1
|
Papachatzi E, Gkentzi D, Tzifas S, Dassios T, Dimitriou G. Daptomycin Use for Persistent Coagulase-Negative Staphylococcal Bacteremia in a Neonatal Intensive Care Unit. Antibiotics (Basel) 2024; 13:254. [PMID: 38534689 DOI: 10.3390/antibiotics13030254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
During the last two decades, the incidence of late-onset sepsis (LOS) has increased due to improved survival of premature neonates. Persistent bacteremia (PB) in LOS is defined as more than two positive blood cultures obtained on different calendar days during the same infectious episode. Although rare, PB should be treated aggressively to prevent adverse outcomes. Daptomycin, a lipopeptide antibiotic, has been used in neonates with persistent coagulase-negative staphylococci (CoNS) bacteremia with promising results, but studies reporting on the efficacy and safety of the agent are scarce. The purpose of this study was to evaluate the efficacy and safety of daptomycin use for persistent CoNS bacteremia in a neonatal cohort. This is a retrospective, observational, single-center study of neonates treated with daptomycin during 2011-2022 in the Tertiary Neonatal Intensive Care Unit (NICU) of the University General Hospital of Patras, Greece. For the years 2011-2022, there were 3.413 admissions to the NICU. During the last 3 years (2020-2022)-the active epidemiological surveillance period-123 infants (out of 851 admissions, 14.4%) developed CoNS bacteremia (LOS). During the study period, twelve infants with PB were treated with daptomycin. They had a median gestational age of 32 weeks (IQR 31-34) and mean (SD) birth weight of 1.840 (867) grams. CoNS bacteremia isolates were s. epidermidis (50%), s. haemolyticus (20%), s. hominis (20%) and s. warneri (10%). The decision to start daptomycin (6 mg/kg/dose twice daily) was taken on median day 10 (ΙQR 7-15) of infection. None of the infants had focal complications or meningitis. Daptomycin therapy caused no renal, hepatic, muscular or gastrointestinal adverse events. One neonate developed seizures, and one death occurred due to multiple complications of prematurity. Most infants (11/12) were successfully treated and eventually had negative blood culture. Daptomycin monotherapy showed an adequate cure rate in premature neonates with persistent CoNS bacteremia in a tertiary NICU. In our study, daptomycin was effective and well tolerated; the safety profile, however, needs to be confirmed in larger studies and randomized controlled trials.
Collapse
Affiliation(s)
- Eleni Papachatzi
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Despoina Gkentzi
- Department of Pediatrics, University General Hospital of Patras, 26504 Patras, Greece
| | - Sotiris Tzifas
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Theodore Dassios
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Gabriel Dimitriou
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
- Department of Pediatrics, University General Hospital of Patras, 26504 Patras, Greece
| |
Collapse
|
2
|
van Donge T, Fuchs A, Leroux S, Pfister M, Rodieux F, Atkinson A, Giannoni E, van den Anker J, Bielicki J. Amoxicillin Dosing Regimens for the Treatment of Neonatal Sepsis: Balancing Efficacy and Neurotoxicity. Neonatology 2020; 117:619-627. [PMID: 32841941 DOI: 10.1159/000509751] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/21/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Large variability in neonatal amoxicillin dosing recommendations may reflect uncertainty about appropriate efficacy and toxicity targets. OBJECTIVE The aim of this study was to model efficacious and safe exposure for current neonatal amoxicillin dosing regimens, given a range of assumptions for minimal inhibitory concentration (MIC), targeted %fT > MIC, and potential for aminopenicillin-related neurotoxicity. METHODS Individual intravenous amoxicillin exposures based on 6 international and 9 Swiss neonatal dosing recommendations, reflecting the range of current dosing approaches, were assessed by a previously developed population pharmacokinetic model informed by neonatal data from an international cohort. Exposure was simulated by attributing each dosing regimen to each patient cohort. End points of interest were %fT > MIC and potential neurotoxicity using Cmax > 140 mg/L as threshold. RESULTS None of the dosing regimens achieved targets of ≥100%fT > MIC at any of the relevant MICs for a desired probability of target attainment (PTA) of ≥90%. All regimens achieved a PTA ≥90% for Streptococcus agalactiae (MIC 0.25 mg/L) and Listeria monocytogenes (MIC 1 mg/L) when targeting ≤70%fT > MIC. In contrast, none of the regimens resulted in a PTA ≥90% targeting ≥70%fT > MIC for enterococci (MIC 4 mg/L). The maximum amoxicillin concentration associated with potential neurotoxicity was exceeded using 4 dosing regimens (100 mg/kg q12, 60/30 mg/kg q12/8, 50 mg/kg q12/8/6, and 50 mg/kg q12/8/4) for ≥10% of neonates. CONCLUSIONS The acceptability of regimens is highly influenced by efficacy and toxicity targets, the selection of which is challenging. Novel randomized trial designs combined with pharmacometric modeling and simulation could assist in selecting optimal dosing regimens in this understudied population.
Collapse
Affiliation(s)
- Tamara van Donge
- Paediatric Pharmacology and Pharmacometrics Research, University Children's Hospital Basel, Basel, Switzerland,
| | - Aline Fuchs
- Paediatric Pharmacology and Pharmacometrics Research, University Children's Hospital Basel, Basel, Switzerland.,Medicines for Malaria Venture, Geneva, Switzerland
| | - Stéphanie Leroux
- Paediatric Pharmacology and Pharmacometrics Research, University Children's Hospital Basel, Basel, Switzerland
| | - Marc Pfister
- Paediatric Pharmacology and Pharmacometrics Research, University Children's Hospital Basel, Basel, Switzerland
| | - Frédérique Rodieux
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive care, and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Andrew Atkinson
- Paediatric Pharmacology and Pharmacometrics Research, University Children's Hospital Basel, Basel, Switzerland
| | - Eric Giannoni
- Clinic of Neonatology, Department Mother-Woman-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - John van den Anker
- Paediatric Pharmacology and Pharmacometrics Research, University Children's Hospital Basel, Basel, Switzerland.,Intensive Care and Department of Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Division of Clinical Pharmacology, Children's National Hospital, Washington, District of Columbia, USA
| | - Julia Bielicki
- Medicines for Malaria Venture, Geneva, Switzerland.,Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| |
Collapse
|
3
|
Suffolk R, Agertoft L, Johansen M, Zachariassen G. Late-onset group B streptococcus infections and severe bronchopulmonary dysplasia in an extremely preterm born infant. BMJ Case Rep 2019; 12:12/7/e229255. [DOI: 10.1136/bcr-2019-229255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
This case report is about a boy born extremely preterm at gestational age of 24 weeks, with extremely low birth weight, developing severe bronchopulmonary dysplasia and in need of mechanical ventilation for 155 days. He also had five recurrent infections with group B streptococcus (GBS) within 4 months from birth, and his respiratory condition clearly deteriorated with every GBS infection. It was difficult to wean him from mechanical ventilation. Finally he was extubated when he was 7 months old and kept out of mechanical ventilation after receiving high-dose methylprednisolone, given according to international recommendations. After GBS was cultured for the fifth time, he received oral rifampicin along with intravenous penicillin and after this treatment, GBS did not occur again. At the age of 22 months, the boy no longer needed any respiratory support and he was about 6 months late in his neurological development.
Collapse
|
4
|
van Donge T, Pfister M, Bielicki J, Csajka C, Rodieux F, van den Anker J, Fuchs A. Quantitative Analysis of Gentamicin Exposure in Neonates and Infants Calls into Question Its Current Dosing Recommendations. Antimicrob Agents Chemother 2018; 62:e02004-17. [PMID: 29358294 PMCID: PMC5913996 DOI: 10.1128/aac.02004-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/09/2018] [Indexed: 12/20/2022] Open
Abstract
Optimal dosing of gentamicin in neonates is still a matter of debate despite its common use. We identified gentamicin dosing regimens from eight international guidelines and seven Swiss neonatal intensive care units. The dose per administration, the dosing interval, the total daily dose, and the demographic characteristics between guidelines were compared. There was considerable variability with respect to dose (4 to 6 mg/kg), dosing interval (24 h to 48 h), total daily dose (2.5 to 6 mg/kg/day), and patient demographic characteristics that were used to calculate individualized dosing regimens. A model-based simulation study in 1071 neonates was performed to determine the achievement of efficacious peak gentamicin concentrations according to predefined MICs (Cmax/MIC ≥ 10) and safe trough concentrations (Cmin ≤ 2 mg/liter) with recommended dosing regimens. MIC targets of 0.5 and 1 mg/liter were used. Dosing optimization was performed giving priority to the first day of treatment and with the goal of simplifying dosing. Current gentamicin neonatal guidelines allow to achieve effective peak concentrations for MICs ≤ 0.5 mg/liter but not higher. Model-based simulations indicate that to attain peak gentamicin concentrations of ≥10 mg/liter, a dose of 7.5 mg/kg should be administered using an extended dosing interval regimen. Trough concentrations of ≤2 mg/liter can be maintained with a dosing interval of 36 to 48 h in neonates according to gestational and postnatal age. For treatment beyond 3 days, therapeutic drug monitoring is advised to maintain adequate serum concentrations.
Collapse
Affiliation(s)
- Tamara van Donge
- Paediatric Pharmacology and Pharmacometrics Research, University of Basel Children's Hospital, Basel, Switzerland
| | - Marc Pfister
- Paediatric Pharmacology and Pharmacometrics Research, University of Basel Children's Hospital, Basel, Switzerland
- Quantitative Solutions, a Certara Company, London, United Kingdom
| | - Julia Bielicki
- Paediatric Pharmacology and Pharmacometrics Research, University of Basel Children's Hospital, Basel, Switzerland
- Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Chantal Csajka
- Service of Clinical Pharmacology, Department of Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Frederique Rodieux
- Service of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Geneva, Switzerland
| | - John van den Anker
- Paediatric Pharmacology and Pharmacometrics Research, University of Basel Children's Hospital, Basel, Switzerland
- Intensive Care and Department of Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
- Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA
| | - Aline Fuchs
- Paediatric Pharmacology and Pharmacometrics Research, University of Basel Children's Hospital, Basel, Switzerland
| |
Collapse
|
5
|
Scheid A, Borriello F, Pietrasanta C, Christou H, Diray-Arce J, Pettengill MA, Joshi S, Li N, Bergelson I, Kollmann T, Dowling DJ, Levy O. Adjuvant Effect of Bacille Calmette-Guérin on Hepatitis B Vaccine Immunogenicity in the Preterm and Term Newborn. Front Immunol 2018; 9:29. [PMID: 29416539 PMCID: PMC5787546 DOI: 10.3389/fimmu.2018.00029] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 01/04/2018] [Indexed: 01/21/2023] Open
Abstract
Immunization is key to protecting term and preterm infants from a heightened risk of infection. However, preterm immunity is distinct from that of the term, limiting its ability to effectively respond to vaccines routinely given at birth, such as hepatitis B vaccine (HBV). As part of the Expanded Program on Immunization, HBV is often given together with the live-attenuated vaccine Bacille Calmette-Guérin (BCG), known to activate multiple pattern-recognition receptors. Of note, some clinical studies suggest BCG can enhance efficacy of other vaccines in term newborns. However, little is known about whether BCG can shape Th-polarizing cytokine responses to HBV nor the age-dependency of such effects, including whether they may extend to the preterm. To characterize the effects of BCG on HBV immunogenicity, we studied individual and combined administration of these vaccines to cord newborn and adult human whole blood and mononuclear cells in vitro and to neonatal and adult mice in vivo. Compared to either BCG or HBV alone, (BCG + HBV) synergistically enhanced in vitro whole blood production of IL-1β, while (BCG + HBV) also promoted production of several cytokines/chemokines in all age groups, age-specific enhancement included IL-12p70 in the preterm and GM-CSF in the preterm and term. In human mononuclear cells, (BCG + HBV) enhanced mRNA expression of several genes including CSF2, which contributed to clustering of genes by vaccine treatment via principle component analysis. To assess the impact of BCG on HBV immunization, mice of three different age groups were immunized subcutaneously with, BCG, HBV, (BCG + HBV) into the same site; or BCG and HBV injected into separate sites. Whether injected into a separate site or at the same site, co-administration of BCG with HBV significantly enhanced anti-HBV IgG titers in mice immunized on day of life-0 or -7, respectively, but not in adult mice. In summary, our data demonstrate that innate and adaptive vaccine responses of preterm and term newborns are immunologically distinct. Furthermore, BCG or "BCG-like" adjuvants should be further studied as a promising adjuvantation approach to enhance immunogenicity of vaccines to protect these vulnerable populations.
Collapse
Affiliation(s)
- Annette Scheid
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Francesco Borriello
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- WAO Center of Excellence, Naples, Italy
| | - Carlo Pietrasanta
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Joann Diray-Arce
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Matthew A. Pettengill
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Sweta Joshi
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Ning Li
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Medical Eli Lilly, Shanghai, China
| | - Ilana Bergelson
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Tobias Kollmann
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, British Columbia Children’s Hospital, Vancouver, BC, Canada
| | - David J. Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| |
Collapse
|
6
|
Pettengill M, Matute JD, Tresenriter M, Hibbert J, Burgner D, Richmond P, Luis Millán J, Ozonoff A, Strunk T, Currie A, Levy O. Human alkaline phosphatase dephosphorylates microbial products and is elevated in preterm neonates with a history of late-onset sepsis. PLoS One 2017; 12:e0175936. [PMID: 28448526 PMCID: PMC5407836 DOI: 10.1371/journal.pone.0175936] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 04/03/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A host defense function for Alkaline phosphatases (ALPs) is suggested by the contribution of intestinal ALP to detoxifying bacterial lipopolysaccharide (endotoxin) in animal models in vivo and the elevation of ALP activity following treatment of human cells with inflammatory stimuli in vitro. However the activity of ALP in human plasma (primarily tissue-nonspecific ALP; TNAP) on lipopolysaccharide and other microbial products has not been assessed, nor has its expression been studied in preterm newborns, a vulnerable population at high risk of sepsis. In this context, the aim of our study was to characterize the activity of TNAP on Toll-like receptor (TLR) agonists and assess the concentrations of plasma ALP during late-onset sepsis in preterm newborns. METHODS Recombinant human TNAP was incubated with microbial products and phosphate release was measured by malachite green assay. Plasma ALP activity was measured serially in a cohort of preterm (N = 129) infants at high risk of late-onset sepsis (LOS). RESULTS TNAP dephosphorylates poly-inosine:cytosine (Toll-like receptor (TLR) 3 agonist) and LPS from Klebsiella pneumoniae and Salmonella minnesota (TLR4 agonists). Plasma ALP significantly increased postnatally over the first 4 weeks of life in preterm and term newborns. Bacteremic LOS in preterm infants (gestational age ≤ 30 weeks) was associated with significantly elevated plasma ALP at 4 weeks postnatal age. CONCLUSIONS TNAP, the main circulating isozyme of ALP, de-phosphorylates TLR agonists, demonstrates a post-natal age dependent increase in preterm and term plasma across the first 4 weeks of life, and is elevated in association with preterm LOS.
Collapse
Affiliation(s)
- Matthew Pettengill
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Juan D. Matute
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Megan Tresenriter
- University of California Davis School of Medicine, Davis, California, United States of America
| | - Julie Hibbert
- The University of Western Australia, Crawley, Western Australia, Australia
| | - David Burgner
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Peter Richmond
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, LaJolla, California, United States of America
| | - Al Ozonoff
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tobias Strunk
- The University of Western Australia, Crawley, Western Australia, Australia
| | - Andrew Currie
- The University of Western Australia, Crawley, Western Australia, Australia
- School of Veterinary & Life Sciences, Murdoch University, Murdoch, Western Australia, Australia
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
7
|
Zambruni M, Villalobos A, Somasunderam A, Westergaard S, Nigalye M, Turin CG, Zegarra J, Bellomo S, Mercado E, Ochoa TJ, Utay NS. Maternal and pregnancy-related factors affecting human milk cytokines among Peruvian mothers bearing low-birth-weight neonates. J Reprod Immunol 2017; 120:20-26. [PMID: 28399439 PMCID: PMC5494983 DOI: 10.1016/j.jri.2017.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/17/2017] [Accepted: 04/03/2017] [Indexed: 12/13/2022]
Abstract
Several cytokines have been detected in human milk but their relative concentrations differ among women and vary over time in the same person. The drivers of such differences have been only partially identified, while the effect of luminal cytokines in the fine-regulation of the intestinal immune system is increasingly appreciated. The aim of this study was to investigate the associations between obstetrical complications and human milk cytokine profiles in a cohort of Peruvian women giving birth to Low Birth Weight (LBW) infants. Colostrum and mature human milk samples were collected from 301 Peruvian women bearing LBW infants. The concentration of twenty-three cytokines was measured using the Luminex platform. Ninety-nine percent of women had at least one identified obstetrical complication leading to intra-uterine growth restriction and/or preterm birth. Median weight at birth was 1,420g; median gestational age 31 weeks. A core of 12 cytokines, mainly involved in innate immunity and epithelial cell integrity, was detectable in most samples. Maternal age, maternal infection, hypertensive disorders, preterm labor, and premature rupture of membranes were associated with specific cytokine profiles both in colostrum and mature human milk. Mothers of Very LBW (VLBW) neonates had significantly higher concentrations of chemokines and growth factor cytokines both in their colostrum and mature milk compared with mothers of larger neonates. Thus, maternal conditions affecting pregnancy duration and in utero growth are also associated with specific human milk cytokine signatures.
Collapse
Affiliation(s)
- Mara Zambruni
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA
| | - Alex Villalobos
- University of Texas Medical Branch (UTMB), School of Medicine, Galveston, TX, USA
| | - Anoma Somasunderam
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA
| | - Sarah Westergaard
- University of Texas Medical Branch (UTMB), School of Medicine, Galveston, TX, USA
| | - Maitreyee Nigalye
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA
| | - Christie G Turin
- Universidad Peruana Cayetano Heredia, Laboratorio de Infectologia Pediatrica, Lima, Peru
| | - Jaime Zegarra
- Universidad Peruana Cayetano Heredia, Department of Pediatrics, Lima, Peru
| | - Sicilia Bellomo
- Universidad Peruana Cayetano Heredia, Department of Pediatrics, Lima, Peru
| | - Erik Mercado
- Universidad Peruana Cayetano Heredia, Laboratorio de Infectologia Pediatrica, Lima, Peru
| | - Theresa J Ochoa
- Universidad Peruana Cayetano Heredia, Department of Pediatrics, Lima, Peru; University of Texas Health Science Center at Houston (UTHSCH), Houston, TX, USA
| | - Netanya S Utay
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA.
| |
Collapse
|
8
|
Nguyen TH, Park MD, Otto M. Host Response to Staphylococcus epidermidis Colonization and Infections. Front Cell Infect Microbiol 2017; 7:90. [PMID: 28377905 PMCID: PMC5359315 DOI: 10.3389/fcimb.2017.00090] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 03/07/2017] [Indexed: 01/11/2023] Open
Abstract
The majority of research in the Staphylococcus field has been dedicated to the understanding of Staphylococcus aureus infections. In contrast, there is limited information on infections by coagulase-negative Staphylococci (CoNS) and how the host responds to them. S. epidermidis, a member of the coagulase-negative Staphylococci, is an important commensal organism of the human skin and mucous membranes; and there is emerging evidence of its benefit for human health in fighting off harmful microorganisms. However, S. epidermidis can cause opportunistic infections, which include particularly biofilm-associated infections on indwelling medical devices. These often can disseminate into the bloodstream; and in fact, S. epidermidis is the most frequent cause of nosocomial sepsis. The increasing use of medical implants and the dramatic shift in the patient demographic population in recent years have contributed significantly to the rise of S. epidermidis infections. Furthermore, treatment has been complicated by the emergence of antibiotic-resistant strains. Today, S. epidermidis is a major nosocomial pathogen posing significant medical and economic burdens. In this review, we present the current understanding of mechanisms of host defense against the prototypical CoNS species S. epidermidis as a commensal of the skin and mucous membranes, and during biofilm-associated infection and sepsis.
Collapse
Affiliation(s)
- Thuan H Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health Bethesda, MD, USA
| | - Matthew D Park
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health Bethesda, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
9
|
Ohlin A, Björkman L, Serenius F, Schollin J, Källén K. Sepsis as a risk factor for neonatal morbidity in extremely preterm infants. Acta Paediatr 2015; 104:1070-6. [PMID: 26118325 DOI: 10.1111/apa.13104] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/28/2015] [Accepted: 06/24/2015] [Indexed: 12/31/2022]
Abstract
AIM This study evaluated sepsis as a risk factor for neonatal morbidities and investigated the association between specific pathogens and neonatal morbidities. METHODS This was a nationwide Swedish prospective cohort study, consisting of the 497 extremely premature children, who were born before 27 weeks of gestation between 2004 and 2007 and survived their first year of life. Neonatal sepsis was evaluated as a risk factor for neonatal morbidity using multiple logistic linear regression analyses. RESULTS We found that 326 (66%) of the infants had at least one sepsis episode and coagulase-negative staphylococci was the most common pathogen. Definite sepsis, with an odds ratio (OR) of 1.6, was associated with severe bronchopulmonary dysplasia, but not clinical sepsis (OR 1.1). Definite sepsis was also associated with a prolonged hospital stay (OR 1.6). Sepsis was not significantly associated with a higher risk of retinopathy of prematurity or intraventricular haemorrhage. CONCLUSION Extremely preterm infants face a great risk of acquiring neonatal sepsis, with coagulase-negative staphylococci being the most common pathogen in this population. Definite sepsis seemed to be a risk factor for severe bronchopulmonary dysplasia and prolonged hospital stay, but the associations were weaker than in previous studies.
Collapse
Affiliation(s)
- Andreas Ohlin
- Department of Pediatrics; Faculty of Medicine and Health; Örebro University; Örebro Sweden
| | - Louise Björkman
- Department of Pediatrics; Faculty of Medicine and Health; Örebro University; Örebro Sweden
| | - Fredrik Serenius
- Department of Women's and Children's Health; Section for Pediatrics; Uppsala University; Uppsala Sweden
- Department of Pediatrics; Institute of Clinical Sciences; Umeå University; Umeå Sweden
| | - Jens Schollin
- Department of Pediatrics; Faculty of Medicine and Health; Örebro University; Örebro Sweden
| | - Karin Källén
- Department of Obstetrics and Gynecology, Clinical Sciences; Centre of Reproduction Epidemiology; University of Lund; Lund Sweden
| |
Collapse
|
10
|
Dong Y, Speer CP. The role of Staphylococcus epidermidis in neonatal sepsis: Guarding angel or pathogenic devil? Int J Med Microbiol 2014; 304:513-20. [DOI: 10.1016/j.ijmm.2014.04.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/23/2014] [Accepted: 04/27/2014] [Indexed: 11/24/2022] Open
|
11
|
Granland C, Strunk T, Hibbert J, Prosser A, Simmer K, Burgner D, Richmond P, Currie AJ. NOD1 and NOD2 expression and function in very preterm infant mononuclear cells. Acta Paediatr 2014; 103:e212-8. [PMID: 24444388 DOI: 10.1111/apa.12559] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/16/2013] [Accepted: 01/14/2014] [Indexed: 12/29/2022]
Abstract
AIM To evaluate mononuclear cell expression and function of the cytosolic nucleotide-binding oligomerization domain-containing receptors, NOD1 and NOD2, in very preterm and full-term infants. METHODS NOD1 and NOD2 gene and protein expression in very preterm infants, term infants and healthy adult, cord and peripheral blood mononuclear cells (C/PBMC) were quantified using qPCR and flow cytometry. Cytokine responses of purified infant and adult monocytes to NOD1- and NOD2-specific agonists were assessed using a multiplex immunoassay (Bioplex). RESULTS NOD1 and NOD2 were expressed by a range of infant and adult mononuclear cell types, including T- and B cells, with highest expression in classical (CD14(++) CD16(-) ) and intermediate (CD14(++) CD16(+) ) monocytes. NOD1 and NOD2 expression levels by monocytes from very preterm infant were similar to those in term infants or adults. Monocyte production of TNFα, IL-6 and IL-1β induced by activation of NOD1 and NOD2 was similar between very preterm infants, term infants and adults. CONCLUSION Monocyte expression and function of NOD1 and NOD2 in very preterm infants are intact and comparable/equivalent to term infants and adults. Functional deficiencies in monocyte NOD signalling pathways are unlikely to contribute to the increased susceptibility to bacterial sepsis in preterm infants.
Collapse
Affiliation(s)
- Caitlyn Granland
- School of Veterinary and Life Sciences; Murdoch University; Perth WA Australia
| | - Tobias Strunk
- Centre for Neonatal Research and Education; University of Western Australia; Perth WA Australia
- School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Julie Hibbert
- School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Amy Prosser
- School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Karen Simmer
- Centre for Neonatal Research and Education; University of Western Australia; Perth WA Australia
- School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - David Burgner
- School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- Murdoch Childrens Research Institute; Royal Children's Hospital; Parkville Vic. Australia
| | - Peter Richmond
- School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Andrew J. Currie
- School of Veterinary and Life Sciences; Murdoch University; Perth WA Australia
- Centre for Neonatal Research and Education; University of Western Australia; Perth WA Australia
| |
Collapse
|
12
|
Laursen IA, Blou L, Sullivan JS, Bang P, Balstrup F, Houen G. Development, manufacturing and characterization of a highly purified, liquid immunoglobulin g preparation from human plasma. Transfus Med Hemother 2014; 41:205-12. [PMID: 25053934 DOI: 10.1159/000357982] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 05/14/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The use of plasma-derived immunoglobulin G (IgG) is increasing, and the number of diseases, including immunodeficiencies, neurological diseases and autoimmune conditions, treated with intravenous IgG (IVIG) is expanding. Consequently, there is a great need for high-yield production processes for plasma-derived IgG. The aim of this work was to develop a high-yield process leading to a highly purified, liquid, ready-to-use IgG for intravenous use. METHODS Plasma from healthy, voluntary, non-remunerated donors was fractionated by ethanol precipitation. IgG was extracted from fraction II + III using a phosphate/acetate buffer, pH 4, and purified by chromatography. RESULTS Precipitation with 6% polyethylene glycol at pH 7 removed high molecular-weight contaminating proteins, aggregates and contaminating viruses. Ion exchange chromatography at pH 5.7 on serially connected anion and cation exchange columns allowed for elution of IgG from the cation exchange column in good yield and high purity. Further safety was achieved by solvent/detergent treatment and repeated ion exchange chromatography. The product consisted of essentially only IgG monomers and dimers, and had a high purity with very low levels of IgM and IgA. CONCLUSION A process providing highly purified IVIG in good yield was developed.
Collapse
Affiliation(s)
- Inga A Laursen
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Lene Blou
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | - Peter Bang
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Flemming Balstrup
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Gunnar Houen
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
13
|
Cruickshank MN, Oshlack A, Theda C, Davis PG, Martino D, Sheehan P, Dai Y, Saffery R, Doyle LW, Craig JM. Analysis of epigenetic changes in survivors of preterm birth reveals the effect of gestational age and evidence for a long term legacy. Genome Med 2013; 5:96. [PMID: 24134860 PMCID: PMC3978871 DOI: 10.1186/gm500] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 09/26/2013] [Indexed: 02/08/2023] Open
Abstract
Background Preterm birth confers a high risk of adverse long term health outcomes for survivors, yet the underlying molecular mechanisms are unclear. We hypothesized that effects of preterm birth can be mediated through measurable epigenomic changes throughout development. We therefore used a longitudinal birth cohort to measure the epigenetic mark of DNA methylation at birth and 18 years comparing survivors of extremely preterm birth with infants born at term. Methods Using 12 extreme preterm birth cases and 12 matched, term controls, we extracted DNA from archived neonatal blood spots and blood collected in a similar way at 18 years of age. DNA methylation was measured at 347,789 autosomal locations throughout the genome using Infinium HM450 arrays. Representative methylation differences were confirmed by Sequenom MassArray EpiTYPER. Results At birth we found 1,555 sites with significant differences in methylation between term and preterm babies. At 18 years of age, these differences had largely resolved, suggesting that DNA methylation differences at birth are mainly driven by factors relating to gestational age, such as cell composition and/or maturity. Using matched longitudinal samples, we found evidence for an epigenetic legacy associated with preterm birth, identifying persistent methylation differences at ten genomic loci. Longitudinal comparisons of DNA methylation at birth and 18 years uncovered a significant overlap between sites that were differentially-methylated at birth and those that changed with age. However, we note that overlapping sites may either differ in the same (300/1,555) or opposite (431/1,555) direction during gestation and aging respectively. Conclusions We present evidence for widespread methylation differences between extreme preterm and term infants at birth that are largely resolved by 18 years of age. These results are consistent with methylation changes associated with blood cell development, cellular composition, immune induction and age at these time points. Finally, we identified ten probes significantly associated with preterm individuals and with greater than 5% methylation discordance at birth and 18 years that may reflect a long term epigenetic legacy of preterm birth.
Collapse
Affiliation(s)
- Mark N Cruickshank
- Early Life Epigenetics Group, Murdoch Childrens Research Institute (MCRI), Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Present address: Telethon Institute for Child Health Research, University of Western Australia, 100 Roberts Road, Subiaco, WA 6008, Australia
| | - Alicia Oshlack
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Bioinformatics Group, MCRI, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Christiane Theda
- Early Life Epigenetics Group, Murdoch Childrens Research Institute (MCRI), Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Neonatal Services, Royal Women's Hospital, Parkville, Victoria 3052, Australia ; Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Peter G Davis
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Neonatal Services, Royal Women's Hospital, Parkville, Victoria 3052, Australia ; Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - David Martino
- Cancer and Developmental Epigenetics Group, MCRI, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Penelope Sheehan
- Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Yun Dai
- Early Life Epigenetics Group, Murdoch Childrens Research Institute (MCRI), Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Richard Saffery
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Cancer and Developmental Epigenetics Group, MCRI, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Lex W Doyle
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Neonatal Services, Royal Women's Hospital, Parkville, Victoria 3052, Australia ; Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Jeffrey M Craig
- Early Life Epigenetics Group, Murdoch Childrens Research Institute (MCRI), Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia ; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| |
Collapse
|
14
|
Yumani DFJ, van den Dungen FAM, van Weissenbruch MM. Incidence and risk factors for catheter-associated bloodstream infections in neonatal intensive care. Acta Paediatr 2013; 102:e293-8. [PMID: 23627968 DOI: 10.1111/apa.12256] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/06/2013] [Accepted: 04/02/2013] [Indexed: 11/29/2022]
Abstract
AIM To determine the incidence and potential novel risk factors for catheter-associated bloodstream infections (CABSI) in neonates. METHODS A retrospective study was conducted for infants admitted to the VU University Medical Center neonatal intensive care unit in 2007. RESULTS One hundred and ninety six infants with a total of 369 central catheters were included. The CABSI rate was 18.1 infections/1000 catheter-days (95% CI 13.7-23.8) according to adjusted criteria used by the Centers for Disease Control and Prevention prior to 2008. Umbilical catheters had a higher infection rate than nonumbilical central catheters: rate ratio (rate ratio 2.4, 95% CI 1.2-4.9). Longer umbilical catheter dwell-time also increased infection rate (p < 0.05). Gestational age, birth weight, duration of parenteral nutrition and the administration of all-in-one feeding mixture versus parenteral nutrition administered in separate components were not related to infection rate in multivariate analysis. CONCLUSION Of all catheter types, umbilical catheters carried the highest infection rate. Longer umbilical catheter dwell-time also increased infection rate. The present data suggest that the impact of gestational age and birth weight on infection rate is mainly due to a prolonged hospital stay. The composition, way of preparation and duration of parenteral nutrition did not seem to influence infection rate.
Collapse
Affiliation(s)
- Dana FJ Yumani
- Division of Neonatology; Department of Paediatrics; VU University Medical Center; Amsterdam; The Netherlands
| | - Frank AM van den Dungen
- Division of Neonatology; Department of Paediatrics; VU University Medical Center; Amsterdam; The Netherlands
| | - Mirjam M van Weissenbruch
- Division of Neonatology; Department of Paediatrics; VU University Medical Center; Amsterdam; The Netherlands
| |
Collapse
|
15
|
Neonatal sepsis due to coagulase-negative staphylococci. Clin Dev Immunol 2013; 2013:586076. [PMID: 23762094 PMCID: PMC3674645 DOI: 10.1155/2013/586076] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/27/2013] [Accepted: 04/27/2013] [Indexed: 02/06/2023]
Abstract
Neonates, especially those born prematurely, are at high risk of morbidity and mortality from sepsis. Multiple factors, including prematurity, invasive life-saving medical interventions, and immaturity of the innate immune system, put these infants at greater risk of developing infection. Although advanced neonatal care enables us to save even the most preterm neonates, the very interventions sustaining those who are hospitalized concurrently expose them to serious infections due to common nosocomial pathogens, particularly coagulase-negative staphylococci bacteria (CoNS). Moreover, the health burden from infection in these infants remains unacceptably high despite continuing efforts. In this paper, we review the epidemiology, immunological risk factors, diagnosis, prevention, treatment, and outcomes of neonatal infection due to the predominant neonatal pathogen CoNS.
Collapse
|
16
|
Chavez-Valdez R, Kovell L, Ahlawat R, McLemore GL, Wills-Karp M, Gauda EB. Opioids and clonidine modulate cytokine production and opioid receptor expression in neonatal immune cells. J Perinatol 2013; 33:374-82. [PMID: 23047422 PMCID: PMC3640758 DOI: 10.1038/jp.2012.124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Opioids and clonidine, used in for sedation, analgesia and control of opioid withdrawal in neonates, directly or indirectly activate opioid receptors (OPRs) expressed in immune cells. Therefore, our objective is to study how clinically relevant concentrations of different opioids and clonidine change cytokine levels in cultured whole blood from preterm and full-term infants. STUDY DESIGN Using blood from preterm (≤ 30 weeks gestational age (GA), n=7) and full-term ( ≥ 37 weeks GA, n=19) infants, we investigated the changes in cytokine profile (IL-1β, IL-6, IL-8, IL-10, IL-12p70 and TNF-α), cyclic adenosine monophosphate (cAMP) levels and μ-, δ- and κ- opioid receptor (OPR) gene and protein expression, following in-vitro exposure to morphine, methadone, fentanyl or clonidine at increasing concentrations ranging from 0 to 1 mM. RESULT Following lipopolysaccharide activation, IL-10 levels were 146-fold greater in cultured blood from full-term than from preterm infants. Morphine and methadone, but not fentanyl, at >10(-5) M decreased all tested cytokines except IL-8. In contrast, clonidine at <10(-9) M increased IL-6, while at >10(-5) M increased IL-1β and decreased TNF-α levels. All cytokine changes followed the same patterns in preterm and full-term infant cultured blood and matched increases in cAMP levels. All three μ-, δ- and κ-OPR genes were expressed in mononuclear cells (MNC) from preterm and full-term infants. Morphine, methadone and clonidine, but not fentanyl, at >10(-5)M decreased the expression of μ-OPR, but not δ- or κ-OPRs. CONCLUSION Generalized cytokine suppression along with downregulation of μ-OPR expression observed in neonatal MNC exposed to morphine and methadone at clinically relevant concentrations contrast with the modest effects observed with fentanyl and clonidine. Therefore, we speculate that fentanyl and clonidine may be safer therapeutic choices for sedation and control of opioid withdrawal and pain in neonates.
Collapse
Affiliation(s)
- Raul Chavez-Valdez
- Department of Pediatrics, Division of Neonatology, Johns Hopkins University - School of Medicine. Baltimore, Maryland 21287, United States, Department of Pediatrics, Division of Neonatology. Texas Tech University – Health Sciences Center. Odessa, Texas 79763, United States
| | - Lara Kovell
- Department of Pediatrics, Division of Neonatology, Johns Hopkins University - School of Medicine. Baltimore, Maryland 21287, United States
| | - Rajni Ahlawat
- Department of Pediatrics, Division of Neonatology, Johns Hopkins University - School of Medicine. Baltimore, Maryland 21287, United States
| | - Gabrielle L. McLemore
- Department of Pediatrics, Division of Neonatology, Johns Hopkins University - School of Medicine. Baltimore, Maryland 21287, United States, Department of Biology, Morgan State University, Baltimore, Maryland 21251Biology, , United States
| | - Marsha Wills-Karp
- Division of Immunobiology. Children's Hospital of Cincinnati. University of Cincinnati College of Medicine. Cincinnati, OH 45229, United States
| | - Estelle B. Gauda
- Department of Pediatrics, Division of Neonatology, Johns Hopkins University - School of Medicine. Baltimore, Maryland 21287, United States
| |
Collapse
|
17
|
Rodriguez-Guerineau L, Salvia-Roigés MD, León-Lozano M, Rodríguez-Miguélez JM, Figueras-Aloy J. Combination of vancomycin and rifampicin for the treatment of persistent coagulase-negative staphylococcal bacteremia in preterm neonates. Eur J Pediatr 2013; 172:693-7. [PMID: 23328960 DOI: 10.1007/s00431-012-1927-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 12/16/2012] [Accepted: 12/24/2012] [Indexed: 11/28/2022]
Abstract
UNLABELLED Coagulase-negative staphylococci are the most common cause of late-onset sepsis in premature neonates. The optimal approach in persistent coagulase-negative staphylococcal bacteremia, despite adequate treatment with glycopeptides, is not well established. A retrospective study was conducted on preterm neonates with persistent coagulase-negative staphylococcal bacteremia treated with the combination of vancomycin-rifampicin. Ten cases were included, with a median gestational age of 26 weeks (range 24 weeks + 3 days-31 weeks + 4 days, interquartile range 25 weeks + 3 days-29 weeks + 3 days) and a median birth weight of 715 g (range 555-2,030). The median age at the onset of infection was 9 days (range 5-37). The most frequent clinical presentation was apnea or increased ventilatory support. Bacteremia persisted for a median of 9 (range 6-19) days until rifampicin initiation. Bacteremia was resolved in all cases on vancomycin-rifampicin with no serious side effects. CONCLUSION Our study provides data supporting the safety and efficacy of vancomycin-rifampicin combination for the treatment of persistent coagulase-negative staphylococcal bacteremia in preterm neonates.
Collapse
Affiliation(s)
- Luciana Rodriguez-Guerineau
- Department of Neonatology, Institut Clínic de Ginecologia, Obstetrícia i Neonatologia, Hospital Clínic- Hospital Sant Joan de Déu, University of Barcelona, Villarroel No. 170, 08036 Barcelona, Spain.
| | | | | | | | | |
Collapse
|
18
|
Abstract
Given the "inborn" nature of the innate immune system, it is surprising to find that innate immune function does in fact change with age. Similar patterns of distinct Toll-like-receptor-mediated immune responses come to light when one contrasts innate immune development at the beginning of life with that toward the end of life. Importantly, these developmental patterns of innate cytokine responses correlate with clinical patterns of susceptibility to disease: A heightened risk of suffering from excessive inflammation is often detected in prematurely born infants, disappears over the first few months of life, and reappears toward the end of life. In addition, risk periods for particular infections in early life reemerge in older adults. The near-mirror-image patterns that emerge in contrasts of early versus late innate immune ontogeny emphasize changes in host-environment interactions as the underlying molecular and teleologic drivers.
Collapse
|
19
|
Sharma AA, Jen R, Butler A, Lavoie PM. The developing human preterm neonatal immune system: a case for more research in this area. Clin Immunol 2012; 145:61-8. [PMID: 22926079 DOI: 10.1016/j.clim.2012.08.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 08/07/2012] [Accepted: 08/08/2012] [Indexed: 12/23/2022]
Abstract
Neonates, particularly those born prematurely, are among the most vulnerable age group for morbidity and mortality due to infections. Immaturity of the innate immune system and a high need for invasive medical procedures in the context of a preterm birth make these infants highly susceptible to common neonatal pathogens. Preterm infants who survive may also suffer permanent disabilities due to organ damage resulting from either the infection itself or from the inflammatory response generated under an oxidative stress. Infections in preterm infants continue to pose important healthcare challenges. Yet, developmental maturation events in the innate immune system that underlie their excessively high vulnerability to infection remain largely understudied. In this review article, we identify pertinent knowledge gaps that must be filled in order to orient future translational research.
Collapse
|
20
|
Strunk T, Prosser A, Levy O, Philbin V, Simmer K, Doherty D, Charles A, Richmond P, Burgner D, Currie A. Responsiveness of human monocytes to the commensal bacterium Staphylococcus epidermidis develops late in gestation. Pediatr Res 2012; 72:10-8. [PMID: 22460219 DOI: 10.1038/pr.2012.48] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Staphylococcus epidermidis (SE) rarely causes infection in term infants but is a leading cause of late-onset sepsis in preterm infants. We hypothesized that the innate immune responses to SE in preterm infants are impaired in a gestational age (GA)-dependent manner. METHODS Cord and peripheral blood mononuclear cells (MNCs) were stimulated with SE bacteria, and a range of innate immune responses were assessed, including phagocytosis, intracellular killing, Toll-like receptor (TLR) pathway transcriptional activation, cytokine production, TLR2 and TLR4 expression, and cell signaling. RESULTS Phagocytosis and intracellular killing of SE bacteria were similar in neonatal and adult monocytes. Cytokine gene expression and protein synthesis increased in a GA-dependent manner, which was confirmed at the single-cell level. These GA-related effects were not associated with differences in expression of TLR2 or TLR4, nor with downstream activation of nuclear factor-κB or mitogen-activated protein kinase pathways. DISCUSSION The expression of TLRs, phagocytic capacity, and intracellular killing by monocytes develops early in fetal development, whereas the ability to mount a bacteria-induced cytokine response requires further maturation. The functional immaturity of monocyte activation pathways in the preterm infant may underpin their particular susceptibility to sepsis with commensal bacteria.
Collapse
Affiliation(s)
- Tobias Strunk
- University of Western Australia Centre for Neonatal Research and Education, Perth, Western Australia, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Briana DD, Liosi S, Gourgiotis D, Boutsikou M, Baka S, Marmarinos A, Hassiakos D, Malamitsi-Puchner A. The potential role of the lectin pathway of complement in the host defence of full-term intrauterine growth restricted neonates at birth. J Matern Fetal Neonatal Med 2012; 25:531-4. [PMID: 22082351 DOI: 10.3109/14767058.2011.636108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To prospectively investigate the potential role of the lectin pathway of complement in intrauterine-growth-restriction (IUGR, associated with impaired immunocompetence and increased risk for neonatal infections), by determining cord blood concentrations of mannose-binding lectin (MBL), H-ficolin and L-ficolin (important mediators of neonatal innate immunity) in IUGR and appropriate for gestational age (AGA) pregnancies. Furthermore, we aimed to describe correlations among cord blood MBL, H- and L-ficolin concentrations and with several demographic parameters of the infants at birth. METHODS Serum MBL, H- and L-ficolin concentrations were determined by ELISA in 154 mixed arteriovenous cord blood samples from IUGR (n = 50) and AGA (n = 104) singleton full-term infants. RESULTS Cord blood MBL concentrations were significantly lower in IUGR cases than AGA controls (p = 0.029). No differences in cord blood H- and L-ficolin concentrations were observed between groups. In the IUGR group, cord blood MBL concentrations negatively correlated with respective L-ficolin ones (r = -0.442, p = 0.001). CONCLUSIONS The relatively decreased MBL expression in IUGR fetuses at term could possibly contribute to IUGR-associated neonatal immunodeficiency, predisposing to increased susceptibility to infections. The negative correlation between MBL and L-ficolin concentrations in the IUGR group might suggest an underlying immune variation and needs to be further investigated.
Collapse
Affiliation(s)
- Despina D Briana
- Neonatal Division, 2nd Department of Obstetrics and Gynecology, Athens University Medical School, Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Schlapbach LJ, Kjaer TR, Thiel S, Mattmann M, Nelle M, Wagner BP, Ammann RA, Aebi C, Jensenius JC. M-ficolin concentrations in cord blood are related to circulating phagocytes and to early-onset sepsis. Pediatr Res 2012; 71:368-74. [PMID: 22391637 DOI: 10.1038/pr.2011.71] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The pattern-recognition molecule M-ficolin is synthesized by monocytes and neutrophils. M-ficolin activates the complement system in a manner similar to mannan-binding lectin (MBL), but little is known about its role in host defense. Neonates are highly vulnerable to bacterial sepsis, in particular, due to their decreased phagocytic function. RESULTS M-ficolin cord blood concentration was positively correlated with the absolute phagocyte count (ρ 0.51, P < 0.001) and with immature/total neutrophil ratio (ρ 0.34, P < 0.001). When comparing infants with sepsis and controls, a high M-ficolin cord blood concentration (>1,000 ng/ml) was associated with early-onset sepsis (EOS) (multivariate odds ratio 10.92, 95% confidence interval 2.21-54.02, P = 0.003). Experimental exposure of phagocytes isolated from adult donors to Escherichia coli resulted in a significant time- and dose-dependent release of M-ficolin. DISCUSSION In conclusion, M-ficolin concentrations were related to circulating phagocytes and EOS. Our results indicate that bacterial sepsis can trigger M-ficolin release by phagocytes. Future studies should investigate whether M-ficolin may be used as a marker of neutrophil activation during invasive infections. METHODS We investigated M-ficolin in 47 infants with culture-positive sepsis during the first 30 days of life (13 with EOS and in 94 matched controls. M-ficolin was measured in cord blood using time-resolved immunofluorometric assay (TRIFMA). Multivariate logistic regression was performed.
Collapse
Affiliation(s)
- Luregn J Schlapbach
- Pediatric Critical Care Research Group, Mater Children's Hospital, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Pinar-Sueiro S, Sota M, Lerchundi TX, Gibelalde A, Berasategui B, Vilar B, Hernandez JL. Dacryocystitis: Systematic Approach to Diagnosis and Therapy. Curr Infect Dis Rep 2012; 14:137-146. [PMID: 22286338 DOI: 10.1007/s11908-012-0238-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The objective of this paper is to review the main findings of the largest studies on the etiopathogenesis and microbiology of the development of dacryocystitis and to formulate clinical and surgical guidelines based on said studies and on our experience at Cruces Hospital, the Basque Country, Spain. The most common sign of this entity is the distal nasolacrimal duct obstruction, and this should be treated to prevent clinical relapse. The time when surgery should be indicated mainly depends on the clinical signs and symptoms, age and general status of a patient. Given the germs isolated in cases of dacryocystitis, antibiotic therapy against Gram positive (S. aureus, S. pneumoniae, S. epidermidis) and Gram negative bacteria (H. influenzae, P. aeruginosa) should be administered, orally in adults and intravenously in pediatric patients, prior to surgery. Gentamicin and amoxicillin-clavulanic acid have been found to be effective against the bacteria commonly implicated in the etiopathogenesis of this entity.
Collapse
Affiliation(s)
- Sergio Pinar-Sueiro
- Department of Ophthalmology, Cruces Hospital, Plaza Cruces s/n, E-48903, Barakaldo, Bizkaia, Spain,
| | | | | | | | | | | | | |
Collapse
|
24
|
Mallard C, Wang X. Infection-induced vulnerability of perinatal brain injury. Neurol Res Int 2011; 2012:102153. [PMID: 22135745 PMCID: PMC3216257 DOI: 10.1155/2012/102153] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/05/2011] [Indexed: 11/21/2022] Open
Abstract
A growing body of evidence demonstrates that susceptibility and progression of both acute and chronic central nervous system disease in the newborn is closely associated with an innate immune response that can manifest from either direct infection and/or infection-triggered damage. A common feature of many of these diseases is the systemic exposure of the neonate to bacterial infections that elicit brain inflammation. In recent years, the importance of innate immune receptors in newborn brain injury, the so-called Toll-like receptors, has been demonstrated. In this paper we will discuss how neonatal sepsis, with particular emphasis on Escherichia coli, coagulase-negative staphylococci, and group B streptococcal infections in preterm infants, and Toll-like receptor-mediated inflammation can increase the vulnerability of the newborn brain to injury.
Collapse
Affiliation(s)
- Carina Mallard
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, P.O. Box 432, 40530 Göteborg, Sweden
| | | |
Collapse
|
25
|
Ho TS, Wang SM, Wu YH, Shen CF, Lin YJ, Lin CH, Liu CC. Long-term characteristics of healthcare-associated infections in a neonatal intensive care unit. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2011; 43:407-15. [PMID: 21075708 DOI: 10.1016/s1684-1182(10)60064-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 06/26/2009] [Accepted: 08/19/2009] [Indexed: 12/20/2022]
Abstract
BACKGROUND/PURPOSE Healthcare-associated infections in neonatal intensive care units (NICUs) are associated with a significant risk of morbidity and mortality. Knowledge regarding pathogens, primary sources of infection and antibiotic resistance in the NICU is essential for developing management strategies. This study aimed to analyze the long-term characteristics of healthcare-associated infections in a tertiary referral center in southern Taiwan. METHODS Infants < 30 days old, with positive blood, cerebrospinal fluid, urine or tissue fluid cultures during hospitalization in the NICU of National Cheng Kung University Hospital from July 1989 to June 2008 were included in the study. RESULTS In total, 1,417 organisms and episodes were identified during the study period. Gram-positive organisms, Gram-negative organisms and fungi constituted 923 (65.1%), 358 (25.3%) and 136 (9.6%) of the pathogens, respectively. Of the Gram-positive organisms, coagulase-negative staphylococci (51.5%), Staphylococcus aureus (34.8%) and Enterococcus spp. (6.1%) were the major pathogens; and 27% of Staphylococcus aureus isolates were oxacillin-resistant. For the Gram-negative organisms, Klebsiella pneumoniae (22%), Pseudomonas aeruginosa (21.8%), Escherichia coli (16.7%) and Enterobacter cloacae (16.7%) were dominant. Also, Candida albicans accounted for 50% of fungal infections. The most common source of infection was bloodstream infection (59.0%), and 5.6% of these were catheter-related. Skin and soft tissue infections were also frequent (26.3%). CONCLUSION Bloodstream and skin/soft tissue infections caused by commensal species play an important role in healthcare-associated infections in the NICU. New measures should be developed in response to the changing patterns in the NICU.
Collapse
Affiliation(s)
- Tzong-Shiann Ho
- Department of Pediatrics, National Cheng Kung University Medical College and Hospital, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
26
|
Du X, Fleiss B, Li H, D'angelo B, Sun Y, Zhu C, Hagberg H, Levy O, Mallard C, Wang X. Systemic stimulation of TLR2 impairs neonatal mouse brain development. PLoS One 2011; 6:e19583. [PMID: 21573120 PMCID: PMC3089625 DOI: 10.1371/journal.pone.0019583] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Accepted: 04/12/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Inflammation is associated with perinatal brain injury but the underlying mechanisms are not completely characterized. Stimulation of Toll-like receptors (TLRs) through specific agonists induces inflammatory responses that trigger both innate and adaptive immune responses. The impact of engagement of TLR2 signaling pathways on the neonatal brain is still unclear. The aim of this study was to investigate the potential effect of a TLR2 agonist on neonatal brain development. METHODOLOGY/PRINCIPAL FINDINGS Mice were injected intraperitoneally (i.p.) once a day from postnatal day (PND) 3 to PND11 with endotoxin-free saline, a TLR2 agonist Pam(3)CSK(4) (5 mg/kg) or Lipopolysaccharide (LPS, 0.3 mg/kg). Pups were sacrificed at PND12 or PND53 and brain, spleen and liver were collected and weighed. Brain sections were stained for brain injury markers. Long-term effects on memory function were assessed using the Trace Fear Conditioning test at PND50. After 9 days of Pam(3)CSK(4) administration, we found a decreased volume of cerebral gray matter, white matter in the forebrain and cerebellar molecular layer that was accompanied by an increase in spleen and liver weight at PND12. Such effects were not observed in Pam3CSK4-treated TLR 2-deficient mice. Pam3CSK4-treated mice also displayed decreased hippocampus neuronal density, and increased cerebral microglia density, while there was no effect on caspase-3 or general cell proliferation at PND12. Significantly elevated levels of IL-1β, IL-6, KC, and MCP-1 were detected after the first Pam3CSK4 injection in brain homogenates of PND3 mice. Pam(3)CSK(4) administration did not affect long-term memory function nor the volume of gray or white matter. CONCLUSIONS/SIGNIFICANCE Repeated systemic exposure to the TLR2 agonist Pam(3)CSK(4) can have a short-term negative impact on the neonatal mouse brain.
Collapse
Affiliation(s)
- Xiaonan Du
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Bobbi Fleiss
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Hongfu Li
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Barbara D'angelo
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yanyan Sun
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Perinatal Center, Department of Obstetrics and Gynecology, University of Gothenburg, Gothenburg, Sweden
- Institute of Reproductive and Developmental Biology, Imperial College, London, United Kingdom
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carina Mallard
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoyang Wang
- Perinatal Center, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
27
|
Jacob J, Sims D, Van de Rostyne C, Schmidt G, O’Leary K. Toward the Elimination of Catheter-Related Bloodstream Infections in a Newborn Intensive Care Unit (NICU). Jt Comm J Qual Patient Saf 2011; 37:211-6, 193. [DOI: 10.1016/s1553-7250(11)37028-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Preterm infants have deficient monocyte and lymphocyte cytokine responses to group B streptococcus. Infect Immun 2011; 79:1588-96. [PMID: 21300777 DOI: 10.1128/iai.00535-10] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Group B streptococcus (GBS) is an important cause of early- and late-onset sepsis in the newborn. Preterm infants have markedly increased susceptibility and worse outcomes, but their immunological responses to GBS are poorly defined. We compared mononuclear cell and whole-blood cytokine responses to heat-killed GBS (HKGBS) of preterm infants (gestational age [GA], 26 to 33 weeks), term infants, and healthy adults. We investigated the kinetics and cell source of induced cytokines and quantified HKGBS phagocytosis. HKGBS-induced tumor necrosis factor (TNF) and interleukin 6 (IL-6) secretion was significantly impaired in preterm infants compared to that in term infants and adults. These cytokines were predominantly monocytic in origin, and production was intrinsically linked to HKGBS phagocytosis. Very preterm infants (GA, <30 weeks) had fewer cytokine-producing monocytes, but nonopsonic phagocytosis ability was comparable to that for term infants and adults. Exogenous complement supplementation increased phagocytosis in all groups, as well as the proportion of preterm monocytes producing IL-6, but for very preterm infants, responses were still deficient. Similar defective preterm monocyte responses were observed in fresh whole cord blood stimulated with live GBS. Lymphocyte-associated cytokines were significantly deficient for both preterm and term infants compared to levels for adults. These findings indicate that a subset of preterm monocytes do not respond to GBS, a defect compounded by generalized weaker lymphocyte responses in newborns. Together these deficient responses may increase the susceptibility of preterm infants to GBS infection.
Collapse
|
29
|
Granslo HN, Klingenberg C, Fredheim EGA, Rønnestad A, Mollnes TE, Flaegstad T. Arginine catabolic mobile element is associated with low antibiotic resistance and low pathogenicity in Staphylococcus epidermidis from neonates. Pediatr Res 2010; 68:237-41. [PMID: 20703143 DOI: 10.1203/pdr.0b013e3181eb01e0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The arginine catabolic mobile element (ACME) in Staphylococci encodes several putative virulence factors. ACME appears to have been transferred from Staphylococcus epidermidis into Staphylococcus aureus and is strongly associated with the epidemic and virulent S. aureus USA300. We sought to determine the distribution of ACME in 128 S. epidermidis blood culture isolates from neonates and to assess ACME's impact on antibiotic resistance, biofilm production, invasive capacity, and host inflammatory response. ACME was detected in 15/64 (23%) invasive blood culture isolates and 26/64 (40%) blood culture contaminants (p = 0.02). ACME-positive S. epidermidis isolates displayed less antibiotic resistance (p < 0.001) and were collected from more mature neonates (p = 0.001). Biofilm production was more prevalent among ACME-negative isolates (61/87) compared with ACME positive (18/41; p = 0.004). Among the 64 children considered having an invasive infection, ACME did not influence the maximum C-reactive protein level. In an in vitro whole-blood sepsis model, there were no differences in the inflammatory response between ACME-positive and ACME-negative isolates. We conclude that ACME in S. epidermidis from neonates was associated with less antibiotic resistance and also does not seem to be associated with increased pathogenicity.
Collapse
|
30
|
Strunk T, Currie A, Richmond P, Simmer K, Burgner D. Innate immunity in human newborn infants: prematurity means more than immaturity. J Matern Fetal Neonatal Med 2010; 24:25-31. [DOI: 10.3109/14767058.2010.482605] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
31
|
Deshpande G, Rao S, Patole S, Bulsara M. Updated meta-analysis of probiotics for preventing necrotizing enterocolitis in preterm neonates. Pediatrics 2010; 125:921-30. [PMID: 20403939 DOI: 10.1542/peds.2009-1301] [Citation(s) in RCA: 337] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Systematic reviews of randomized, controlled trials (RCTs) indicate lower mortality and necrotizing enterocolitis (NEC) and shorter time to full feeds after probiotic supplementation in preterm (<34 weeks' gestation) very low birth weight (VLBW; birth weight <1500 g) neonates. The objective of this study was to update our 2007 systematic review of RCTs of probiotic supplementation for preventing NEC in preterm VLBW neonates. METHODS We searched in March 2009 the Cochrane Central register; Medline, Embase, and Cinahl databases; and proceedings of the Pediatric Academic Society meetings and gastroenterology conferences. Cochrane Neonatal Review Group search strategy was followed. Selection criteria were RCTs of any enteral probiotic supplementation that started within first 10 days and continued for > or =7 days in preterm VLBW neonates and reported on stage 2 NEC or higher (Modified Bell Staging). RESULTS A total of 11 (N = 2176), including 4 new (n = 783), trials were eligible for inclusion in the meta-analysis by using a fixed-effects model. The risk for NEC and death was significantly lower. Risk for sepsis did not differ significantly. No significant adverse effects were reported. Trial sequential analysis) showed 30% reduction in the incidence of NEC (alpha = .05 and .01; power: 80%). CONCLUSIONS The results confirm the significant benefits of probiotic supplements in reducing death and disease in preterm neonates. The dramatic effect sizes, tight confidence intervals, extremely low P values, and overall evidence indicate that additional placebo-controlled trials are unnecessary if a suitable probiotic product is available.
Collapse
Affiliation(s)
- Girish Deshpande
- Department of Neonatal Paediatrics, King Edward Memorial Hospital for Women, Perth, Western Australia, Australia
| | | | | | | |
Collapse
|
32
|
Strunk T, Power Coombs MR, Currie AJ, Richmond P, Golenbock DT, Stoler-Barak L, Gallington LC, Otto M, Burgner D, Levy O. TLR2 mediates recognition of live Staphylococcus epidermidis and clearance of bacteremia. PLoS One 2010; 5:e10111. [PMID: 20404927 PMCID: PMC2852418 DOI: 10.1371/journal.pone.0010111] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Accepted: 03/19/2010] [Indexed: 12/12/2022] Open
Abstract
Background Staphylococcus epidermidis (SE) is a nosocomial pathogen that causes catheter-associated bacteremia in the immunocompromised, including those at the extremes of age, motivating study of host clearance mechanisms. SE-derived soluble components engage TLR2; but additional signaling pathways have also been implicated, and TLR2 can play complex, at times detrimental, roles in host defense against other Staphylococcal spp. The role of TLR2 in responses of primary blood leukocytes to live SE and in clearance of SE bacteremia, the most common clinical manifestation of SE infection, is unknown. Methodology/Principal Findings We studied TLR2-mediated recognition of live clinical SE strain 1457 employing TLR2-transfected cells, neutralizing anti-TLR antibodies and TLR2-deficient mice. TLR2 mediated SE-induced cytokine production in human embryonic kidney cells, human whole blood and murine primary macrophages, in part via recognition of a soluble TLR2 agonist. After i.v. challenge with SE, early (1 h) cytokine/chemokine production and subsequent clearance of bacteremia (24–48 h) were markedly impaired in TLR2-deficient mice. Conclusions/Significance TLR2 mediates recognition of live SE and clearance of SE bacteremia in vivo.
Collapse
Affiliation(s)
- Tobias Strunk
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - Melanie R. Power Coombs
- Division Infectious Diseases, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Andrew J. Currie
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - Peter Richmond
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - Douglas T. Golenbock
- University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Liat Stoler-Barak
- Division Infectious Diseases, Children's Hospital Boston, Boston, Massachusetts, United States of America
| | - Leighanne C. Gallington
- Division Infectious Diseases, Children's Hospital Boston, Boston, Massachusetts, United States of America
| | - Michael Otto
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David Burgner
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
- Murdoch Childrens Research Institute, Royal Childrens Hospital, Parkville, Victoria, Australia
| | - Ofer Levy
- Division Infectious Diseases, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
33
|
Nelson A, Hultenby K, Hell E, Riedel HM, Brismar H, Flock JI, Lundahl J, Giske CG, Marchini G. Staphylococcus epidermidis isolated from newborn infants express pilus-like structures and are inhibited by the cathelicidin-derived antimicrobial peptide LL37. Pediatr Res 2009; 66:174-8. [PMID: 19390494 DOI: 10.1203/pdr.0b013e3181a9d80c] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Coagulase-negative staphylococci and its subtype Staphylococcus epidermidis are major indigenous Gram-positive inhabitants of the human skin. Colonization occurs in direct connection with birth and terrestrial adaptation. This study focuses on factors that may influence skin colonization of the newborn infant that relates to the immune status of both the bacteria and the host. Skin is an effective barrier against bacteria, and this function is partly mediated by the presence of antimicrobial peptides including human cathelicidin peptide LL37. Gram-positive bacteria have been described to have adhesive pili on their surface that mediates specific attachment to the host. Here, we identify, by negative staining transmission electron microscopy (EM), two different types of pilus-like structures commonly expressed on S. epidermidis isolated from newborn infants. We also show that the cathelicidin antimicrobial peptide LL37, constitutively expressed in the skin barrier of the newborn, significantly inhibited growth of S. epidermidis indicating its importance for the ecological stability of the skin microbiota. Further studies are required to elucidate molecular mechanisms of host-microbe interactions, both for the maintenance of a mutually beneficial homeostatic relationship and for the protection of self when it results in overt disease.
Collapse
Affiliation(s)
- Annika Nelson
- Department of Woman and Child Health, Karolinska Institutet, Stockholm 17176, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Schlapbach LJ, Kessler U, Thiel S, Hansen AG, Nelle M, Ammann RA, Aebi C, Jensenius JC. M-ficolin in the neonatal period: Associations with need for mechanical ventilation and mortality in premature infants with necrotising enterocolitis. Mol Immunol 2009; 46:2597-603. [PMID: 19539995 DOI: 10.1016/j.molimm.2009.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 04/21/2009] [Accepted: 05/02/2009] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Necrotising enterocolitis (NEC) causes significant mortality in premature infants. The involvement of the innate immune system in the pathogenesis of NEC remains unclear. M-, L- and H-ficolins recognize microorganisms and activate the complement system, but their role in host defense is largely unknown. This study investigated whether ficolin concentrations are associated with NEC. STUDY DESIGN Case-control study including 30 premature infants with NEC and 60 controls. M-, L- and H-ficolins were measured in cord blood using time-resolved immunofluorometric assays. Multivariate logistic regression was performed. RESULTS Of the 30 NEC cases (median gestational age, 29.5 weeks), 12 (40%) were operated and 4 (13%) died. No difference regarding ficolin concentration was found when comparing NEC cases versus controls (p>0.05). However, infants who died of NEC had significantly lower M-ficolin cord blood concentrations than NEC survivors (for M-ficolin <300ng/ml; multivariate OR 12.35, CI 1.03-148.59, p=0.048). In the entire study population, M-, L- and H-ficolins were positively correlated with gestational age (p<0.001) and birth weight (p<0.001). Infants with low M-ficolin required significantly more often mechanical ventilation after birth multivariate (OR 10.55, CI 2.01-55.34, p=0.005). CONCLUSIONS M-, L- and H-ficolins are already present in cord blood and increase with gestational age. Low cord blood concentration of M-ficolin was associated with higher NEC-associated fatality and with increased need for mechanical ventilation. Future studies need to assess whether M-ficolin is involved in multiorgan failure and pulmonary disease.
Collapse
MESH Headings
- Blotting, Western
- Case-Control Studies
- Cell Line
- Enterocolitis, Necrotizing/blood
- Enterocolitis, Necrotizing/mortality
- Enterocolitis, Necrotizing/therapy
- Fetal Blood/chemistry
- Humans
- Infant Mortality
- Infant, Newborn
- Infant, Premature/blood
- Lectins/analysis
- Lectins/blood
- Lectins/genetics
- Logistic Models
- Macrophages/cytology
- Macrophages/metabolism
- Macrophages, Alveolar/cytology
- Macrophages, Alveolar/metabolism
- Monocytes/cytology
- Monocytes/metabolism
- Multivariate Analysis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Respiration, Artificial
- Reverse Transcriptase Polymerase Chain Reaction
- Ficolins
Collapse
|
35
|
Phase 1/2 double-blind, placebo-controlled, dose escalation, safety, and pharmacokinetic study of pagibaximab (BSYX-A110), an antistaphylococcal monoclonal antibody for the prevention of staphylococcal bloodstream infections, in very-low-birth-weight neonates. Antimicrob Agents Chemother 2009; 53:2879-86. [PMID: 19380597 DOI: 10.1128/aac.01565-08] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcal sepsis is a major cause of morbidity and mortality in very-low-birth-weight (VLBW) infants. A human chimeric monoclonal antibody, pagibaximab, was developed against staphylococcal lipoteichoic acid. We evaluated the safety, tolerability, and pharmacokinetics of pagibaximab in VLBW neonates. A phase 1/2, randomized, double-blind, placebo-controlled, dose escalation study was conducted in VLBW infants (700 to 1,300 g) 3 to 7 days old. Patients received two doses 14 days apart of intravenous pagibaximab (10, 30, 60, or 90 mg/kg of body weight) or placebo in a 2:1 ratio. Blood and urine samples were obtained pre- and postinfusion for analysis of safety and pharmacokinetics, and data on adverse events were gathered. Staphylococcal organisms causing sepsis were collected and evaluated. Fifty-three patients received at least one dose of pagibaximab or placebo. The average gestational age was 27.6 weeks; the average birth weight was 1,003 g. All serious adverse events were deemed unrelated or probably not drug related. Morbidity and mortality were similar across treatment groups. No evidence of immunogenicity of pagibaximab was detected. Pagibaximab pharmacokinetics was linear. The mean clearance (CL), volume of distribution, and elimination half-life of pagibaximab were independent of dose. The serum half-life was 20.5 +/- 6.8 days. Pagibaximab enhanced serum opsonophagocytic activity. All staphylococci causing sepsis were opsonizable by pagibaximab. Two infusions of pagibaximab, administered 2 weeks apart to high-risk neonates appeared safe and tolerable, and pharmacokinetics were linear. Evaluation of more frequent doses, at the highest doses tested, in neonates at high-risk of staphylococcal sepsis, is warranted.
Collapse
|
36
|
Shah PS, Kaufman DA. Antistaphylococcal immunoglobulins to prevent staphylococcal infection in very low birth weight infants. Cochrane Database Syst Rev 2009:CD006449. [PMID: 19370635 DOI: 10.1002/14651858.cd006449.pub2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Nosocomial infection is a major problem affecting the immediate health and long-term outcome of preterm and very low birth weight neonates. More than half of these infections are caused by staphylococci. Various type specific antibodies targeted at different antigenic markers of Staphylococcus have been developed and have shown promise in animal studies. OBJECTIVES To evaluate the efficacy and safety of antistaphylococcal immunoglobulins in the prevention of Staphylococcal infection in very low birth weight infants. SEARCH STRATEGY Medline, Embase, CINAHL, Cochrane Central Register of Controlled Trials (The Cochrane Library) were searched from their inception until February 2009. In addition, abstracts of major pediatric meetings of last seven years were searched. SELECTION CRITERIA Randomized and quasi-randomized studies of antistaphylococcal immunoglobulins for the prevention of staphylococcal infections in preterm or very low birth weight neonates were reviewed by both authors for their eligibility for inclusion. Studies of any dose and/or route were included. Quality of studies was evaluated using criteria of masking of randomization, masking of intervention, completeness of follow-up and masking of outcome assessment by both review authors. DATA COLLECTION AND ANALYSIS Data from the primary author were obtained where published data provided inadequate information for the review or where relevant data could not be abstracted. Data were abstracted independently by both review authors. Statistical methods included calculation of relative risk (RR), risk difference (RD), number needed to treat (NNT) and weighted mean difference (WMD) when appropriate. Ninety five percent confidence intervals (CI) was used for these estimates of treatment effects. A fixed effect model was used for meta-analyses. MAIN RESULTS Three eligible studies were included (two studies of INH A-21 and one study of Altastaph involving a total of 2,701 neonates). Three reports of Pagibaximab were published as abstracts and will be considered for inclusion when further information is obtained. There were no significant differences noted in the risk of Staphylococcal infection between INH A-21 vs. placebo (typical RR 1.07, 95% CI 0.94, 1.22) or Altastaph vs. placebo (RR 0.86, 95% CI 0.32, 2.28); the risk of other bacterial infection between INH A-21 vs. placebo (typical RR 0.87, 95% CI 0.72, 1.06) or Altastaph vs. placebo (RR 0.93, 95% CI 0.53, 1.64); or the risk of any infection between INH A-21 vs. placebo (RR 1.00, 95% CI 0.91, 1.09) or Altastaph vs. placebo (RR 0.93, 95% CI 0.54, 1.62). There was no significant difference in the incidence of relevant secondary outcomes (chronic lung disease at 28 days, patent ductus arteriosus, necrotizing enterocolitis, intraventricular hemorrhage, retinopathy of prematurity or duration of antibiotic and vancomycin use). AUTHORS' CONCLUSIONS Antistaphylococcal immunoglobulins (INH A-21 and Altastaph) are not recommended for prevention of staphylococcal infections in preterm or VLBW neonates. Further research to investigate the efficacy of other products such as Pagibaximab is needed.
Collapse
Affiliation(s)
- Prakeshkumar S Shah
- Department of Paediatrics and Department of Health Policy, Management and Evaluation, Rm 775A, University of Toronto, 600 University Avenue, Toronto, Ontario, Canada, M5G 1XB.
| | | |
Collapse
|
37
|
Schlapbach LJ, Aebi C, Fisch U, Ammann RA, Otth M, Bigler S, Nelle M, Berger S, Kessler U. Higher cord blood levels of mannose-binding lectin-associated serine protease-2 in infants with necrotising enterocolitis. Pediatr Res 2008; 64:562-6. [PMID: 18596574 DOI: 10.1203/pdr.0b013e3181841335] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Necrotising enterocolitis (NEC) causes significant morbidity and mortality in premature infants. The role of innate immunity in the pathogenesis of NEC remains unclear. Mannose-binding lectin (MBL) recognizes microorganisms and activates the complement system via MBL-associated serine protease-2 (MASP-2). The aim of this study was to investigate whether MBL and MASP-2 are associated with NEC. This observational case-control study included 32 infants with radiologically confirmed NEC and 64 controls. MBL and MASP-2 were measured in cord blood using ELISA. Multivariate logistic regression was performed. Of the 32 NEC cases (median gestational age, 30.5 wk), 13 (41%) were operated and 5 (16%) died. MASP-2 cord blood concentration ranged from undetectable (<10 ng/mL) to 277 ng/mL. Eighteen of 32 (56%) NEC cases had higher MASP-2 levels (> or =30 ng/mL) compared with 22 of 64 (34%) controls (univariate OR 2.46; 95% CI 1.03-5.85; p = 0.043). Higher cord blood MASP-2 levels were significantly associated with an increased risk of NEC in multivariate analysis (OR 3.00; 95% CI 1.17-7.93; p = 0.027). MBL levels were not associated with NEC (p = 0.64). In conclusion, infants later developing NEC had significantly higher MASP-2 cord blood levels compared with controls. Higher MASP-2 may favor complement-mediated inflammation and could thereby predispose to NEC.
Collapse
|
38
|
Baskin DE, Reddy AK, Chu YI, Coats DK. The timing of antibiotic administration in the management of infant dacryocystitis. J AAPOS 2008; 12:456-9. [PMID: 18595757 DOI: 10.1016/j.jaapos.2008.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 04/18/2008] [Accepted: 04/19/2008] [Indexed: 11/30/2022]
Abstract
PURPOSE To report (1) the prevalence of bacteremia among infants with dacryocystitis and (2) the influence of timing of antibiotic administration on the need for repeat probing in the management of these patients. METHODS A retrospective analysis of the hospital records of 25 infants < or =6 weeks of age treated for acute dacryocystitis was conducted, including analysis of laboratory data and outcomes. RESULTS Of 22 infants who underwent blood cultures, 5 (22.7%) were bacteremic. Twenty-one of the 25 infants underwent nasolacrimal duct probing. Infants who received preoperative antibiotics were less likely to require a repeat probing than those who did not (6% vs. 80%), and this difference was statistically significant (p = 0.004). CONCLUSIONS The high rate of bacteremia in this series of patients and the significantly lower incidence of repeat probing among infants who received preprocedural antibiotics suggests that blood cultures and subsequent administration of intravenous antibiotics should be considered prior to probing of infants with dacryocystitis.
Collapse
Affiliation(s)
- Darrell E Baskin
- Department of Ophthalmology, Baylor College of Medicine Texas Children's Hospital, Houston, Texas, USA
| | | | | | | |
Collapse
|
39
|
Castro BA. The immunocompromised pediatric patient and surgery. Best Pract Res Clin Anaesthesiol 2008; 22:611-26. [DOI: 10.1016/j.bpa.2008.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Recent trends and prevention of infection in the neonatal intensive care unit. Curr Opin Infect Dis 2008; 21:350-6. [DOI: 10.1097/qco.0b013e3283013af4] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Shah DK, Doyle LW, Anderson PJ, Bear M, Daley AJ, Hunt RW, Inder TE. Adverse neurodevelopment in preterm infants with postnatal sepsis or necrotizing enterocolitis is mediated by white matter abnormalities on magnetic resonance imaging at term. J Pediatr 2008; 153:170-5, 175.e1. [PMID: 18534228 DOI: 10.1016/j.jpeds.2008.02.033] [Citation(s) in RCA: 304] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2007] [Revised: 02/04/2008] [Accepted: 02/18/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVES To test the hypothesis that the impact of postnatal sepsis/necrotizing enterocolitis (NEC) on neurodevelopment may be mediated by white matter abnormality (WMA), which can be demonstrated with magnetic resonance imaging (MRI). STUDY DESIGN A prospective cohort of 192 unselected preterm infants (gestational age <30 weeks), who were evaluated for sepsis and NEC, underwent imaging at term-equivalent age and neurodevelopmental outcome at 2 years corrected age with the Bayley Scales of Infant Development. RESULTS Sixty-eight preterm (35%) infants had 100 episodes of confirmed sepsis, and 9 (5%) infants had confirmed NEC. Coagulase-negative staphylococci accounted for 73% (73/100) of the episodes of confirmed sepsis. Infants with sepsis/NEC had significantly more WMA on MRI at term compared with infants in the no-sepsis/NEC group. They also had poorer psychomotor development that persisted after adjusting for potential confounders but which became nonsignificant after adjusting for WMA. CONCLUSIONS Preterm infants with sepsis/NEC are at greater risk of motor impairment at 2 years, which appears to be mediated by WMA. These findings may assist in defining a neuroprotective target in preterm infants with sepsis/NEC.
Collapse
Affiliation(s)
- Divyen K Shah
- Department of Pediatrics, Washington University, St Louis, MO 63105, USA.
| | | | | | | | | | | | | |
Collapse
|