Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(6): 622-632, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28459433

RESUMEN

The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.


Asunto(s)
Calgranulina A/inmunología , Calgranulina B/inmunología , Inmunidad Innata/inmunología , Monocitos/inmunología , Sepsis Neonatal/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Animales Recién Nacidos , Calgranulina A/efectos de los fármacos , Calgranulina B/efectos de los fármacos , Epigénesis Genética , Sangre Fetal , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Immunoblotting , Recién Nacido , Inflamación , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Monocitos/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Sepsis Neonatal/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 4/inmunología
2.
Proc Natl Acad Sci U S A ; 117(14): 7941-7949, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32179676

RESUMEN

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.


Asunto(s)
Traslocación Bacteriana/inmunología , Receptores ErbB/metabolismo , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Microbioma Gastrointestinal/inmunología , Leche Humana/inmunología , Sepsis Neonatal/inmunología , Animales , Animales Recién Nacidos , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Lactancia Materna , Colon/metabolismo , Colon/microbiología , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Escherichia coli/aislamiento & purificación , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Heces/química , Heces/microbiología , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Transgénicos , Leche Humana/metabolismo , Sepsis Neonatal/metabolismo , Sepsis Neonatal/microbiología , Transducción de Señal/inmunología , Factores de Tiempo
3.
Pediatr Res ; 91(1): 230-234, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33731804

RESUMEN

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.


Asunto(s)
Recien Nacido Extremadamente Prematuro , Inflamación/complicaciones , Sepsis Neonatal/complicaciones , Enfermedades Uterinas/complicaciones , Femenino , Humanos , Inmunidad Innata , Recién Nacido , Inflamación/inmunología , Sepsis Neonatal/inmunología , Estudios Retrospectivos , Factores de Riesgo , Enfermedades Uterinas/inmunología
4.
J Immunol ; 205(1): 56-66, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32444390

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Sangre Fetal/citología , Granulocitos/inmunología , Sepsis Neonatal/inmunología , Proteína S100A12/metabolismo , Inmunidad Adaptativa , Adulto , Antígenos CD/análisis , Antígenos CD/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/metabolismo , Comunicación Celular/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Sangre Fetal/inmunología , Citometría de Flujo , Proteínas Ligadas a GPI/análisis , Proteínas Ligadas a GPI/metabolismo , Granulocitos/metabolismo , Voluntarios Sanos , Humanos , Inmunidad Innata , Recién Nacido , Recuento de Leucocitos , Masculino , Sepsis Neonatal/sangre , Cultivo Primario de Células , Proteína S100A12/análisis , Factores Sexuales
5.
Am J Physiol Regul Integr Comp Physiol ; 321(6): R879-R902, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34612068

RESUMEN

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.


Asunto(s)
Antiinflamatorios/uso terapéutico , Sistema Inmunológico/efectos de los fármacos , Mediadores de Inflamación/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Sepsis Neonatal/tratamiento farmacológico , Receptores Toll-Like/efectos de los fármacos , Factores de Edad , Animales , Antiinflamatorios/efectos adversos , Desarrollo Infantil , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad Innata/efectos de los fármacos , Recién Nacido , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Terapia Molecular Dirigida , Sepsis Neonatal/genética , Sepsis Neonatal/inmunología , Sepsis Neonatal/metabolismo , Factores Sexuales , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
6.
Int J Mol Sci ; 22(13)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34208904

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/patogenicidad , Células Supresoras de Origen Mieloide/metabolismo , Sepsis Neonatal/microbiología , Animales , Animales Recién Nacidos , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Recién Nacido , Ratones , Sepsis Neonatal/genética , Sepsis Neonatal/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Receptores Toll-Like/genética
7.
Infect Immun ; 88(3)2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31818960

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Inmunidad Activa/inmunología , Interleucina-27/metabolismo , Sepsis Neonatal/inmunología , Animales , Modelos Animales de Enfermedad , Ratones
8.
Pediatr Res ; 87(2): 399-405, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31689710

RESUMEN

The first days of postnatal life are energetically demanding as metabolic functions change dramatically to accommodate drastic environmental and physiologic transitions after birth. It is increasingly appreciated that metabolic pathways are not only crucial for nutrition but also play important roles in regulating inflammation and the host response to infection. Neonatal susceptibility to infection is increased due to a functionally distinct immune response characterized by high reliance on innate immune mechanisms. Interactions between metabolism and the immune response are increasingly recognized, as changes in metabolic pathways drive innate immune cell function and activation and consequently host response to pathogens. Moreover, metabolites, such as acetyl-coenzyme A (acetyl-CoA) and succinate have immunoregulatory properties and serve as cofactors for enzymes involved in epigenetic reprogramming or "training" of innate immune cells after an initial infectious exposure. Highly sensitive metabolomic approaches allow us to define alterations in metabolic signatures as they change during ontogeny and as perturbed by immunization or infection, thereby linking metabolic pathways to immune cell effector functions. Characterizing the ontogeny of immunometabolism will offer new opportunities to prevent, diagnose, and treat neonatal sepsis.


Asunto(s)
Metabolismo Energético , Inmunidad Innata , Sepsis Neonatal/inmunología , Sepsis Neonatal/metabolismo , Animales , Alimentación con Biberón , Lactancia Materna , Extracción de Leche Materna , Nutrición Enteral , Humanos , Fórmulas Infantiles , Recién Nacido , Metabolómica , Leche Humana/inmunología , Leche Humana/metabolismo , Sepsis Neonatal/diagnóstico , Sepsis Neonatal/terapia , Valor Nutritivo , Valor Predictivo de las Pruebas , Pronóstico
9.
Infect Immun ; 87(5)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30833331

RESUMEN

Gastrointestinal (GI) colonization of 2-day-old (P2) rat pups with Escherichia coli K1 results in translocation of the colonizing bacteria across the small intestine, bacteremia, and invasion of the meninges, with animals frequently succumbing to lethal infection. Infection, but not colonization, is strongly age dependent; pups become progressively less susceptible to infection over the P2-to-P9 period. Colonization leads to strong downregulation of the gene encoding trefoil factor 2 (Tff2), preventing maturation of the protective mucus barrier in the small intestine. Trefoil factors promote mucosal homeostasis. We investigated the contribution of Tff2 to protection of the neonatal rat from E. coli K1 bacteremia and tissue invasion. Deletion of tff2, using clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9, sensitized P9 pups to E. coli K1 bacteremia. There were no differences between tff2-/- homozygotes and the wild type with regard to the dynamics of GI colonization. Loss of the capacity to elaborate Tff2 did not impact GI tract integrity or the thickness of the small-intestinal mucus layer but, in contrast to P9 wild-type pups, enabled E. coli K1 bacteria to gain access to epithelial surfaces in the distal region of the small intestine and exploit an intracellular route across the epithelial monolayer to enter the blood circulation via the mesenteric lymphatic system. Although primarily associated with the mammalian gastric mucosa, we conclude that loss of Tff2 in the developing neonatal small intestine enables the opportunistic neonatal pathogen E. coli K1 to enter the compromised mucus layer in the distal small intestine prior to systemic invasion and infection.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Inmunidad Innata/inmunología , Sepsis Neonatal/inmunología , Factor Trefoil-2/inmunología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Humanos , Ratas
10.
J Pediatr Gastroenterol Nutr ; 69(1): 126-130, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30964820

RESUMEN

OBJECTIVE: The purpose of this study was to evaluate the effects of oropharyngeal colostrum administration in the incidence of late-onset clinical and proven sepsis and in concentrations of immunoglobulin A (IgA) in very-low-birth-weight (VLBW) infants. METHODS: We conducted a double-blinded, randomized, placebo-controlled trial and assigned 113 VLBW infants to receive 0.2 mL of maternal colostrum or sterile water (placebo) via oropharyngeal route every 2 hours for 48 hours, beginning in the first 48 to 72 hours of life. Neonates of both groups were fed breast milk from the first 3 days of life until a volume of at least 100 mL ·â€Škg ·â€Šday. IgA was measured in serum and urine before and after treatment. Clinical data during hospitalization were collected. RESULTS: We found no statistically significant differences between colostrum and placebo groups in the incidence of late-onset clinical sepsis (odds ratio 0.7602; CI 95% 0.3-1.6) and proven sepsis (odds ratio 0.7028; CI 95% 0.3-1.6). The measurement of IgA was similar in serum before (P value 0.87) and after treatment (P value 0.26 day 4 and 0.77 day 18). No differences were also observed in IgA in urine before (P value 0.8) and after treatment (P value 0.73 day 4 and 0.52). CONCLUSIONS: This study could not confirm the hypothesis that oropharyngeal administration of maternal colostrum to VLBW could reduce the incidence of late-onset sepsis and increase the levels of IgA. We believe that this finding can be justified by the practice of feeding VLBW infants exclusively with breast milk in the first days of life and reinforces the prior knowledge of the importance of early nutrition, especially, with human milk. It also suggests that oropharyngeal administration of colostrum should be reserved for neonates who cannot be fed in first few days of life.


Asunto(s)
Calostro/inmunología , Nutrición Enteral/métodos , Sepsis Neonatal/dietoterapia , Lactancia Materna/métodos , Método Doble Ciego , Femenino , Edad Gestacional , Humanos , Inmunoglobulina G/inmunología , Recién Nacido , Recién Nacido de muy Bajo Peso , Masculino , Leche Humana/inmunología , Sepsis Neonatal/inmunología , Sepsis Neonatal/mortalidad
12.
Proc Natl Acad Sci U S A ; 113(19): E2627-35, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27114524

RESUMEN

Interleukin (IL)-18 is an important effector of innate and adaptive immunity, but its expression must also be tightly regulated because it can potentiate lethal systemic inflammation and death. Healthy and septic human neonates demonstrate elevated serum concentrations of IL-18 compared with adults. Thus, we determined the contribution of IL-18 to lethality and its mechanism in a murine model of neonatal sepsis. We find that IL-18-null neonatal mice are highly protected from polymicrobial sepsis, whereas replenishing IL-18 increased lethality to sepsis or endotoxemia. Increased lethality depended on IL-1 receptor 1 (IL-1R1) signaling but not adaptive immunity. In genome-wide analyses of blood mRNA from septic human neonates, expression of the IL-17 receptor emerged as a critical regulatory node. Indeed, IL-18 administration in sepsis increased IL-17A production by murine intestinal γδT cells as well as Ly6G(+) myeloid cells, and blocking IL-17A reduced IL-18-potentiated mortality to both neonatal sepsis and endotoxemia. We conclude that IL-17A is a previously unrecognized effector of IL-18-mediated injury in neonatal sepsis and that disruption of the deleterious and tissue-destructive IL-18/IL-1/IL-17A axis represents a novel therapeutic approach to improve outcomes for human neonates with sepsis.


Asunto(s)
Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Interleucina-18/inmunología , Sepsis Neonatal/inmunología , Sepsis Neonatal/terapia , Tasa de Supervivencia , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales/uso terapéutico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Sepsis Neonatal/patología , Resultado del Tratamiento
13.
Scand J Immunol ; 86(5): 418-423, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28891256

RESUMEN

T cell receptor (TCR) diversity is clearly related to protection from infection. However, the characteristics of TCR diversity in neonates are not clear. In this study, we investigated the TCR diversity of neonates with sepsis. Twenty neonates with severe sepsis and eight matched neonates without infection were enrolled in the study. For the neonates with sepsis, EDTA-anticoagulated blood was collected on day 1 after the diagnosis of sepsis and on day 7 of treatment. For the neonates without infection, blood was collected one time. DNA was extracted from peripheral blood mononuclear cells. The complementarity determining region 3 (CDR3) gene was analysed by multiplex PCR and high-throughput sequencing. The CDR3 types and lengths were similar in patients and healthy controls. There was a significant difference in VJ gene usage among the three groups. Compared to the healthy neonates, the neonates with sepsis had different VJ pairs and generated different clonotypes. Although the TCR ß-chain diversity was generally lower in the neonates with sepsis, there was no significant difference in TCR ß-chain diversity between the patients and the healthy controls. Our data showed the characteristics of the TCR repertoire in neonates with sepsis, which represents a potentially valuable data set. This result is useful for understanding neonatal susceptibility to infection.


Asunto(s)
Regiones Determinantes de Complementariedad/genética , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Sepsis Neonatal/genética , Sepsis Neonatal/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Estudios de Casos y Controles , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Recién Nacido
14.
Med Sci Monit ; 22: 2374-8, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-27389933

RESUMEN

BACKGROUND As a major cause of mortality in neonates, neonatal sepsis is often accompanied by immune dysfunctions, which are frequently caused by dysregulated T cell sub-populations. The role of regulatory B cells in neonatal sepsis, however, remains unknown. Therefore, this study investigated the percentage and functional variation of CD19+CD24hiCD38hi regulatory B cells in peripheral blood of neonatal sepsis patients in an attempt to elucidate the role of these regulatory B cells in pathogenesis of sepsis. MATERIAL AND METHODS Flow cytometry was used to quantify the percentage of CD19+CD24hiCD38hi regulatory B cells from peripheral blood samples. The correlation between B cell percentage and C reactive protein (CRP) level was analyzed. Secretion level of interleukin-10 (IL-10) and effects on the proliferation of naïve CD4+ T cells were further analyzed. RESULTS The percentage of CD19+CD24hiCD38hi regulatory B cells in neonatal sepsis patients was significantly higher compared to healthy controls (p<0.05), and was positively correlated with serum CRP level. The percentage of IL-10+ CD19+CD24hiCD38hi regulatory B cells was also higher in sepsis patients, and also had more potent inhibition on naïve CD4+ T cells (p<0.01). CONCLUSIONS The elevation of CD19+CD24hiCD38hi regulatory B cells in neonatal sepsis can inhibit body immune function and thus may participate in the pathogenesis of sepsis.


Asunto(s)
ADP-Ribosil Ciclasa 1/sangre , Antígenos CD19/sangre , Antígeno CD24/sangre , Glicoproteínas de Membrana/sangre , Sepsis Neonatal/inmunología , Linfocitos B Reguladores/inmunología , Linfocitos B Reguladores/patología , Proteína C-Reactiva/metabolismo , Femenino , Citometría de Flujo , Humanos , Lactante , Recién Nacido , Interleucina-10/sangre , Masculino , Sepsis Neonatal/sangre , Sepsis Neonatal/patología , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/patología , Trombocitopenia/sangre , Trombocitopenia/patología
15.
Front Immunol ; 15: 1365174, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38774873

RESUMEN

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.


Asunto(s)
Animales Recién Nacidos , Sepsis Neonatal , Transducción de Señal , Animales , Ratones , Sepsis Neonatal/inmunología , Sepsis Neonatal/mortalidad , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Modelos Animales de Enfermedad , Femenino , Cardiopatías/etiología , Cardiopatías/inmunología , Pulmón/inmunología , Pulmón/patología , Sepsis/inmunología , Sepsis/metabolismo
16.
J Exp Med ; 221(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38753245

RESUMEN

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.


Asunto(s)
Antígeno CD83 , Sepsis Neonatal , Receptores de Antígenos de Linfocitos T gamma-delta , Adulto , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Antígenos CD/metabolismo , Antígenos CD/genética , Estudios de Cohortes , Recien Nacido Prematuro/inmunología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Sepsis Neonatal/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
18.
Biomed Pharmacother ; 145: 112444, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34808550

RESUMEN

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.


Asunto(s)
Epigénesis Genética , MicroARNs/genética , Sepsis Neonatal/genética , Animales , Biomarcadores/metabolismo , Regulación de la Expresión Génica , Humanos , Recién Nacido , Sepsis Neonatal/diagnóstico , Sepsis Neonatal/inmunología , Pronóstico
19.
J Immunol Res ; 2021: 4884537, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34676267

RESUMEN

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.


Asunto(s)
Proteína C-Reactiva/análisis , Interleucina-10/análisis , Volúmen Plaquetario Medio , Sepsis Neonatal/diagnóstico , Saliva/química , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Humanos , Recién Nacido , Masculino , Sepsis Neonatal/sangre , Sepsis Neonatal/inmunología , Estudios Prospectivos , Curva ROC , Saliva/inmunología
20.
Mol Immunol ; 134: 129-140, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33773156

RESUMEN

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.


Asunto(s)
Células Dendríticas/metabolismo , Sepsis Neonatal/metabolismo , Receptores Inmunológicos/metabolismo , Biomarcadores/metabolismo , Células Dendríticas/inmunología , Femenino , Humanos , Recién Nacido , Masculino , Células Mieloides/inmunología , Células Mieloides/metabolismo , Sepsis Neonatal/inmunología , Receptores Inmunológicos/inmunología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA