Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Front Immunol ; 15: 1365174, 2024.
Article in English | MEDLINE | ID: mdl-38774873

ABSTRACT

Introduction: Sepsis remains a major source of morbidity and mortality in neonates, and characterization of immune regulation in the neonatal septic response remains limited. HVEM is a checkpoint regulator which can both stimulate or inhibit immune responses and demonstrates altered expression after sepsis. We hypothesized that signaling via HVEM would be essential for the neonatal response to sepsis, and that therefore blockade of this pathway would improve survival to septic challenge. Methods: To explore this, neonatal mice were treated with cecal slurry (CS), CS with Anti-HVEM antibody (CS-Ab) or CS with isotype (CS-IT) and followed for 7-day survival. Mice from all treatment groups had thymus, lung, kidney and peritoneal fluid harvested, weighed, and stained for histologic evaluation, and changes in cardiac function were assessed with echocardiography. Results: Mortality was significantly higher for CS-Ab mice (72.2%) than for CS-IT mice (22.2%). CS resulted in dysregulated alveolar remodeling, but CS-Ab lungs demonstrated significantly less dysfunctional alveolar remodeling than CS alone (MCL 121.0 CS vs. 87.6 CS-Ab), as well as increased renal tubular vacuolization. No morphologic differences in alveolar septation or thymic karyorrhexis were found between CS-Ab and CS-IT. CS-Ab pups exhibited a marked decrease in heart rate (390.3 Sh vs. 342.1 CS-Ab), stroke volume (13.08 CS-IT vs. 8.83 CS-Ab) and ultimately cardiac output (4.90 Sh vs. 3.02 CS-Ab) as well as a significant increase in ejection fraction (73.74 Sh vs. 83.75 CS-Ab) and cardiac strain (40.74 Sh vs. 51.16 CS-Ab) as compared to CS-IT or Sham animals. Discussion: While receptor ligation of aspects of HVEM signaling, via antibody blockade, appears to mitigate aspects of lung injury and thymic involution, stimulatory signaling via HVEM still seems to be necessary for vascular and hemodynamic resilience and overall neonatal mouse survival in response to this experimental polymicrobial septic insult. This dissonance in the activity of anti-HVEM neutralizing antibody in neonatal animals speaks to the differences in how septic cardiac dysfunction should be considered and approached in the neonatal population.


Subject(s)
Animals, Newborn , Neonatal Sepsis , Signal Transduction , Animals , Mice , Neonatal Sepsis/immunology , Neonatal Sepsis/mortality , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Tumor Necrosis Factor, Member 14/immunology , Disease Models, Animal , Female , Heart Diseases/etiology , Heart Diseases/immunology , Lung/immunology , Lung/pathology , Sepsis/immunology , Sepsis/metabolism
2.
J Exp Med ; 221(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38753245

ABSTRACT

Preterm infants are at high risk of developing neonatal sepsis. γδ T cells are thought to be an important set of effector cells in neonates. Here, γδ T cells were investigated in a longitudinal cohort of preterm neonates using next-generation sequencing, flow cytometry, and functional assays. During the first year of life, the Vγ9Vδ2 T cell subset showed dynamic phenotypic changes and elevated levels of fetal-derived Vγ9Vδ2 T cells were evident in infants with sepsis. Single-cell transcriptomics identified HLA-DRhiCD83+ γδ T cells in neonatal sepsis, which expressed genes related to antigen presentation. In vitro assays showed that CD83 was expressed on activated Vγ9Vδ2 T cells in preterm and term neonates, but not in adults. In contrast, activation of adult Vγ9Vδ2 T cells enhanced CD86 expression, which was presumably the key receptor to induce CD4 T cell proliferation. Together, we provide a map of the maturation of γδ T cells after preterm birth and highlight their phenotypic diversity in infections.


Subject(s)
Antigens, CD , CD83 Antigen , Infant, Premature , Receptors, Antigen, T-Cell, gamma-delta , Humans , Infant, Newborn , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Infant, Premature/immunology , Antigens, CD/metabolism , Antigens, CD/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Female , Male , Sepsis/immunology , Cohort Studies , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adult , Lymphocyte Activation/immunology , Neonatal Sepsis/immunology , Infant
3.
Pediatr Res ; 91(1): 230-234, 2022 01.
Article in English | MEDLINE | ID: mdl-33731804

ABSTRACT

BACKGROUND: Late-onset sepsis is an important cause of mortality and morbidity in preterm infants. As these infants rely mostly on their innate immune system to fight off infection, enhancing this immune system by appropriate stimuli may prevent late-onset sepsis. However, it remains unclear which stimuli can enhance the neonatal immune system. This study aims to investigate the influence of intrauterine inflammation on late-onset sepsis. METHODS: This is a retrospective cohort study in a Neonatal Intensive Care Unit in the Netherlands. Between 2005 and 2016, 1014 infants with ≤32 weeks gestational age and/or with a birth weight ≤1500 g were included. Intrauterine inflammation was subdivided into histological chorioamnionitis, fetal inflammatory response, and funisitis. Logistic and Cox regression analyses were performed to investigate the influence of intrauterine inflammation on late-onset sepsis. RESULTS: Thirty-six percent of the included infants developed late-onset sepsis; 24% of placentas showed intrauterine inflammation. Late-onset sepsis incidence did not differ between infants with or without exposure to intrauterine inflammation after adjustment for gestational age (histological chorioamnionitis aHR 0.928 [CI: 0.727-1.185], p = 0.551; fetal inflammatory response aHR 1.011 [CI: 0.793-1.288], p = 0.930); funisitis aHR 0.965 [CI: 0.738-1.263], p = 0.797). CONCLUSIONS: Late-onset sepsis in very preterm infants seems not to be associated with intrauterine inflammation. IMPACT: Intrauterine inflammation is not protective of developing late-onset sepsis in premature infants. A large cohort study on the effect of intrauterine inflammation on neonatal outcome. This study adds to existing knowledge on finding appropriate stimuli to enhance the immune system of premature infants to improve neonatal outcome.


Subject(s)
Infant, Extremely Premature , Inflammation/complications , Neonatal Sepsis/complications , Uterine Diseases/complications , Female , Humans , Immunity, Innate , Infant, Newborn , Inflammation/immunology , Neonatal Sepsis/immunology , Retrospective Studies , Risk Factors , Uterine Diseases/immunology
4.
Biomed Pharmacother ; 145: 112444, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34808550

ABSTRACT

Neonatal sepsis constitutes a highly relevant public health challenge and is the most common cause of infant morbidity and mortality worldwide. Recent studies have demonstrated that during infection epigenetic changes may occur leading to reprogramming of gene expression. Post-transcriptional regulation by short non-coding RNAs (e.g., microRNAs) have recently acquired special relevance because of their role in the regulation of the pathophysiology of sepsis and their potential clinical use as biomarkers. ~22-nucleotide of microRNAs are not only involved in regulating multiple relevant cellular and molecular functions, such as immune cell function and inflammatory response, but have also been proposed as good candidates as biomarkers in sepsis. Nevertheless, establishing clinical practice guidelines based on microRNA patterns as biomarkers for diagnosis and prognosis in neonatal sepsis has yet to be achieved. Given their differential expression across tissues in neonates, the release of specific microRNAs to blood and their expression pattern can differ compared to sepsis in adult patients. Further in-depth research is necessary to fully understand the biological relevance of microRNAs and assess their potential use in clinical settings. This review provides a general overview of microRNAs, their structure, function and biogenesis before exploring their potential clinical interest as diagnostic and prognostic biomarkers of neonatal sepsis. An important part of the review is focused on immune and inflammatory aspects of selected microRNAs that may become biomarkers for clinical use and therapeutic intervention.


Subject(s)
Epigenesis, Genetic , MicroRNAs/genetics , Neonatal Sepsis/genetics , Animals , Biomarkers/metabolism , Gene Expression Regulation , Humans , Infant, Newborn , Neonatal Sepsis/diagnosis , Neonatal Sepsis/immunology , Prognosis
5.
Am J Physiol Regul Integr Comp Physiol ; 321(6): R879-R902, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34612068

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Immune System/drug effects , Inflammation Mediators/antagonists & inhibitors , Inflammation/drug therapy , Neonatal Sepsis/drug therapy , Toll-Like Receptors/drug effects , Age Factors , Animals , Anti-Inflammatory Agents/adverse effects , Child Development , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Female , Gene Expression Regulation, Developmental , Humans , Immune System/immunology , Immune System/metabolism , Immunity, Innate/drug effects , Infant, Newborn , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Molecular Targeted Therapy , Neonatal Sepsis/genetics , Neonatal Sepsis/immunology , Neonatal Sepsis/metabolism , Sex Factors , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
6.
J Immunol Res ; 2021: 4884537, 2021.
Article in English | MEDLINE | ID: mdl-34676267

ABSTRACT

Salivary markers could serve as potential noninvasive markers in the diagnosis of neonatal infections. We aimed to investigate the diagnostic role of salivary and serum interleukin 10 (IL-10), C-reactive protein (CRP), mean platelet volume (MPV), and CRP/MPV ratio in the diagnosis of late-onset neonatal sepsis in full-term neonates. Seventy full-term neonates were enrolled in this prospective case-control study, 35 with late-onset neonatal sepsis, and 35 controls. Salivary IL-10, serum IL-10, and CRP concentrations were measured by enzyme-linked immunosorbent assay (ELISA). Complete blood (CBC) count was measured by an automated blood cell counter. The salivary IL-10, serum IL-10, CRP, MPV, and CRP/MPV ratio levels were much higher in neonates with late-onset sepsis than in control (220 ± 150 vs. 18 ± 9 pg/ml, P < 0.001), (316 ± 198 vs. 23.7 ± 14 pg/ml, P < 0.001), (78.2 ± 34 vs. 3.3 ± 1.7 mg/L, P < 0.001), (11.2 ± 0.9 vs. 8.6 ± 0.4 fL), and (7.08 ± 3.3 vs. 0.4 ± 0.2, P < 0.001), respectively. At the cutoff point of >31 pg/ml, salivary IL-10 showed 97.1% sensitivity and 94.3% specificity. Serum IL-10 at a cutoff value of ≥33.6 pg/ml had a sensitivity of 97.1% and specificity of 80%. MPV showed a sensitivity of 100% and specificity of 94.4% at a cutoff value ≥ 9.2 fL. CRP/MPV ratio showed a sensitivity of 100% and specificity of 97.1% at a cutoff value > 0.9. Salivary and serum IL-10 showed a positive correlation with CRP and CRP/MPV ratio in septic neonates. The current study shows for the first time that both salivary IL-10 and CRP/MPV showed statistically significant differences between neonates with late-onset sepsis and controls. Accordingly, salivary IL-10 could serve as a potential noninvasive biomarker for the diagnosis of late-onset sepsis in full-term neonates.


Subject(s)
C-Reactive Protein/analysis , Interleukin-10/analysis , Mean Platelet Volume , Neonatal Sepsis/diagnosis , Saliva/chemistry , Biomarkers/blood , Case-Control Studies , Female , Humans , Infant, Newborn , Male , Neonatal Sepsis/blood , Neonatal Sepsis/immunology , Prospective Studies , ROC Curve , Saliva/immunology
7.
Inflammation ; 44(6): 2395-2406, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34523052

ABSTRACT

Neonatal sepsis (NS) is a severe syndrome in newborns that is induced by infections, and the initiation and development of NS are closely associated with the function of miRs. In the current study, the effects of berberine, which is a functional component in traditional Chinese medicine (TCM), against NS were assessed by focusing on the interaction of berberine with miR-132-3p-mediated signaling. An NS model was induced using cecal slurry (CS) in vivo and LPS in vitro, and berberine treatment was applies. The changes in survival rate, intestinal structure, and systemic inflammation in mice and the viability, apoptosis, and inflammatory response in intestinal cells were measured. At the molecular level, miR-132-3p levels and the activities of the FOXA1 and NF-κB pathways were analyzed. The data showed that berberine increased the survival rates of CS-induced mice. The intestinal injuries induced by CS were also attenuated by berberine, which was associated with inhibition of the production of systemic IL-6, IL-1ß, and TNF-α. At the molecular level, the expression of miR-132-3p was upregulated, suppressing the expression of FOXA1, p-IκBα, and p65 while inducing the expression of IκBα. The effects of berberine on NS-induced impairments were blocked by the injection of the miR-132-3p antagomir, which exacerbated intestinal injuries, induced systemic inflammation, and reactivated the FOXA1 and NF-κB pathways. The findings in the in vivo model were validated with in vitro assays. Collectively, the findings outlined in the current study indicated that berberine had solid protective effects against NS-induced symptoms in newborn mice, and the effects depended on the upregulation of miR-132-3p.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Berberine/pharmacology , Hepatocyte Nuclear Factor 3-alpha/metabolism , Inflammation/prevention & control , Intestines/drug effects , MicroRNAs/metabolism , NF-kappa B/metabolism , Neonatal Sepsis/prevention & control , Animals , Animals, Newborn , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Intestines/immunology , Intestines/metabolism , Intestines/pathology , Mice, Inbred C57BL , MicroRNAs/genetics , Neonatal Sepsis/genetics , Neonatal Sepsis/immunology , Neonatal Sepsis/metabolism , Signal Transduction , Up-Regulation
9.
Int J Mol Sci ; 22(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34208904

ABSTRACT

Neonates are at an increased risk of an infectious disease. This is consistent with an increased abundance of myeloid-derived suppressor cells (MDSCs) compared with older children and adults. Using a murine model of neonatal bacterial sepsis, we demonstrate that MDSCs modulate their activity during an infection to enhance immune suppressive functions. A gene expression analysis shows that MDSCs increased NOS2, Arg-1 and IL-27p28 expression in vitro and in vivo in response to Escherichia coli O1:K1:H7 and this is regulated at the level of the gene expression. Changes in the effector gene expression are consistent with increased enzymatic activity and cytokine secretion. The neonatal MDSCs express toll-like receptor (TLR) 2, 4 and 5 capable of recognizing pathogen-associated molecular patterns (PAMPS) on E. coli. However, a variable level of effector expression was achieved in response to LPS, peptidoglycan or flagellin. Individual bacterial PAMPs did not stimulate the expression of Arg-l and IL-27p28 equivalently to E. coli. However, the upregulation of NOS2 was achieved in response to LPS, peptidoglycan and flagella. The increased immune suppressive profile translated to an enhanced suppression of CD4+ T cell proliferation. Collectively, these findings increase our understanding of the dynamic nature of MDSC activity and suggest that these cells abundant in early life can acquire activity during an infection that suppresses protective immunity.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/pathogenicity , Myeloid-Derived Suppressor Cells/metabolism , Neonatal Sepsis/microbiology , Animals , Animals, Newborn , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Infant, Newborn , Mice , Neonatal Sepsis/genetics , Neonatal Sepsis/immunology , Nitric Oxide Synthase Type II/genetics , Toll-Like Receptors/genetics
10.
Mol Immunol ; 134: 129-140, 2021 06.
Article in English | MEDLINE | ID: mdl-33773156

ABSTRACT

Dentritic cells (DCs) dysfunction has been verified detrimental for sepsis and B and T lymphocyte attenuator (BTLA) is an immune-regulatory receptor shown to be associated with DCs dysfunction. However, the role of BTLA expression in myeloid DCs (mDCs) in neonatal sepsis is unknown. In the current study, we found BTLA-expressing mDCs were elevated in neonates with sepsis and the BTLA expression level in mDCs was positively correlated to the severity of sepsis. The presence of BTLA negatively regulated the phagocytosis capacity and bactericidal ability of mDCs as well as the maturation markers expression of mDCs. Our data also showed BTLA+mDCs shifted into an anti-inflammatory phenotype with decreased expression of IL-6, TNF-α and IL-12, but increased IL-10. in addition, we found BTLA expression indeedly altered the mDCs allo-stimulatory capacity. Therefore, BTLA expression in mDCs could be a useful predictive marker for neonatal sepsis and targeting BTLA expression in mDCs may be a new therapeutic strategy.


Subject(s)
Dendritic Cells/metabolism , Neonatal Sepsis/metabolism , Receptors, Immunologic/metabolism , Biomarkers/metabolism , Dendritic Cells/immunology , Female , Humans , Infant, Newborn , Male , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neonatal Sepsis/immunology , Receptors, Immunologic/immunology , Up-Regulation
11.
Front Immunol ; 12: 634529, 2021.
Article in English | MEDLINE | ID: mdl-33746973

ABSTRACT

Morbidity and mortality associated with neonatal sepsis remains a healthcare crisis. PD1-/- neonatal mice endured experimental sepsis, in the form of cecal slurry (CS), and showed improved rates of survival compared to wildtype (WT) counterparts. End-organ injury, particularly of the lung, contributes to the devastation set forth by neonatal sepsis. PDL1-/- neonatal mice, in contrast to PD1-/- neonatal mice did not have a significant improvement in survival after CS. Because of this, we focused subsequent studies on the impact of PD1 gene deficiency on lung injury. Here, we observed that at 24 h post-CS (but not at 4 or 12 h) there was a marked increase in pulmonary edema (PE), neutrophil influx, myeloperoxidase (MPO) levels, and cytokine expression sham (Sh) WT mice. Regarding pulmonary endothelial cell (EC) adhesion molecule expression, we observed that Zona occludens-1 (ZO-1) within the cell shifted from a membranous location to a peri-nuclear location after CS in WT murine cultured ECs at 24hrs, but remained membranous among PD1-/- lungs. To expand the scope of this inquiry, we investigated human neonatal lung tissue. We observed that the lungs of human newborns exposed to intrauterine infection had significantly higher numbers of PD1+ cells compared to specimens who died from non-infectious causes. Together, these data suggest that PD1/PDL1, a pathway typically thought to govern adaptive immune processes in adult animals, can modulate the largely innate neonatal pulmonary immune response to experimental septic insult. The potential future significance of this area of study includes that PD1/PDL1 checkpoint proteins may be viable therapeutic targets in the septic neonate.


Subject(s)
B7-H1 Antigen/metabolism , Lung Injury/etiology , Lung/metabolism , Neonatal Sepsis/complications , Programmed Cell Death 1 Receptor/metabolism , Animals , Animals, Newborn , B7-H1 Antigen/genetics , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Humans , Immunity, Innate , Infant, Newborn , Lung/immunology , Lung/pathology , Lung Injury/immunology , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neonatal Sepsis/immunology , Neonatal Sepsis/metabolism , Neonatal Sepsis/microbiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Programmed Cell Death 1 Receptor/genetics , Pulmonary Edema/etiology , Pulmonary Edema/immunology , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Zonula Occludens-1 Protein/metabolism
12.
Biomolecules ; 10(11)2020 11 20.
Article in English | MEDLINE | ID: mdl-33233724

ABSTRACT

Neonatal sepsis is a life-threatening condition and Staphylococcus aureus is one of its major causes. However, to date, no rapid and sensitive diagnostic tool has been developed for its direct detection. Bioinformatics analyses identified a surface-exposed 112-amino acid polypeptide of the cell wall protein NWMN_1649, a surface protein involved in cell aggregation and biofilm formation, as being a species-specific and highly conserved moiety. The polypeptide was cloned, purified, and used to immunize mice to raise specific immunoglobulins. The purified antibodies were conjugated to gold nano-particles and used to assemble an immunochromatographic strip (ICS). The developed prototype ICS detected as low as 5 µg purified polypeptide and 102 CFU/mL S. aureus within 15 min. The strip showed superior ability to directly detect S. aureus in neonatal sepsis blood specimens without prior sample processing. Moreover, it showed no cross-reaction in specimens infected with two other major causes of neonatal sepsis; coagulase-negative staphylococci and Klebsiella pneumoniae. The selected NWMN_1649-derived polypeptide demonstrates success as a promising biomolecule upon which a prototype ICS has been developed. This ICS provides a rapid, direct, sensitive, and specific option for the detection of S. aureus causing neonatal sepsis. Such a tool is urgently needed especially in resources-limited countries.


Subject(s)
Chromatography, Affinity/methods , Neonatal Sepsis/diagnosis , Neonatal Sepsis/immunology , Peptides/chemistry , Peptides/immunology , Staphylococcal Infections/diagnosis , Staphylococcal Infections/immunology , Animals , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Computational Biology , Computer Simulation , Female , Humans , Infant, Newborn , Metal Nanoparticles/chemistry , Mice, Inbred BALB C , Neonatal Sepsis/blood , Neonatal Sepsis/microbiology , Peptide Biosynthesis/immunology , Peptides/isolation & purification , Sensitivity and Specificity , Staphylococcal Infections/blood , Staphylococcus aureus/cytology , Staphylococcus aureus/immunology
13.
Front Immunol ; 11: 577878, 2020.
Article in English | MEDLINE | ID: mdl-33072121

ABSTRACT

Introduction: Neonatal sepsis triggers an inflammatory response that contributes to mortality and multiple organ injury. Pentoxifylline (PTX), a phosphodiesterase inhibitor which suppresses pro-inflammatory cytokines, is a candidate adjunctive therapy for newborn sepsis. We hypothesized that administration of PTX in addition to antibiotics decreases live bacteria-induced pro-inflammatory and/or enhances anti-inflammatory cytokine production in septic neonatal mice without augmenting bacterial growth. Methods: Newborn C57BL/6J mice (< 24 h old) were injected intravenously with 105 colony forming units (CFUs)/g weight of a bioluminescent derivative of the encapsulated clinical isolate Escherichia coli O18:K1. Adequacy of intravenous injections was validated using in vivo bioluminescence imaging and Evans blue. Pups were treated with gentamicin (GENT), PTX, (GENT + PTX) or saline at 0, 1.5, or 4 h after sepsis initiation, and euthanized after an additional 4 h. CFUs and cytokines were measured from blood and homogenized organ tissues. Results: GENT alone inhibited bacterial growth, IL-1ß, and IL-6 production in blood and organs. Addition of PTX to GENT profoundly inhibited E. coli-induced TNF and enhanced IL-10 in blood of newborn mice at all timepoints, whereas it primarily upregulated IL-10 production in peripheral organs (lung, spleen, brain). PTX, whether alone or adjunctive to GENT, did not increase microbial colony counts in blood and organs. Conclusion: Addition of PTX to antibiotics in murine neonatal E. coli sepsis promoted an anti-inflammatory milieu through inhibition of plasma TNF and enhancement of IL-10 production in plasma and organs without increasing bacterial growth, supporting its utility as a potential adjunctive agent for newborn sepsis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Cytokines/blood , Escherichia coli Infections/drug therapy , Escherichia coli/drug effects , Gentamicins/pharmacology , Inflammation Mediators/blood , Neonatal Sepsis/drug therapy , Pentoxifylline/pharmacology , Animals , Animals, Newborn , Disease Models, Animal , Drug Therapy, Combination , Escherichia coli/growth & development , Escherichia coli/pathogenicity , Escherichia coli Infections/blood , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Mice, Inbred C57BL , Neonatal Sepsis/blood , Neonatal Sepsis/immunology , Neonatal Sepsis/microbiology , Phosphodiesterase Inhibitors/pharmacology
14.
Biosci Rep ; 40(7)2020 07 31.
Article in English | MEDLINE | ID: mdl-32618342

ABSTRACT

Neonatal septicemia is a serious infectious disease in the neonatal period. MicroRNAs (miRNAs) have been reported to participate in the inflammatory responses in neonatal sepsis. The aim of the present study was to explore the effects and molecular mechanism of miR-96-5p on regulating the inflammatory responses in neonatal sepsis. MiR-96-5p was low expressed while nicotinamide phosphoribosyltransferase (NAMPT) was high expressed in the serum of neonatal septicemia patients. The expression of miR-96-5p was decreased in LPS-induced inflammatory responses. Besides, miR-95-5p relieved LPS-induced inflammatory responses in RAW264.7 cells. NAMPT was demonstrated as a potential target of miR-96-5p, and knockdown of NAMPT reduced inflammatory in RAW264.7 cells stimulated with LPS. Moreover, overexpression of NAMPT reversed the effects of miR-96-5p on LPS-induced inflammatory responses. In addition, miR-96-5p inhibited nuclear factor (NF)-κB signaling pathway in RAW264.7 cells stimulated with LPS. MiR-96-5p alleviated inflammatory responses via targeting NAMPT and inhibiting NF-κB pathway in neonatal sepsis.


Subject(s)
Cytokines/genetics , MicroRNAs/metabolism , Neonatal Sepsis/immunology , Nicotinamide Phosphoribosyltransferase/genetics , Animals , Case-Control Studies , Cytokines/blood , Disease Models, Animal , Gene Knockdown Techniques , HEK293 Cells , Humans , Infant, Newborn , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Lipopolysaccharides/immunology , Mice , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/blood , NF-kappa B/metabolism , Neonatal Sepsis/blood , Neonatal Sepsis/genetics , Nicotinamide Phosphoribosyltransferase/blood , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
15.
Front Immunol ; 11: 972, 2020.
Article in English | MEDLINE | ID: mdl-32582153

ABSTRACT

Chorioamnionitis (CA) is considered a key risk factor for very preterm birth and for developing early onset sepsis (EOS) in preterm infants, but recent data suggest that CA might be protective against late onset sepsis (LOS). We performed a systematic review and meta-analysis of studies exploring the association between CA and sepsis. A comprehensive literature search was performed in PubMed/MEDLINE and EMBASE, from their inception to December 1, 2018. A random-effects model was used to calculate odds ratios (OR) and 95% confidence intervals (CI). Sources of heterogeneity were analyzed by subgroup and meta-regression analyses. The following categories of sepsis were analyzed: EOS, LOS, unspecified onset sepsis (UOS), culture-proven, and clinical sepsis. CA was subdivided into clinical and histological chorioamnionitis. Funisitis was also analyzed. We found 3,768 potentially relevant studies, of which 107 met the inclusion criteria (387,321 infants; 44,414 cases of CA). Meta-analysis showed an association between any CA and any EOS (OR 4.29, CI 3.63-5.06), any LOS (OR 1.29, CI 1.11-1.54), and any UOS (OR 1.59, CI 1.11-1.54). Subgroup analysis showed that CA was associated with culture-proven EOS (OR 4.69, CI 3.91-5.56), clinical EOS (OR 3.58, CI 1.90-6.76), and culture-proven LOS (OR 1.31, CI 1.12-1.53), but not with clinical LOS (OR 1.52, CI 0.78-2.96). The presence of funisitis did not increase the risk of either EOS or LOS when compared with CA without funisitis. CA-exposed infants had lower gestational age (-1.11 weeks, CI -1.37 to -0.84) than the infants not exposed to CA. Meta-regression analysis showed that the lower gestational age of the CA group correlated with the association between CA and LOS but not with the association between CA and EOS. In conclusion, our data suggest that the positive association between chorioamnionitis and LOS may be modulated by the effect of chorioamnionitis on gestational age.


Subject(s)
Chorioamnionitis/epidemiology , Infant, Premature , Neonatal Sepsis/epidemiology , Premature Birth/epidemiology , Age Factors , Chorioamnionitis/immunology , Chorioamnionitis/pathology , Female , Gestational Age , Humans , Infant, Newborn , Neonatal Sepsis/diagnosis , Neonatal Sepsis/immunology , Observational Studies as Topic , Pregnancy , Premature Birth/diagnosis , Premature Birth/immunology , Prognosis , Risk Assessment , Risk Factors
16.
PLoS One ; 15(6): e0233841, 2020.
Article in English | MEDLINE | ID: mdl-32479514

ABSTRACT

BACKGROUND: Host immune responses during late-onset sepsis (LOS) in very preterm infants are poorly characterised due to a complex and dynamic pathophysiology and challenges in working with small available blood volumes. We present here an unbiased transcriptomic analysis of whole peripheral blood from very preterm infants at the time of LOS. METHODS: RNA-Seq was performed on peripheral blood samples (6-29 days postnatal age) taken at the time of suspected LOS from very preterm infants <30 weeks gestational age. Infants were classified based on blood culture positivity and elevated C-reactive protein concentrations as having confirmed LOS (n = 5), possible LOS (n = 4) or no LOS (n = 9). Bioinformatics and statistical analyses performed included pathway over-representation and protein-protein interaction network analyses. Plasma cytokine immunoassays were performed to validate differentially expressed cytokine pathways. RESULTS: The blood leukocyte transcriptional responses of infants with confirmed LOS differed significantly from infants without LOS (1,317 differentially expressed genes). However, infants with possible LOS could not be distinguished from infants with no LOS or confirmed LOS. Transcriptional alterations associated with LOS included genes involved in pathogen recognition (mainly TLR pathways), cytokine signalling (both pro-inflammatory and inhibitory responses), immune and haematological regulation (including cell death pathways), and metabolism (altered cholesterol biosynthesis). At the transcriptional-level cytokine responses during LOS were characterised by over-representation of IFN-α/ß, IFN-γ, IL-1 and IL-6 signalling pathways and up-regulation of genes for inflammatory responses. Infants with confirmed LOS had significantly higher levels of IL-1α and IL-6 in their plasma. CONCLUSIONS: Blood responses in very preterm infants with LOS are characterised by altered host immune responses that appear to reflect unbalanced immuno-metabolic homeostasis.


Subject(s)
Infant, Extremely Premature , Neonatal Sepsis/immunology , Transcriptome , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Infant, Newborn , Male , Neonatal Sepsis/blood , Neonatal Sepsis/genetics , Signal Transduction
17.
J Immunol ; 205(1): 56-66, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32444390

ABSTRACT

Although substantial progress has been achieved concerning neonatal sepsis, its lethality remains considerably high, and further insights into peculiarities and malfunctions of neonatal immunity are needed. This study aims to contribute to a better understanding of the role of human neonatal granulocyte subpopulations and calgranulin C (S100A12). For this purpose, we gathered 136 human cord blood (CB) samples. CD66b+ CB low-density granulocytes (LDG) and CB normal-density granulocytes were isolated and functionally and phenotypically compared with healthy adult control granulocytes. We could identify CB-LDG as CD66bbright CD64high CD16low CD35low CD10low S100A12med-low and, based on these markers, recovered in whole CB stainings. Consistent with flow cytometric findings, microscopic imaging supported an immature phenotype of CB-LDG with decreased S100A12 expression. In CB serum of healthy neonates, S100A12 was found to be higher in female newborns when compared with males. Additionally, S100A12 levels correlated positively with gestational age independently from sex. We could solidify functional deficits of CB-LDG concerning phagocytosis and generation of neutrophil extracellular traps. Our study reveals that previously described suppressive effects of CB-LDG on CD4+ T cell proliferation are exclusively due to phagocytosis of stimulation beads used in cocultures and absent when using soluble or coated Abs. In conclusion, we characterize CB-LDG as immature neutrophils with functional deficits and decreased expression and storage of S100A12. Concerning their cross-talk with the adaptive immunity, we found no direct inhibitory effect of LDG. Neonatal LDG may thus represent a distinct population that differs from LDG populations found in adults.


Subject(s)
Cell Differentiation/immunology , Fetal Blood/cytology , Granulocytes/immunology , Neonatal Sepsis/immunology , S100A12 Protein/metabolism , Adaptive Immunity , Adult , Antigens, CD/analysis , Antigens, CD/metabolism , Biomarkers/analysis , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Cell Communication/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Female , Fetal Blood/immunology , Flow Cytometry , GPI-Linked Proteins/analysis , GPI-Linked Proteins/metabolism , Granulocytes/metabolism , Healthy Volunteers , Humans , Immunity, Innate , Infant, Newborn , Leukocyte Count , Male , Neonatal Sepsis/blood , Primary Cell Culture , S100A12 Protein/analysis , Sex Factors
18.
Proc Natl Acad Sci U S A ; 117(14): 7941-7949, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32179676

ABSTRACT

Late-onset sepsis (LOS) is a highly consequential complication of preterm birth and is defined by a positive blood culture obtained after 72 h of age. The causative bacteria can be found in patients' intestinal tracts days before dissemination, and cohort studies suggest reduced LOS risk in breastfed preterm infants through unknown mechanisms. Reduced concentrations of epidermal growth factor (EGF) of maternal origin within the intestinal tract of mice correlated to the translocation of a gut-resident human pathogen Escherichia coli, which spreads systemically and caused a rapid, fatal disease in pups. Translocation of Escherichia coli was associated with the formation of colonic goblet cell-associated antigen passages (GAPs), which translocate enteric bacteria across the intestinal epithelium. Thus, maternally derived EGF, and potentially other EGFR ligands, prevents dissemination of a gut-resident pathogen by inhibiting goblet cell-mediated bacterial translocation. Through manipulation of maternally derived EGF and alteration of the earliest gut defenses, we have developed an animal model of pathogen dissemination which recapitulates gut-origin neonatal LOS.


Subject(s)
Bacterial Translocation/immunology , ErbB Receptors/metabolism , Escherichia coli Infections/immunology , Escherichia coli/immunology , Gastrointestinal Microbiome/immunology , Milk, Human/immunology , Neonatal Sepsis/immunology , Animals , Animals, Newborn , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Breast Feeding , Colon/metabolism , Colon/microbiology , Disease Models, Animal , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Feces/chemistry , Feces/microbiology , Female , Humans , Infant, Newborn , Infant, Premature/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice , Mice, Transgenic , Milk, Human/metabolism , Neonatal Sepsis/metabolism , Neonatal Sepsis/microbiology , Signal Transduction/immunology , Time Factors
19.
Expert Opin Pharmacother ; 21(5): 549-556, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32011188

ABSTRACT

Introduction: Mortality due to sepsis is still prevalent, peaking at extreme ages of life including infancy. Despite many efforts, the peculiarity of the infant immune system has limited further advances in its treatment. Indeed, neonates experience a dramatic physiological transition from immune tolerance to the maternal antigens to functional maturity. Such a transition is extremely dynamic, as is the pathophysiology of infant sepsis, which is dependent on many infant, maternal, and environmental factors.Areas covered: In this review, the authors critically update and summarize the current paradigm of immunomodulation in infant sepsis. They confirm how exogenous stimulation of the immune system through intravenous immunoglobulin, colony stimulating factors, and granulocyte transfusion have failed to impact on the prognosis of infant sepsis. They also strongly support the beneficial effects of supplementation/replacement therapies with products naturally contained within maternal milk as well as antioxidant compounds.Expert opinion: Breastfeeding is beneficial against sepsis. Knowledge of the neonatal immune system is indeed too limited to effectively strengthen immune response by exogenous interventions, especially in preterm and low-birth-weight infants. Awareness of this limitation should pave the way for future studies (e.g. gender- and omics-based) aimed at better characterizing the infant immune system and promoting a more tailored approach.


Subject(s)
Infant, Premature, Diseases/drug therapy , Neonatal Sepsis/drug therapy , Adaptive Immunity/drug effects , Antioxidants/therapeutic use , Breast Feeding , Humans , Immunity, Innate/drug effects , Immunoglobulins/therapeutic use , Infant , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/immunology , Milk, Human/immunology , Neonatal Sepsis/immunology , Sex Characteristics , Treatment Outcome
20.
Infect Immun ; 88(3)2020 02 20.
Article in English | MEDLINE | ID: mdl-31818960

ABSTRACT

Neonates are at increased risk for bacterial sepsis. We established that the immune-suppressive cytokine interleukin-27 (IL-27) is elevated in neonatal mice. Similarly, human cord blood-derived macrophages express IL-27 genes and secrete more cytokine than macrophages from adults. In the present work, we hypothesized that increased levels of IL-27 predispose neonatal mice to more severe infection during Gram-negative sepsis. Serum IL-27 levels continued to rise during infection. Peripheral tissue analysis revealed systemic IL-27 expression, while myeloid cell profiling identified Gr-1- and F4/80-expressing cells as the most abundant producers of IL-27 during infection. Increased IL-27 levels were consistent with increased mortality that was improved in IL-27 receptor α (IL-27Rα)-/- mice that lack a functional IL-27 receptor. Infected IL-27Rα-/- pups also exhibited improved weight gain and reduced morbidity. This was consistent with reduced bacterial burdens and more efficient bacterial killing by Ly6B.2+ myeloid cells and macrophages compared to WT neonates. Live animal imaging further supported a more severe and disseminated infection in WT neonates. This is the first report to describe the impact of elevated early-life IL-27 on the host response in a neonatal infection model while also defining the cell and tissue sources of cytokine. IL-27 is frequently associated with suppressed inflammation. In contrast, our findings demonstrate that IL-27 indirectly promotes an inflammatory cytokine response during neonatal sepsis by directly compromising control of bacteria that drive the inflammatory response. Collectively, our results suggest that IL-27 represents a therapeutic target to limit susceptibility and improve infectious outcomes in neonatal sepsis.


Subject(s)
Escherichia coli Infections/immunology , Immunity, Active/immunology , Interleukin-27/metabolism , Neonatal Sepsis/immunology , Animals , Disease Models, Animal , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...