1
|
Gleeson B, Ferreyra C, Palamountain K, Jacob ST, Spotswood N, Kissoon N, Nisar YB, Fitzgerald F, Murless-Collins S, Okomo U, Cross JH, Molyneux E, Piriou E, Iloh KK, Data S, Goldfarb D, Stevenson A, Kirby R, Nichols BE, Blumel B, Kelly-Cirino C, Walsh T, Lloyd L, Liaghati-Mobarhan S. A call to bridge the diagnostic gap: diagnostic solutions for neonatal sepsis in low- and middle-income countries. BMJ Glob Health 2024; 9:e015862. [PMID: 39260829 PMCID: PMC11404204 DOI: 10.1136/bmjgh-2024-015862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/19/2024] [Indexed: 09/13/2024] Open
Affiliation(s)
| | | | | | | | - Naomi Spotswood
- Burnet Institute, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne VCCC, Parkville, Victoria, Australia
| | - Niranjan Kissoon
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Yasir Bin Nisar
- Department of Maternal, Newborn, Child and Adolescent Health and Ageing, World Health Organization, Geneve, Switzerland
| | - Felicity Fitzgerald
- Imperial College London, London, UK
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Sarah Murless-Collins
- Maternal Adolescent Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Uduak Okomo
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - James H Cross
- Maternal Adolescent Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Erwan Piriou
- Médecins Sans Frontières, Amsterdam, The Netherlands
| | - Kenechukwu K Iloh
- University of Nigeria Teaching Hospital, Ituku Ozalla, Enugu, Nigeria
| | - Santorino Data
- Pediatrics and Child Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - David Goldfarb
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | - Lizel Lloyd
- Stellenbosch University, Stellenbosch, South Africa
| | | | - Neonatal Sepsis Diagnostic Working Group
- FIND, Geneva, Switzerland
- Northwestern University, Evanston, Illinois, USA
- Liverpool School of Tropical Medicine, Liverpool, UK
- Burnet Institute, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne VCCC, Parkville, Victoria, Australia
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
- Department of Maternal, Newborn, Child and Adolescent Health and Ageing, World Health Organization, Geneve, Switzerland
- Imperial College London, London, UK
- Biomedical Research and Training Institute, Harare, Zimbabwe
- Maternal Adolescent Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, UK
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, Gambia
- Kamuzu University of Health Sciences, Blantyre, Southern Region, Malawi
- Médecins Sans Frontières, Amsterdam, The Netherlands
- University of Nigeria Teaching Hospital, Ituku Ozalla, Enugu, Nigeria
- Pediatrics and Child Health, Mbarara University of Science and Technology, Mbarara, Uganda
- University of Zimbabwe, Harare, Zimbabwe
- University of Oxford, Oxford, UK
- Stellenbosch University, Stellenbosch, South Africa
- UNICEF Supply Division, Copenhagen, Denmark
| |
Collapse
|
2
|
Alsultan A, Aldawsari MR, Alturaiq NK, Syed SA, Alsubai A, Kurdee Z, Alsubaie S, Alqahtani S, Abouelkheir M. Evaluation of pharmacokinetic pharmacodynamic target attainment of meropenem in pediatric patients. Pediatr Neonatol 2024; 65:386-390. [PMID: 38218717 DOI: 10.1016/j.pedneo.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/27/2023] [Accepted: 09/08/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Meropenem is a widely used carbapenem for treating severe pediatric infections. However, few studies have assessed its pharmacokinetics/pharmacodynamics (PK/PD) in pediatric patients. This study aimed to evaluate the proportion of Saudi pediatric patients achieving the PK/PD target of meropenem. METHODS A prospective observational study was conducted at King Saud University Medical City from July to September 2022. Pediatric patients receiving meropenem for suspected or proven infections were included in the study. The primary outcome was the percentage of patients achieving the recommended PK/PD target for critically ill or non-critically ill pediatric patients. RESULTS The study included 30 patients (nine neonates and 21 older pediatric patients). All neonates were critically ill. Among them, 55 % achieved the PK/PD target of 100 % free time above the MIC. In older ICU pediatric patients, only 11 % attained this target, whereas 58 % of older pediatrics in the general wards achieved the PK/PD target of 50 % free time above the MIC. Augmented renal clearance (ARC) was identified in 57 % of our pediatric patient population, none of whom achieved the recommended PK/PD targets. The median trough concentrations in patients with and without ARC were 0.75 and 1.3 μg/mL, respectively (P < 0.05). CONCLUSIONS The majority of patients in our cohort did not achieve the PK/PD target for meropenem. ARC emerged as a major risk factor for target attainment failure in both critically ill and non-critically ill pediatric patients.
Collapse
Affiliation(s)
- Abdullah Alsultan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia.
| | - Maram R Aldawsari
- Department of Pharmacy, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Nujood Khaled Alturaiq
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saeed Ali Syed
- Department of Pharmaceutical, Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz Alsubai
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Zeyad Kurdee
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Saudi Arabia
| | - Sarah Alsubaie
- Pediatric Infectious Disease Unit, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Saeed Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Manal Abouelkheir
- Department of Clinical Pharmacy, Faculty of Pharmacy, Misr International University, Cairo, Egypt.
| |
Collapse
|
3
|
Fanous MS, de la Cruz JE, Michael OS, Afolabi JM, Kumar R, Adebiyi A. EARLY FLUID PLUS NOREPINEPHRINE RESUSCITATION DIMINISHES KIDNEY HYPOPERFUSION AND INFLAMMATION IN SEPTIC NEWBORN PIGS. Shock 2024; 61:885-893. [PMID: 38662580 PMCID: PMC11251746 DOI: 10.1097/shk.0000000000002343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
ABSTRACT Sepsis is the most frequent risk factor for acute kidney injury (AKI) in critically ill infants. Sepsis-induced dysregulation of kidney microcirculation in newborns is unresolved. The objective of this study was to use the translational swine model to evaluate changes in kidney function during the early phase of sepsis in newborns and the impact of fluid plus norepinephrine resuscitation. Newborn pigs (3-7-day-old) were allocated randomly to three groups: 1) sham, 2) sepsis (cecal ligation and puncture) without subsequent resuscitation, and 3) sepsis with lactated Ringer plus norepinephrine resuscitation. All animals underwent standard anesthesia and mechanical ventilation. Cardiac output and glomerular filtration rate were measured noninvasively. Mean arterial pressure, total renal blood flow, cortical perfusion, medullary perfusion, and medullary tissue oxygen tension (mtPO 2 ) were determined for 12 h. Cecal ligation and puncture decreased mean arterial pressure and cardiac output by more than 50%, with a proportional increase in renal vascular resistance and a 60-80% reduction in renal blood flow, cortical perfusion, medullary perfusion, and mtPO 2 compared to sham. Cecal ligation and puncture also decreased glomerular filtration rate by ~79% and increased AKI biomarkers. Isolated foci of tubular necrosis were observed in the septic piglets. Except for mtPO 2 , changes in all these parameters were ameliorated in resuscitated piglets. Resuscitation also attenuated sepsis-induced increases in the levels of plasma C-reactive protein, proinflammatory cytokines, lactate dehydrogenase, alanine transaminase, aspartate aminotransferase, and renal NLRP3 inflammasome. These data suggest that newborn pigs subjected to cecal ligation and puncture develop hypodynamic septic AKI. Early implementation of resuscitation lessens the degree of inflammation, AKI, and liver injury.
Collapse
Affiliation(s)
- Mina S. Fanous
- Stormont Vail Pediatric Critical Care, Topeka, Kansas
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
| | - Julia E. de la Cruz
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Olugbenga S. Michael
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jeremiah M. Afolabi
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
| | - Ravi Kumar
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Adebowale Adebiyi
- Department of Physiology, University of TN Health Science Center, Memphis, Tennessee
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
4
|
Boll EJ, Lopez DV, Terne M, Hessing S, Parschat K, Jensen SR. Human milk oligosaccharides differentially support gut barrier integrity and enhance Th1 and Th17 cell effector responses in vitro. Front Immunol 2024; 15:1359499. [PMID: 38510254 PMCID: PMC10950922 DOI: 10.3389/fimmu.2024.1359499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Human milk oligosaccharides (HMOs) can modulate the intestinal barrier and regulate immune cells to favor the maturation of the infant intestinal tract and immune system, but the precise functions of individual HMOs are unclear. To determine the structure-dependent effects of individual HMOs (representing different structural classes) on the intestinal epithelium as well as innate and adaptive immune cells, we assessed fucosylated (2'FL and 3FL), sialylated (3'SL and 6'SL) and neutral non-fucosylated (LNT and LNT2) HMOs for their ability to support intestinal barrier integrity, to stimulate the secretion of chemokines from intestinal epithelial cells, and to modulate cytokine release from LPS-activated dendritic cells (DCs), M1 macrophages (MØs), and co-cultures with naïve CD4+ T cells. The fucosylated and neutral non-fucosylated HMOs increased barrier integrity and protected the barrier following an inflammatory insult but exerted minimal immunomodulatory activity. The sialylated HMOs enhanced the secretion of CXCL10, CCL20 and CXCL8 from intestinal epithelial cells, promoted the secretion of several cytokines (including IL-10, IL-12p70 and IL-23) from LPS-activated DCs and M1 MØs, and increased the secretion of IFN-γ and IL-17A from CD4+ T cells primed by LPS-activated DCs and MØs while reducing the secretion of IL-13. Thus, 3'SL and 6'SL supported Th1 and Th17 responses while reducing Th2 responses. Collectively, our data show that HMOs exert structure-dependent effects on the intestinal epithelium and possess immunomodulatory properties that confer benefits to infants and possibly also later in life.
Collapse
Affiliation(s)
| | | | - Mandy Terne
- Chr. Hansen A/S, Applied HMOs, Hoersholm, Denmark
| | - Sara Hessing
- Chr. Hansen A/S, Applied HMOs, Hoersholm, Denmark
| | | | | |
Collapse
|
5
|
Borghesi A. Life-threatening infections in human newborns: Reconciling age-specific vulnerability and interindividual variability. Cell Immunol 2024; 397-398:104807. [PMID: 38232634 DOI: 10.1016/j.cellimm.2024.104807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
In humans, the interindividual variability of clinical outcome following exposure to a microorganism is immense, ranging from silent infection to life-threatening disease. Age-specific immune responses partially account for the high incidence of infection during the first 28 days of life and the related high mortality at population level. However, the occurrence of life-threatening disease in individual newborns remains unexplained. By contrast, inborn errors of immunity and their immune phenocopies are increasingly being discovered in children and adults with life-threatening viral, bacterial, mycobacterial and fungal infections. There is a need for convergence between the fields of neonatal immunology, with its in-depth population-wide characterization of newborn-specific immune responses, and clinical immunology, with its investigations of infections in patients at the cellular and molecular levels, to facilitate identification of the mechanisms of susceptibility to infection in individual newborns and the design of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia, EU, Italy; School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland.
| |
Collapse
|
6
|
Doğan G, Karagenç N, Esmen K, Kul BÇ, Yeşilkaya H, Akgün Ş, Orman MN, Sandıkçı M, Eren Ü, Ünsal H, Karagenç L. Expression of Toll-Like Receptors in the Lung Tissue of Mouse Fetuses Generated by in vitro Embryo Culture and Embryo Transfer. Cells Tissues Organs 2023; 213:181-202. [PMID: 37105136 DOI: 10.1159/000529974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 02/27/2023] [Indexed: 04/29/2023] Open
Abstract
Mouse fetuses generated by in vitro embryo culture and embryo transfer exhibit impaired lung development, altered composition of pulmonary epithelial cells associated with downregulation of several genes involved in lung development and toll-like receptor (TLR) signaling pathway. The aims of the present study were to determine the expression of all TLRs and to examine if the expression of TLRs, along with genes involved in TLR signaling pathway, is altered in the lung tissue of mouse fetuses generated through embryo culture and embryo transfer. Two experimental (EGs) and one control (CG) group were included in the study. Embryos cultured at 5% CO2-95% air for 95 h or less than 24 h were transferred to pseudo-pregnant females to obtain fetuses comprising EGin vitro (n = 18) and EGin vivo (n = 18), respectively. Fetuses obtained from naturally ovulating females on day 18 of pregnancy served as the CG (n = 18). Western blot and immunohistochemistry were used to determine the expression of TLR proteins. The expression of transcripts encoding TLRs, and the genes involved in TLR signaling pathway (Lbp, Pik3r1, Pik3cb, Nfkbia, and Fos), was determined using qRT-PCR. While all TLRs were expressed by cells lining the bronchial/bronchiolar epithelium of lung tissues in all groups, some of the TLRs were expressed in a specific pattern. When compared to CG, the expression of transcripts encoding TLR-2, -3, -4, -5, -7, -8, -9, -12, -13, Lbp, Pik3r1, Pik3cb, Nfkbia, and Fos was significantly downregulated in both EGs. It appears that stress imposed on embryos at preimplantation stages of development is associated with downregulation of TLRs, along with some of the genes involved in TLR signaling pathway, in the lung tissue during the perinatal period. It remains to be determined if downregulation of TLRs, along with the genes involved in TLR signaling pathway, has any functional consequences in the adult lung tissue.
Collapse
Affiliation(s)
- Göksel Doğan
- Department of Histology-Embryology, Faculty of Veterinary Medicine, Adnan Menderes University, Aydın, Turkey
| | - Nedim Karagenç
- Department of Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Kerem Esmen
- Department of Medical, Faculty of Medicine, Dokuz Eylül University, İzmir, Turkey
| | - Bengi Çınar Kul
- Department of Genetics, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Hasan Yeşilkaya
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Şakir Akgün
- Department of Medical Biology, Faculty of Medicine, Kafkas University, Kars, Turkey
| | - Mehmet Nurullah Orman
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Ege University, İzmir, Turkey
| | - Mustafa Sandıkçı
- Department of Histology-Embryology, Faculty of Veterinary Medicine, Adnan Menderes University, Aydın, Turkey
| | - Ülker Eren
- Department of Histology-Embryology, Faculty of Veterinary Medicine, Adnan Menderes University, Aydın, Turkey
| | - Hümeyra Ünsal
- Department of Physiology, Faculty of Veterinary Medicine, Adnan Menderes University, Aydın, Turkey
| | - Levent Karagenç
- Department of Histology-Embryology, Faculty of Veterinary Medicine, Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
7
|
Lubin JB, Green J, Maddux S, Denu L, Duranova T, Lanza M, Wynosky-Dolfi M, Flores JN, Grimes LP, Brodsky IE, Planet PJ, Silverman MA. Arresting microbiome development limits immune system maturation and resistance to infection in mice. Cell Host Microbe 2023; 31:554-570.e7. [PMID: 36996818 PMCID: PMC10935632 DOI: 10.1016/j.chom.2023.03.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 01/09/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023]
Abstract
Disruptions to the intestinal microbiome during weaning lead to negative effects on host immune function. However, the critical host-microbe interactions during weaning that are required for immune system development remain poorly understood. We find that restricting microbiome maturation during weaning stunts immune system development and increases susceptibility to enteric infection. We developed a gnotobiotic mouse model of the early-life microbiome Pediatric Community (PedsCom). These mice develop fewer peripheral regulatory T cells and less IgA, hallmarks of microbiota-driven immune system development. Furthermore, adult PedsCom mice retain high susceptibility to Salmonella infection, which is characteristic of young mice and children. Altogether, our work illustrates how the post-weaning transition in microbiome composition contributes to normal immune maturation and protection from infection. Accurate modeling of the pre-weaning microbiome provides a window into the microbial requirements for healthy development and suggests an opportunity to design microbial interventions at weaning to improve immune development in human infants.
Collapse
Affiliation(s)
- Jean-Bernard Lubin
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jamal Green
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Maddux
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lidiya Denu
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tereza Duranova
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew Lanza
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | | | - Julia N Flores
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Logan P Grimes
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Igor E Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA; Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul J Planet
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Michael A Silverman
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immunology Research Unit, GlaxoSmithKline, Collegeville, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
8
|
Parra-Llorca A, Pinilla-Gonzlez A, Torrejón-Rodríguez L, Lara-Cantón I, Kuligowski J, Collado MC, Gormaz M, Aguar M, Vento M, Serna E, Cernada M. Effects of Sepsis on Immune Response, Microbiome and Oxidative Metabolism in Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2023; 10:602. [PMID: 36980160 PMCID: PMC10046958 DOI: 10.3390/children10030602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
This is a narrative review about the mechanisms involved in bacterial sepsis in preterm infants, which is an illness with a high incidence, morbidity, and mortality. The role of the innate immune response and its relationship with oxidative stress in the pathogenesis are described as well as their potential implementation as early biomarkers. Moreover, we address the impact that all the mechanisms triggered by sepsis have on the dysbiosis and the changes on neonatal microbiota.
Collapse
Affiliation(s)
- Anna Parra-Llorca
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Alejandro Pinilla-Gonzlez
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Laura Torrejón-Rodríguez
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Inmaculada Lara-Cantón
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - María Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - María Gormaz
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Marta Aguar
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Máximo Vento
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Eva Serna
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - María Cernada
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| |
Collapse
|
9
|
Dual inhibition of complement C5 and CD14 attenuates inflammation in a cord blood model. Pediatr Res 2023:10.1038/s41390-023-02489-2. [PMID: 36725909 DOI: 10.1038/s41390-023-02489-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 12/20/2022] [Accepted: 01/13/2023] [Indexed: 02/03/2023]
Abstract
BACKGROUND Escherichia coli and Group B streptococci (GBS) are the main causes of neonatal early-onset sepsis (EOS). Despite antibiotic therapy, EOS is associated with high morbidity and mortality. Dual inhibition of complement C5 and the Toll-like receptor co-factor CD14 has in animal studies been a promising novel therapy for sepsis. METHODS Whole blood was collected from the umbilical cord after caesarean section (n = 30). Blood was anti-coagulated with lepirudin. C5 inhibitor (eculizumab) and anti-CD14 was added 8 min prior to, or 15 and 30 min after adding E. coli or GBS. Total bacterial incubation time was 120 min (n = 16) and 240 min (n = 14). Cytokines and the terminal complement complex (TCC) were measured using multiplex technology and ELISA. RESULTS Dual inhibition significantly attenuated TCC formation by 25-79% when adding inhibitors with up to 30 min delay in both E. coli- and GBS-induced inflammation. TNF, IL-6 and IL-8 plasma concentration were significantly reduced by 28-87% in E. coli-induced inflammation when adding inhibitors with up to 30 min delay. The dual inhibition did not significantly reduce TNF, IL-6 and IL-8 plasma concentration in GBS-induced inflammation. CONCLUSION Dual inhibition of C5 and CD14 holds promise as a potential future treatment for severe neonatal EOS. IMPACT Neonatal sepsis can cause severe host inflammation with high morbidity and mortality, but there are still no effective adjunctive immunologic interventions available. Adding CD14 and complement C5 inhibitors up to 30 min after incubation of E. coli or Group B streptococci in a human umbilical cord blood model significantly reduced complement activation and cytokine release. Dual inhibition of C5 and CD14 is a potential future therapy to modulate systemic inflammation in severe cases of neonatal sepsis.
Collapse
|
10
|
Kou C, Li DF, Tang BH, Dong L, Yao BF, van den Anker J, You DP, Wu YE, Zhao W. Clinical Utility of A Model-based Amoxicillin Dosage Regimen in Neonates with Early-Onset Sepsis. Br J Clin Pharmacol 2022; 88:4950-4955. [PMID: 36057912 DOI: 10.1111/bcp.15521] [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: 01/13/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/26/2022] Open
Abstract
Early-onset sepsis (EOS) is one of the most significant causes of morbidity and mortality in neonates. Currently, amoxicillin is empirically used to treat neonates with EOS. However, data on its effectiveness in neonates with EOS are still limited. Therefore, we aimed to evaluate the pharmacodynamics (PD) target attainment and effectiveness of a model-based amoxicillin dosage regimen in these neonates. We used a previously developed model and collected additional clinical data from the EOS neonates who used the model-based dosage regimen (25 mg/kg q12h). The primary outcomes were PD target attainment (free drug concentration above MIC during 70% of the dosing interval) and treatment failure rate. The secondary endpoints were length of amoxicillin treatment, duration of hospitalization, etc. Seventy-five neonates (postmenstrual age 28.4-41.6 weeks) were enrolled. A total of 70 (93.3%) neonates reached their PD target using 1 mg/L as the MIC breakpoint. The treatment failure rate was 10.7%.
Collapse
Affiliation(s)
- Chen Kou
- Department of Neonatology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Di-Fei Li
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Dong
- Department of Pharmacy, Children's Hospital of Hebei Province affiliated to Hebei Medical University, Shijiazhuang, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA.,Departments of Pediatrics, Pharmacology & Physiology, Genomics and Precision Medicine, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Switzerland
| | - Dian-Ping You
- Pediatric Research Institute, Children's Hospital of Hebei Province affiliated to Hebei Medical University, Shijiazhuang, China
| | - Yue-E Wu
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pharmacy, Clinical Trial Center, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
11
|
Qiao Y, Zhang K, Zhang Z, Zhang C, Sun Y, Feng Z. Fermented soybean foods: A review of their functional components, mechanism of action and factors influencing their health benefits. Food Res Int 2022; 158:111575. [PMID: 35840260 DOI: 10.1016/j.foodres.2022.111575] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/27/2022]
Abstract
After thousands of years of evolution and development, traditional fermented soybean foods, with their unique charm, have gained a stable place in the global market. With the explosive development of modern biological technologies, some traditional fermented soybean foods that possess health-promoting benefits are gradually appearing. Physiologically active substances in fermented soybean foods have received extensive attention in recent decades. This review addresses the potential health benefits of several representative fermented soybean foods, as well as the action mechanism and influencing factors of their functional components. Phenolic compounds, low-molecular-weight peptides, melanoidins, furanones and 3-hydroxyanthranilic acid are the antioxidative components predominantly found in fermented soybean foods. Angiotensin I-converting enzyme inhibitory peptides and γ-aminobutyric acid isolated from fermented soy foods provide potential selectivity for hypertension therapy. The potential anti-inflammatory bioactive components in fermented soybean foods include γ-linolenic acid, butyric acid, soy sauce polysaccharides, 2S albumin and isoflavone glycones. Deoxynojirimycin, genistein, and betaine possess high activity against α-glucosidase. Additionally, fermented soybean foods contain neuroprotective constituents, including indole alkaloids, nattokinase, arbutin, and isoflavone vitamin B12. The anticancer activities of fermented soybean foods are associated with surfactin, isolavone, furanones, trypsin inhibitors, and 3-hydroxyanthranilic acid. Nattokinase is highly correlated with antioxidant activity. And a high level of menaquinones-7 is linked to protection against neurodegenerative diseases. Sufficiently recognizing and exploiting the health benefits and functional components of traditional fermented soybean foods could provide a new strategy in the development of the food fermentation industry.
Collapse
Affiliation(s)
- Yali Qiao
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Kenan Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Zongcai Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Chao Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Yan Sun
- Heilongjiang Tobacco Industry Co., Ltd. Harbin Cigarette Factory, Harbin 150027, China
| | - Zhen Feng
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China; Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China.
| |
Collapse
|
12
|
Yang Z, Liu X, Wu Y, Peng J, Wei H. Effect of the Microbiome on Intestinal Innate Immune Development in Early Life and the Potential Strategy of Early Intervention. Front Immunol 2022; 13:936300. [PMID: 35928828 PMCID: PMC9344006 DOI: 10.3389/fimmu.2022.936300] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022] Open
Abstract
Early life is a vital period for mammals to be colonized with the microbiome, which profoundly influences the development of the intestinal immune function. For neonates to resist pathogen infection and avoid gastrointestinal illness, the intestinal innate immune system is critical. Thus, this review summarizes the development of the intestinal microbiome and the intestinal innate immune barrier, including the intestinal epithelium and immune cells from the fetal to the weaning period. Moreover, the impact of the intestinal microbiome on innate immune development and the two main way of early-life intervention including probiotics and fecal microbiota transplantation (FMT) also are discussed in this review. We hope to highlight the crosstalk between early microbial colonization and intestinal innate immunity development and offer some information for early intervention.
Collapse
Affiliation(s)
- Zhipeng Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiangchen Liu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yanting Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
13
|
Zhou J, Jiang L, Zhang ZL, Wang ZR, Zhang YX, Lin X, Tang BH, Yao BF, Guo ZX, Yang JJ, Van Den Anker J, Wu YE, Zhao W. Population pharmacokinetics and dosing optimization of mezlocillin in neonates and young infants. J Antimicrob Chemother 2022; 77:2238-2244. [PMID: 35662337 DOI: 10.1093/jac/dkac176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/07/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Mezlocillin is used in the treatment of neonatal infectious diseases. However, due to the absence of population pharmacokinetic studies in neonates and young infants, dosing regimens differ considerably in clinical practice. Hence, this study aimed to describe the pharmacokinetic characteristics of mezlocillin in neonates and young infants, and propose the optimal dosing regimen based on the population pharmacokinetic model of mezlocillin. METHODS A prospective, open-label pharmacokinetic study of mezlocillin was carried out in newborns. Blood samples were collected using an opportunistic sampling method. HPLC was used to measure the plasma drug concentrations. A population pharmacokinetic model was developed using NONMEM software. RESULTS Ninety-five blood samples from 48 neonates and young infants were included. The ranges of postmenstrual age and birth weight were 29-40 weeks and 1200-4000 g, respectively, including term and preterm infants. A two-compartment model with first-order elimination was developed to describe the population pharmacokinetics of mezlocillin. Postmenstrual age, current weight and serum creatinine concentration were the most important covariates. Monte Carlo simulation results indicated that the current dose of 50 mg/kg q12h resulted in 89.2% of patients achieving the therapeutic target, when the MIC of 4 mg/L was used as the breakpoint. When increasing the dosing frequency to q8h, a dose of 20 mg/kg resulted in 74.3% of patients achieving the therapeutic target. CONCLUSIONS A population pharmacokinetic model of mezlocillin in neonates and young infants was established. Optimal dosing regimens based on this model were provided for use in neonatal infections.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Li Jiang
- Department of Pediatrics, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Zhi-Ling Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Zhao-Rui Wang
- Department of Pediatrics, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Yan-Xiu Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Xu Lin
- Department of Pediatrics, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zi-Xuan Guo
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing-Jing Yang
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - John Van Den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA.,Departments of Pediatrics, Pharmacology & Physiology, Genomics and Precision Medicine, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Switzerland
| | - Yue-E Wu
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| |
Collapse
|
14
|
Jankauskaite L, Malinauskas M, Mickeviciute GC. HMGB1: A Potential Target of Nervus Vagus Stimulation in Pediatric SARS-CoV-2-Induced ALI/ARDS. Front Pediatr 2022; 10:884539. [PMID: 35633962 PMCID: PMC9132499 DOI: 10.3389/fped.2022.884539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 12/19/2022] Open
Abstract
From the start of pandemics, children were described as the ones who were less affected by SARS-Cov-2 or COVID-19, which was mild in most of the cases. However, with the growing vaccination rate of the adult population, children became more exposed to the virus and more cases of severe SARS-CoV-2-induced ARDS are being diagnosed with the disabling consequences or lethal outcomes associated with the cytokine storm. Thus, we do hypothesize that some of the children could benefit from nervus vagus stimulation during COVID-19 ARDS through the inhibition of HMGB1 release and interaction with the receptor, resulting in decreased neutrophil accumulation, oxidative stress, and coagulopathy as well as lung vascular permeability. Moreover, stimulation through alpha-7 nicotinic acetylcholine receptors could boost macrophage phagocytosis and increase the clearance of DAMPs and PAMPs. Further rise of FGF10 could contribute to lung stem cell proliferation and potential regeneration of the injured lung. However, this stimulation should be very specific, timely, and of proper duration, as it could lead to such adverse effects as increased viral spread and systemic infection, especially in small children or infants due to specific pediatric immunity state and anatomical features of the respiratory system.
Collapse
Affiliation(s)
- Lina Jankauskaite
- Lithuanian University of Health Sciences, Medical Academy, Pediatric Department, Kaunas, Lithuania
- Lithuanian University of Health Sciences, Medical Academy, Institute of Physiology and Pharmacology, Kaunas, Lithuania
| | - Mantas Malinauskas
- Lithuanian University of Health Sciences, Medical Academy, Institute of Physiology and Pharmacology, Kaunas, Lithuania
| | - Goda-Camille Mickeviciute
- Lithuanian University of Health Sciences, Medical Academy, Pediatric Department, Kaunas, Lithuania
- Lithuanian University of Health Sciences, Medical Academy, Institute of Physiology and Pharmacology, Kaunas, Lithuania
- Rehabilitation Center “Palangos Linas”, Palanga, Lithuania
| |
Collapse
|
15
|
Raza MA, Yao B, Shi H, Xu H, Hao G, Anker J, Zhao W. Optimal Dose of Meropenem for the Treatment of Neonatal Sepsis: Dosing Guideline Variations and Clinical Practice Deviations. Br J Clin Pharmacol 2022; 88:3483-3489. [DOI: 10.1111/bcp.15308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 11/27/2022] Open
Affiliation(s)
- Muhammad Ahmer Raza
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University Jinan China
| | - Bu‐Fan Yao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University Jinan China
| | - Hai‐Yan Shi
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy Jinan China
| | - Hai‐Yan Xu
- Department of Neonatology The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital Jinan China
| | - Guo‐Xiang Hao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University Jinan China
| | - John Anker
- Division of Clinical Pharmacology, Children’s National Medical Center Washington DC USA
- Departments of Pediatrics, Pharmacology & Physiology, Genomics & Precision Medicine the George Washington University School of Medicine and Health Sciences Washington DC USA
- Department of Paediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel University of Basel Switzerland
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University Jinan China
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy Jinan China
| |
Collapse
|
16
|
Dias ML, O'Connor KM, Dempsey EM, O'Halloran KD, McDonald FB. Targeting the Toll-like receptor pathway as a therapeutic strategy for neonatal infection. Am J Physiol Regul Integr Comp Physiol 2021; 321:R879-R902. [PMID: 34612068 DOI: 10.1152/ajpregu.00307.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Toll-like receptors (TLRs) are crucial transmembrane receptors that form part of the innate immune response. They play a role in the recognition of various microorganisms and their elimination from the host. TLRs have been proposed as vital immunomodulators in the regulation of multiple neonatal stressors that extend beyond infection such as oxidative stress and pain. The immune system is immature at birth and takes some time to become fully established. As such, babies are especially vulnerable to sepsis at this early stage of life. Findings suggest a gestational age-dependent increase in TLR expression. TLRs engage with accessory and adaptor proteins to facilitate recognition of pathogens and their activation of the receptor. TLRs are generally upregulated during infection and promote the transcription and release of proinflammatory cytokines. Several studies report that TLRs are epigenetically modulated by chromatin changes and promoter methylation upon bacterial infection that have long-term influences on immune responses. TLR activation is reported to modulate cardiorespiratory responses during infection and may play a key role in driving homeostatic instability observed during sepsis. Although complex, TLR signaling and downstream pathways are potential therapeutic targets in the treatment of neonatal diseases. By reviewing the expression and function of key Toll-like receptors, we aim to provide an important framework to understand the functional role of these receptors in response to stress and infection in premature infants.
Collapse
Affiliation(s)
- Maria L Dias
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Eugene M Dempsey
- Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland.,Department of Pediatrics and Child Health, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland.,Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland
| | - Fiona B McDonald
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland.,Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland
| |
Collapse
|
17
|
Ding X, Kambara H, Guo R, Kanneganti A, Acosta-Zaldívar M, Li J, Liu F, Bei T, Qi W, Xie X, Han W, Liu N, Zhang C, Zhang X, Yu H, Zhao L, Ma F, Köhler JR, Luo HR. Inflammasome-mediated GSDMD activation facilitates escape of Candida albicans from macrophages. Nat Commun 2021; 12:6699. [PMID: 34795266 PMCID: PMC8602704 DOI: 10.1038/s41467-021-27034-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is the most common cause of fungal sepsis. Inhibition of inflammasome activity confers resistance to polymicrobial and LPS-induced sepsis; however, inflammasome signaling appears to protect against C. albicans infection, so inflammasome inhibitors are not clinically useful for candidiasis. Here we show disruption of GSDMD, a known inflammasome target and key pyroptotic cell death mediator, paradoxically alleviates candidiasis, improving outcomes and survival of Candida-infected mice. Mechanistically, C. albicans hijacked the canonical inflammasome-GSDMD axis-mediated pyroptosis to promote their escape from macrophages, deploying hyphae and candidalysin, a pore-forming toxin expressed by hyphae. GSDMD inhibition alleviated candidiasis by preventing C. albicans escape from macrophages while maintaining inflammasome-dependent but GSDMD-independent IL-1β production for anti-fungal host defenses. This study demonstrates key functions for GSDMD in Candida's escape from host immunity in vitro and in vivo and suggests that GSDMD may be a potential therapeutic target in C. albicans-induced sepsis.
Collapse
Affiliation(s)
- Xionghui Ding
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
- Department of Burn and Plastic Surgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China
| | - Hiroto Kambara
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Rongxia Guo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, CAMS Key laboratory for prevention and control of hematological disease treatment related infection, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Apurva Kanneganti
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Maikel Acosta-Zaldívar
- Division of Infectious Diseases, Boston Children's Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Jiajia Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, CAMS Key laboratory for prevention and control of hematological disease treatment related infection, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Fei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, CAMS Key laboratory for prevention and control of hematological disease treatment related infection, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Ting Bei
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Wanjun Qi
- Division of Infectious Diseases, Boston Children's Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Xuemei Xie
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Wenli Han
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Ningning Liu
- Division of Infectious Diseases, Boston Children's Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Cunling Zhang
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Xiaoyu Zhang
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Hongbo Yu
- VA Boston Healthcare System, Department of Pathology and Laboratory Medicine, 1400 VFW Parkway West Roxbury, Boston, MA, 02132, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Li Zhao
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA
| | - Fengxia Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, CAMS Key laboratory for prevention and control of hematological disease treatment related infection, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Julia R Köhler
- Division of Infectious Diseases, Boston Children's Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Hongbo R Luo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA, 02115, USA.
| |
Collapse
|
18
|
Peterson LS, Hedou J, Ganio EA, Stelzer IA, Feyaerts D, Harbert E, Adusumelli Y, Ando K, Tsai ES, Tsai AS, Han X, Ringle M, Houghteling P, Reiss JD, Lewis DB, Winn VD, Angst MS, Aghaeepour N, Stevenson DK, Gaudilliere B. Single-Cell Analysis of the Neonatal Immune System Across the Gestational Age Continuum. Front Immunol 2021; 12:714090. [PMID: 34497610 PMCID: PMC8420969 DOI: 10.3389/fimmu.2021.714090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/02/2021] [Indexed: 12/21/2022] Open
Abstract
Although most causes of death and morbidity in premature infants are related to immune maladaptation, the premature immune system remains poorly understood. We provide a comprehensive single-cell depiction of the neonatal immune system at birth across the spectrum of viable gestational age (GA), ranging from 25 weeks to term. A mass cytometry immunoassay interrogated all major immune cell subsets, including signaling activity and responsiveness to stimulation. An elastic net model described the relationship between GA and immunome (R=0.85, p=8.75e-14), and unsupervised clustering highlighted previously unrecognized GA-dependent immune dynamics, including decreasing basal MAP-kinase/NFκB signaling in antigen presenting cells; increasing responsiveness of cytotoxic lymphocytes to interferon-α; and decreasing frequency of regulatory and invariant T cells, including NKT-like cells and CD8+CD161+ T cells. Knowledge gained from the analysis of the neonatal immune landscape across GA provides a mechanistic framework to understand the unique susceptibility of preterm infants to both hyper-inflammatory diseases and infections.
Collapse
Affiliation(s)
- Laura S Peterson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Julien Hedou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ina A Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Eliza Harbert
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Yamini Adusumelli
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Eileen S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Amy S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Megan Ringle
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Pearl Houghteling
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Jonathan D Reiss
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - David B Lewis
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, United States
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Nima Aghaeepour
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States.,Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - David K Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Brice Gaudilliere
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States.,Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
19
|
Wu YE, Wang T, Yang HL, Tang BH, Kong L, Li X, Gao Q, Li X, Yao BF, Shi HY, Huang X, Wang WQ, Jacqz-Aigrain E, Allegaert K, van den Anker J, Tian XY, Zhao W. Population pharmacokinetics and dosing optimization of azlocillin in neonates with early-onset sepsis: a real-world study. J Antimicrob Chemother 2021; 76:699-709. [PMID: 33188385 DOI: 10.1093/jac/dkaa468] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Nowadays, real-world data can be used to improve currently available dosing guidelines and to support regulatory approval of drugs for use in neonates by overcoming practical and ethical hurdles. This proof-of-concept study aimed to assess the population pharmacokinetics of azlocillin in neonates using real-world data, to make subsequent dose recommendations and to test these in neonates with early-onset sepsis (EOS). METHODS This prospective, open-label, investigator-initiated study of azlocillin in neonates with EOS was conducted using an adaptive two-step design. First, a maturational pharmacokinetic-pharmacodynamic model of azlocillin was developed, using an empirical dosing regimen combined with opportunistic samples resulting from waste material. Second, a Phase II clinical trial (ClinicalTrials.gov: NCT03932123) of this newly developed model-based dosing regimen of azlocillin was conducted to assure optimized target attainment [free drug concentration above MIC during 70% of the dosing interval ('70% fT>MIC')] and to investigate the tolerance and safety in neonates. RESULTS A one-compartment model with first-order elimination, using 167 azlocillin concentrations from 95 neonates (31.7-41.6 weeks postmenstrual age), incorporating current weight and renal maturation, fitted the data best. For the second step, 45 neonates (30.3-41.3 weeks postmenstrual age) were subsequently included to investigate target attainment, tolerance and safety of the pharmacokinetic-pharmacodynamic model-based dose regimen (100 mg/kg q8h). Forty-three (95.6%) neonates reached their pharmacokinetic target and only two neonates experienced adverse events (feeding intolerance and abnormal liver function), possibly related to azlocillin. CONCLUSIONS Target attainment, tolerance and safety of azlocillin was shown in neonates with EOS using a pharmacokinetic-pharmacodynamic model developed with real-world data.
Collapse
Affiliation(s)
- Yue-E Wu
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tao Wang
- Department of Pharmacy, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China
| | - Hua-Liang Yang
- Department of Pharmacy, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Li Kong
- Department of Neonatology, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China
| | - Xin Li
- Department of Neonatology, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China
| | - Qi Gao
- Department of Neonatology, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China.,Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China
| | - Xue Li
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hai-Yan Shi
- Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xin Huang
- Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Wen-Qi Wang
- Clinical Research Centre, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Evelyne Jacqz-Aigrain
- Department of Paediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, APHP, Paris, France.,Clinical Investigation Centre CIC1426, Hôpital Robert Debré, Paris, France.,University of Paris, Paris, France
| | - Karel Allegaert
- Department of Development and Regeneration and Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Department of Clinical Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA.,Departments of Pediatrics, Pharmacology & Physiology, Genomics and Precision Medicine, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.,Department of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Basel, Switzerland
| | - Xiu-Ying Tian
- Department of Neonatology, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China.,Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.,Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Clinical Research Centre, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
20
|
Crocker LW, White A, Heaton PA, Horta DP, Paul SP. Recognition and management of neonatal sepsis. ACTA ACUST UNITED AC 2021; 30:410-415. [PMID: 33830803 DOI: 10.12968/bjon.2021.30.7.410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neonatal sepsis results from acute bacterial or viral infection occurring in the first 28 days of life. It causes significant morbidity and mortality, although the outcome can be improved by early recognition and prompt treatment by health professionals. This article describes the most common causes of sepsis, and explains why neonates are particularly vulnerable to infection. It highlights the non-specific way in which an infant with a serious infection may present, indicating the crucial features to elicit during history taking and examination, and emphasising the 'red-flag' signs and symptoms that should increase suspicion of a serious illness. The authors have adapted National Institute for Health and Care Excellence guidelines to produce an evidence-based approach to the management of an infant with suspected sepsis, and describe the roles of nurses in ensuring effective treatment and best outcomes for these babies.
Collapse
Affiliation(s)
| | - Ayesha White
- 5th Year Medical Student, University of Bristol, Bristol
| | | | | | | |
Collapse
|
21
|
Belizario JA, Lopes LG, Pires RH. Fungi in the indoor air of critical hospital areas: a review. AEROBIOLOGIA 2021; 37:379-394. [PMID: 34007098 PMCID: PMC8119621 DOI: 10.1007/s10453-021-09706-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/17/2021] [Indexed: 05/31/2023]
Abstract
Invasive fungal infection is an important cause of mortality and morbidity in neonates, especially in low-birthweight neonates. The contribution of fungi in the indoor air to the incidence of mucocutaneous colonization and to the risk of invasive fungal infection in this population is uncertain. This review aimed to identify and to summarize the best available evidence on the fungal contamination in the indoor air of critical hospital areas with an emphasis on pediatric/neonatal ICUs. Publications from 2005 to 2019 were searched in the databases Scientific Electronic Library Online (SciELO), US National Library of Medicine National Institutes of Health Search (PubMed), and Latin American Caribbean Health Sciences (LILACS). Descriptors in Health Sciences (DeCS) were used. Research papers published in Portuguese, English, and Spanish were included. Twenty-nine papers on all continents except Australia were selected. The results showed that the air mycobiota contained several fungal species, notably Aspergillus, Penicillium, Cladosporium, Fusarium, and yeast (Candida) species. The selected papers point out the risks that fungi pose to neonates, who have immature immune system, and describe simultaneous external factors (air humidity, seasonality, air and people flow, use of particulate filters, and health professionals' hand hygiene) that contribute to indoor air contamination with fungi. Improving communication among health professionals is a great concern because this can prevent major health complications in neonates, especially in low-birthweight neonates. The results reinforced the need to monitor environmental fungi more frequently and efficiently in hospitals, especially in neonatal ICUs.
Collapse
Affiliation(s)
- Jenyffie A. Belizario
- Universidade de Franca, Av. Dr. Armando Salles de Oliveira, 201, Parque Universitário, Franca, São Paulo 14404-600 Brazil
| | - Leonardo G. Lopes
- Universidade de Franca, Av. Dr. Armando Salles de Oliveira, 201, Parque Universitário, Franca, São Paulo 14404-600 Brazil
| | - Regina H. Pires
- Universidade de Franca, Av. Dr. Armando Salles de Oliveira, 201, Parque Universitário, Franca, São Paulo 14404-600 Brazil
| |
Collapse
|
22
|
Kuti BP, Ogunlesi TA, Oduwole O, Oringanje C, Udoh EE, Meremikwu MM. Hand hygiene for the prevention of infections in neonates. Cochrane Database Syst Rev 2021; 1:CD013326. [PMID: 33471367 PMCID: PMC8094276 DOI: 10.1002/14651858.cd013326.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Annually, infections contribute to approximately 25% of the 2.8 million neonatal deaths worldwide. Over 95% of sepsis-related neonatal deaths occur in low- and middle-income countries. Hand hygiene is an inexpensive and cost-effective method of preventing infection in neonates, making it an affordable and practicable intervention in low- and middle-income settings. Therefore, hand hygiene practices may hold strong prospects for reducing the occurrence of infection and infection-related neonatal death. OBJECTIVES To determine the effectiveness of different hand hygiene agents for preventing neonatal infection in community and health facility settings. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2019, Issue 5), in the Cochrane Library; MEDLINE via PubMed (1966 to 10 May 2019); Embase (1980 to 10 May 2019); and the Cumulative Index to Nursing and Allied Health Literature (CINAHL) (1982 to 10 May 2019). We also searched clinical trials databases and the reference lists of retrieved articles for randomised controlled trials (RCTs) and quasi-randomised trials. Searches were updated 1 June 2020. SELECTION CRITERIA We included RCTs, cross-over trials, and quasi-RCTs that included pregnant women, mothers, other caregivers, and healthcare workers who received interventions within the community or in health facility settings DATA COLLECTION AND ANALYSIS: We used standard methodological procedures expected by Cochrane and the GRADE approach to assess the certainty of evidence. Primary outcomes were incidence of (study author-defined) suspected infection within the first 28 days of life, bacteriologically confirmed infection within the first 28 days of life, all-cause mortality within the first seven days of life (early neonatal death), and all-cause mortality from the 8th to the 28th day of life (late neonatal death). MAIN RESULTS Our review included five studies: one RCT, one quasi-RCT, and three cross-over trials with a total of more than 5450 neonates (two studies included all neonates but did not report the actual number of neonates involved). Four studies involved 279 nurses working in neonatal intensive care units and all neonates on admission. The fifth study did not clearly state how many nurses were included in the study. Studies examined the effectiveness of different hand hygiene practices for the incidence of (study author-defined) suspected infection within the first 28 days of life. Two studies were rated as low risk for selection bias, another two were rated as high risk, and one study was rated as unclear risk. One study was rated as low risk for allocation bias, and four were rated as high risk. Only one of the five studies was rated as low risk for performance bias. 4% chlorhexidine gluconate (CHG) compared to plain liquid soap We are uncertain whether plain soap is better than 4% chlorhexidine gluconate (CHG) for nurses' skin based on very low-certainty evidence (mean difference (MD) -1.75, 95% confidence interval (CI) -3.31 to -0.19; 16 participants, 1 study; very low-certainty evidence). We identified no studies that reported on other outcomes for this comparison. 4% chlorhexidine gluconate compared to triclosan 1% One study compared 1% w/v triclosan with 4% chlorhexidine gluconate and suggests that 1% w/v triclosan may reduce the incidence of suspected infection (risk ratio (RR) 1.04, 95% CI 0.19 to 5.60; 1916 participants, 1 study; very low-certainty evidence). There may be fewer cases of infection in the 1% w/v triclosan group compared to the 4% chlorhexidine gluconate group (RR 6.01, 95% CI 3.56 to 10.14; 1916 participants, 1 study; very low-certainty evidence); however, we are uncertain of the available evidence. We identified no study that reported on all-cause mortality, duration of hospital stay, and adverse events for this comparison. 2% CHG compared to alcohol hand sanitiser (61% alcohol and emollients) We are uncertain whether 2% chlorhexidine gluconate reduces the risk of all infection in neonates compared to 61% alcohol hand sanitiser with regards to the incidence of all bacteriologically confirmed infection within the first 28 days of life (RR 2.19, 95% CI 1.79 to 2.69; 2932 participants, 1 study; very low-certainty evidence) in the 2% chlorhexidine gluconate group, but the evidence is very uncertain. The adverse outcome was reported as mean visual scoring on the skin. There may be little to no difference between the effects of 2% CHG on nurses' skin compared to alcohol hand sanitiser based on very low-certainty evidence (MD 0.80, 95% CI 0.01 to 1.59; 118 participants, 1 study; very low-certainty evidence). We identified no study that reported on all-cause mortality and other outcomes for this comparison. None of the included studies assessed all-cause mortality within the first seven days of life nor duration of hospital stay. AUTHORS' CONCLUSIONS: We are uncertain as to the superiority of one hand hygiene agent over another because this review included very few studies with very serious study limitations.
Collapse
Affiliation(s)
- Bankole Peter Kuti
- Department of Paediatrics and Child Health, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Tinuade A Ogunlesi
- Department of Paediatrics (Neonatal Unit), Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu, Nigeria
| | - Olabisi Oduwole
- Department of Medical Laboratory Science, Achievers University, Owo, Nigeria
| | - Chukwudi Oringanje
- Institute of Tropical Diseases Research and Prevention, University of Calabar Teaching Hospital (ITDR/P), Calabar, Nigeria
| | - Ekong E Udoh
- Department of Paediatrics, University of Uyo Teaching Hospital, Uyo, Nigeria
| | - Martin M Meremikwu
- Department of Paediatrics, University of Calabar Teaching Hospital, Calabar, Nigeria
| |
Collapse
|
23
|
McCulloh RJ, Commers T, Williams DD, Michael J, Mann K, Newland JG. Effect of Combined Clinical Practice Guideline and Electronic Order Set Implementation on Febrile Infant Evaluation and Management. Pediatr Emerg Care 2021; 37:e25-e31. [PMID: 32221058 DOI: 10.1097/pec.0000000000002012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Management of febrile infants 60 days and younger for suspected serious infection varies widely. Clinical practice guidelines (CPGs) are intended to improve clinician adherence to evidence-based practices. In 2011, a CPG for managing febrile infants was implemented in an urban children's hospital with simultaneous release of an electronic order set and algorithm to guide clinician decisions for managing infants for suspected serious bacterial infection. The objective of the present study was to determine the association of CPG implementation with order set use, clinical practices, and clinical outcomes. METHODS Records of febrile infants 60 days and younger from February 1, 2009, to January 31, 2013, were retrospectively reviewed. Clinical documentation, order set use, clinical management practices, and outcomes were compared pre-CPG and post-CPG release. RESULTS In total, 1037 infants pre-CPG and 930 infants post-CPG implementation were identified. After CPG release, more infants 29 to 60 days old underwent lumbar puncture (56% vs 62%, P = 0.02). Overall antibiotic use and duration of antibiotic use decreased for infants 29 to 60 days (57% vs 51%, P = 0.02). Blood culture and urine culture obtainment remained unchanged for older infants. Diagnosed infections, hospital readmissions, and length of stay were unchanged. Electronic order sets were used in 80% of patient encounters. CONCLUSIONS Antibiotic use and lumbar puncture performance modestly changed in accordance with CPG recommendations provided in the electronic order set and algorithm, suggesting that the presence of embedded prompts may affect clinician decision-making. Our results highlight the potential usefulness of these decision aids to improve adherence to CPG recommendations.
Collapse
Affiliation(s)
| | | | - David D Williams
- Division of Health Services and Outcomes Research, Children's Mercy Kansas City, Kansas City
| | | | | | | |
Collapse
|
24
|
Gilbert R, Brown M, Faria R, Fraser C, Donohue C, Rainford N, Grosso A, Sinha AK, Dorling J, Gray J, Muller-Pebody B, Harron K, Moitt T, McGuire W, Bojke L, Gamble C, Oddie SJ. Antimicrobial-impregnated central venous catheters for preventing neonatal bloodstream infection: the PREVAIL RCT. Health Technol Assess 2020; 24:1-190. [PMID: 33174528 DOI: 10.3310/hta24570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Clinical trials show that antimicrobial-impregnated central venous catheters reduce catheter-related bloodstream infection in adults and children receiving intensive care, but there is insufficient evidence for use in newborn babies. OBJECTIVES The objectives were (1) to determine clinical effectiveness by conducting a randomised controlled trial comparing antimicrobial-impregnated peripherally inserted central venous catheters with standard peripherally inserted central venous catheters for reducing bloodstream or cerebrospinal fluid infections (referred to as bloodstream infections); (2) to conduct an economic evaluation of the costs, cost-effectiveness and value of conducting additional research; and (3) to conduct a generalisability analysis of trial findings to neonatal care in the NHS. DESIGN Three separate studies were undertaken, each addressing one of the three objectives. (1) This was a multicentre, open-label, pragmatic randomised controlled trial; (2) an analysis was undertaken of hospital care costs, lifetime cost-effectiveness and value of information from an NHS perspective; and (3) this was a retrospective cohort study of bloodstream infection rates in neonatal units in England. SETTING The randomised controlled trial was conducted in 18 neonatal intensive care units in England. PARTICIPANTS Participants were babies who required a peripherally inserted central venous catheter (of 1 French gauge in size). INTERVENTIONS The interventions were an antimicrobial-impregnated peripherally inserted central venous catheter (coated with rifampicin-miconazole) or a standard peripherally inserted central venous catheter, allocated randomly (1 : 1) using web randomisation. MAIN OUTCOME MEASURE Study 1 - time to first bloodstream infection, sampled between 24 hours after randomisation and 48 hours after peripherally inserted central venous catheter removal. Study 2 - cost-effectiveness of the antimicrobial-impregnated peripherally inserted central venous catheter compared with the standard peripherally inserted central venous catheters. Study 3 - risk-adjusted bloodstream rates in the trial compared with those in neonatal units in England. For study 3, the data used were as follows: (1) case report forms and linked death registrations; (2) case report forms and linked death registrations linked to administrative health records with 6-month follow-up; and (3) neonatal health records linked to infection surveillance data. RESULTS Study 1, clinical effectiveness - 861 babies were randomised (antimicrobial-impregnated peripherally inserted central venous catheter, n = 430; standard peripherally inserted central venous catheter, n = 431). Bloodstream infections occurred in 46 babies (10.7%) randomised to antimicrobial-impregnated peripherally inserted central venous catheters and in 44 (10.2%) babies randomised to standard peripherally inserted central venous catheters. No difference in time to bloodstream infection was detected (hazard ratio 1.11, 95% confidence interval 0.73 to 1.67; p = 0.63). Secondary outcomes of rifampicin resistance in positive blood/cerebrospinal fluid cultures, mortality, clinical outcomes at neonatal unit discharge and time to peripherally inserted central venous catheter removal were similar in both groups. Rifampicin resistance in positive peripherally inserted central venous catheter tip cultures was higher in the antimicrobial-impregnated peripherally inserted central venous catheter group (relative risk 3.51, 95% confidence interval 1.16 to 10.57; p = 0.02) than in the standard peripherally inserted central venous catheter group. Adverse events were similar in both groups. Study 2, economic evaluation - the mean cost of babies' hospital care was £83,473. Antimicrobial-impregnated peripherally inserted central venous catheters were not cost-effective. Given the increased price, compared with standard peripherally inserted central venous catheters, the minimum reduction in risk of bloodstream infection for antimicrobial-impregnated peripherally inserted central venous catheters to be cost-effective was 3% and 15% for babies born at 23-27 and 28-32 weeks' gestation, respectively. Study 3, generalisability analysis - risk-adjusted bloodstream infection rates per 1000 peripherally inserted central venous catheter days were similar among babies in the trial and in all neonatal units. Of all bloodstream infections in babies receiving intensive or high-dependency care in neonatal units, 46% occurred during peripherally inserted central venous catheter days. LIMITATIONS The trial was open label as antimicrobial-impregnated and standard peripherally inserted central venous catheters are different colours. There was insufficient power to determine differences in rifampicin resistance. CONCLUSIONS No evidence of benefit or harm was found of peripherally inserted central venous catheters impregnated with rifampicin-miconazole during neonatal care. Interventions with small effects on bloodstream infections could be cost-effective over a child's life course. Findings were generalisable to neonatal units in England. Future research should focus on other types of antimicrobial impregnation of peripherally inserted central venous catheters and alternative approaches for preventing bloodstream infections in neonatal care. TRIAL REGISTRATION Current Controlled Trials ISRCTN81931394. FUNDING This project was funded by the National Institute for Health Research Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 24, No. 57. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Ruth Gilbert
- UCL Great Ormond Street Institute of Child Health, Faculty of Population Health Sciences, University College London, London, UK.,Health Data Research UK, London, UK
| | - Michaela Brown
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, UK
| | - Rita Faria
- Centre for Health Economics, University of York, York, UK
| | - Caroline Fraser
- UCL Great Ormond Street Institute of Child Health, Faculty of Population Health Sciences, University College London, London, UK
| | - Chloe Donohue
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, UK
| | - Naomi Rainford
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, UK
| | | | | | - Jon Dorling
- Division of Neonatal-Perinatal Medicine, Dalhousie University IWK Health Centre, Halifax, NS, Canada
| | - Jim Gray
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | | | - Katie Harron
- UCL Great Ormond Street Institute of Child Health, Faculty of Population Health Sciences, University College London, London, UK
| | - Tracy Moitt
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Laura Bojke
- Centre for Health Economics, University of York, York, UK
| | - Carrol Gamble
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, UK
| | - Sam J Oddie
- Centre for Reviews and Dissemination, University of York, York, UK.,Bradford Neonatology, Bradford Royal Infirmary, Bradford, UK
| |
Collapse
|
25
|
Meena R, Meena KK, Athwani V, Gothwal S, Bairwa GS, Sitaraman S. Umbilical Cord Blood Culture in Diagnosis of Early Onset Neonatal Sepsis. Indian J Pediatr 2020; 87:793-797. [PMID: 32468390 DOI: 10.1007/s12098-020-03345-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/11/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To evaluate the use of umbilical cord blood culture (UCBC) as compared to neonatal blood culture (NBC) in the diagnosis of early onset neonatal sepsis (EONS). METHODS This hospital based prospective observational study was conducted in the Department of Pediatrics and Department of Obstetrics & Gynaecology at a tertiary care centre of North India from 2017 through 2018. A total of 80 newborns with presence of two or more risk factors for sepsis were included in the study. Blood culture samples were collected from the umbilical cord at delivery and from the neonate within 1 h of birth. RESULTS UCBC was positive in 17 (21.2%) neonates and NBC was positive in 15 (18.7%) neonates. Out of these positive cases, 10 (45.4%) neonates had similar bacteriological profile in both UCBC and NBC which included Burkholderia cepacia, Acinetobacter species, Enterobacter cloacae and Coagulase-negative Staphylococcus. UCBC had a sensitivity of 66.7%, a specificity of 89.2%, a positive predictive value 58.8% and a negative predictive value 92.1% for the diagnosis of EONS in high risk neonates, considering NBC as the gold standard test. CONCLUSIONS UCBC is a reliable alternative to NBC for early etiological diagnosis of EONS in high risk neonates. Additional blood sampling from the newborn is suggested if there is any clinical sign of sepsis.
Collapse
Affiliation(s)
- Ramraj Meena
- Department of Pediatrics, SMS Medical College and J K Lon Hospital, Jaipur, Rajasthan, India
| | - Kailash Kumar Meena
- Department of Pediatrics, SMS Medical College and J K Lon Hospital, Jaipur, Rajasthan, India
| | - Vivek Athwani
- Department of Pediatrics, SMS Medical College and J K Lon Hospital, Jaipur, Rajasthan, India
| | - Sunil Gothwal
- Department of Pediatrics, SMS Medical College and J K Lon Hospital, Jaipur, Rajasthan, India.
| | - Ghan Shyam Bairwa
- Department of Pediatrics, Jaipuria Hospital and RUHS, Jaipur, Rajasthan, India
| | - Sadasivam Sitaraman
- Department of Pediatrics, SMS Medical College and J K Lon Hospital, Jaipur, Rajasthan, India
| |
Collapse
|
26
|
Petrova VN, Sawatsky B, Han AX, Laksono BM, Walz L, Parker E, Pieper K, Anderson CA, de Vries RD, Lanzavecchia A, Kellam P, von Messling V, de Swart RL, Russell CA. Incomplete genetic reconstitution of B cell pools contributes to prolonged immunosuppression after measles. Sci Immunol 2020; 4:4/41/eaay6125. [PMID: 31672862 DOI: 10.1126/sciimmunol.aay6125] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022]
Abstract
Measles is a disease caused by the highly infectious measles virus (MeV) that results in both viremia and lymphopenia. Lymphocyte counts recover shortly after the disappearance of measles-associated rash, but immunosuppression can persist for months to years after infection, resulting in increased incidence of secondary infections. Animal models and in vitro studies have proposed various immunological factors underlying this prolonged immune impairment, but the precise mechanisms operating in humans are unknown. Using B cell receptor (BCR) sequencing of human peripheral blood lymphocytes before and after MeV infection, we identified two immunological consequences from measles underlying immunosuppression: (i) incomplete reconstitution of the naïve B cell pool leading to immunological immaturity and (ii) compromised immune memory to previously encountered pathogens due to depletion of previously expanded B memory clones. Using a surrogate model of measles in ferrets, we investigated the clinical consequences of morbillivirus infection and demonstrated a depletion of vaccine-acquired immunity to influenza virus, leading to a compromised immune recall response and increased disease severity after secondary influenza virus challenge. Our results show that MeV infection causes changes in naïve and memory B lymphocyte diversity that persist after the resolution of clinical disease and thus contribute to compromised immunity to previous infections or vaccinations. This work highlights the importance of MeV vaccination not only for the control of measles but also for the maintenance of herd immunity to other pathogens, which can be compromised after MeV infection.
Collapse
Affiliation(s)
| | - Bevan Sawatsky
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines and DZIF TTU Emerging Infections, Langen, Germany
| | - Alvin X Han
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Brigitta M Laksono
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Lisa Walz
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines and DZIF TTU Emerging Infections, Langen, Germany
| | - Edyth Parker
- Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Kathrin Pieper
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Carl A Anderson
- Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Paul Kellam
- Department of Medicine, Division of Infectious Diseases, Imperial College Faculty of Medicine, Wright Fleming Institute, St Mary's Campus, London, UK.,Kymab Ltd., The Bennet Building, Babraham Research Campus, Cambridge, UK
| | - Veronika von Messling
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines and DZIF TTU Emerging Infections, Langen, Germany
| | - Rik L de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Colin A Russell
- Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
27
|
Sato Y, Ochiai D, Abe Y, Masuda H, Fukutake M, Ikenoue S, Kasuga Y, Shimoda M, Kanai Y, Tanaka M. Prophylactic therapy with human amniotic fluid stem cells improved survival in a rat model of lipopolysaccharide-induced neonatal sepsis through immunomodulation via aggregates with peritoneal macrophages. Stem Cell Res Ther 2020; 11:300. [PMID: 32690106 PMCID: PMC7370504 DOI: 10.1186/s13287-020-01809-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/06/2020] [Accepted: 07/03/2020] [Indexed: 01/15/2023] Open
Abstract
Background Despite recent advances in neonatal care, sepsis remains a leading cause of mortality in neonates. Mesenchymal stem cells derived from various tissues, such as bone marrow, umbilical cord, and adipose tissue, have beneficial effects on adult sepsis. Although human amniotic fluid stem cells (hAFSCs) have mesenchymal stem cell properties, the efficacy of hAFSCs on neonatal sepsis is yet to be elucidated. This study aimed to investigate the therapeutic potential of hAFSCs on neonatal sepsis using a rat model of lipopolysaccharide (LPS)-induced sepsis. Methods hAFSCs were isolated as CD117-positive cells from human amniotic fluid. Three-day-old rat pups were intraperitoneally treated with LPS to mimic neonatal sepsis. hAFSCs were administered either 3 h before or at 0, 3, or 24 h after LPS exposure. Serum inflammatory cytokine levels, gene expression profiles from spleens, and multiple organ damage were analyzed. hAFSC localization was determined in vivo. In vitro LPS stimulation tests were performed using neonatal rat peritoneal macrophages co-cultured with hAFSCs in a cell-cell contact-dependent/independent manner. Immunoregulation in the spleen was determined using a DNA microarray analysis. Results Prophylactic therapy with hAFSCs improved survival in the LPS-treated rats while the hAFSCs transplantation after LPS exposure did not elicit a therapeutic response. Therefore, hAFSC pretreatment was used for all subsequent studies. Inflammatory cytokine levels were elevated after LPS injection, which was attenuated by hAFSC pretreatment. Subsequently, inflammation-induced damages in the brain, lungs, and liver were ameliorated. hAFSCs aggregated with peritoneal macrophages and/or transiently accumulated in the liver, mesentery, and peritoneum. Paracrine factors released by hAFSCs induced M1-M2 macrophage polarization in a cell-cell contact-independent manner. Direct contact between hAFSCs and peritoneal macrophages further enhanced the polarization. Microarray analysis of the spleen showed that hAFSC pretreatment reduced the expression of genes involved in apoptosis and inflammation and subsequently suppressed toll-like receptor 4 signaling pathways. Conclusions Prophylactic therapy with hAFSCs improved survival in a rat model of LPS-induced neonatal sepsis. These effects might be mediated by a phenotypic switch from M1 to M2 in peritoneal macrophages, triggered by hAFSCs in a cell-cell contact-dependent/independent manner and the subsequent immunomodulation of the spleen.
Collapse
Affiliation(s)
- Yu Sato
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Daigo Ochiai
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan.
| | - Yushi Abe
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Hirotaka Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Marie Fukutake
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Satoru Ikenoue
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Yoshifumi Kasuga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| |
Collapse
|
28
|
Chen YS, Chen IB, Pham G, Shao TY, Bangar H, Way SS, Haslam DB. IL-17-producing γδ T cells protect against Clostridium difficile infection. J Clin Invest 2020; 130:2377-2390. [PMID: 31990686 PMCID: PMC7190913 DOI: 10.1172/jci127242] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 01/17/2020] [Indexed: 01/04/2023] Open
Abstract
Colitis caused by Clostridium difficile infection is a growing cause of human morbidity and mortality, especially after antibiotic use in health care settings. The natural immunity of newborn infants and protective host immune mediators against C. difficile infection are not fully understood, with data suggesting that inflammation can be either protective or pathogenic. Here, we show an essential role for IL-17A produced by γδ T cells in host defense against C. difficile infection. Fecal extracts from children with C. difficile infection showed increased IL-17A and T cell receptor γ chain expression, and IL-17 production by intestinal γδ T cells was efficiently induced after infection in mice. C. difficile-induced tissue inflammation and mortality were markedly increased in mice deficient in IL-17A or γδ T cells. Neonatal mice, with naturally expanded RORγt+ γδ T cells poised for IL-17 production were resistant to C. difficile infection, whereas elimination of γδ T cells or IL-17A each efficiently overturned neonatal resistance against infection. These results reveal an expanded role for IL-17-producing γδ T cells in neonatal host defense against infection and provide a mechanistic explanation for the clinically observed resistance of infants to C. difficile colitis.
Collapse
MESH Headings
- Animals
- Clostridioides difficile/immunology
- Enterocolitis, Pseudomembranous/genetics
- Enterocolitis, Pseudomembranous/immunology
- Enterocolitis, Pseudomembranous/pathology
- Enterocolitis, Pseudomembranous/prevention & control
- Female
- Humans
- Interleukin-17/genetics
- Interleukin-17/immunology
- Male
- Mice
- Mice, Knockout
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
Collapse
Affiliation(s)
- Yee-Shiuan Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Iuan-Bor Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Giang Pham
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tzu-Yu Shao
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Hansraj Bangar
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sing Sing Way
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - David B. Haslam
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
29
|
Angulo M, Reyes-Becerril M, Cepeda-Palacios R, Angulo C. Oral administration of Debaryomyces hansenii CBS8339-β-glucan induces trained immunity in newborn goats. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 105:103597. [PMID: 31883447 DOI: 10.1016/j.dci.2019.103597] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/26/2019] [Accepted: 12/21/2019] [Indexed: 06/10/2023]
Abstract
Beta-glucans from yeast can induce trained immunity in in vitro and in vivo models. Intraperitoneal doses of β-glucans in mammals have shown to induce trained immunity, but the training effects of orally administering β-glucans are unknown. Newborn goats are susceptible to infections in the neonatal stage, so the induction of trained immunity could improve animal survival. This study aimed to describe the in vitro effects of immunological training by β-glucan from Debaryomyces hansenii (β-Dh) on caprine monocytes, as well as its in vivo effects using oral doses on newborn goats upon challenge with lipopolysaccharide (LPS). Hence in vitro, goat monocytes trained with β-Dh up-regulated the gene expression of macrophage surface markers (CD11b and F4/80) whereas enhanced cell survival and high phagocytic ability was found upon LPS challenge. In the in vivo experiment, newborn goats stimulated with two doses (day -7 and - 4) of β-Dh (50 mg/kg) and challenged (day 0) with LPS showed an increase in respiratory burst activity, IL-1β, IL-6, and TNFα production in plasma, and transcription of the macrophage surface markers. This study has demonstrated for the first time that trained immunity was induced with oral doses of β-glucan upon LPS challenge in mammals using newborn goats.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste (CIBNOR), Av. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Martha Reyes-Becerril
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste (CIBNOR), Av. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Ramón Cepeda-Palacios
- Laboratorio de Sanidad Animal, Universidad Autónoma de Baja California Sur, Carretera al Sur km. 5.5, Col. Mezquitito, La Paz, B.C.S., 23080, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste (CIBNOR), Av. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico.
| |
Collapse
|
30
|
Khalil RH, Al-Humadi N. Types of acute phase reactants and their importance in vaccination. Biomed Rep 2020; 12:143-152. [PMID: 32190302 PMCID: PMC7054702 DOI: 10.3892/br.2020.1276] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023] Open
Abstract
Vaccines are considered to be one of the most cost-effective life-saving interventions in human history. The body's inflammatory response to vaccines has both desired effects (immune response), undesired effects [(acute phase reactions (APRs)] and trade-offs. Trade-offs are more potent immune responses which may be potentially difficult to separate from potent acute phase reactions. Thus, studying acute phase proteins (APPs) during vaccination may aid our understanding of APRs and homeostatic changes which can result from inflammatory responses. Depending on the severity of the response in humans, these reactions can be classified as major, moderate or minor. In this review, types of APPs and their importance in vaccination will be discussed.
Collapse
Affiliation(s)
- Rafaat H Khalil
- Department of Biology, College of Science and Technology, Florida Agricultural and Mechanical University, Tallahassee, FL 32307, USA
| | - Nabil Al-Humadi
- Office of Vaccines, Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD 20993, USA
| |
Collapse
|
31
|
Short SS, Papillon SC, Ford HR. Sepsis. PEDIATRIC SURGERY 2020:461-476. [DOI: 10.1007/978-3-662-43588-5_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
32
|
Gródecka-Szwajkiewicz D, Ulańczyk Z, Zagrodnik E, Łuczkowska K, Rogińska D, Kawa MP, Stecewicz I, Safranow K, Ustianowski P, Szymański S, Machaliński B. Comparative Analysis of Global Gene Expression and Complement Components Levels in Umbilical Cord Blood from Preterm and Term Neonates: Implications for Significant Downregulation of Immune Response Pathways related to Prematurity. Int J Med Sci 2020; 17:1840-1853. [PMID: 32714087 PMCID: PMC7378668 DOI: 10.7150/ijms.46339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/03/2020] [Indexed: 01/03/2023] Open
Abstract
Background: Preterm birth is the most frequent cause of neonatal death, but its aetiology remains unclear. It has been suggested that the imbalance of immunological mechanisms responsible for maintaining pregnancy is contributing to preterm birth pathogenesis. We aimed to investigate global gene expression and the levels of several complement system components in umbilical cord blood samples from preterm neonates and compare them to term newborns. We sought to examine how differentially expressed genes could affect various immune-related pathways that are believed to be crucial factors in preterm birth. Material and methods: We enrolled 27 preterm infants (<37 weeks GA) and 52 term infants (>37 weeks GA), from which umbilical cord blood samples were collected. From these samples, peripheral blood mononuclear cells were isolated and subsequent RNA isolation was performed. We used Affymetrix Human Gene 2.1 ST Array Strip for microarray experiment and DAVID resources for bioinformatics analysis of the obtained data. Concentrations of C2, C3a, C5/C5a, C9, FactorD, Properdin were measured in umbilical cord blood plasma samples using multiplex fluorescent bead-based immunoassays using Luminex technology. Results: The levels of C3a and C5/5a were significantly elevated in preterm neonates compared to term babies, whereas C9 concentration was evidently increased in term infants. The expression of 250 genes was upregulated at least 2-fold and 3781 genes were downregulated at least 2-fold in preterm neonates in comparison with term infants. Functional annotation analysis revealed that in preterm infants in comparison to term babies there was a significant downregulation of genes encoding several Toll-like receptors, interleukins and genes involved in major signalling pathways (e.g. NF-κB, MAPK, TNF, Notch, JAK) and vital cellular processes (e.g. intracellular signal transduction, protein ubiquitination, protein transport, RNA splicing, DNA-templated transcription). Conclusions: Preterm birth results in immediate and long-term complications. Our results indicate that infants born prematurely show significant differences in complement components concentration and a downregulation of over 3,000 genes, involved mainly in various immune-related pathways, including innate immune response, phagocytosis and TLR function, when compared to full-term babies. Further studies on larger cohorts are needed to elucidate the role of immunity in prematurity.
Collapse
Affiliation(s)
| | - Zofia Ulańczyk
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Edyta Zagrodnik
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Miłosz P Kawa
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Iwona Stecewicz
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Przemysław Ustianowski
- Department of Perinatology, Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Sławomir Szymański
- Department of Obstetrics and Pathology of Pregnancy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
33
|
Tsafaras GP, Ntontsi P, Xanthou G. Advantages and Limitations of the Neonatal Immune System. Front Pediatr 2020; 8:5. [PMID: 32047730 PMCID: PMC6997472 DOI: 10.3389/fped.2020.00005] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/07/2020] [Indexed: 12/30/2022] Open
Abstract
During early post-natal life, neonates must adjust to the transition from the sheltered intra-uterine environment to the microbe-laden external world, wherein they encounter a constellation of antigens and the colonization by the microbiome. At this vulnerable stage, neonatal immune responses are considered immature and present significant differences to those of adults. Pertinent to innate immunity, functional and quantitative deficiencies in antigen-presenting cells and phagocytes are often documented. Exposure to environmental antigens and microbial colonization is associated with epigenetic immune cell reprogramming and activation of effector and regulatory mechanisms that ensure age-depended immune system maturation and prevention of tissue damage. Moreover, neonatal innate immune memory has emerged as a critical mechanism providing protection against infectious agents. Still, in neonates, inexperience to antigenic exposure, along with enhancement of tissue-protective immunosuppressive mechanisms are often associated with severe immunopathological conditions, including sepsis and neurodevelopmental disorders. Despite significant advances in the field, adequate vaccination in newborns is still in its infancy due to elemental restrictions associated also with defective immune responses. In this review, we provide an overview of neonatal innate immune cells, highlighting phenotypic and functional disparities with their adult counterparts. We also discuss the effects of epigenetic modifications and microbial colonization on the regulation of neonatal immunity. A recent update on mechanisms underlying dysregulated neonatal innate immunity and linked infectious and neurodevelopmental diseases is provided. Understanding of the mechanisms that augment innate immune responsiveness in neonates may facilitate the development of improved vaccination protocols that can protect against pathogens and organ damage.
Collapse
Affiliation(s)
- George P Tsafaras
- Cellular Immunology Lab, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Polyxeni Ntontsi
- Second Respiratory Medicine Department, 'Attikon' University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Georgina Xanthou
- Cellular Immunology Lab, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
34
|
Alkie TN, Yitbarek A, Hodgins DC, Kulkarni RR, Taha-Abdelaziz K, Sharif S. Development of innate immunity in chicken embryos and newly hatched chicks: a disease control perspective. Avian Pathol 2019; 48:288-310. [PMID: 31063007 DOI: 10.1080/03079457.2019.1607966] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Newly hatched chickens are confronted by a wide array of pathogenic microbes because their adaptive immune defences have limited capabilities to control these pathogens. In such circumstances, and within this age group, innate responses provide a degree of protection. Moreover, as the adaptive immune system is relatively naïve to foreign antigens, synergy with innate defences is critical. This review presents knowledge on the ontogeny of innate immunity in chickens pre-hatch and early post-hatch and provides insights into possible interventions to modulate innate responses early in the life of the bird. As in other vertebrate species, the chicken innate immune system which include cellular mediators, cytokine and chemokine repertoires and molecules involved in antigen detection, develop early in life. Comparison of innate immune systems in newly hatched chickens and mature birds has revealed differences in magnitude and quality, but responses in younger chickens can be boosted using innate immune system modulators. Functional expression of pattern recognition receptors and several defence molecules by innate immune system cells of embryos and newly hatched chicks suggests that innate responses can be modulated at this stage of development to combat pathogens. Improved understanding of innate immune system ontogeny and functionality in chickens is critical for the implementation of sound and safe interventions to provide long-term protection against pathogens. Next-generation tools for studying genetic and epigenetic regulation of genes, functional metagenomics and gene knockouts can be used in the future to explore and dissect the contributions of signalling pathways of innate immunity and to devise more efficacious disease control strategies.
Collapse
Affiliation(s)
- Tamiru N Alkie
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Alexander Yitbarek
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Douglas C Hodgins
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Raveendra R Kulkarni
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Khaled Taha-Abdelaziz
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada.,b Pathology Department, Faculty of Veterinary Medicine , Beni-Suef University , Beni-Suef , Egypt
| | - Shayan Sharif
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| |
Collapse
|
35
|
Wisgrill L, Wessely I, Netzl A, Pummer L, Sadeghi K, Spittler A, Berger A, Förster‐Waldl E. Diminished secretion and function of IL-29 is associated with impaired IFN-α response of neonatal plasmacytoid dendritic cells. J Leukoc Biol 2019; 106:1177-1185. [PMID: 31211458 PMCID: PMC6852569 DOI: 10.1002/jlb.4a0518-189r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 03/20/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are key players in the antiviral immune response and type III IFNs such as IL-29 appear to play a pivotal role in pDC function. Pronounced susceptibility to viral infections in neonates is partly resulting from diminished antiviral immune mechanisms. Accordingly, the aim of the present study was to investigate the impact of IL-29 in the altered immune response of neonatal pDCs. PBMCs of adult and term newborns were stimulated with CpG-ODN2216 in the presence or absence of IL-29 and assessed for IFN-α production, downstream-signaling, and activation marker expression. A significantly lower IL-29 production after TLR9-specific stimulation was demonstrated in neonatal pDCs. IL-29 enhanced the IFN-α production of pDCs in adults compared to newborns. Newborn pDCs displayed a significantly lower surface expression of IL-10 and IL-28Rα receptor resulting in diminished STAT1 and IRF7 activation. Interestingly, concomitant stimulation with CpG-ODN2216/IL-29 had no impact on the expression of surface activation and maturation markers of pDCs in neither population. The diminished antiviral immune response of neonatal pDCs is associated with reduced production and cellular responses toward IL-29. Potential therapeutic agents enhancing the IL-29 response in neonatal pDCs possibly augment viral protection in newborns.
Collapse
Affiliation(s)
- Lukas Wisgrill
- Department of Pediatrics and Adolescent MedicineDivision of NeonatologyPediatric Intensive Care & NeuropediatricsMedical University of ViennaViennaAustria
| | - Isabelle Wessely
- Department of Pediatrics and Adolescent MedicineDivision of NeonatologyPediatric Intensive Care & NeuropediatricsMedical University of ViennaViennaAustria
| | - Antonia Netzl
- Department of Pediatrics and Adolescent MedicineDivision of NeonatologyPediatric Intensive Care & NeuropediatricsMedical University of ViennaViennaAustria
| | - Linda Pummer
- Department of Pediatrics and Adolescent MedicineDivision of NeonatologyPediatric Intensive Care & NeuropediatricsMedical University of ViennaViennaAustria
| | - Kambis Sadeghi
- Department of Pediatrics and Adolescent MedicineDivision of NeonatologyPediatric Intensive Care & NeuropediatricsMedical University of ViennaViennaAustria
| | - Andreas Spittler
- Department of Surgery & Core Facility Flow CytometryMedical University of ViennaViennaAustria
| | - Angelika Berger
- Department of Pediatrics and Adolescent MedicineDivision of NeonatologyPediatric Intensive Care & NeuropediatricsMedical University of ViennaViennaAustria
| | - Elisabeth Förster‐Waldl
- Department of Pediatrics and Adolescent MedicineDivision of NeonatologyPediatric Intensive Care & NeuropediatricsMedical University of ViennaViennaAustria
- Center for Congenital ImmunodeficienciesMedical University of ViennaViennaAustria
| |
Collapse
|
36
|
Şah İpek M. Neonatal Bacterial Meningitis. NEONATAL MEDICINE 2019. [DOI: 10.5772/intechopen.87118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
37
|
Biondi EA, McCulloh R, Staggs VS, Garber M, Hall M, Arana J, Barsotti B, Natt BC, Schroeder AR, Schroeder L, Wylie T, Ralston SL. Reducing Variability in the Infant Sepsis Evaluation (REVISE): A National Quality Initiative. Pediatrics 2019; 144:peds.2018-2201. [PMID: 31434688 DOI: 10.1542/peds.2018-2201] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/30/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Substantial variability exists in the care of febrile, well-appearing infants. We aimed to assess the impact of a national quality initiative on appropriate hospitalization and length of stay (LOS) in this population. METHODS The initiative, entitled Reducing Variability in the Infant Sepsis Evaluation (REVISE), was designed to standardize care for well-appearing infants ages 7 to 60 days evaluated for fever without an obvious source. Twelve months of baseline and 12 months of implementation data were collected from emergency departments and inpatient units. Ill-appearing infants and those with comorbid conditions were excluded. Participating sites received change tools, run charts, a mobile application, live webinars, coaching, and a LISTSERV. Analyses were performed via statistical process control charts and interrupted time series regression. The 2 outcome measures were the percentage of hospitalized infants who were evaluated and hospitalized appropriately and the percentage of hospitalized infants who were discharged with an appropriate LOS. RESULTS In total, 124 hospitals from 38 states provided data on 20 570 infants. The median site improvement in percentages of infants who were evaluated and hospitalized appropriately and in those with appropriate LOS was 5.3% (interquartile range = -2.5% to 13.7%) and 15.5% (interquartile range = 2.9 to 31.3), respectively. Special cause variation toward the target was identified for both measures. There was no change in delayed treatment or missed bacterial infections (slope difference 0.1; 95% confidence interval, -8.3 to 9.1). CONCLUSIONS Reducing Variability in the Infant Sepsis Evaluation noted improvement in key aspects of febrile infant management. Similar projects may be used to improve care in other clinical conditions.
Collapse
Affiliation(s)
- Eric A Biondi
- Department of Pediatrics, Johns Hopkins Children's Center, Baltimore, Maryland;
| | | | - Vincent S Staggs
- Children's Mercy Hospital, Kansas City, Missouri.,University of Missouri-Kansas City, Kansas City, Missouri
| | - Matthew Garber
- College of Medicine - Jacksonville, University of Florida, Jacksonville, Florida
| | - Matt Hall
- Children's Hospital Association, Overland Park, Kansas
| | - Julia Arana
- East Tennessee Children's Hospital, Knoxville, Tennessee
| | - Benj Barsotti
- Virginia Mason Memorial Hospital, Yakima, Washington
| | - Beth C Natt
- Connecticut Children's Medical Center, Hartford, Connecticut
| | - Alan R Schroeder
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, California; and
| | | | - Todd Wylie
- College of Medicine - Jacksonville, University of Florida, Jacksonville, Florida
| | - Shawn L Ralston
- Department of Pediatrics, Children's Hospital at Dartmouth-Hitchcock, Lebanon, New Hampshire
| | | |
Collapse
|
38
|
Plotnikov EY, Pevzner IB, Zorova LD, Chernikov VP, Prusov AN, Kireev II, Silachev DN, Skulachev VP, Zorov DB. Mitochondrial Damage and Mitochondria-Targeted Antioxidant Protection in LPS-Induced Acute Kidney Injury. Antioxidants (Basel) 2019; 8:antiox8060176. [PMID: 31197113 PMCID: PMC6617298 DOI: 10.3390/antiox8060176] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/16/2022] Open
Abstract
Induced and frequently unwanted alterations in the mitochondrial structure and functions are a key component of the pathological cascade in many kidney pathologies, including those associated with acute damage. One of the principal pathogenic elements causing mitochondrial dysfunction in Acute Kidney Injury (AKI) is oxidative stress. After ischemia and nephrotoxic action of drugs, sepsis and systemic inflammation are the most frequent causes of AKI. As the kidney suffers from oxidative stress during sepsis, one of the most promising approaches to alleviate such damaging consequences is the use of antioxidants. Considering administration of lipopolysaccharide (LPS) as a model of sepsis, we demonstrate that the mitochondria of neonatal renal tissue are severely affected by LPS-induced AKI, with pathological ultrastructural changes observed in both the mitochondria of the renal tubular epithelium and the vascular endothelium. Upon mitochondrial damage, we evaluated the effect of the mitochondria-targeted antioxidant plastoquinol decylrhodamine 19 (SkQR1) on the development of acute renal failure in newborn rats associated with systemic inflammation induced by the administration of LPS. We found that SkQR1 administration 3 h before LPS led to decreased urinal expression of the AKI marker neutrophil gelatinase-associated lipocalin 2 (NGAL), in addition to a decrease in urea and creatinine levels in the blood. Additionally, an observed impairment of proliferative activity in the neonatal kidney caused by LPS treatment was also prevented by the treatment of rat pups with SkQR1. Thus, one of the key events for renal tissue damage in neonatal sepsis is an alteration in the structure and function of the mitochondria and the mitochondria-targeted antioxidant SkQR1 is an effective nephroprotective agent, which protects the neonatal kidney from sepsis-induced AKI.
Collapse
Affiliation(s)
- Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 11797, Russia.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Irina B Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 11797, Russia.
| | - Ljubava D Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 11797, Russia.
| | | | - Andrey N Prusov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Igor I Kireev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Denis N Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 11797, Russia.
| | - Vladimir P Skulachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Dmitry B Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia.
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 11797, Russia.
| |
Collapse
|
39
|
Kuti BP, Ogunlesi TA, Oduwole O, Oringanje C, Udoh EE, Meremikwu MM. Hand hygiene for the prevention of infections in neonates. Cochrane Database Syst Rev 2019. [DOI: 10.1002/14651858.cd013326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Bankole Peter Kuti
- Obafemi Awolowo University; Department of Paediatrics and Child Health; Ile-Ife Osun State Nigeria
| | - Tinuade A Ogunlesi
- Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University; Department of Paediatrics (Neonatal Unit); Sagamu Ogun State Nigeria 121001NG
| | - Olabisi Oduwole
- Achievers University; Department of Medical Laboratory Science; Owo Nigeria
| | - Chukwudi Oringanje
- University of Calabar Teaching Hospital (ITDR/P); Institute of Tropical Diseases Research and Prevention; Moore Road Calabar Cross River Nigeria
| | - Ekong E Udoh
- University of Calabar Teaching Hospital; Department of Paediatrics; Calabar Nigeria
| | - Martin M Meremikwu
- University of Calabar Teaching Hospital; Department of Paediatrics; Calabar Nigeria
| |
Collapse
|
40
|
Gilbert R, Brown M, Rainford N, Donohue C, Fraser C, Sinha A, Dorling J, Gray J, McGuire W, Gamble C, Oddie SJ. Antimicrobial-impregnated central venous catheters for prevention of neonatal bloodstream infection (PREVAIL): an open-label, parallel-group, pragmatic, randomised controlled trial. THE LANCET CHILD & ADOLESCENT HEALTH 2019; 3:381-390. [PMID: 31040096 DOI: 10.1016/s2352-4642(19)30114-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/01/2019] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Bloodstream infection is associated with high mortality and serious morbidity in preterm babies. Evidence from clinical trials shows that antimicrobial-impregnated central venous catheters (CVCs) reduce catheter-related bloodstream infection in adults and children receiving intensive care, but there is a paucity of similar evidence for babies receiving neonatal intensive care. METHODS This open-label, parallel-group, pragmatic, randomised controlled trial was done in 18 neonatal intensive care units in England. Newborn babies who needed a peripherally inserted CVC (PICC) were allocated randomly (1:1) to receive either a PICC impregnated with miconazole and rifampicin or a standard (non-antimicrobial-impregnated) PICC. Random allocation was done with a web-based program, which was centrally controlled to ensure allocation concealment. Randomisation sequences were computer-generated in random blocks of two and four, and stratified by site. Masking of clinicians to PICC allocation was impractical because rifampicin caused brown staining of the antimicrobial-impregnated PICC. However, participant inclusion in analyses and occurrence of outcome events were determined following an analysis plan that was specified before individuals saw the unblinded data. The primary outcome was the time from random allocation to first microbiologically confirmed bloodstream or cerebrospinal fluid (CSF) infection between 24 h after randomisation and 48 h after PICC removal or death. We analysed outcome data according to the intention-to-treat principle. We excluded babies for whom a PICC was not inserted from safety analyses, as these analyses were done with groups defined by the PICC used. This trial is registered with ISRCTN, number 81931394. FINDINGS Between Aug 12, 2015, and Jan 11, 2017, we randomly assigned 861 babies (754 [88%] born before 32 weeks of gestation) to receive an antimicrobial-impregnated PICC (430 babies) or standard PICC (431 babies). The median time to PICC removal was 8·20 days (IQR 4·77-12·13) in the antimicrobial-impregnated PICC group versus 7·86 days (5·00-12·53) days in the standard PICC group (hazard ratio [HR] 1·03, 95% CI 0·89-1·18, p=0·73), with 46 (11%) of 430 babies versus 44 (10%) of 431 babies having a microbiologically confirmed bloodstream or CSF infection. The time from random allocation to first bloodstream or CSF infection was similar between the two groups (HR 1·11, 95% CI 0·73-1·67, p=0·63). Secondary outcomes relating to infection, rifampicin resistance in positive blood or CSF cultures, mortality, clinical outcomes at neonatal unit discharge, and time to PICC removal were similar between the two groups, although rifampicin resistance in positive cultures of PICC tips was higher in the antimicrobial-impregnated PICC group (relative risk 3·51, 95% CI 1·16-10·57, p=0·018). 60 adverse events were reported from 49 (13%) patients in the antimicrobial-impregnated PICC group and 50 events from 45 (10%) babies in the standard PICC group. INTERPRETATION We found no evidence of benefit or harm associated with miconazole and rifampicin-impregnated PICCs compared with standard PICCs for newborn babies. Future research should focus on other types of antimicrobial impregnation of PICCs and alternative approaches for preventing infection. FUNDING UK National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Ruth Gilbert
- UCL Great Ormond Street Institute of Child Health, London, UK; Health Data Research UK, London, UK.
| | - Michaela Brown
- Clinical Trials Research Centre, Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Naomi Rainford
- Clinical Trials Research Centre, Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Chloe Donohue
- Clinical Trials Research Centre, Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Caroline Fraser
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Ajay Sinha
- Barts Health NHS Trust, London, UK; Blizard Institute, Queen Mary University of London, London, UK
| | - Jon Dorling
- Division of Neonatal-Perinatal Medicine, Dalhousie University IWK Health Centre, Halifax, NS, Canada
| | - Jim Gray
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Carrol Gamble
- Clinical Trials Research Centre, Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Sam J Oddie
- Centre for Reviews and Dissemination, University of York, York, UK; Bradford Institute for Health Research, Bradford Royal Infirmary, Bradford, UK
| | | |
Collapse
|
41
|
Tang BH, Wu YE, Kou C, Qi YJ, Qi H, Xu HY, Leroux S, Huang X, Zhou Y, Zheng Y, Jacqz-Aigrain E, Shen AD, Zhao W. Population Pharmacokinetics and Dosing Optimization of Amoxicillin in Neonates and Young Infants. Antimicrob Agents Chemother 2019; 63:e02336-18. [PMID: 30509939 PMCID: PMC6355568 DOI: 10.1128/aac.02336-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/27/2018] [Indexed: 12/19/2022] Open
Abstract
Amoxicillin is widely used to treat bacterial infections in neonates. However, considerable intercenter variability in dosage regimens of antibiotics exists in clinical practice. The pharmacokinetics of amoxicillin has been described in only a few preterm neonates. Thus, we aimed to evaluate the population pharmacokinetics of amoxicillin through a large sample size covering the entire age range of neonates and young infants and to establish evidence-based dosage regimens based on developmental pharmacokinetics-pharmacodynamics. This is a prospective, multicenter, pharmacokinetic study using an opportunistic sampling design. Amoxicillin plasma concentrations were determined using high-performance liquid chromatography. Population pharmacokinetic analysis was performed using NONMEM. A total of 224 pharmacokinetic samples from 187 newborns (postmenstrual age range, 28.4 to 46.3 weeks) were available for analysis. A two-compartment model with first-order elimination was used to describe population pharmacokinetics. Covariate analysis showed that current weight, postnatal age, and gestational age were significant covariates. The final model was further validated for predictive performance in an independent cohort of patients. Monte Carlo simulation demonstrated that for early-onset sepsis, the currently used dosage regimen (25 mg/kg twice daily [BID]) resulted in 99.0% of premature neonates and 87.3% of term neonates achieving the pharmacodynamic target (percent time above MIC), using a MIC breakpoint of 1 mg/liter. For late-onset sepsis, 86.1% of premature neonates treated with 25 mg/kg three times a day (TID) and 79.0% of term neonates receiving 25 mg/kg four times a day (QID) reached the pharmacodynamic target, using a MIC breakpoint of 2 mg/liter. The population pharmacokinetics of amoxicillin was assessed in neonates and young infants. A dosage regimen was established based on developmental pharmacokinetics-pharmacodynamics.
Collapse
Affiliation(s)
- Bo-Hao Tang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yue-E Wu
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Chen Kou
- Department of Neonatology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yu-Jie Qi
- Neonatal Intensive Care Unit, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hui Qi
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hai-Yan Xu
- Department of Neonatology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | | | - Xin Huang
- Department of Pharmacy, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Evelyne Jacqz-Aigrain
- Department of Pediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, APHP, Paris, France
| | - A-Dong Shen
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Zhao
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
42
|
Fraser C, Harron K, Dalton L, Gilbert R, Oddie SJ. Variation in infection prevention practices for peripherally inserted central venous catheters: A survey of neonatal units in England and Wales. PLoS One 2018; 13:e0204894. [PMID: 30383769 PMCID: PMC6211675 DOI: 10.1371/journal.pone.0204894] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 09/17/2018] [Indexed: 12/29/2022] Open
Abstract
Background There is uncertainty about the variation in infection prevention practices for central venous catheters (CVC) in neonatal units (NNUs) and how practices relate to national guidance. Aim To evaluate evidence supporting infection prevention practices for CVCs recommended in national guidelines and to compare with reported practices for peripherally inserted central catheters (PICC), a type of CVC widely used in NNUs. Design We searched national guidelines for neonates and children to identify infection prevention practices for CVCs and conducted an overview of studies to determine the quality of evidence underpinning recommendations. We surveyed 134 NNUs in England and Wales to ascertain reported practice. Results We found low quality evidence supporting CVC care bundles and use of 2% alcoholic chlorhexidine to decontaminate catheter ports and skin before insertion. Moderate quality evidence supported recommendations against routinely replacing CVCs and against chlorhexidine-impregnated dressings. 90% (44/49) of NICUs and 40% (34/85) of LNUs responded. 66% (48/73) of NNUs reported using CVC care bundles for insertion; 62% (45/73) used bundles for maintenance. 63% (32/51) of those using bundles reported monitoring adherence. 85% (61/72) of NNUs did not routinely replace PICCs and 89% (63/71) did not use chlorhexidine-impregnated dressings. Antiseptic use varied with alcoholic 2% chlorhexidine used for skin preparation in 33% (23/71) of NNUs and for catheter ports in 52% (37/71). Conclusions Lack of consistency across NNUs in antiseptic use and low rates of reported CVC care bundle use may reflect the low quality of evidence of the effectiveness and safety of these interventions in NNUs. Clinical trials are needed to quantify benefits and harms of infection prevention practices in NNUs.
Collapse
Affiliation(s)
- Caroline Fraser
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- * E-mail:
| | - Katie Harron
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Laura Dalton
- Bradford Neonatology, Bradford Royal Infirmary, Bradford, United Kingdom
| | - Ruth Gilbert
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Sam J. Oddie
- Bradford Neonatology, Bradford Royal Infirmary, Bradford, United Kingdom
- Centre for reviews and dissemination, University of York, York, United Kingdom
| | | |
Collapse
|
43
|
Deficiency of receptor-interacting protein kinase 3 (RIPK3) attenuates inflammation and organ injury in neonatal sepsis. J Pediatr Surg 2018; 53:1699-1705. [PMID: 29248164 PMCID: PMC5966335 DOI: 10.1016/j.jpedsurg.2017.11.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/10/2017] [Accepted: 11/18/2017] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Sepsis is the third leading cause of morbidity and mortality in neonates. Sepsis in neonates is characterized as the systemic inflammation owing to infection within the first 28days after birth. The molecular mechanism causing the exaggerated inflammation phenotype in neonates has not been completely elucidated. Receptor interacting protein kinase 3 (RIPK3) is a protein identified as a mediator in programmed necrosis or necroptosis. We hypothesize that RIPK3 could be responsible for the inflammatory response in neonates and that deficiency in the RIPK3 protein attenuates inflammation and organ injury in neonatal sepsis. METHODS Male and female C57BL6 wild-type (WT) and RIPK3 knock-out (KO) newborn mice aged 5-7days (3-4g body weight) were injected intraperitoneally with 0.9mg/g cecal slurry (CS). At 10h after injection, the newborns were euthanized and blood, the lungs and gut tissues were collected. RESULTS At 10h after CS injection, serum cytokines IL-6 and IL-1β in the WT mice were increased by 511- and 43-fold whereas in KO mice, these levels were increased by 166-fold and 22-fold, respectively. Lung IL-1β in the WT mice increased by 7-fold after CS injection whereas only a 4-fold increase was seen in the KO mice. In the lungs of CS injected KO mice, the injury score, MIP-2 mRNA, myeloperoxidase (MPO) activity and TUNEL staining were significantly reduced by 76%, 70%, 26% and 74%, respectively compared to the CS WT mice. Gut TUNEL staining was also reduced by 80%. CONCLUSION The deficiency in RIPK3 attenuated serum and lung cytokines, lung injury and neutrophil infiltration and lung and gut apoptosis. These data suggest that RIPK3, in part, is responsible for the systemic inflammatory response in neonatal sepsis.
Collapse
|
44
|
SslE (YghJ), a Cell-Associated and Secreted Lipoprotein of Neonatal Septicemic Escherichia coli, Induces Toll-Like Receptor 2-Dependent Macrophage Activation and Proinflammation through NF-κB and MAP Kinase Signaling. Infect Immun 2018; 86:IAI.00399-18. [PMID: 29891541 DOI: 10.1128/iai.00399-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
SslE (YghJ), a cell surface-associated and secreted lipoprotein, was identified as a potential vaccine candidate for extraintestinal pathogenic Escherichia coli, providing nearly complete protection from sepsis in a mouse model. We earlier found that SslE from neonatal septicemic E. coli could trigger the secretion of various proinflammatory cytokines in murine macrophages, the signaling pathway of which is still obscure. In this study, we showed that SslE specifically binds to Toll-like receptor 2 (TLR2)/TLR1 heterodimers and recruits downstream adaptors MyD88, TIRAP, and TRAF6. In addition, SslE stimulates nuclear translocation of NF-κB and activates different mitogen-activated protein (MAP) kinase signaling cascades specific to the secretion of each cytokine in murine macrophages, which becomes impaired in TLR2 small interfering RNA (siRNA)-transfected cells and in cells blocked with a monoclonal antibody (MAb) against TLR2, suggesting the involvement of TLR2 in NF-κB and MAP kinase activation and subsequent cytokine secretion. Furthermore, our study is the first to show that SslE can stimulate TLR2-dependent production of other proinflammatory hallmarks, such as reactive nitrogen and oxygen species as well as type 1 chemokines, which contribute to the anti-infection immune response of the host. Also, the overexpression of major histocompatibility complex class II (MHC II) and other costimulatory molecules (CD80 and CD86) in macrophages essentially indicates that SslE promotes macrophage activation and M1 polarization, which are crucial in framing the host's innate immune response to this protein, and hence, SslE could be a potent immunotherapeutic target against E. coli sepsis.
Collapse
|
45
|
Nestander M, Dintaman J, Susi A, Gorman G, Hisle-Gorman E. Immunization Completion in Infants Born at Low Birth Weight. J Pediatric Infect Dis Soc 2018; 7:e58-e64. [PMID: 29036471 DOI: 10.1093/jpids/pix079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/21/2017] [Indexed: 11/13/2022]
Abstract
BACKGROUND Low birth weight (LBW) has been associated with underimmunization. We sought to understand the effect of LBW on immunization completion after controlling for previously hypothesized mediators, including prematurity, neonatal illness, well-child care, non-well-child visits, and provider consistency. METHODS We formed a retrospective cohort of infants born between 2008 and 2011 with ≥2 years of military healthcare follow-up. International Classification of Diseases, Ninth Revision codes were used to identify LBW, preterm birth, neonatal illnesses, well-child visits, non-well-child visits, provider consistency, and parental rank in the inpatient and outpatient records. Immunization records were extracted from both records. Logistic regression determined the odds of immunization completion and well-child care completion (ie, having had ≥6 WCC visits by 15 months of age). RESULTS Of 135964 included infants, 116521 (85.7%) were completely immunized at the age of 2 years. In adjusted analysis, the odds of immunization completion were significantly decreased in infants born at LBW (odds ratio [OR], 0.88 [95% confidence interval (CI), 0.79-0.97]), very LBW (OR, 0.61 [95% CI, 0.48-0.77]), or extremely LBW (OR, 0.45 [95% CI, 0.33-0.63]) or at ≤32 weeks' gestation (OR, 0.76 [95% CI, 0.63-0.92]), infants with chronic lung disease (OR, 0.63 [95% CI, 0.45-0.88]), male infants (OR, 0.96 [95% CI, 0.93-0.99]), and infants who experienced decreased provider consistency (OR, 0.92 [95% CI, 0.91-0.92]). The rate of immunization completion increased with the overall number of healthcare visits (OR, 1.02 [95% CI, 1.02-1.02]) and complete well-child care (OR, 1.80 [95% CI, 1.75-1.86]). However, children born LBW or preterm were significantly less likely to have complete well-child care. CONCLUSIONS After adjustment for preterm birth, comorbid neonatal conditions, and early childhood patterns of healthcare use, LBW was significantly associated with immunization noncompletion in a universal healthcare system. Provider consistency and well-child care seem important for increasing immunization completion in LBW infants.
Collapse
Affiliation(s)
- Matt Nestander
- Department of Pediatrics, Madigan Army Medical Center, Tacoma, Washington
| | | | - Apryl Susi
- Department of Pediatrics, Uniformed Services University, Bethesda, Maryland
| | - Gregory Gorman
- Department of Pediatrics, Uniformed Services University, Bethesda, Maryland.,Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, Maryland
| | | |
Collapse
|
46
|
Mechanisms of LPS-Induced Acute Kidney Injury in Neonatal and Adult Rats. Antioxidants (Basel) 2018; 7:antiox7080105. [PMID: 30096767 PMCID: PMC6115895 DOI: 10.3390/antiox7080105] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/28/2018] [Accepted: 08/06/2018] [Indexed: 01/03/2023] Open
Abstract
Neonatal sepsis is one of the major causes of mortality and morbidity in newborns, greatly associated with severe acute kidney injury (AKI) and failure. Handling of newborns with kidney damage can be significantly different compared to adults, and it is necessary to consider the individuality of an organism's response to systemic inflammation. In this study, we used lipopolysaccharide (LPS)-mediated acute kidney injury model to study mechanisms of kidney cells damage in neonatal and adult rats. We found LPS-associated oxidative stress was more severe in adults compared to neonates, as judged by levels of carbonylated proteins and products of lipids peroxidation. In both models, LPS-mediated septic simulation caused apoptosis of kidney cells, albeit to a different degree. Elevated levels of proliferating cell nuclear antigen (PCNA) in the kidney dropped after LPS administration in neonates but increased in adults. Renal fibrosis, as estimated by smooth muscle actin levels, was significantly higher in adult kidneys, whereas these changes were less profound in LPS-treated neonatal kidneys. We concluded that in LPS-mediated AKI model, renal cells of neonatal rats were more tolerant to oxidative stress and suffered less from long-term pathological consequences, such as fibrosis. In addition, we assume that by some features LPS administration simulates the conditions of accelerated aging.
Collapse
|
47
|
El Shimy MS, El-Raggal NM, El-Farrash RA, Shaaban HA, Mohamed HE, Barakat NM, Farag AS, El Zohiery AK, Shaaban MAA, Salama DH. Cerebral blood flow and serum neuron-specific enolase in early-onset neonatal sepsis. Pediatr Res 2018; 84:261-266. [PMID: 29907848 DOI: 10.1038/s41390-018-0062-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/17/2018] [Accepted: 05/10/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND OBJECTIVES Sepsis leads to systemic inflammatory response with cerebral blood flow (CBF) alteration and blood-brain barrier disruption that contribute to sepsis-associated encephalopathy (SAE). We aimed to evaluate cord blood neuron-specific enolase (cNSE) and CBF in early-onset neonatal sepsis (EONS) as predictors of SAE and to define short-term neurodevelopmental outcomes among survivors. METHODS cNSE was measured in 200 neonates with antenatal risk factors for EONS, stratified into two groups: sepsis (n = 96) and no-sepsis (n = 104). Trans-cranial Doppler of peak systolic velocities (PSV), end diastolic velocities (EDV) and resistive indices (RI) of anterior (ACA) and middle (MCA) cerebral arteries recorded on day 1 postnatal. Griffiths mental developmental scale (GMDS) was assessed at 6 months. RESULTS Increased cNSE, PSV, EDV, and decreased RI of both ACA and MCA were found in sepsis group compared to no-sepsis group (p < 0.001 for all). Patients with SAE (n = 34) had higher NSE, PSV, and EDV as well as lower RI of ACA and MCA compared to those without (p < 0.01 for all). SAE neonates had lower GMDS than those without. ACA RI of ≤0.61 was the best predictor of SAE. CONCLUSION High CBF and cNSE could be useful markers for prediction of SAE. SAE impairs neurodevelopmental scales at 6 months.
Collapse
Affiliation(s)
| | | | | | | | | | - Noha M Barakat
- Departments of Pediatrics, Ain Shams University, Cairo, Egypt
| | - Ahmed S Farag
- Obstetric and Gynecology, Ain Shams University, Cairo, Egypt
| | - Abeer K El Zohiery
- Physical Medicine and Rehabilitation, Ain Shams University, Cairo, Egypt
| | | | - Dina H Salama
- Radiodiagnosis Department, National Center for Radiation Research and Technology of the Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
48
|
Harbeson D, Ben-Othman R, Amenyogbe N, Kollmann TR. Outgrowing the Immaturity Myth: The Cost of Defending From Neonatal Infectious Disease. Front Immunol 2018; 9:1077. [PMID: 29896192 PMCID: PMC5986917 DOI: 10.3389/fimmu.2018.01077] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/30/2018] [Indexed: 12/28/2022] Open
Abstract
Newborns suffer high rates of mortality due to infectious disease-this has been generally regarded to be the result of an "immature" immune system with a diminished disease-fighting capacity. However, the immaturity dogma fails to explain (i) greater pro-inflammatory responses than adults in vivo and (ii) the ability of neonates to survive a significantly higher blood pathogen burden than of adults. To reconcile the apparent contradiction of clinical susceptibility to disease and the host immune response findings when contrasting newborn to adult, it will be essential to capture the entirety of available host-defense strategies at the newborn's disposal. Adults focus heavily on the disease resistance approach: pathogen reduction and elimination. Newborn hyperactive innate immunity, sensitivity to immunopathology, and the energetic requirements of growth and development (immune and energy costs), however, preclude them from having an adult-like resistance response. Instead, newborns also may avail themselves of disease tolerance (minimizing immunopathology without reducing pathogen load), as a disease tolerance approach provides a counterbalance to the dangers of a heightened innate immunity and has lower-associated immune costs. Further, disease tolerance allows for the establishment of a commensal bacterial community without mounting an unnecessarily dangerous immune resistance response. Since disease tolerance has its own associated costs (immune suppression leading to unchecked pathogen proliferation), it is the maintenance of homeostasis between disease tolerance and disease resistance that is critical to safe and effective defense against infections in early life. This paradigm is consistent with nearly all of the existing evidence.
Collapse
Affiliation(s)
- Danny Harbeson
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Nelly Amenyogbe
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias R. Kollmann
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
49
|
Martin SL, Desai S, Nanavati R, Colah RB, Ghosh K, Mukherjee MB. Innate immune gene polymorphisms and their association with neonatal sepsis. INFECTION GENETICS AND EVOLUTION 2018; 62:205-210. [PMID: 29715526 DOI: 10.1016/j.meegid.2018.04.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 11/27/2022]
Affiliation(s)
- Snehal L Martin
- Department of Haematogenetics, National Institute of Immunohaematology (ICMR), KEM Hospital Campus, Parel, Mumbai, India
| | - Saumil Desai
- Department of Neonatology, KEM Hospital, Parel, Mumbai, India
| | - Ruchi Nanavati
- Department of Neonatology, KEM Hospital, Parel, Mumbai, India
| | - Roshan B Colah
- Department of Haematogenetics, National Institute of Immunohaematology (ICMR), KEM Hospital Campus, Parel, Mumbai, India
| | - Kanjaksha Ghosh
- Department of Haematogenetics, National Institute of Immunohaematology (ICMR), KEM Hospital Campus, Parel, Mumbai, India
| | - Malay B Mukherjee
- Department of Haematogenetics, National Institute of Immunohaematology (ICMR), KEM Hospital Campus, Parel, Mumbai, India.
| |
Collapse
|
50
|
Vermillion MS, Klein SL. Pregnancy and infection: using disease pathogenesis to inform vaccine strategy. NPJ Vaccines 2018; 3:6. [PMID: 29423318 PMCID: PMC5794984 DOI: 10.1038/s41541-017-0042-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/29/2017] [Accepted: 12/11/2017] [Indexed: 02/03/2023] Open
Abstract
Vaccination is the mainstay of preventative medicine for many infectious diseases. Pregnant women, unborn fetuses, and neonates represent three at-risk populations that can be simultaneously protected by strategic vaccination protocols. Because the pathogenesis of different infectious microbes varies based on tissue tropism, timing of infection, and host susceptibility, the goals of immunization are not uniform across all vaccines. Mechanistic understanding of infectious disease pathogenesis and immune responses is therefore essential to inform vaccine design and the implementation of appropriate immunization protocols that optimize protection of pregnant women, fetuses, and neonates.
Collapse
Affiliation(s)
- Meghan S. Vermillion
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205 USA
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205 USA
| |
Collapse
|