open
Abstract
Streptococcus agalactiae or group B streptococcus (GBS) is a commensal of the gastrointestinal and genitourinary tracts of healthy women and an important cause of neonatal invasive infections worldwide. Transmission of bacteria to the newborn occurs at birth and can be prevented by intrapartum antibiotic prophylaxis. However, this not available in resource limited settings in Africa, which carries a particular high burden of disease. Serotype based vaccines are in development and present a suitable alternative to prevent neonatal infections. To be able to assess vaccine efficacy, knowledge and surveillance of GBS epidemiological data are required. This review summarizes investigations about the serotype distribution and the multi-locus sequence types (MLST) found in different African countries. While most serotypes and MLST data are comparable to findings from other continents, some specific differences exist. Serotype V is predominant among colonizing maternal strains in many different African countries. Serotypes that are rarely detected in western industrialized nations, such as serotypes VI, VII and IX, are prevalent in studies from Ghana and Egypt. Moreover, some specific MLST sequence types that seem to be more or less unique to Africa have been detected. However, overall, the data confirm that a hexavalent vaccine can provide broad coverage for the African continent and that a protein vaccine could represent a promising alternative.
Collapse
Affiliation(s)
- Sarah Shabayek
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Patricia Ferrieri
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, 89081 Ulm, Germany
- Correspondence:
| |
Collapse
|
60
|
Kam KQ, Thoon KC, Tee WSN, Ang MLT, Tan NWH, Yeo KT, Li J, Chong CY. Serotype distribution and incidence of invasive early onset and late onset group B streptococcal disease amongst infants in Singapore. BMC Infect Dis 2021; 21:1221. [PMID: 34876053 PMCID: PMC8650237 DOI: 10.1186/s12879-021-06891-1] [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: 02/05/2020] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Background The current group B streptococcal (GBS) preventive measures had reduced invasive GBS early onset disease (EOD) incidences worldwide, but the late onset disease (LOD) incidences had remained unchanged. Administration of a safe and effective GBS vaccine in addition to the current strategies were thought to be the next steps in reducing the incidences of invasive GBS infection especially LOD. In this study, we aimed to examine the causative GBS serotypes in invasive GBS disease, determine the incidences of EOD and LOD, and compare the risk factors between EOD and LOD. Methods A retrospective study of infants ≤ 90-day-old over an 8-year period (2010–2017). The incidences of EOD and LOD were obtained by using patients with EOD and LOD who were born in our institution as the numerator and the live births in our institution per year of the study period as the denominator. Available GBS isolates were serotyped by the National Public Health Laboratory using capsular serotyping methods. The risk factors of EOD and LOD were compared. Results A total of 71 infants were identified; 16 (22.5%) and 55 (77.5%) of them had EOD and LOD, respectively. Serotype III (n = 42, 71.2%) was the most common serotype amongst the 59 isolates available for serotyping. Serotypes Ia, Ib, II, III, and V accounted for 98.3% (n = 58) of the invasive GBS diseases. The overall incidence was 0.42 per 1000 live births. The mean incidences of EOD and LOD were 0.13 per 1000 live births and 0.29 per 1000 live births, respectively. On multivariate analysis, risk factors for LOD as compared to EOD were: Chinese ethnicity (OR 27.1, 95% CI 3.0–243.1, p = 0.003) and negative/unknown maternal GBS status (OR 20.0, 95% CI 2.0–250.0, p = 0.012). Prematurity and intrapartum risk factors (peripartum maternal pyrexia, prolonged rupture of membrane) of EOD were not associated with LOD. Conclusions The LOD incidence had remained higher than EOD incidence in our cohort. A GBS vaccine that covers the major causative serotypes found in our cohort can potentially reduce the overall GBS disease burden in the country.
Collapse
Affiliation(s)
- Kai-Qian Kam
- Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah, Singapore City, 229899, Singapore. .,Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore. .,Duke-National University of Singapore Medical School, Singapore City, Singapore. .,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore City, Singapore.
| | - Koh Cheng Thoon
- Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah, Singapore City, 229899, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore.,Duke-National University of Singapore Medical School, Singapore City, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore City, Singapore
| | - Wen Sim Nancy Tee
- Department of Laboratory Medicine, National University Health System, Singapore City, Singapore
| | - Michelle Lay Teng Ang
- National Public Health Laboratory, National Centre of Infectious Diseases, Singapore City, Singapore
| | - Natalie Woon Hui Tan
- Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah, Singapore City, 229899, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore.,Duke-National University of Singapore Medical School, Singapore City, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore City, Singapore
| | - Kee Thai Yeo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore.,Duke-National University of Singapore Medical School, Singapore City, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore City, Singapore.,Department of Neonatology, KK Women's and Children's Hospital, Singapore City, Singapore
| | - Jiahui Li
- Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah, Singapore City, 229899, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore.,Duke-National University of Singapore Medical School, Singapore City, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore City, Singapore
| | - Chia Yin Chong
- Infectious Disease Service, Department of Pediatrics, KK Women's and Children's Hospital, 100 Bukit Timah, Singapore City, 229899, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore.,Duke-National University of Singapore Medical School, Singapore City, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore City, Singapore
| |
Collapse
|
61
|
Scheftelowitz Cohen R, Chodik G, Eisenberg VH. Re-evaluating Perinatal Group B Streptococcal screening in Israel - Is it time for a change in policy? Prev Med 2021; 153:106716. [PMID: 34245779 DOI: 10.1016/j.ypmed.2021.106716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/10/2021] [Accepted: 07/03/2021] [Indexed: 10/20/2022]
Abstract
Group B streptococcal early-onset disease (EOGBSD) is a significant cause of morbidity and fatality in newborns. Current policy in Israel is risk-based management. Our aim was to re-evaluate the current screening policy for Group B Streptococcus (GBS), considering colonization and prevalence rates and costs estimates. This was a retrospective cohort study including term pregnancies between 2015 and 2016 insured by Maccabi Healthcare Services (MHS). A costs estimation model was performed comparing three approaches: universal culture-based screening, current policy in Israel and the current clinical scenario. Out of 54,759 pregnancies, 46.3% women undergo GBS culture-based screening. Overall GBS colonization rates in screened women were 21%. Six EOGBSD cases were identified, all offspring of mothers who were not screened. EOGBSD prevalence rate was 11 per 100,000. Universal culture-based screening was found to be 50% less costly than the current risk-based policy, and would have prevented 20.29 per 100,000 cases. Universal GBS culture-based screening was found to be more cost-effective, compared to the current policy and screening behaviors. Due to the clinical and economic benefits, we recommend that a change in policy should be considered.
Collapse
Affiliation(s)
| | - Gabriel Chodik
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Maccabitech, Maccabi Health Services, Tel Aviv, Israel
| | - Vered H Eisenberg
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel.
| |
Collapse
|
62
|
McLeod E, Mason C, Swainston K. Parents experiences of having an infant with early onset group B streptococcus infection. Br J Health Psychol 2021; 27:777-788. [PMID: 34806260 DOI: 10.1111/bjhp.12572] [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: 12/02/2020] [Revised: 10/20/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The study explores parents lived experience of having an infant with early onset group B streptococcus (GBS). DESIGN The study adopts a qualitative approach and a phenomenological framework with written autobiographical accounts as the method of data collection. METHODS Twenty-seven parents wrote first-hand accounts of their experience of having an infant with early onset GBS. Participants documented their experiences in their own way, reporting their thoughts and feelings, experiences, and events that were meaningful to them. RESULTS Four themes were developed from data analysis: 'bonding'; 'grief'; 'communication and information provision'; and 'future family'. CONCLUSIONS The study findings demonstrate the complexity of emotions within parent's experiences and highlight grief and loss as a core component of these experiences. Medical intervention, while acknowledged as being vital and in many cases lifesaving, was viewed as a disruption to early bonding experiences resulting in sadness and guilt. Variation in information provision, communication about this infection, and feeling that their infant's illness and/or death were preventable added to the sense of loss. Breakdowns in interpersonal communications with partners and family were commonly described and experiences of early onset GBS had implications for decision-making around future pregnancies.
Collapse
Affiliation(s)
- Emma McLeod
- School of Social Sciences, Humanities and Law, Teesside University, Middlesbrough, UK
| | - Celia Mason
- School of Social Sciences, Humanities and Law, Teesside University, Middlesbrough, UK
| | - Katherine Swainston
- School of Social Sciences, Humanities and Law, Teesside University, Middlesbrough, UK
| |
Collapse
|
63
|
Maternal vaccination with a type-III glycoconjugate protects mouse neonates against Group B Streptococcus intranasal infection. Sci Rep 2021; 11:21384. [PMID: 34725414 PMCID: PMC8560876 DOI: 10.1038/s41598-021-00941-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/12/2021] [Indexed: 11/15/2022] Open
Abstract
Group B Streptococcus (GBS) is generally an asymptomatic colonizer of human mucosa but it occasionally infects pregnant women and neonates through vertical transmission, causing disease during the first weeks of life with frequent and severe complications. Preclinical studies have shown that maternal vaccination with polysaccharide-based vaccines protects mothers and offspring from GBS mucosal colonization and consecutive infection. In these models, bacteria were inoculated in mouse either intravaginally in the last trimester of pregnancy or systemically in pups. Here, we investigated whether maternal vaccination with glycoconjugate vaccines may also prevent GBS-mediated colonization and disease in neonates using an infection route that more closely mimics inhalation or ingestion of bacteria during human delivery. To address this point, mice aged less than two days were intranasally challenged with epidemiologically relevant GBS strains. Bacteria were found to colonize nose and intestine, reaching in some cases lungs and blood during the first days of life. Bacteria were also found in vagina of a fraction of colonized female mice within the first month of life. GBS-specific IgG induced by maternal vaccination with a glycoconjugate vaccine formulation were found in blood and mucosal tissues of newborns. Finally, when intranasally challenged with GBS serotype III strains, pups delivered by vaccinated mothers were partially protected against mucosal colonization and deeper infection.
Collapse
|
64
|
Eto SF, Fernandes DC, Baldassi AC, Balbuena TS, da Costa Alecrim JV, Almeida de Carvalho FC, Lima C, Lopes-Ferreira M, Pizauro JM. Proteomic analysis capsule synthesis and redox mechanisms in the intracellular survival of group B Streptococcus in fish microglia. FISH & SHELLFISH IMMUNOLOGY 2021; 118:34-50. [PMID: 34464686 DOI: 10.1016/j.fsi.2021.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/20/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
Group B Streptococcus (GBS) causes meningitis in neonates and Nile tilapia (Oreochromis niloticus). The molecular mechanisms regulating the intracellular survival of this pathogen in the host cell are complex and crucial for the progression of infection. Thus, we propose the use of GBS-infected Nile tilapia microglia as an in vitro model system simulating infection caused by homologous bacteria in humans. We used this model to evaluate the phagocytic activity, as well as the functional aspects of the capsular proteins A, B, C, and D and the major redox enzymes, and the synergistic role of mechanisms/proteins involved in blocking phagocytic process. We observed that in the intracellular phase, GBS showed enhanced synthesis of the polysaccharide capsule and used superoxide dismutase, thioredoxin, NADH oxidase, and alkyl hydroperoxide reductase to scavenge reactive oxygen species and reactive nitrogen species produced by the host cell. Furthermore, although these virulence mechanisms were effective during the initial hours of infection, they were not able to subvert microglial responses, which partially neutralized the infection. Altogether, our findings provided important information regarding the intracellular survival mechanisms of GBS and perspectives for the production of new drugs and vaccines, through the druggability analysis of specific proteins. In conclusion, tilapia microglia serve as a potent in vitro experimental model for the study of meningitis.
Collapse
Affiliation(s)
- Silas Fernandes Eto
- Department of Postgraduate in Health Sciences-PROCISA, Federal University of Roraima (UFRR), Boa Vista, 69310-000, Brazil.
| | - Dayanne Carla Fernandes
- Immunochemistry Laboratory, Butantan Institute, (CeTICs/FAPESP), Vital Brazil Avenue, 1500, Butantã, 05503-009, São Paulo, Brazil
| | - Amanda Cristina Baldassi
- Department of Technology, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, 14884-900, Sao Paulo/ SP, Brazil
| | - Thiago Santana Balbuena
- Department of Technology, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, 14884-900, Sao Paulo/ SP, Brazil
| | - João Victor da Costa Alecrim
- Department of Postgraduate in Health Sciences-PROCISA, Federal University of Roraima (UFRR), Boa Vista, 69310-000, Brazil
| | | | - Carla Lima
- Immunoregulation Unit of the Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, Vital Brazil Avenue, 1500, Butantã, 05503-009, São Paulo, Brazil
| | - Monica Lopes-Ferreira
- Immunoregulation Unit of the Laboratory of Applied Toxinology (CeTICs/FAPESP), Butantan Institute, Vital Brazil Avenue, 1500, Butantã, 05503-009, São Paulo, Brazil
| | - João Martins Pizauro
- Department of Technology, School of Agrarian and Veterinary Sciences, Sao Paulo State University (Unesp), Jaboticabal, 14884-900, Sao Paulo/ SP, Brazil
| |
Collapse
|
65
|
Complicated Streptococcus agalactiae Sepsis with/without Meningitis in Young Infants and Newborns: The Clinical and Molecular Characteristics and Outcomes. Microorganisms 2021; 9:microorganisms9102094. [PMID: 34683413 PMCID: PMC8540989 DOI: 10.3390/microorganisms9102094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/13/2021] [Accepted: 10/01/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Streptococcus agalactiae (also known as group B streptococcus, GBS) is associated with high mortality and morbidity rates in infants, especially those with complicated GBS sepsis, defined as those with meningitis, severe sepsis and/or septic shock. We aimed to characterize the clinical and molecular characteristics and risk factors for adverse outcomes of neonates with invasive GBS diseases. Methods: From 2003 to 2020, all neonates with invasive GBS diseases who were hospitalized in a tertiary-level neonatal intensive care unit (NICU) were enrolled. The GBS isolates underwent serotyping, multilocus sequence typing (MLST) and antibiotic susceptibility testing. We compared cases of complicated GBS sepsis with uncomplicated GBS bacteremia. Results: During the study period, a total of 188 neonates (aged less than 6 months old) with invasive GBS diseases were identified and enrolled. Among them, 119 (63.3%) had uncomplicated GBS bacteremia and 69 (36.7%) neonates had complicated GBS sepsis, including meningitis (25.5%, n = 48) and severe sepsis or septic shock. Among neonates with complicated GBS sepsis, 45 (65.2%) had neurological complications, and 21 (42.0%) of 50 survivors had neurological sequelae at discharge. The overall final mortality rate was 10.1% (19 neonates died). Type III/ST-17 GBS isolates accounted for 56.5% of all complicated GBS sepsis and 68.8% of all GBS meningitis, but this strain was not significantly associated with worse outcomes. The antimicrobial resistance rate among the invasive GBS isolates was obviously increasing in the past two decades. After multivariate logistic regression analysis, neonates with thrombocytopenia and respiratory failure were independently associated with final adverse outcomes. Conclusions: a total of 36.7% of all neonatal invasive GBS diseases were associated with complicated sepsis with/without meningitis. Given the high mortality and morbidity rates in neonates with complicated GBS sepsis, further studies for early identification of specific strains, risk factors or genetic mechanisms that will cause complicated GBS sepsis are urgently needed in the future.
Collapse
|
66
|
Coppolino F, Romeo L, Pietrocola G, Lentini G, De Gaetano GV, Teti G, Galbo R, Beninati C. Lysine Residues in the MK-Rich Region Are Not Required for Binding of the PbsP Protein From Group B Streptococci to Plasminogen. Front Cell Infect Microbiol 2021; 11:679792. [PMID: 34568085 PMCID: PMC8455988 DOI: 10.3389/fcimb.2021.679792] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Binding to plasminogen (Plg) enables bacteria to associate with and invade host tissues. The cell wall protein PbsP significantly contributes to the ability of group B streptococci, a frequent cause of invasive infection, to bind Plg. Here we sought to identify the molecular regions involved in the interactions between Plg and PbsP. The K4 Kringle domain of the Plg molecule was required for binding of Plg to whole PbsP and to a PbsP fragment encompassing a region rich in methionine and lysine (MK-rich domain). These interactions were inhibited by free L-lysine, indicating the involvement of lysine binding sites in the Plg molecule. However, mutation to alanine of all lysine residues in the MK-rich domain did not decrease its ability to bind Plg. Collectively, our data identify a novel bacterial sequence that can interact with lysine binding sites in the Plg molecule. Notably, such binding did not require the presence of lysine or other positively charged amino acids in the bacterial receptor. These data may be useful for developing alternative therapeutic strategies aimed at blocking interactions between group B streptococci and Plg.
Collapse
Affiliation(s)
- Francesco Coppolino
- Department of Biomedical, Dental and Imaging Sciences, University of Messina, Messina, Italy
| | - Letizia Romeo
- Department of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Giampiero Pietrocola
- Department Molecular Medicine, Biochemistry Section, University of Pavia, Pavia, Italy
| | - Germana Lentini
- Department of Human Pathology and Medicine, University of Messina, Messina, Italy
| | | | | | - Roberta Galbo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Concetta Beninati
- Department of Human Pathology and Medicine, University of Messina, Messina, Italy.,Scylla Biotech Srl, Messina, Italy
| |
Collapse
|
67
|
Ghia C, Rambhad G. Disease burden due to Group B Streptococcus in the Indian population and the need for a vaccine - a narrative review. Ther Adv Infect Dis 2021; 8:20499361211045253. [PMID: 34540226 PMCID: PMC8445532 DOI: 10.1177/20499361211045253] [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: 06/08/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
Streptococcus agalactiae, a Gram-positive bacterium, causes invasive infection known as Group B streptococcal disease (GBS). It is a leading cause of neonatal death and complications prior to delivery. The burden of GBS is unknown in India despite the high incidence of preterm and stillbirths. In this study, we performed a narrative review of the available literature (published in the last 10 years) on the epidemiology of GBS, using PubMed and Google Scholar, to understand its impact in India and evaluate potential strategies to prevent the disease in the high-risk population, that is, neonates. The review showed that the incidence of early- and late-onset GBS in neonates (per 1000 live births) was in the ranges of 0.090-0.68 and 0.0-0.07 respectively. The overall case fatality rate reported in only one study was 0.63. In pregnant women, the prevalence of GBS colonization was 2-62% and its transmission to their newborns varied from 6.7% to 11.1%. The serotype distribution of GBS is unclear, but some studies reported the distribution of types Ia, Ib, II, III, V, VII among pregnant women in India. The associated risk factors for GBS colonization in pregnant women are unclear but a few studies suggest the role of age and multigravida, while the risk factors in neonates include preterm birth, prolonged rupture of membrane (⩾18 h), maternal fever, obstetric complications, and prolonged labor >18 h. Screening of GBS is not a routine practice in India and intrapartum antibiotics prophylaxis is limited to only in risk conditions to prevent neonatal disease transmission. A few studies also suggest that high birth rate, poor detection methods, and financial constraints limit routine GBS screening in a developing country such as India. Hence, maternal vaccination is the most promising strategy to prevent neonatal GBS and Pfizer's hexavalent GBS conjugate vaccine (GBS6) is being developed for GBS neonatal disease.
Collapse
Affiliation(s)
- Canna Ghia
- Pfizer Ltd, The Capital, 1802, 18th Floor, Plot No. C-70, G Block, Bandra Kurla Complex, Bandra (East), Mumbai, Maharashtra 400051, India
| | - Gautam Rambhad
- Pfizer Ltd, The Capital, 1802, 18th Floor, Plot No. C-70, G Block, Bandra Kurla Complex, Bandra (East), Mumbai, 400051, India
| |
Collapse
|
68
|
Mazzuoli MV, Daunesse M, Varet H, Rosinski-Chupin I, Legendre R, Sismeiro O, Gominet M, Kaminski PA, Glaser P, Chica C, Trieu-Cuot P, Firon A. The CovR regulatory network drives the evolution of Group B Streptococcus virulence. PLoS Genet 2021; 17:e1009761. [PMID: 34491998 PMCID: PMC8448333 DOI: 10.1371/journal.pgen.1009761] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/17/2021] [Accepted: 08/09/2021] [Indexed: 01/31/2023] Open
Abstract
Virulence of the neonatal pathogen Group B Streptococcus is under the control of the master regulator CovR. Inactivation of CovR is associated with large-scale transcriptome remodeling and impairs almost every step of the interaction between the pathogen and the host. However, transcriptome analyses suggested a plasticity of the CovR signaling pathway in clinical isolates leading to phenotypic heterogeneity in the bacterial population. In this study, we characterized the CovR regulatory network in a strain representative of the CC-17 hypervirulent lineage responsible of the majority of neonatal meningitis. Transcriptome and genome-wide binding analysis reveal the architecture of the CovR network characterized by the direct repression of a large array of virulence-associated genes and the extent of co-regulation at specific loci. Comparative functional analysis of the signaling network links strain-specificities to the regulation of the pan-genome, including the two specific hypervirulent adhesins and horizontally acquired genes, to mutations in CovR-regulated promoters, and to variability in CovR activation by phosphorylation. This regulatory adaptation occurs at the level of genes, promoters, and of CovR itself, and allows to globally reshape the expression of virulence genes. Overall, our results reveal the direct, coordinated, and strain-specific regulation of virulence genes by the master regulator CovR and suggest that the intra-species evolution of the signaling network is as important as the expression of specific virulence factors in the emergence of clone associated with specific diseases. Streptococcus agalactiae, commonly known as the Group B Streptococcus (GBS), is a commensal bacterium of the intestinal and vaginal tracts found in approximately 30% of healthy adults. However, GBS is also an opportunistic pathogen and the leading cause of neonatal invasive infections. Epidemiologic data have identified a particular GBS clone, designated the CC-17 hypervirulent clonal complex, as responsible for the overwhelming majority of neonatal meningitis. The hypervirulence of CC-17 has been linked to the expression of two specific surface proteins increasing their abilities to cross epithelial and endothelial barriers. In this study, we characterized the role of the major regulator of virulence gene expression, the CovR response regulator, in a representative hypervirulent strain. Transcriptome and genome-wide binding analysis reveal the architecture of the CovR signaling network characterized by the direct repression of a large array of virulence-associated genes, including the specific hypervirulent adhesins. Comparative analysis in a non-CC-17 wild type strain demonstrates a high level of plasticity of the regulatory network, allowing to globally reshape pathogen-host interaction. Overall, our results suggest that the intra-species evolution of the regulatory network is an important factor in the emergence of GBS clones associated with specific pathologies.
Collapse
Affiliation(s)
- Maria-Vittoria Mazzuoli
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
- Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Maëlle Daunesse
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Isabelle Rosinski-Chupin
- Unité Écologie et Évolution de la Résistance aux Antibiotiques, CNRS UMR3525, Institut Pasteur, Paris, France
| | - Rachel Legendre
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Odile Sismeiro
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Myriam Gominet
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
| | - Pierre Alexandre Kaminski
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
| | - Philippe Glaser
- Unité Écologie et Évolution de la Résistance aux Antibiotiques, CNRS UMR3525, Institut Pasteur, Paris, France
| | - Claudia Chica
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
| | - Patrick Trieu-Cuot
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
| | - Arnaud Firon
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
- * E-mail:
| |
Collapse
|
69
|
Evaluation of Xpert GBS assay and Xpert GBS LB assay for detection of Streptococcus agalactiae. Ann Clin Microbiol Antimicrob 2021; 20:62. [PMID: 34488790 PMCID: PMC8419907 DOI: 10.1186/s12941-021-00461-8] [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: 01/22/2021] [Accepted: 08/16/2021] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Group B Streptococcal (GBS) infection is the primary agent of neonatal morbidity and mortality. Rapid and simple methods to detect GBS are Xpert GBS and GBS LB assays based on real-time polymerase chain reaction (PCR). However, since the diagnostic accuracy of the two techniques in diagnosing GBS remains unclear, we designed this study to appraise the diagnostic accuracy of the aforementioned. METHODS A systematic search of all literature published before July 16, 2020 was conducted using Embase, PubMed, Web of Science, and Cochrane Library. The study quality was evaluated through Review Manager 5.3. Accordingly, data extracted in the included studies were analyzed using Meta-DiSc 1.4 and Stata 12.0 software. The diagnosis odds ratio (DOR) and bivariate boxplot were utilized to evaluate the heterogeneity. Publication bias was appraised by using Deeks' funnel plot. RESULTS A total of 13 studies were adopted and only 19 sets of data met the criteria. The sensitivity and specificity of Xpert GBS were 0.91 (95% CI 0.89-0.92) and 0.93 (95% CI 0.92-0.94). The area under the curve (AUC) was 0.9806. The sensitivity and specificity results of Xpert GBS LB were 0.96 (95% CI 0.95-0.98) and 0.94 (95% CI 0.92-0.95), respectively. The AUC was 0.9950. No publication bias was found. CONCLUSIONS The Xpert GBS and GBS LB assays are valuable alternative methods with high sensitivity and specificity. However, determining whether they can be used as clinical diagnostic standards for GBS is essential for the future.
Collapse
|
70
|
Baeringsdottir B, Erlendsdottir H, Bjornsdottir ES, Martins ER, Ramirez M, Haraldsson A, Thorkelsson T. Group B streptococcal infections in infants in Iceland: clinical and microbiological factors. J Med Microbiol 2021; 70:001426. [PMID: 34554080 PMCID: PMC8697508 DOI: 10.1099/jmm.0.001426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/19/2021] [Indexed: 02/04/2023] Open
Abstract
Introduction. Group B streptococcus (GBS) is a leading cause of invasive neonatal infections. These have been divided into early-onset disease (EOD; <7 days) and late-onset disease (LOD; 7-89 days), with different GBS clonal complexes (CCs) associated with different disease presentations.Hypothesis. Different GBS CCs are associated with timing of infection (EOD or LOD) and clinical presentation (sepsis, meningitis or pneumonia).Aim. To study infant GBS infections in Iceland from 1975 to 2019. Are specific GBS CCs related to disease presentation? Is CC17 overrepresented in infant GBS infections in Iceland?Methodology. All culture-confirmed invasive GBS infections in infants (<90 days) in Iceland from 1975 to 2019 were included. Clinical information was gathered from medical records.Results. A total of 127 invasive GBS infections in infants were diagnosed, but 105 infants were included in the study. Of these, 56 had EOD and 49 had LOD. The incidence of GBS infections declined from 2000 onwards but increased again at the end of the study period. Furthermore, there was a significant increase in LOD over the study period (P=0.0001). The most common presenting symptoms were respiratory difficulties and fever and the most common presentation was sepsis alone. Approximately one-third of the cases were caused by GBS CC17 of serotype III with surface protein RIB and pili PI-1+PI-2b or PI-2b. CC17 was significantly associated with LOD (P<0.001).Conclusion. CC17 is a major cause of GBS infection in infants in Iceland. This clone is associated with LOD, which has been increasing in incidence. Because intrapartum antibiotic prophylaxis only prevents EOD, it is important to continue the development of a GBS vaccine in order to prevent LOD infections.
Collapse
Affiliation(s)
| | - Helga Erlendsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Clinical Microbiology, Landspitali University Hospital, Reykjavik, Iceland
| | | | - Elisabete R. Martins
- Instituto de Microbiologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mário Ramirez
- Instituto de Microbiologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Asgeir Haraldsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- The Children’s Hospital, Landspitali University Hospital, Reykjavik, Iceland
| | - Thordur Thorkelsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- The Children’s Hospital, Landspitali University Hospital, Reykjavik, Iceland
| |
Collapse
|
71
|
Takahashi K, Sato Y, Ikeda K. Group B streptococcus neonatal umbilical colonization managed by dry cord care in nurseries: A retrospective cohort study. Pediatr Neonatol 2021; 62:506-511. [PMID: 34088650 DOI: 10.1016/j.pedneo.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/19/2021] [Accepted: 05/10/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Screening-based intrapartum antibiotic prophylaxis (IAP) has reduced the prevalence of early-onset group B Streptococcus (GBS) infection in newborns. Nevertheless, early-onset disease still occurs despite IAP, and IAP is not effective in preventing late-onset disease. This study aimed to determine the prevalence and risk factors of GBS neonatal umbilical colonization managed by dry cord care in Japan. METHODS Of 735 healthy newborns in the well-baby nursery at Saitama City Hospital, 353 from whom umbilical bacterial swabs were obtained before discharge were included in the analysis. Maternal and neonatal clinical characteristics were retrospectively reviewed. RESULTS GBS was detected in 4.2% (15/353) of umbilical swabs; 13/15 (86.8%) were born to GBS-negative mothers. The median (IQR) age at umbilical swab collection was 4.0 (4.0-5.0) days. Comparison of clinical characteristics between GBS-positive and negative neonates revealed a significant difference in the proportion of vaginal deliveries (15/15 [100%] in GBS-positive neonates vs. 115/338 [34.0%] in GBS-negative neonates, p < 0.0001). Of 15 GBS-positive neonates, 10 (66.7%) were also co-colonized with other enteric bacteria such as Escherichia coli. CONCLUSION Vaginal delivery was a risk factor associated with GBS neonatal umbilical colonization in Japanese neonates. Co-colonization with multiple enteric bacterial species implicates vertical transmission of GBS from undetected carrier mothers during passage through the birth canal.
Collapse
Affiliation(s)
- Keigo Takahashi
- Division of Neonatology, Department of Pediatrics, Saitama City Hospital, Saitama, Japan
| | - Yuka Sato
- Department of Central Clinical Laboratory, Saitama City Hospital, Saitama, Japan
| | - Kazushige Ikeda
- Division of Neonatology, Department of Pediatrics, Saitama City Hospital, Saitama, Japan.
| |
Collapse
|
72
|
Ching NS, Buttery JP, Lai E, Steer AC, Standish J, Ziffer J, Daley AJ, Doherty R. Breastfeeding and Risk of Late-Onset Group B Streptococcal Disease. Pediatrics 2021; 148:peds.2020-049561. [PMID: 34385351 DOI: 10.1542/peds.2020-049561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Group B Streptococcus (GBS) is a major contributor to neonatal sepsis worldwide. Late-onset group B Streptococcus disease (LOGBS) and its risk factors remain poorly understood. The isolation of GBS from breast milk has been described in cases of LOGBS. This potential association has raised concerns for mothers and clinicians regarding the safety of ongoing breastfeeding. In this study, we aimed to investigate whether exposure to breast milk is associated with increased risk of LOGBS. METHODS A case-control study of LOGBS was conducted across 4 hospital networks in Victoria, Australia, including the 2 major tertiary pediatric centers in the state, to evaluate 11 years of data (2007-2017). Cases were captured initially from microbiology databases and recaptured with International Classification of Diseases discharge coding. Each case patient was matched with 4 controls to assess feeding status. Patients were matched for chronological age, gestation, discharge status, recruitment site, and calendar year. RESULTS We identified 92 cases of LOGBS: 73 cases on initial capture and 76 cases on the recapture analysis. Case patients were matched with 368 controls: 4 controls to each patient. Seventy-two patients were exposed to breast milk at the time of LOGBS (78.3%), compared with 274 controls (74.5%; odds ratio 1.2 [95% confidence interval 0.7-2.3]). CONCLUSIONS Breastfeeding was not associated with increased risk of LOGBS. Breast milk should not be tested for GBS during a first episode of LOGBS.
Collapse
Affiliation(s)
- Natasha S Ching
- Departments of Infection and Immunity .,Department of Paediatrics, Monash University, Clayton, Victoria, Australia.,General Paediatrics, Monash Children's Hospital, Monash Health, Clayton, Victoria, Australia
| | - Jim P Buttery
- Departments of Infection and Immunity.,Department of Paediatrics, Monash University, Clayton, Victoria, Australia.,Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine.,Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Emily Lai
- Departments of Infection and Immunity.,Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew C Steer
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia.,Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Jane Standish
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia.,Department of Children's Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia
| | - Joel Ziffer
- Department of Paediatrics, Bendigo Health, Bendigo, Victoria, Australia
| | - Andrew J Daley
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia.,Laboratory Services.,Department of Microbiology and Infectious Diseases, The Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Richard Doherty
- Departments of Infection and Immunity.,Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
73
|
Garrido F, Allegaert K, Arribas C, Villamor E, Raffaeli G, Paniagua M, Cavallaro G. Variations in Antibiotic Use and Sepsis Management in Neonatal Intensive Care Units: A European Survey. Antibiotics (Basel) 2021; 10:1046. [PMID: 34572631 PMCID: PMC8469483 DOI: 10.3390/antibiotics10091046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Management of neonatal sepsis and the use of antimicrobials have an important impact on morbidity and mortality. However, there is no recent background on which antibiotic regimens are used in different European neonatal intensive care units (NICUs). Our study aimed to describe the use of antibiotics and other aspects of early- and late-onset sepsis (EOS and LOS, respectively) management by European NICUs. We conducted an online survey among NICUs throughout Europe to collect information about antibiotic stewardship, antibiotic regimens, and general aspects of managing neonatal infections. NICUs from up to 38 European countries responded, with 271 valid responses. Most units had written clinical guidelines for EOS (92.2%) and LOS (81.1%) management. For EOS, ampicillin, penicillin, gentamicin, and amikacin were the most commonly used antibiotics. Analysis of the combinations of EOS regimens showed that the most frequently used was ampicillin plus gentamicin (54.6%). For LOS, the most frequently used antibiotics were vancomycin (52.4%), gentamicin (33.9%), cefotaxime (28%), and meropenem (15.5%). Other aspects of the general management of sepsis have also been analyzed. The management of neonatal sepsis in European NICUs is diverse. There was high self-reported adherence to the local clinical guidelines. There was homogeneity in the combination of antibiotics in EOS but less in LOS.
Collapse
Affiliation(s)
- Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain; (C.A.); (M.P.)
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Cristina Arribas
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain; (C.A.); (M.P.)
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), 6229 Maastricht, The Netherlands;
| | - Genny Raffaeli
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, NICU, 20122 Milan, Italy; (G.R.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Miren Paniagua
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain; (C.A.); (M.P.)
| | - Giacomo Cavallaro
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, NICU, 20122 Milan, Italy; (G.R.); (G.C.)
| | | |
Collapse
|
74
|
Berardi A, Trevisani V, Di Caprio A, Bua J, China M, Perrone B, Pagano R, Lucaccioni L, Fanaro S, Iughetti L, Lugli L, Creti R. Understanding Factors in Group B Streptococcus Late-Onset Disease. Infect Drug Resist 2021; 14:3207-3218. [PMID: 34429620 PMCID: PMC8380284 DOI: 10.2147/idr.s291511] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/14/2021] [Indexed: 12/22/2022] Open
Abstract
Group B streptococcus (GBS) infection remains a leading cause of sepsis, pneumonia, and meningitis in infants. Rates of GBS early onset disease have declined following the widcespread use of intrapartum antibiotic prophylaxis; hence, late-onset infections (LOGBS) are currently a common presentation of neonatal GBS dicsease. The pathogenesis, mode of transmission, and risk factors associated with LOGBS are unclear, which interfere with effective prevention efforts. GBS may be transmitted from the mother to the infant at the time of delivery or during the postpartum period via contaminated breast milk, or as nosocomial or community-acquired infection. Maternal GBS colonization, prematurity, young maternal age, HIV exposure, and ethnicity (Black) are identified as risk factors for LOGBS disease; however, further studies are necessary to confirm additional risk factors, if any, for the implementation of effective prevention strategies. This narrative review discusses current and previous studies that have reported LOGBS. Few well-designed studies have described this condition; therefore, reliable assessment of maternal GBS colonization, breastfeeding, and twin delivery as risk factors for LOGBS remains limited.
Collapse
Affiliation(s)
- Alberto Berardi
- Terapia Intensiva Neonatale, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Viola Trevisani
- Scuola di Specializzazione in Pediatria, Università di Modena & Reggio Emilia, Modena, Italy
| | - Antonella Di Caprio
- Scuola di Specializzazione in Pediatria, Università di Modena & Reggio Emilia, Modena, Italy
| | - Jenny Bua
- Terapia Intensiva Neonatale, IRCCS Azienda Ospedaliero Universitaria “Burlo Garofalo”, Trieste, Italy
| | | | - Barbara Perrone
- Terapia Intensiva Neonatale, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - Rossella Pagano
- Unità Operativa di Pediatria, Civile Sassuolo, Sassuolo, Italy
| | - Laura Lucaccioni
- Unità Operativa di Pediatria, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Silvia Fanaro
- Terapia Intensiva Neonatale, Azienda Ospedaliero-Universitaria S. Anna, Ferrara, Italy
| | - Lorenzo Iughetti
- Scuola di Specializzazione in Pediatria, Università di Modena & Reggio Emilia, Modena, Italy
- Unità Operativa di Pediatria, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Licia Lugli
- Terapia Intensiva Neonatale, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Roberta Creti
- Reparto di Antibiotico Resistenza e Patogeni Speciali (AR-PS), Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
75
|
Travier L, Alonso M, Andronico A, Hafner L, Disson O, Lledo PM, Cauchemez S, Lecuit M. Neonatal susceptibility to meningitis results from the immaturity of epithelial barriers and gut microbiota. Cell Rep 2021; 35:109319. [PMID: 34192531 DOI: 10.1016/j.celrep.2021.109319] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/25/2021] [Accepted: 06/07/2021] [Indexed: 01/06/2023] Open
Abstract
Neonates are highly susceptible to bacterial meningitis as compared to children and adults. Group B streptococcus (GBS) is a major cause of neonatal meningitis. Neonatal meningitis can result from GBS intestinal colonization and translocation across the intestinal barrier (IB). Here, we show that the immaturity of the neonatal intestinal microbiota leads to low resistance to GBS intestinal colonization and permissiveness of the gut-vascular barrier. Moreover, the age-dependent but microbiota-independent Wnt activity in intestinal and choroid plexus (CP) epithelia results in a lower degree of cell-cell junctions' polarization, which favors bacterial translocation. This study thus reveals that neonatal susceptibility to GBS meningitis results from the age-dependent immaturity of the intestinal microbiota and developmental pathways associated with neonatal tissue growth, which both concur to GBS gut colonization, systemic dissemination, and neuroinvasion. Whereas the activation of developmental pathways is intrinsic to neonates, interventions aimed at maturing the microbiota may help prevent neonatal meningitis.
Collapse
Affiliation(s)
- Laetitia Travier
- Institut Pasteur, Biology of Infection Unit, Paris, France; Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Mariana Alonso
- Laboratory for Perception and Memory, Institut Pasteur, Paris, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Paris, France
| | - Alessio Andronico
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Paris, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 2000, Paris, France
| | - Lukas Hafner
- Institut Pasteur, Biology of Infection Unit, Paris, France; Institut National de la Santé et de la Recherche Médicale U1117, Paris, France; Université de Paris, Paris, France
| | - Olivier Disson
- Institut Pasteur, Biology of Infection Unit, Paris, France; Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Pierre-Marie Lledo
- Laboratory for Perception and Memory, Institut Pasteur, Paris, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3571, Paris, France
| | - Simon Cauchemez
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Paris, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 2000, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France; Institut National de la Santé et de la Recherche Médicale U1117, Paris, France; Université de Paris, Paris, France; National Reference Centre and WHO Collaborating Centre Listeria, Institut Pasteur, Paris, France; Necker-Enfants Malades University Hospital, Department of Infectious Diseases and Tropical Medicine, APHP, Institut Imagine, Paris, France.
| |
Collapse
|
76
|
Gonçalves BP, Procter SR, Clifford S, Koukounari A, Paul P, Lewin A, Jit M, Lawn J. Estimation of country-level incidence of early-onset invasive Group B Streptococcus disease in infants using Bayesian methods. PLoS Comput Biol 2021; 17:e1009001. [PMID: 34125829 PMCID: PMC8202927 DOI: 10.1371/journal.pcbi.1009001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/25/2021] [Indexed: 01/30/2023] Open
Abstract
Neonatal invasive disease caused by Group B Streptococcus (GBS) is responsible for much acute mortality and long-term morbidity. To guide development of better prevention strategies, including maternal vaccines that protect neonates against GBS, it is necessary to estimate the burden of this condition globally and in different regions. Here, we present a Bayesian model that estimates country-specific invasive GBS (iGBS) disease incidence in children aged 0 to 6 days. The model combines different types of epidemiological data, each of which has its own limitations: GBS colonization prevalence in pregnant women, risk of iGBS disease in children born to GBS-colonized mothers and direct estimates of iGBS disease incidence where available. In our analysis, we present country-specific maternal GBS colonization prevalence after adjustment for GBS detection assay used in epidemiological studies. We then integrate these results with other epidemiological data and estimate country-level incidence of iGBS disease including in countries with no studies that directly estimate incidence. We are able to simultaneously estimate two key epidemiological quantities: the country-specific incidence of early-onset iGBS disease, and the risk of iGBS disease in babies born to GBS-colonized women. Overall, we believe our method will contribute to a more comprehensive quantification of the global burden of this disease, inform cost-effectiveness assessments of potential maternal GBS vaccines and identify key areas where data are necessary. Invasive disease caused by Group B Streptococcus (GBS) in young infants continues to be a major public health problem in both developed and developing countries. However, data on the incidence of this infection during the first week of life are only available for a small number of countries, which has complicated the quantification of the burden of this disease globally. In this paper, we develop a Bayesian framework to estimate the incidence of invasive GBS infection that combines data from multiple types of epidemiological studies, with adjustment for relevant factors such as diagnostic methods used in the studies. We present estimates from a series of models, and our results highlight the potential weaknesses of different types of studies and the importance to consider the entire evidence when estimating global burden of invasive neonatal infections. We believe this model is a step toward better quantification of the number of cases in different regions.
Collapse
Affiliation(s)
- Bronner P. Gonçalves
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
- * E-mail:
| | - Simon R. Procter
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Sam Clifford
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Artemis Koukounari
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Proma Paul
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Alexandra Lewin
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Joy Lawn
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
77
|
Yuan XY, Liu HZ, Liu JF, Sun Y, Song Y. Pathogenic mechanism, detection methods and clinical significance of group B Streptococcus. Future Microbiol 2021; 16:671-685. [PMID: 34098731 DOI: 10.2217/fmb-2020-0189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Group B Streptococcus (GBS) is the main pathogen of perinatal infection. It can lead to adverse pregnancy, maternal infection, premature delivery, abortion, stillbirth and a series of adverse maternal and infant outcomes such as neonatal sepsis, meningitis or pneumonia during delivery. In order to reduce the infection of perinatal pregnant and the adverse pregnancy outcome, more attention should be paid in the clinical practice, screening efforts, universal detection of GBS infection for pregnant women and preventive treatment for the possible mother infant infection. In this study, the biological characteristics, immunophenotype, major pathogenic mechanism, laboratory test methods and clinical significance of GBS are summarized.
Collapse
Affiliation(s)
- Xiao-Yan Yuan
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Hai-Zhu Liu
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Jia-Fei Liu
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China.,Department of Medical Laboratory Sciences, Weifang Medical University, Weifang, Shandong, 261000, PR China
| | - Yong Sun
- Department of Clinical Lab, Yantai Laiyang Central Hospital, Yantai, Shandong, 264200, PR China
| | - Yu Song
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| |
Collapse
|
78
|
Pediatric sepsis cases diagnosed with group B streptococcal meningitis using next-generation sequencing: a report of two cases. BMC Infect Dis 2021; 21:531. [PMID: 34090359 PMCID: PMC8180162 DOI: 10.1186/s12879-021-06231-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/24/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Group B Streptococcus (GBS) is an important cause of invasive infection in neonates and infants. Cerebrospinal fluid (CSF) findings and culture may not show evidence of infection early in GBS meningitis. Next-generation sequencing (NGS) has the potential to detect microbial genetic material in patients with infectious diseases. We report two cases of infantile sepsis of GBS meningitis with negative results for CSF culture tests, but positive results for NGS analysis. CASE PRESENTATION Patient 1 was a 22-day-old male infant diagnosed with sepsis and meningitis. His CSF findings showed pleocytosis, decreased glucose, and increased protein levels. However, CSF and blood culture results at admission were negative. He received a total of 3 weeks of treatment with ampicillin and cefotaxime, and showed clinical improvement. GBS was detected through NGS analysis of CSF collected at admission. Patient 2 was a 51-day-old male infant with sepsis. CSF findings on admission were normal, and blood and CSF cultures were also negative. Intravenous ampicillin and cefotaxime treatment were initiated. Treatment was de-escalated to ampicillin alone because Enterococcus faecalis was cultured from urine. He was discharged after a total of 1 week of antibiotic treatment. Six days after discharge, he was re-hospitalized for sepsis. Blood and CSF cultures were negative, and E. faecalis was again cultured from urine. He received a total of 3 weeks of ampicillin treatment for enterococcal-induced nephritis and did not relapse thereafter. NGS pathogen searches were retrospectively performed on both blood and CSF collected at the first and second admission. GBS was detected in the CSF collected at the first admission, but no significant pathogen was detected in the other samples. Inadequate treatment for GBS meningitis at the first admission may have caused the recurrence of the disease. CONCLUSION Infantile sepsis may present bacterial meningitis that is not diagnosed by either culture testing or CSF findings. NGS analysis for CSF may be useful for confirming the diagnosis of bacterial meningitis.
Collapse
|
79
|
Chen J, Yang S, Li W, Yu W, Fan Z, Wang M, Feng Z, Tong C, Song B, Ma J, Cui Y. IL-17A Secreted by Th17 Cells Is Essential for the Host against Streptococcus agalactiae Infections. J Microbiol Biotechnol 2021; 31:667-675. [PMID: 33879639 PMCID: PMC9706036 DOI: 10.4014/jmb.2103.03053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022]
Abstract
Streptococcus agalactiae is an important bacterial pathogen and causative agent of diseases including neonatal sepsis and meningitis, as well as infections in healthy adults and pregnant women. Although antibiotic treatments effectively relieve symptoms, the emergence and transmission of multidrug-resistant strains indicate the need for an effective immunotherapy. Effector T helper (Th) 17 cells are a relatively newly discovered subpopulation of helper CD4+ T lymphocytes, and which, by expressing interleukin (IL)-17A, play crucial roles in host defenses against a variety of pathogens, including bacteria and viruses. However, whether S. agalactiae infection can induce the differentiation of CD4+ T cells into Th17 cells, and whether IL-17A can play an effective role against S. agalactiae infections, are still unclear. In this study, we analyzed the responses of CD4+ T cells and their defensive effects after S. agalactiae infection. The results showed that S. agalactiae infection induces not only the formation of Th1 cells expressing interferon (IFN)-γ, but also the differentiation of mouse splenic CD4+ T cells into Th17 cells, which highly express IL-17A. In addition, the bacterial load of S. agalactiae was significantly increased and decreased in organs as determined by antibody neutralization and IL-17A addition experiments, respectively. The results confirmed that IL-17A is required by the host to defend against S. agalactiae and that it plays an important role in effectively eliminating S. agalactiae. Our findings therefore prompt us to adopt effective methods to regulate the expression of IL-17A as a potent strategy for the prevention and treatment of S. agalactiae infection.
Collapse
Affiliation(s)
- Jing Chen
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China,College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Siyu Yang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Wanyu Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Wei Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Zhaowei Fan
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Mengyao Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Zhenyue Feng
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Baifen Song
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Jinzhu Ma
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | - Yudong Cui
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China,College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China,Corresponding author Phone: +13836962508 Fax: +0459-6031177 E-mail:
| |
Collapse
|
80
|
Development and Validation of Enzyme-Linked Immunosorbent Assay for Group B Streptococcal Polysaccharide Vaccine. Vaccines (Basel) 2021; 9:vaccines9060545. [PMID: 34064299 PMCID: PMC8224333 DOI: 10.3390/vaccines9060545] [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: 03/04/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/20/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus, GBS) is a leading cause of neonatal sepsis and meningitis in infants. Limitations of prenatal GBS screening and intrapartum antibiotic prophylaxis render developing GBS vaccines a high priority. In this study, we developed an enzyme-linked immunosorbent assay (ELISA) for the practical and large-scale evaluation of GBS capsular polysaccharide (PS) vaccine immunogenicity against three main serotypes, Ia, III, and V. GBS-ELISA was developed and subsequently validated using a standardized curve-fitting four-parameter logistic method. Specificity was measured using adsorption of serum with homologous and heterologous PS. Homologous adsorption showed a ≥75% inhibition of all three serotypes, whereas with heterologous PS, IgG GBS-ELISA inhibited only ≤25% of serotypes III and V. However, with serotype Ia, IgG antibody levels decreased by >50%, even after adsorption with heterologous PS (III or V). In comparison, the inhibition opsonophagocytic killing assay (OPA) of serotypes Ia GBS exhibited a reduction in opsonophagocytic activity of only 20% and 1.1% for serotypes III and V GBS, respectively. The precision of the GBS-ELISA was assessed in five independent experiments using four serum samples. The coefficient of variation was <5% for all three serotypes. This standardized GBS-ELISA would be useful for GBS vaccine development and its evaluation.
Collapse
|
81
|
Braye K, Ferguson J, Ball J, Foureur M. Intrapartum antibiotic prophylaxis for women who are screened positive for group B streptococcal colonisation: Clinical compliance with the guideline. THE AUSTRALIAN & NEW ZEALAND JOURNAL OF OBSTETRICS & GYNAECOLOGY 2021; 61:870-875. [PMID: 33987829 DOI: 10.1111/ajo.13370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/27/2021] [Accepted: 04/07/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND In high income countries, ~30% of pregnant women are provided with intrapartum antibiotic prophylaxis (IAP) for early-onset group B streptococcal infection (EOGBSI). The infection rate is low, 0.2/1000 livebirths in our jurisdiction, and others. We hypothesised factors, other than IAP alone, were reasons for the low rate of EOGBSI. AIMS Compliance with our local guideline, referred to here as 'the guideline'. METHOD Compliance was defined as an initial dose of benzylpenicillin IAP followed by four-hourly doses until birth. The study population was drawn from 4098 women who had 4100 pregnancies resulting in 4200 babies in an Australian birth setting from 1/1/2016 to 31/12/2016. Most, 93%, were eligible for universal GBS screening, 67% were reported as screened and 90% of these had a result documented; 23% were positive for GBS. A random sample (n = 223) was taken for further analysis. RESULTS The adjusted odds of receiving benzylpenicillin IAP in accord with the guideline were three times higher among primiparous compared to multiparous women (P < 0.001, odds ratio (OR) = 3.4, 95% CI 1.7-6.7) and three times higher among women experiencing induction of labour compared to women who commenced labour spontaneously (P < 0.001, OR = 3.4, 95% CI 1.8-6.3). Of the 223 women, 188 received IAP: 176 received benzylpenicillin IAP, 31% (or 24% of the total sample) received this intervention in accord with the guideline, 24% received benzylpenicillin ≥4 h before birth but not in accord with the guideline and 44% received benzylpenicillin <4 h before birth. CONCLUSION We conclude that sub-optimal compliance was largely a consequence of an unrealistic guideline.
Collapse
Affiliation(s)
- Kathryn Braye
- Faculty of Health, University of Technology Sydney, Sydney, New South Wales, Australia.,Department of Maternity and gynaecology, John Hunter Hospital, Hunter New England Local Health District, New Lambton, New South Wales, Australia
| | - John Ferguson
- NSW Health Pathology, John Hunter Hospital, Hunter New England Local Health District, New Lambton, New South Wales, Australia.,The University of Newcastle Faculty of Health and Medicine, Callaghan, New South Wales, Australia
| | - Jean Ball
- Clinical Research Design IT and Statistical Support Unit (CReDITSS), The University of Newcastle, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Maralyn Foureur
- Hunter New England Local Health District, Nurses and Midwives Research Centre, The University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
82
|
Hulbah M, Croxen MA, Tyrrell GJ. Phenotypic changes in group B streptococci grown in the presence of the polyols, erythritol, sorbitol and mannitol. BMC Microbiol 2021; 21:145. [PMID: 33985431 PMCID: PMC8117501 DOI: 10.1186/s12866-021-02208-z] [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: 06/22/2020] [Accepted: 04/29/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Group B streptococci (GBS) are important neonatal bacterial pathogens that can cause severe invasive disease in the newborn. It is thought that in many cases of invasive neonatal GBS disease, the bacteria ascend the vagina into the uterus and infect the amniotic fluid surrounding the fetus. Important constituents of this environment include the polyols or sugar alcohols of which erythritol, sorbitol and mannitol are examples. The aim of our study was to investigate the effect of polyols on GBS grown in media containing these sugar alcohols. RESULTS GBS incubated in varying concentrations of polyols (erythritol, sorbitol or mannitol) did not display any significant enhancement or inhibition of bacterial growth. However, growth of GBS in the presence of erythritol significantly increased the surface expression of GBS-PGK (a plasminogen binding protein) 1.25 to 1.5-fold depending on the erythritol concentration and significantly enhanced the survival in human blood 3X to 18X depending on the concentration of polyol used. Interestingly, GBS grown in 1% erythritol significantly increased invasion by the bacteria of HeLa cells (epithelial cell line) (150% vs 100%) however, at higher concentrations (2% or 4% of polyol) the number of CFUs was significantly reduced (55-75% vs 100%) suggesting higher concentrations of polyols may inhibit invasion. Erythritol also increased GBS hemolytic activity as well as enhancing biofilm formation 1.4X to 3.3X depending on the concentration of polyol used. CONCLUSIONS GBS grown in the presence of polyols alters the bacteria's phenotype resulting in changes associated with GBS virulence. This effect was greatest for the polyol erythritol.
Collapse
Affiliation(s)
- Maram Hulbah
- Division of Diagnostic and Applied Microbiology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Matthew A Croxen
- Division of Diagnostic and Applied Microbiology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
- Alberta Precision Laboratories-Public Health, Edmonton, Alberta, T6G 2J2, Canada
| | - Gregory J Tyrrell
- Division of Diagnostic and Applied Microbiology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada.
- Alberta Precision Laboratories-Public Health, Edmonton, Alberta, T6G 2J2, Canada.
| |
Collapse
|
83
|
Bedeley E, Gori A, Yeboah-Manu D, Diallo K. Control of Streptococcal Infections: Is a Common Vaccine Target Achievable Against Streptococcus agalactiae and Streptococcus pneumoniae. Front Microbiol 2021; 12:658824. [PMID: 33967998 PMCID: PMC8103614 DOI: 10.3389/fmicb.2021.658824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/30/2021] [Indexed: 12/21/2022] Open
Abstract
Both Streptococcus agalactiae [group B streptococcus (GBS)] and Streptococcus pneumoniae (pneumococcus) remain significant pathogens as they cause life threatening infections mostly in children and the elderly. The control of diseases caused by these pathogens is dependent on antibiotics use and appropriate vaccination. The introduction of the pneumococcal conjugate vaccines (PCVs) against some serotypes has led to reduction in pneumococcal infections, however, the subsequent serotype switching, and replacement has been a serious challenge. On the other hand, no vaccine is yet licensed for use in the control of GBS diseases. In this review, we provide an overview of the history and global disease burden, disease pathophysiology and management, vaccines update, and the biology of both pathogens. Furthermore, we address recent findings regarding structural similarities that could be explored for vaccine targets across both mucosal pathogens. Finally, we conclude by proposing future genomic sequence comparison using the wealth of available sequences from both species and the possibility of identifying more related structural components that could be exploited for pan-pathogen vaccine development.
Collapse
Affiliation(s)
- Edmund Bedeley
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Andrea Gori
- NIHR Global Health Research Unit on Mucosal Pathogens, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dorothy Yeboah-Manu
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Kanny Diallo
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Centre Suisse de Recherche Scientifique de Côte d’Ivoire, Abidjan, Côte d’Ivoire
| |
Collapse
|
84
|
Pastuszka A, Beauruelle C, Camiade E, Rousseau GM, Moineau S, Mereghetti L, Horvath P, Lanotte P. Functional Study of the Type II-A CRISPR-Cas System of Streptococcus agalactiae Hypervirulent Strains. CRISPR J 2021; 4:233-242. [PMID: 33876956 DOI: 10.1089/crispr.2020.0145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Nearly all strains of Streptococcus agalactiae, the leading cause of invasive infections in neonates, encode a type II-A clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system. Interestingly, S. agalactiae strains belonging to the hypervirulent Sequence Type 17 (ST17) contain significantly fewer spacers in their CRISPR locus than other lineages, which could be the result of a less functional CRISPR-Cas system. Here, we revealed one large deletion in the ST17 cas promoter region and we evaluated its impact on the transcription of cas genes as well as the functionalities of the CRISPR-Cas system. We demonstrated that Cas9 interference is functional and that the CRISPR-Cas system of ST17 strains can still acquire new spacers, despite the absence of a regular cas promoter. We demonstrated that a promoter sequence upstream of srn036, a small RNA partially overlapping the antisense tracrRNA, is responsible for the ST17 CRISPR-Cas adaptation and interference activities.
Collapse
Affiliation(s)
- Adeline Pastuszka
- ISP, Université de Tours, INRAE, Tours, France; Dangé-Saint-Romain, France.,Service de Bactériologie-Virologie-Hygiène Hospitalière, CHRU de Tours, Tours, France; Dangé-Saint-Romain, France
| | - Clémence Beauruelle
- Département de Bactériologie-Virologie, Hygiène Hospitalière et Parasitologie-Mycologie, Centre Hospitalier Régional Universitaire (CHRU) de Brest, Brest, France; Dangé-Saint-Romain, France.,Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest, France; Dangé-Saint-Romain, France
| | - Emilie Camiade
- ISP, Université de Tours, INRAE, Tours, France; Dangé-Saint-Romain, France
| | - Geneviève M Rousseau
- Département de Biochimie, de Microbiologie, et de Bio-informatique, Faculté des Sciences et de Génie, Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec City, Canada; Dangé-Saint-Romain, France
| | - Sylvain Moineau
- Département de Biochimie, de Microbiologie, et de Bio-informatique, Faculté des Sciences et de Génie, Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec City, Canada; Dangé-Saint-Romain, France.,Félix d'Hérelle Reference Center for Bacterial Viruses, Université Laval, Québec City, Canada; and Dangé-Saint-Romain, France
| | - Laurent Mereghetti
- ISP, Université de Tours, INRAE, Tours, France; Dangé-Saint-Romain, France.,Service de Bactériologie-Virologie-Hygiène Hospitalière, CHRU de Tours, Tours, France; Dangé-Saint-Romain, France
| | | | - Philippe Lanotte
- ISP, Université de Tours, INRAE, Tours, France; Dangé-Saint-Romain, France.,Service de Bactériologie-Virologie-Hygiène Hospitalière, CHRU de Tours, Tours, France; Dangé-Saint-Romain, France
| |
Collapse
|
85
|
Prevalence of group B streptococcal colonization in the healthy non-pregnant population: a systematic review and meta-analysis. Clin Microbiol Infect 2021; 27:968-980. [PMID: 33813109 DOI: 10.1016/j.cmi.2021.03.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/17/2021] [Accepted: 03/06/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Colonization and transmission precede invasive group B streptococcal (GBS) disease. Data on GBS colonization prevalence, detection methods and risk factors for carriage are relevant for vaccine development and to understand GBS pathogenesis. OBJECTIVES To evaluate GBS colonization prevalence after the first week of life in the healthy non-pregnant population. DATA SOURCES Pubmed/Medline, Embase, Latin American and Caribbean Health Sciences Literature, World Health Organization Library Information System, and Scopus. Search performed 12 January 2021 with search terms related to 'GBS' and 'colonization, epidemiology, prevalence or screening' without restrictions. STUDY ELIGIBILITY CRITERIA All studies that reported prevalence of GBS colonization (any site) in the healthy population. PARTICIPANTS All individuals (>6 days of age), with no indication of pregnancy, invasive disease or severe underlying immunological co-morbidities. METHODS Logit transformation and a random effects model (DerSimonian and Laird) were used to pool colonization estimates. Subgroup analysis and meta-regression on a priori determined subgroups were performed. RESULTS We included 98 studies with 43 112 participants. Our search identified 9309 studies of which 8831 were excluded based on title and abstract and 380 after reading the full text. Colonization rates varied considerably between studies (I2 = 97%), which could be partly explained by differences in culture methods (R2 = 27%), culture sites (R2 = 24%), continent (R2 = 10%) and participant's age (R2 = 6%). Higher prevalence was found with selective culture methods (19%, 95% CI 16%-23% versus non-selective methods 8%, 95% CI 6%-9%; p < 0.0001). Colonization rates were highest in rectum (19%, 95% CI 15%-24%), vagina (14%, 95% CI 12%-17%) and urethra (9%, 95% CI 5%-18%). In participants with negative rectal cultures, 7% (95% CI 5%-9%) had GBS cultured from another niche. Colonization prevalence was lower in children (6 months to 16 years; 3%, 95% CI 2%-5%) compared with adults (16%, 95% CI 14%-20%; p < 0.0001). Using selective culture methods in adults resulted in a prevalence of 26% (95% CI 19%-33%) rectal, 21% (95% CI 17%-25%) vaginal and 9% (95% CI 6%-14%) urethral colonization. CONCLUSION The rectum is the most common body site colonized by GBS. The best approach to screen for any GBS colonization is to screen multiple body sites using selective culture methods.
Collapse
|
86
|
van Sorge NM, Bonsor DA, Deng L, Lindahl E, Schmitt V, Lyndin M, Schmidt A, Nilsson OR, Brizuela J, Boero E, Sundberg EJ, van Strijp JAG, Doran KS, Singer BB, Lindahl G, McCarthy AJ. Bacterial protein domains with a novel Ig-like fold target human CEACAM receptors. EMBO J 2021; 40:e106103. [PMID: 33522633 PMCID: PMC8013792 DOI: 10.15252/embj.2020106103] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 01/19/2023] Open
Abstract
Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed β protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in β represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.
Collapse
Affiliation(s)
- Nina M van Sorge
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
- Present address:
Department of Medical Microbiology,Infection Prevention and Netherlands Reference Laboratory for Bacterial MeningitisAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Daniel A Bonsor
- Institute of Human VirologyUniversity of Maryland School of MedicineUniversity of MarylandBaltimoreMDUSA
| | - Liwen Deng
- Department of Immunology & MicrobiologyUniversity of Colorado Anschutz Medical CampusAuroraCOUSA
| | - Erik Lindahl
- Department of Biochemistry and BiophysicsScience for Life LaboratoryStockholm UniversityStockholmSweden
| | - Verena Schmitt
- Institute of AnatomyMedical Faculty, University Duisburg‐EssenEssenGermany
| | - Mykola Lyndin
- Institute of AnatomyMedical Faculty, University Duisburg‐EssenEssenGermany
- Department of PathologySumy State UniversitySumyUkraine
| | - Alexej Schmidt
- Department of Medical BiosciencesUmeå UniversityPathology, UmeåSweden
| | - Olof R Nilsson
- Department of Laboratory MedicineDivision of Medical MicrobiologyLund UniversityLundSweden
| | - Jaime Brizuela
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology & InfectionImperial College LondonLondonUK
| | - Elena Boero
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Eric J Sundberg
- Institute of Human VirologyUniversity of Maryland School of MedicineUniversity of MarylandBaltimoreMDUSA
- Department of BiochemistryEmory University School of MedicineAtlantaGAUSA
| | - Jos A G van Strijp
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Kelly S Doran
- Department of Immunology & MicrobiologyUniversity of Colorado Anschutz Medical CampusAuroraCOUSA
| | - Bernhard B Singer
- Institute of AnatomyMedical Faculty, University Duisburg‐EssenEssenGermany
| | - Gunnar Lindahl
- Department of Laboratory MedicineDivision of Medical MicrobiologyLund UniversityLundSweden
- Department of ChemistryDivision of Applied MicrobiologyLund UniversityLundSweden
| | - Alex J McCarthy
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology & InfectionImperial College LondonLondonUK
| |
Collapse
|
87
|
Ren J, Qiang Z, Li YY, Zhang JN. Biomarkers for a histological chorioamnionitis diagnosis in pregnant women with or without group B streptococcus infection: a case-control study. BMC Pregnancy Childbirth 2021; 21:250. [PMID: 33765949 PMCID: PMC7993527 DOI: 10.1186/s12884-021-03731-7] [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] [Received: 09/22/2020] [Accepted: 03/17/2021] [Indexed: 01/12/2023] Open
Abstract
Background Chorioamnionitis may cause serious perinatal and neonatal adverse outcomes, and group B streptococcus (GBS) is one of the most common bacteria isolated from human chorioamnionitis. The present study analyzed the impact of GBS infection and histological chorioamnionitis (HCA) on pregnancy outcomes and the diagnostic value of various biomarkers. Methods Pregnant women were grouped according to GBS infection and HCA detection. Perinatal and neonatal adverse outcomes were recorded with a follow-up period of 6 weeks. The white blood cell count (WBC), neutrophil ratio, and C-reactive protein (CRP) level from peripheral blood and soluble intercellular adhesion molecule-1 (sICAM-1), interleukin 8 (IL-8), and tumor necrosis factor α (TNF-α) levels from cord blood were assessed. Results A total of 371 pregnant women were included. Pregnant women with GBS infection or HCA had a higher risk of pathological jaundice and premature rupture of membranes and higher levels of sICAM-1, IL-8, and TNF-α in umbilical cord blood. Univariate and multivariate regression analysis revealed that sICMA-1, IL-8, TNF-α, WBC, and CRP were significantly related to an increased HCA risk. For all included pregnant women, TNF-α had the largest receiver operating characteristic (ROC) area (area: 0.841; 95% CI: 0.778–0.904) of the biomarkers analyzed. TNF-α still had the largest area under the ROC curve (area: 0.898; 95% CI: 0.814–0.982) for non-GBS-infected pregnant women, who also exhibited a higher neutrophil ratio (area: 0.815; 95% CI: 0.645–0.985) and WBC (area: 0.849; 95% CI: 0.72–0.978), but all biomarkers had lower value in the diagnosis of HCA in GBS-infected pregnant women. Conclusion GBS infection and HCA correlated with several perinatal and neonatal adverse outcomes. TNF-α in cord blood and WBCs in peripheral blood had diagnostic value for HCA in non-GBS-infected pregnant women but not GBS-infected pregnant women. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-021-03731-7.
Collapse
Affiliation(s)
- Jie Ren
- Second Department of Obstetrics, The Fourth Hospital of Shijiazhuang, No.206, Zhongshan East Road, Chang'an District, Shijiazhuang, Hebei, People's Republic of China, 050011
| | - Zhe Qiang
- Second Department of Obstetrics, The Fourth Hospital of Shijiazhuang, No.206, Zhongshan East Road, Chang'an District, Shijiazhuang, Hebei, People's Republic of China, 050011.
| | - Yuan-Yuan Li
- Perinatal center, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, People's Republic of China, 050000
| | - Jun-Na Zhang
- Second Department of Obstetrics, The Fourth Hospital of Shijiazhuang, No.206, Zhongshan East Road, Chang'an District, Shijiazhuang, Hebei, People's Republic of China, 050011
| |
Collapse
|
88
|
Bacterial Meningitis in Children: Neurological Complications, Associated Risk Factors, and Prevention. Microorganisms 2021; 9:microorganisms9030535. [PMID: 33807653 PMCID: PMC8001510 DOI: 10.3390/microorganisms9030535] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 01/07/2023] Open
Abstract
Bacterial meningitis is a devastating infection, with a case fatality rate of up to 30% and 50% of survivors developing neurological complications. These include short-term complications such as focal neurological deficit and subdural effusion, and long-term complications such as hearing loss, seizures, cognitive impairment and hydrocephalus. Complications develop due to bacterial toxin release and the host immune response, which lead to neuronal damage. Factors associated with increased risk of developing neurological complications include young age, delayed presentation and Streptococcus pneumoniae as an etiologic agent. Vaccination is the primary method of preventing bacterial meningitis and therefore its complications. There are three vaccine preventable causes: Haemophilus influenzae type b (Hib), S. pneumoniae, and Neisseria meningitidis. Starting antibiotics without delay is also critical to reduce the risk of neurological complications. Additionally, early adjuvant corticosteroid use in Hib meningitis reduces the risk of hearing loss and severe neurological complications.
Collapse
|
89
|
Perme T, Golparian D, Unemo M, Jeverica S. Lack of diagnostic-escape mutants of group B streptococcus in Slovenia. Clin Microbiol Infect 2021; 27:1054-1055. [PMID: 33540114 DOI: 10.1016/j.cmi.2021.01.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Tina Perme
- Department of Perinatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Daniel Golparian
- WHO Collaborating Centre for Gonorrhoea and other STIs, National Reference Laboratory for STIs, Department of Laboratory Medicine, Microbiology, Örebro University, Örebro, Sweden
| | - Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and other STIs, National Reference Laboratory for STIs, Department of Laboratory Medicine, Microbiology, Örebro University, Örebro, Sweden
| | - Samo Jeverica
- National Laboratory of Health, Environment and Food, Maribor, Slovenia.
| |
Collapse
|
90
|
Schmitt C, Novy M, Hascoët JM. Term newborns at risk for early-onset neonatal sepsis: Clinical surveillance versus systematic paraclinical test. Arch Pediatr 2021; 28:117-122. [PMID: 33446431 DOI: 10.1016/j.arcped.2020.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/06/2020] [Accepted: 11/21/2020] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Early-onset neonatal sepsis is a rare but potentially lethal infection that is very often suspected in daily practice. Previous national guidelines recommended the use of systematic paraclinical tests for healthy term newborns with suspected infection. These guidelines were updated in 2017 by the French Health Authority (Haute Autorité de santé), and promote initial clinical monitoring taking into account the infectious risk level for term and near-term born infants. OBJECTIVES To assess the impact of the new recommendations on antibiotic therapy prescription and invasive tests, and on the outcomes of infants born from 36weeks' gestation. MATERIALS AND METHODS This study compared the management and the outcome of neonates born from 36weeks' gestation at the level III University Hospital of Nancy, according to their infectious risk level during two periods, before and after the update of national recommendations: from July 1 to December 31, 2017, versus July 1 to December 31, 2018. Data were retrospectively collected from the infants' files. This study compared the number and length of antibiotic treatment and the number of invasive tests, the number of documented infections, the number and length of hospitalization, and mortality between the two periods. RESULTS During the first period, among 1248 eligible newborns, 643 presented an infectious risk factor, versus 1152 newborns with 343 having an infectious risk factor during the second period. Antibiotic treatment was initiated for 18 newborns during the first period (1.4%) and for nine during the second (0.8%) (P=0.13). The mean (SD) duration of the antibiotic treatment was longer in the first than in the second period: 6.3±2days vs. 3.1±2.3days (P=0.003). There was no death related to neonatal infection. A total of 1052 blood samples were collected during the first period versus 51 during the second (P<0.01). There was no documented infection. In the first period, there were 18 newborns (1.4%) hospitalized for suspected infection versus nine (0.8%) in the second period (P=0.13). The duration of hospitalization was 5.7±1.7days in the first period versus 5.2±3days in the second (P=0.33). CONCLUSION In this study, the application of the new guidelines enabled a reduction of antibiotic exposure and a reduction of invasive tests without additional risk.
Collapse
Affiliation(s)
- C Schmitt
- Neonatal Intensive Care Unit, Maternité Régionale, CHRU Nancy, 54000 Nancy, France.
| | - M Novy
- Neonatal Intensive Care Unit, Maternité Régionale, CHRU Nancy, 54000 Nancy, France
| | - J-M Hascoët
- Neonatal Intensive Care Unit, Maternité Régionale, CHRU Nancy, 54000 Nancy, France; DevAH, Lorraine University, 54000 Nancy, France
| |
Collapse
|
91
|
Leroux-Roels G, Bebia Z, Maes C, Aerssens A, De Boever F, Grassano L, Buffi G, Margarit I, Karsten A, Cho S, Slobod K, Corsaro B, Henry O. Safety and Immunogenicity of a Second Dose of an Investigational Maternal Trivalent Group B Streptococcus Vaccine in Nonpregnant Women 4-6 Years After a First Dose: Results From a Phase 2 Trial. Clin Infect Dis 2021; 70:2570-2579. [PMID: 31394574 PMCID: PMC7286364 DOI: 10.1093/cid/ciz737] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/09/2019] [Indexed: 12/23/2022] Open
Abstract
Background Maternal immunization against group B streptococcus (GBS) could protect infants from invasive GBS disease. Additional doses in subsequent pregnancies may be needed. We evaluated the safety and immunogenicity of a second dose of an investigational trivalent CRM197-glycoconjugate GBS vaccine (targeting serotypes Ia/Ib/III), administered to nonpregnant women 4–6 years postdose 1. Methods Healthy women either previously vaccinated with 1 dose of trivalent GBS vaccine 4–6 years before enrollment (n = 53) or never GBS vaccinated (n = 27) received a single trivalent GBS vaccine injection. Adverse events (AEs) were recorded. Serotype-specific (Ia/Ib/III) anti-GBS antibodies were measured by multiplex immunoassay prevaccination and 30/60 days postvaccination. Results AEs were reported with similar rates after a first or second dose; none were serious. Of previously GBS-vaccinated women, 92%–98% had anti-GBS concentrations that exceeded an arbitrary threshold (8 µg/mL) for each serotype 60 days postdose 2 vs 36%–56% postdose 1 in previously non–GBS-vaccinated women. Of previously GBS-vaccinated women with undetectable baseline (predose 1) anti-GBS levels, 90%–98% reached this threshold postdose 2. For each serotype, anti-GBS geometric mean concentrations (GMCs) 30/60 days postdose 2 in previously GBS-vaccinated women were ≥200-fold higher than baseline GMCs. Among women with undetectable baseline anti-GBS levels, postdose 2 GMCs in previously GBS-vaccinated women exceeded postdose 1 GMCs in previously non–GBS-vaccinated women (≥7-fold). Conclusions A second trivalent GBS vaccine dose administered 4–6 years postdose 1 was immunogenic with a favorable safety profile. Women with undetectable preexisting anti-GBS concentrations may benefit from a sufficiently spaced second vaccine dose. Clinical Trials Registration NCT02690181
Collapse
Affiliation(s)
- Geert Leroux-Roels
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | | | - Cathy Maes
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | - Annelies Aerssens
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | - Fien De Boever
- Center for Vaccinology, Ghent University and Ghent University Hospital, Belgium
| | | | | | | | | | - Stephen Cho
- Novartis, Cambridge, Massachusetts.,GSK, Cambridge, Massachusetts
| | - Karen Slobod
- Novartis, Cambridge, Massachusetts.,GSK, Cambridge, Massachusetts
| | | | | |
Collapse
|
92
|
Lohrmann F, Berg A, Wicker E, Imm A, Krause G, Zürn K, Berner R, Hufnagel M, Lander F. Prevalence of Capsular Serotype, Pilus Island Distribution, and Antibiotic Resistance in Pediatric and Adult Invasive Group B Streptococcus Isolates: Data From a Nationwide Prospective Surveillance Study in Germany. Pediatr Infect Dis J 2021; 40:76-82. [PMID: 33201062 DOI: 10.1097/inf.0000000000002943] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
For neonates, group B Streptococcus is life threatening. Current prevention strategies remain insufficient, especially for cases of late-onset sepsis, where intrapartum antibiotic prophylaxis has demonstrated no benefit. One promising approach is the vaccination of pregnant women, which offers protective immunity via transplacental transmission of neutralizing antibodies. Our nationwide, prospective surveillance study aimed to characterize the prevalence of pilus antigen, capsular polysaccharide serotypes, and antibiotic resistance from invasive GBS infections in neonates and compare these results with those from children and adults in Germany. Our study includes 173 neonatal isolates of a total of 450 reported cases during the study period (incidence: 0.34/1000 live births), in addition to 2 pediatric and 803 adult isolates. The comparison between neonatal and adult isolates reveals age-dependent differences in capsular serotype and pilus type distribution and differences in antibiotic resistance patterns.
Collapse
Affiliation(s)
- Florens Lohrmann
- From the Department of Pediatrics and Adolescent Medicine, University Medical Center, Medical Faculty
- Spemann Graduate School of Biology and Medicine (SGBM), Faculty of Biology
- IMM-PACT Clinician Scientist Program, Faculty of Medicine, Freiburg, Germany
| | - Angela Berg
- From the Department of Pediatrics and Adolescent Medicine, University Medical Center, Medical Faculty
- Department for Gynecology and Obstetrics, DRK Hospital Westend, Berlin, Germany
| | - Esther Wicker
- Department of Anaesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anita Imm
- From the Department of Pediatrics and Adolescent Medicine, University Medical Center, Medical Faculty
| | - Gérard Krause
- Epidemiology Department, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Infectious Disease Epidemiology, TWINCORE, Hannover, Germany
- Translational Infrastructure Epidemiology, German Centre for Infection Research (DZIF), Braunschweig, Germany
| | - Katharina Zürn
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Reinhard Berner
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Markus Hufnagel
- From the Department of Pediatrics and Adolescent Medicine, University Medical Center, Medical Faculty
| | - Fabian Lander
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
93
|
Abu-Raya B, Maertens K. Protection of the Newborn Through Vaccination in Pregnancy. Neoreviews 2021; 22:e25-e39. [PMID: 33386312 DOI: 10.1542/neo.22-1-e25] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Newborns and infants are at risk for severe infections with some pathogens (eg, Bordetella pertussis, influenza, respiratory syncytial virus, group B Streptococcus) during early life. To decrease this window of high susceptibility to some infections during early life and protect young infants, vaccination in pregnancy against some vaccine-preventable diseases (eg, influenza, pertussis, tetanus) has been recommended in an increasing number of countries with notable success. In addition, recent advances have been made in developing vaccines for pregnant women with the aim of reducing the respiratory syncytial virus and group B Streptococcus burden in infancy. In this article, we review the vaccines currently recommended during pregnancy and their benefits to newborns and infants. We also discuss progress made in the development of other vaccines that are expected to be evaluated in pregnant women in the near future.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Vaccine Evaluation Center, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kirsten Maertens
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Diseases Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
94
|
Lee J, Naiduvaje K, Chew KL, Charan N, Chan YH, Lin RTP, Yong EL. Preventing early-onset group B streptococcal sepsis: clinical risk factor-based screening or culture-based screening? Singapore Med J 2021; 62:34-38. [PMID: 33619578 PMCID: PMC8027162 DOI: 10.11622/smedj.2019155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Two strategies are available for prevention of early-onset group B streptococcal (GBS) sepsis - clinical risk factor-based screening and routine culture-based screening of pregnant women for GBS colonisation. In our hospital, we switched from the former to the latter approach in 2014. METHODS We compared the incidence of early-onset GBS sepsis during 2001-2015 between infants born to pregnant women who were screened for GBS colonisation and those born to women who were not screened. RESULTS Among 41,143 live births, there were nine cases of early-onset GBS sepsis. All infants with GBS sepsis were born to pregnant women who were not screened for GBS colonisation. The incidence of early-onset GBS sepsis among infants of women who were not screened was 0.41 per 1,000 live births (95% confidence interval [CI] 0.19-0.77) when compared to infants of women who were screened, for whom the sepsis incidence was zero per 1,000 live births (95% CI 0-0.19; p = 0.005). CONCLUSION Our data suggests that routine culture-based screening of pregnant women for GBS colonisation is a better preventive strategy for early-onset GBS sepsis in neonates when compared to clinical risk factor-based screening.
Collapse
Affiliation(s)
- Jiun Lee
- Department of Neonatology, National University Health System, Singapore
| | | | - Ka Lip Chew
- Department of Laboratory Medicine, National University Health System, Singapore
| | - Natasha Charan
- Department of Neonatology, National University Health System, Singapore
| | - Yiong Huak Chan
- Biostatistics Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Raymond Tzer-Pin Lin
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Eu Leong Yong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
95
|
Perme T, Golparian D, Bombek Ihan M, Rojnik A, Lučovnik M, Kornhauser Cerar L, Fister P, Lozar Krivec J, Grosek Š, Ihan A, Jeverica S, Unemo M. Genomic and phenotypic characterisation of invasive neonatal and colonising group B Streptococcus isolates from Slovenia, 2001-2018. BMC Infect Dis 2020; 20:958. [PMID: 33327946 PMCID: PMC7739447 DOI: 10.1186/s12879-020-05599-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/09/2020] [Indexed: 11/30/2022] Open
Abstract
Background Group B Streptococcus (GBS) is the leading cause of invasive neonatal disease in the industrialized world. We aimed to genomically and phenotypically characterise invasive GBS isolates in Slovenia from 2001 to 2018 and contemporary colonising GBS isolates from screening cultures in 2018. Methods GBS isolates from 101 patients (invasive isolates) and 70 pregnant women (colonising isolates) were analysed. Basic clinical characteristics of the patients were collected from medical records. Antimicrobial susceptibility and phenotypic capsular serotype were determined. Whole-genome sequencing was performed to assign multilocus sequence types (STs), clonal complexes (CCs), pathogenicity/virulence factors, including capsular genotypes, and genome-based phylogeny. Results Among invasive neonatal disease patients, 42.6% (n = 43) were females, 41.5% (n = 39/94) were from preterm deliveries (< 37 weeks gestation), and 41.6% (n = 42) had early-onset disease (EOD). All isolates were susceptible to benzylpenicillin with low minimum inhibitory concentrations (MICs; ≤0.125 mg/L). Overall, 7 serotypes were identified (Ia, Ib, II-V and VIII); serotype III being the most prevalent (59.6%). Twenty-eight MLST STs were detected that clustered into 6 CCs. CC-17 was the most common CC overall (53.2%), as well as among invasive (67.3%) and non-invasive (32.9%) isolates (p < 0.001). CC-17 was more common among patients with late-onset disease (LOD) (81.4%) compared to EOD (47.6%) (p < 0.001). The prevalence of other CCs was 12.9% (CC-23), 11.1% (CC-12), 10.5% (CC-1), 8.2% (CC-19), and 1.8% (CC-498). Of all isolates, 2.3% were singletons. Conclusions A high prevalence of hypervirulent CC-17 isolates, with low genomic diversity and characteristic profile of pathogenicity/virulence factors, was detected among invasive neonatal and colonising GBS isolates from pregnant women in Slovenia. This is the first genomic characterisation of GBS isolates in Slovenia and provides valuable microbiological and genomic baseline data regarding the invasive and colonising GBS population nationally. Continuous genomic surveillance of GBS infections is crucial to analyse the impact of IND prevention strategies on the population structure of GBS locally, nationally, and internationally. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-020-05599-y.
Collapse
Affiliation(s)
- Tina Perme
- Department of Perinatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Daniel Golparian
- WHO Collaborating Centre for Gonorrhoea and other STIs, National Reference Laboratory for STIs, Department of Laboratory Medicine, Microbiology, Örebro University, SE-70185, Örebro, Sweden
| | - Maja Bombek Ihan
- National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Andrej Rojnik
- National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Miha Lučovnik
- Department of Perinatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | - Petja Fister
- Department of Neonatology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jana Lozar Krivec
- Department of Neonatology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Štefan Grosek
- Department of Perinatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Chair of Pediatrics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Institute for Microbiology and Immunology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Samo Jeverica
- National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and other STIs, National Reference Laboratory for STIs, Department of Laboratory Medicine, Microbiology, Örebro University, SE-70185, Örebro, Sweden.
| |
Collapse
|
96
|
Carboni F, Adamo R. Structure-based glycoconjugate vaccine design: The example of Group B Streptococcus type III capsular polysaccharide. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 35-36:23-33. [PMID: 33388125 DOI: 10.1016/j.ddtec.2020.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 06/12/2023]
Abstract
Microbial surface polysaccharides are important virulence factors and targets for vaccine development. Glycoconjugate vaccines, obtained by covalently linking carbohydrates and proteins, are well established tools for prevention of bacterial infections. Elucidation of the minimal portion involved in the interactions with functional antibodies is of utmost importance for the understanding of their mechanism of induction of protective immune responses and the design of synthetic glycan based vaccines. Typically, this is achieved by combination of different techniques, which include ELISA, glycoarray, Surface Plasmon Resonance in conjunction with approaches for mapping at atomic level the position involved in binding, such as Saturation Transfer NMR and X-ray crystallography. This review provides an overview of the structural studies performed to map glycan epitopes (glycotopes), with focus on the highly complex structure of Group B Streptococcus type III (GBSIII) capsular polysaccharide. Furthermore, it describes the rational process followed to translate the obtained information into the design of a protective glycoconjugate vaccine based on a well-defined synthetic glycan epitope.
Collapse
|
97
|
Manzer HS, Nobbs AH, Doran KS. The Multifaceted Nature of Streptococcal Antigen I/II Proteins in Colonization and Disease Pathogenesis. Front Microbiol 2020; 11:602305. [PMID: 33329493 PMCID: PMC7732690 DOI: 10.3389/fmicb.2020.602305] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022] Open
Abstract
Streptococci are Gram-positive bacteria that belong to the natural microbiota of humans and animals. Certain streptococcal species are known as opportunistic pathogens with the potential to cause severe invasive disease. Antigen I/II (AgI/II) family proteins are sortase anchored cell surface adhesins that are nearly ubiquitous across streptococci and contribute to many streptococcal diseases, including dental caries, respiratory tract infections, and meningitis. They appear to be multifunctional adhesins with affinities to various host substrata, acting to mediate attachment to host surfaces and stimulate immune responses from the colonized host. Here we will review the literature including recent work that has demonstrated the multifaceted nature of AgI/II family proteins, focusing on their overlapping and distinct functions and their important contribution to streptococcal colonization and disease.
Collapse
Affiliation(s)
- Haider S. Manzer
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angela H. Nobbs
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Kelly S. Doran
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
98
|
Gyllensvärd J, Ingemansson F, Hentz E, Studahl M, Elfvin A. C-reactive protein- and clinical symptoms-guided strategy in term neonates with early-onset sepsis reduced antibiotic use and hospital stay: a quality improvement initiative. BMC Pediatr 2020; 20:531. [PMID: 33218324 PMCID: PMC7678045 DOI: 10.1186/s12887-020-02426-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/10/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Early-onset sepsis (EOS) is a potentially life-threatening complication of birth. Clinical symptoms are often unspecific and biomarkers have low predictive values for EOS. Therefore, clinical suspicion often leads to antibiotic therapy in neonates with a negative blood culture. In the study we evaluated if a quality improvement initiative could reduce unwarranted antibiotic use in a safe way in term neonates with culture-negative sepsis. METHODS The quality improvement initiative included new treatment guidelines and were introduced on 11 June 2018. The guidelines included C-reactive protein- and clinical symptoms-guided decision-making and shorter intravenous antibiotic therapy. All term neonates treated for EOS at Ryhov Hospital, Jönköping, Sweden were studied before (period 1: 2016-2017) and after the introduction of the new guidelines (period 2: 11 June 2018 to 30 Sept 2019). Laboratory and clinical data were analysed. RESULTS There were 7618 term neonates in period 1 and 5005 term neonates in period 2. We identified 140 (1.8%) EOS in period 1 and 97 (1.9%) EOS in period 2. During period 1 and 2, there were 61 (61/140, 44%) and 59 (59/97, 61%) EOS neonates, respectively, who met the criteria for shorter antibiotic treatment. The number of positive blood cultures were seven (0.92/1000 live births) and five (1.0/1000 live births) in period 1 and 2. The median C-reactive protein were 52 mg/L (37-62) in period 1 and 42 mg/L (31-56) in period 2 in the group who met the criteria of the guidelines. The duration of antibiotic therapy (Median: seven vs. five days, p < 0.001) and hospital stay (Median: seven vs. five days, p < 0.001) as well as healthcare costs (decreased by €122,000/year) was reduced in the group who met the criteria after the introduction of the guidelines. CONCLUSION C-reactive protein- and clinical symptoms-guided decision-making for EOS significantly decreased the duration of antibiotic therapy and hospital stay, and hence reduced healthcare costs, with no reinfection in a cohort of term infants. TRIAL REGISTRATION Trial registration number: ISRCTN29535824 . Date of registration: 28 May 2020. Retrospectively registered.
Collapse
Affiliation(s)
- Johan Gyllensvärd
- Department of Pediatrics, Region Jönköping County, Jönköping, Sweden S- 553 05, Jönköping, Sweden.
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Fredrik Ingemansson
- Department of Pediatrics, Region Jönköping County, Jönköping, Sweden S- 553 05, Jönköping, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Elisabet Hentz
- Department of Pediatrics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marie Studahl
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Infectious Diseases, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Elfvin
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
99
|
Lichvariková A, Soltys K, Szemes T, Slobodnikova L, Bukovska G, Turna J, Drahovska H. Characterization of Clinical and Carrier Streptococcus agalactiae and Prophage Contribution to the Strain Variability. Viruses 2020; 12:v12111323. [PMID: 33217933 PMCID: PMC7698700 DOI: 10.3390/v12111323] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus, GBS) represents a leading cause of invasive bacterial infections in newborns and is also responsible for diseases in older and immunocompromised adults. Prophages represent an important factor contributing to the genome plasticity and evolution of new strains. In the present study, prophage content was analyzed in human GBS isolates. Thirty-seven prophages were identified in genomes of 20 representative sequenced strains. On the basis of the sequence comparison, we divided the prophages into eight groups named A–H. This division also corresponded to the clustering of phage integrase, even though several different integration sites were observed in some relative prophages. Next, PCR method was used for detection of the prophages in 123 GBS strains from adult hospitalized patients and from pregnancy screening. At least one prophage was present in 105 isolates (85%). The highest prevalence was observed for prophage group A (71%) and satellite prophage group B (62%). Other groups were detected infrequently (1–6%). Prophage distribution did not differ between clinical and screening strains, but it was unevenly distributed in MLST (multi locus sequence typing) sequence types. High content of full-length and satellite prophages detected in present study implies that prophages could be beneficial for the host bacterium and could contribute to evolution of more adapted strains.
Collapse
Affiliation(s)
- Aneta Lichvariková
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 841 15 Bratislava, Slovakia; (A.L.); (K.S.); (T.S.); (J.T.)
- Comenius University Science Park, Ilkovicova 8, 841 04 Bratislava, Slovakia
| | - Katarina Soltys
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 841 15 Bratislava, Slovakia; (A.L.); (K.S.); (T.S.); (J.T.)
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 841 15 Bratislava, Slovakia
| | - Tomas Szemes
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 841 15 Bratislava, Slovakia; (A.L.); (K.S.); (T.S.); (J.T.)
- Comenius University Science Park, Ilkovicova 8, 841 04 Bratislava, Slovakia
| | - Livia Slobodnikova
- Institute of Microbiology, Medical Faculty, Comenius University in Bratislava, 813 72 Bratislava, Slovakia;
| | - Gabriela Bukovska
- Institute of Molecular Biology, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia;
| | - Jan Turna
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 841 15 Bratislava, Slovakia; (A.L.); (K.S.); (T.S.); (J.T.)
| | - Hana Drahovska
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 841 15 Bratislava, Slovakia; (A.L.); (K.S.); (T.S.); (J.T.)
- Correspondence:
| |
Collapse
|
100
|
Progressive Control of Streptococcus agalactiae-Induced Innate Inflammatory Response Is Associated with Time Course Expression of MicroRNA-223 by Neutrophils. Infect Immun 2020; 88:IAI.00563-20. [PMID: 32958526 DOI: 10.1128/iai.00563-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
Group B streptococcus (GBS) is a human-pathogenic bacterium inducing a strong inflammatory response that may be detrimental for host tissues if not finely regulated. The inflammatory response can be modulated by different molecular mechanisms, among which growing evidence points toward the crucial role of microRNAs (miRNAs). Regarding innate inflammatory response, studies have reported that miR-223 is essential for the control of granulocyte proliferation and activation. Moreover, a number of investigations on miRNA expression profiling performed in various inflammatory settings have revealed that miR-223 is among the top differentially expressed miRNAs. Yet the dynamic pattern of expression of miR-223 in vivo with respect to the evolution of the inflammatory process, especially in microbial infection, remains elusive. In this study, we analyzed the kinetic expression of miR-223 in an inflammatory model of GBS-induced murine pneumonia and looked for correlates with inflammatory markers, including innate cell infiltrates. We found that miR-223 expression is rapidly induced at very early time points (3 to 6 h postinfection [p.i.]) mainly by lung-infiltrating neutrophils. Interestingly, the level of miR-223 accumulating in the lungs remains higher at later stages of infection (24 h and 48 h p.i.), and this correlates with reduced expression of primary inflammatory cytokines and chemokines and with a shift in infiltrating monocyte and macrophage subtypes toward a regulatory phenotype. Transient inhibition of miR-223 by an antagomir resulted in significant increase of CXCL2 expression and partial enhancement of infiltrating neutrophils in GBS-infected lung tissues. This suggests the potential contribution of miR-223 to the resolution phase of GBS-induced acute inflammation.
Collapse
|