Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 318
Filtrar
1.
Glia ; 70(9): 1699-1719, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35579329

RESUMEN

Preterm infants often show pathologies of the cerebellum, which are associated with impaired motor performance, lower IQ and poor language skills at school ages. Using a mouse model of inflammation-induced encephalopathy of prematurity driven by systemic administration of pro-inflammatory IL-1ß, we sought to uncover causes of cerebellar damage. In this model, IL-1ß is administered between postnatal day (P) 1 to day 5, a timing equivalent to the last trimester for brain development in humans. Structural MRI analysis revealed that systemic IL-1ß treatment induced specific reductions in gray and white matter volumes of the mouse cerebellar lobules I and II (5% false discovery rate [FDR]) from P15 onwards. Preceding these MRI-detectable cerebellar volume changes, we observed damage to oligodendroglia, with reduced proliferation of OLIG2+ cells at P10 and reduced levels of the myelin proteins myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) at P10 and P15. Increased density of IBA1+ cerebellar microglia were observed both at P5 and P45, with evidence for increased microglial proliferation at P5 and P10. Comparison of the transcriptome of microglia isolated from P5 cerebellums and cerebrums revealed significant enrichment of pro-inflammatory markers in microglia from both regions, but cerebellar microglia displayed a unique type I interferon signaling dysregulation. Collectively, these data suggest that perinatal inflammation driven by systemic IL-1ß leads to specific cerebellar volume deficits, which likely reflect oligodendrocyte pathology downstream of microglial activation. Further studies are now required to confirm the potential of protective strategies aimed at preventing sustained type I interferon signaling driven by cerebellar microglia as an important therapeutic target.


Asunto(s)
Enfermedades Cerebelosas , Enfermedades del Prematuro , Inflamación , Interferón Tipo I , Interleucina-1beta , Microglía , Animales , Encefalopatías/inducido químicamente , Encefalopatías/inmunología , Encefalopatías/patología , Enfermedades Cerebelosas/inducido químicamente , Enfermedades Cerebelosas/inmunología , Enfermedades Cerebelosas/patología , Cerebelo/efectos de los fármacos , Cerebelo/inmunología , Cerebelo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inducido químicamente , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/patología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Interferón Tipo I/inmunología , Interleucina-1beta/efectos adversos , Interleucina-1beta/farmacología , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/patología , Embarazo
2.
BMC Microbiol ; 21(1): 225, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34362295

RESUMEN

BACKGROUND: Necrotising enterocolitis (NEC) is a devastating bowel disease, primarily affecting premature infants, with a poorly understood aetiology. Prior studies have found associations in different cases with an overabundance of particular elements of the faecal microbiota (in particular Enterobacteriaceae or Clostridium perfringens), but there has been no explanation for the different results found in different cohorts. Immunological studies have indicated that stimulation of the TLR4 receptor is involved in development of NEC, with TLR4 signalling being antagonised by the activated TLR9 receptor. We speculated that differential stimulation of these two components of the signalling pathway by different microbiota might explain the dichotomous findings of microbiota-centered NEC studies. Here we used shotgun metagenomic sequencing and qPCR to characterise the faecal microbiota community of infants prior to NEC onset and in a set of matched controls. Bayesian regression was used to segregate cases from control samples using both microbial and clinical data. RESULTS: We found that the infants suffering from NEC fell into two groups based on their microbiota; one with low levels of CpG DNA in bacterial genomes and the other with high abundances of organisms expressing LPS. The identification of these characteristic communities was reproduced using an external metagenomic validation dataset. We propose that these two patterns represent the stimulation of a common pathway at extremes; the LPS-enriched microbiome suggesting overstimulation of TLR4, whilst a microbial community with low levels of CpG DNA suggests reduction of the counterbalance to TLR4 overstimulation. CONCLUSIONS: The identified microbial community patterns support the concept of NEC resulting from TLR-mediated pathways. Identification of these signals suggests characteristics of the gastrointestinal microbial community to be avoided to prevent NEC. Potential pre- or pro-biotic treatments may be designed to optimise TLR signalling.


Asunto(s)
Enterocolitis Necrotizante/microbiología , Células Epiteliales/inmunología , Microbioma Gastrointestinal/genética , Enfermedades del Prematuro/microbiología , Receptor Toll-Like 4/inmunología , Teorema de Bayes , ADN Bacteriano/genética , Enterocolitis Necrotizante/inmunología , Células Epiteliales/microbiología , Heces/microbiología , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inmunología , Metagenómica , ARN Ribosómico 16S/genética , Receptor Toll-Like 4/genética
3.
Respir Res ; 22(1): 208, 2021 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-34273977

RESUMEN

BACKGROUND: Very preterm infants are at high risk of developing chronic lung disease, which requires respiratory support and might have long-term sequelae. As lung inflammation plays an important role in pathogenesis, antileukotrienes have been explored in both clinical and animal studies. We aimed to assess the benefits and harms of antileukotrienes for the prevention and treatment of respiratory morbidity and mortality in very preterm newborns. METHODS: In this systematic review, we included randomized trials and non-randomized studies in humans and animals reporting the effects of antileukotrienes in very preterm infants or other mammals within 10 days of birth. Our pre-specified primary outcomes were all-cause mortality and any harm, and, for the clinical studies, incidence of chronic lung disease. Included studies underwent risk of bias-assessment and data extraction performed by two authors independently. There were no language restrictions. RESULTS: Fifteen studies totally met our inclusion criteria: one randomized trial and four non-randomized studies in humans and 10 animal studies (five in rodents, two in lambs and one in either guinea pigs, rabbits or caprinae). All five clinical studies used montelukast and had a small sample size, ranging from 4 to 77 infants. The randomized trial (n = 60) found no difference in the incidence of chronic lung disease between the groups. Only one clinical study, which enrolled four very preterm infants and had a critical overall risk of bias, reported long-term outcomes. All other studies had unclear or greater overall risk of bias and meta-analyses were therefore deemed unfeasible. Eight of ten animal studies used leukotriene receptor antagonists as antileukotriene (montelukast in three of ten studies) and seven had an experimental study design (i.e. some animals were not exposed to antileukotrienes but no randomization). Three of the ten animal studies assessed different doses. Animal studies found no effect on the outcomes mortality, growth, or lung function related surrogate outcomes. CONCLUSIONS: Use of antileukotrienes in very preterm infants to prevent or treat chronic lung disease is not supported by the available evidence. Large randomized trials focusing on outcomes relevant to patients, including long-term outcomes, are needed. Studies should also minimize risk of bias.


Asunto(s)
Modelos Animales de Enfermedad , Recien Nacido Extremadamente Prematuro/fisiología , Enfermedades del Prematuro/tratamiento farmacológico , Antagonistas de Leucotrieno/uso terapéutico , Enfermedades Pulmonares/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Animales , Animales Recién Nacidos , Enfermedad Crónica , Humanos , Recién Nacido , Enfermedades del Prematuro/diagnóstico , Enfermedades del Prematuro/inmunología , Enfermedades Pulmonares/diagnóstico , Enfermedades Pulmonares/inmunología
4.
Semin Fetal Neonatal Med ; 25(4): 101146, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-33164775

RESUMEN

The fetus can deploy a local or systemic inflammatory response when exposed to microorganisms or, alternatively, to non-infection-related stimuli (e.g., danger signals or alarmins). The term "Fetal Inflammatory Response Syndrome" (FIRS) was coined to describe a condition characterized by evidence of a systemic inflammatory response, frequently a result of the activation of the innate limb of the immune response. FIRS can be diagnosed by an increased concentration of umbilical cord plasma or serum acute phase reactants such as C-reactive protein or cytokines (e.g., interleukin-6). Pathologic evidence of a systemic fetal inflammatory response indicates the presence of funisitis or chorionic vasculitis. FIRS was first described in patients at risk for intraamniotic infection who presented preterm labor with intact membranes or preterm prelabor rupture of the membranes. However, FIRS can also be observed in patients with sterile intra-amniotic inflammation, alloimmunization (e.g., Rh disease), and active autoimmune disorders. Neonates born with FIRS have a higher rate of complications, such as early-onset neonatal sepsis, intraventricular hemorrhage, periventricular leukomalacia, and death, than those born without FIRS. Survivors are at risk for long-term sequelae that may include bronchopulmonary dysplasia, neurodevelopmental disorders, such as cerebral palsy, retinopathy of prematurity, and sensorineuronal hearing loss. Experimental FIRS can be induced by intra-amniotic administration of bacteria, microbial products (such as endotoxin), or inflammatory cytokines (such as interleukin-1), and animal models have provided important insights about the mechanisms responsible for multiple organ involvement and dysfunction. A systemic fetal inflammatory response is thought to be adaptive, but, on occasion, may become dysregulated whereby a fetal cytokine storm ensues and can lead to multiple organ dysfunction and even fetal death if delivery does not occur ("rescued by birth"). Thus, the onset of preterm labor in this context can be considered to have survival value. The evidence so far suggests that FIRS may compound the effects of immaturity and neonatal inflammation, thus increasing the risk of neonatal complications and long-term morbidity. Modulation of a dysregulated fetal inflammatory response by the administration of antimicrobial agents, anti-inflammatory agents, or cell-based therapy holds promise to reduce infant morbidity and mortality.


Asunto(s)
Corioamnionitis/inmunología , Corioamnionitis/fisiopatología , Trabajo de Parto Prematuro/fisiopatología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Adulto , Corioamnionitis/diagnóstico , Corioamnionitis/terapia , Citocinas/sangre , Femenino , Feto , Humanos , Recién Nacido , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/fisiopatología , Interleucina-6/sangre , Embarazo , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico
5.
PLoS One ; 15(5): e0232933, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32407417

RESUMEN

INTRODUCTION: Deficiencies in innate immune responses may contribute to the increased susceptibility to infection in preterm infants. In vivo cytokine profiles in response to sepsis in very preterm infants are not fully understood. AIMS: To characterise plasma pro- and anti-inflammatory cytokine concentrations and pre-defined ratios in very preterm infants with late-onset sepsis (LOS). METHODS: In this observational study, peripheral blood samples were collected at the time of evaluation for suspected LOS from 31 preterm infants (<30 weeks gestational age). Plasma cytokine concentrations were determined by 12-plex immunoassay. RESULTS: IL-10, IFN-γ, IL-12p70, IP-10, IL-6 and CCL2 were elevated in the majority infants with LOS (n = 12) compared to those without LOS (n = 19). There was no difference in TNF-α, IL-1ß, IL-17AF, IL-8 and IL-15 concentrations between groups. IL-10/TNF-α ratios were increased, while CCL2/IL-10 and IL-12p70/IL-10 ratios were decreased in infants with LOS compared to those without. CONCLUSION: Very preterm infants have a marked innate inflammatory response at the time of LOS. The increase in IL-10/TNF-α ratio may indicate early immune hypo-responsiveness. Longitudinal studies with a larger number of participants are required to understand immune responses and clinical outcomes following LOS in preterm infants.


Asunto(s)
Biomarcadores/sangre , Citocinas/sangre , Enfermedades del Prematuro/diagnóstico , Recien Nacido Prematuro/inmunología , Recién Nacido de muy Bajo Peso/inmunología , Inflamación/diagnóstico , Sepsis/diagnóstico , Australia/epidemiología , Estudios de Casos y Controles , Femenino , Edad Gestacional , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro/sangre , Enfermedades del Prematuro/sangre , Enfermedades del Prematuro/epidemiología , Enfermedades del Prematuro/inmunología , Recién Nacido de muy Bajo Peso/sangre , Inflamación/sangre , Inflamación/epidemiología , Inflamación/inmunología , Masculino , Estudios Prospectivos , Sepsis/sangre , Sepsis/epidemiología , Sepsis/inmunología
6.
Semin Fetal Neonatal Med ; 25(4): 101110, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32303463

RESUMEN

The fetal inflammatory response syndrome (FIRS) is characterized by umbilical cord inflammation and elevated fetal pro-inflammatory cytokines. Surviving neonates, especially very preterm infants, have increased rates of neonatal morbidity including neurodevelopmental impairment. The mechanism of brain injury in FIRS is complex and may involve "multiple hits." Exposure to in utero inflammation initiates a cascade of the fetal immune response, where pro-inflammatory cytokines can cause direct injury to oligodendrocytes and neurons. Activation of microglia results in further injury to vulnerable pre-myelinating oligodendrocytes and influences the integrity of the fetal and newborn's blood-brain barrier, resulting in further exposure of the brain to developmental insults. Newborns exposed to FIRS are frequently exposed to additional perinatal and postnatal insults that can result in further brain injury. Future directions should include evaluations for new therapeutic interventions aimed at reducing brain injury by dampening FIRS, inhibition of microglial activation, and regeneration of immature oligodendrocytes.


Asunto(s)
Lesiones Encefálicas/inmunología , Corioamnionitis/inmunología , Recien Nacido Prematuro/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Citocinas/inmunología , Femenino , Sangre Fetal/inmunología , Humanos , Recién Nacido de Bajo Peso/inmunología , Recién Nacido , Enfermedades del Prematuro/inmunología , Embarazo
7.
Expert Opin Pharmacother ; 21(5): 549-556, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32011188

RESUMEN

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.


Asunto(s)
Enfermedades del Prematuro/tratamiento farmacológico , Sepsis Neonatal/tratamiento farmacológico , Inmunidad Adaptativa/efectos de los fármacos , Antioxidantes/uso terapéutico , Lactancia Materna , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunoglobulinas/uso terapéutico , Lactante , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inmunología , Leche Humana/inmunología , Sepsis Neonatal/inmunología , Caracteres Sexuales , Resultado del Tratamiento
8.
Dig Dis Sci ; 65(3): 789-796, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32008132

RESUMEN

Necrotizing enterocolitis (NEC) is a poorly defined disease that primarily affects preterm infants. There has not been much progress in the prevention or treatment of NEC since it became recognized as a common problem in preterm infants. Reasons for this lack of progress include the likelihood that different diseases are being put under the same moniker of "NEC," similar to using "diabetes" for the different diseases it represents. In order to make progress, better delineation of the phenotypes that present as NEC will be necessary to clearly establish their pathophysiology, find specific and sensitive biomarkers, and establish preventative regimens. In this review, we summarize some of the entities that are being called NEC, discuss the pathophysiology of the most classic form of NEC, and provide an overview of how we might proceed in the future to make progress in this field.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/microbiología , Microbioma Gastrointestinal/fisiología , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/microbiología , Enterocolitis Necrotizante/diagnóstico , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/diagnóstico , Microbiota/fisiología
9.
Curr Pediatr Rev ; 16(2): 148-155, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31951184

RESUMEN

Infants born prematurely (before completion of 37 weeks of gestation) are at increased risk of morbidity and mortality due to vaccine preventable diseases, mostly because of their immunological immaturity and failure of transfer of maternal protective antibodies. Despite their great need of being vaccinated, concerns on vaccine safety and efficacy, constitute the main reasons for which vaccinations are often delayed in this group. In this review we summarize the latest evidence on vaccine safety, efficacy and immunogenicity in preterm infants which is similar to full-term infants. Therefore there is no reason for delaying vaccination in this population.


Asunto(s)
Enfermedades del Prematuro/prevención & control , Vacunación/efectos adversos , Vacunación/métodos , Vacunas/administración & dosificación , Vacunas/efectos adversos , Virosis/prevención & control , Factores de Edad , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inmunología , Seguridad del Paciente , Vacunas/inmunología , Virosis/inmunología
10.
J Neonatal Perinatal Med ; 13(3): 373-380, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31985475

RESUMEN

BACKGROUND: Differences in the susceptibility of preterm infants to develop necrotizing enterocolitis (NEC) implicate potential genetic differences in response to the inflammatory stimuli leading to NEC. Dual specificity phosphatases (DUSPs) are a key suppressor pathway of the mitogen-activated protein kinase (MAPK) pro-inflammatory signaling pathway. We hypothesized that inherited single nucleotide polymorphisms (SNPs) in DUSP genes contribute to NEC susceptibility in premature infants. METHODS: Patients admitted between 2010 and 2015 born at <  32 weeks GA and≤1,500 g BW with stage II+NEC (cases; n = 50) and age, weight-matched controls (n = 38) were included. Blood samples were collected for DNA isolation. Agena Mass Array assay was used to examine 31 SNPs in 9 different DUSP genes. Calculated minor allele frequencies (MAF) for cases and controls were compared using χ2 and logistic regression. RESULTS: The presence of the rs704074 SNP was associated with a 48% decreased risk of developing NEC (OR 0.52; 95% CI 0.27- 1.01, p = 0.04). The odds of surgical NEC decreased by 78% (OR 0.22; 95% CI 0.06- 0.84, p = 0.027) for each copy of rs704074/G allele in patients with NEC. CONCLUSION: In this small single-center pilot study, DUSP-6 SNP (rs704074) was associated with a lower risk of developing NEC and surgical NEC, the most severe form of NEC, in preterm infants.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/genética , Enterocolitis Necrotizante , Enfermedades del Prematuro , Recien Nacido Prematuro/fisiología , Enterocolitis Necrotizante/diagnóstico , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/inmunología , Femenino , Microbioma Gastrointestinal/inmunología , Humanos , Recién Nacido , Enfermedades del Prematuro/diagnóstico , Enfermedades del Prematuro/genética , Enfermedades del Prematuro/inmunología , Mucosa Intestinal/inmunología , Sistema de Señalización de MAP Quinasas/genética , Masculino , Polimorfismo de Nucleótido Simple , Estados Unidos/epidemiología
11.
J Neonatal Perinatal Med ; 13(3): 381-386, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31771081

RESUMEN

BACKGROUND: Neonatal infections are a leading cause of morbi-mortality despite advances in antimicrobials and neonatal care. Preterm infants have greater susceptibility to sepsis due to an immature immune system and lower immunoglobulin levels. Intravenous immunoglobulins (IVIG) have been used in several studies as an adjuvant treatment to improve this physiological immune deficiency, with different outcomes. METHODS: Very low birth weight (VLBW) infants who developed sepsis in the neonatal ICU were studied. They were randomly divided in 2 groups: one group was treated with antibiotics (Group I), and the other received antibiotics plus a 500 mg/kg/day of IVIG during 7 days (Group II). Serum IgG concentration was determined at initiation, during and after treatment Group I, and daily during the 7 days of therapy in Group II. RESULTS: The baseline IgG concentration in group II was 486 g/dL, and increased to 852 mg/dL after the first dose of IVIG (p < 0.01). After the seventh day of infusion a mean IgG level of 1898 mg/dL was achieved. A direct correlation (r = 0.94) between IgG concentration and days of treatment was observed. Blood cultures were positive in 70% of the infants in group I and 75.5% in group II. Staphylococcus epidermidis was the most frequent isolated bacteria in blood cultures. The lethality rate was 25.0% in group I and 5.0% in Group II (p < 0.03). We did not observe collateral effects with the administration of IVIG. CONCLUSIONS: Prolonged therapy with IVIG seems to be safe and effective as an adjuvant treatment in VLBW infants with sepsis.


Asunto(s)
Duración de la Terapia , Inmunoglobulinas Intravenosas/administración & dosificación , Enfermedades del Prematuro , Recien Nacido Prematuro/inmunología , Sepsis Neonatal , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Inmunoglobulina G/sangre , Factores Inmunológicos/administración & dosificación , Lactante , Recién Nacido , Enfermedades del Prematuro/diagnóstico , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/microbiología , Enfermedades del Prematuro/terapia , Recién Nacido de muy Bajo Peso/fisiología , Masculino , Sepsis Neonatal/diagnóstico , Sepsis Neonatal/inmunología , Sepsis Neonatal/microbiología , Sepsis Neonatal/terapia , Staphylococcus epidermidis/aislamiento & purificación , Resultado del Tratamiento
12.
Hosp Pediatr ; 8(11): 693-698, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30373887

RESUMEN

OBJECTIVES: Immunizations provide important protection from serious childhood illnesses. Infant chronic lung disease (CLD) is a serious complication of prematurity and predisposes premature infants to respiratory morbidity, rehospitalization, and mortality. This high-risk group is especially vulnerable to infections, such as invasive pneumococcal disease, influenza, and bronchiolitis. Our purpose for this project was to increase 2-, 4-, and 6-month immunization rates in eligible infants with CLD in the NICU by 30% through December 2016. METHODS: A multidisciplinary team developed weekly targeted rounds to identify eligible patients with outstanding immunizations. Exclusion criteria included the following: (1) a fraction of inspired oxygen requirement of >80%, (2) pulmonary hypertensive crisis, (3) positive blood culture results or if within 48 hours of a sepsis evaluation, (4) if within 5 days of a surgical or interventional procedure, (5) receiving steroid treatment (not including a physiologic hydrocortisone dose for adrenal insufficiency), (6) a CLD team consensus of contraindication, and (7) parental refusal. RESULTS: The project managed 60 patients from March 2016 to December 2016. Immunization of eligible patients increased from 44% to 75% and was sustained for the next 6 months. The average number of days from admission to immunization record review decreased from 71 days at baseline to 27 days. CONCLUSIONS: The implementation of (1) an in-hospital immunization record review, (2) an e-mail reminder, (3) a weekly multidisciplinary eligibility discussion, and (4) an updated rounding tool was successful in increasing and sustaining immunization rates in this population of infants with CLD. The multidisciplinary CLD meeting was a novel opportunity to discuss immunization eligibility and safety monitoring.


Asunto(s)
Enfermedad Crónica/prevención & control , Inmunización/estadística & datos numéricos , Enfermedades del Prematuro/prevención & control , Recien Nacido Prematuro , Enfermedades Pulmonares/prevención & control , Vacunación/estadística & datos numéricos , Dexametasona , Femenino , Glucocorticoides , Investigación sobre Servicios de Salud , Humanos , Lactante , Recién Nacido , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/fisiopatología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/fisiopatología , Masculino , Guías de Práctica Clínica como Asunto , Mejoramiento de la Calidad
13.
J Pediatr Gastroenterol Nutr ; 66(6): e153-e157, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29470288

RESUMEN

OBJECTIVES: The aim of this study was to determine the effect of digested whole human milk (HM; first sample available after birth from mothers of premature infants) on inflammation, oxidative stress, and cytotoxicity in Caco-2 human intestinal epithelial cells stimulated with lipopolysaccharides or tumor necrosis factor (TNF) to mimic the potential in vivo insults facing the premature infant's gastrointestinal tract. METHODS: Fully differentiated Caco-2 cells were exposed to digested HM (n = 10; samples from 10 different individuals) before stimulation with lipopolysaccharides, TNF, or no stimulation overnight. Inflammation was determined by production of interleukin-8, oxidative stress by levels of F2-isoprostane, and cytotoxicity by released lactate dehydrogenase. RESULTS: HM significantly suppressed interleukin-8 production and cytotoxicity in TNF-stimulated cells, while also suppressing cell death under baseline conditions. Individual HM samples differed widely in their ability to modulate cellular responses. CONCLUSIONS: Results from this study provide evidence that digested HM can reduce both an exaggerated inflammatory response and intestinal damage that contribute to the pathogenesis of necrotizing enterocolitis.


Asunto(s)
Muerte Celular/inmunología , Enterocolitis Necrotizante/prevención & control , Enfermedades del Prematuro/prevención & control , Recien Nacido Prematuro/inmunología , Inflamación/prevención & control , Mucosa Intestinal/inmunología , Leche Humana/inmunología , Biomarcadores/metabolismo , Células CACO-2 , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Humanos , Recién Nacido , Recien Nacido Prematuro/metabolismo , Enfermedades del Prematuro/etiología , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/metabolismo , Inflamación/etiología , Inflamación/inmunología , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Lipopolisacáridos , Leche Humana/metabolismo , Estrés Oxidativo/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
14.
Arch Dis Child Fetal Neonatal Ed ; 103(4): F391-F394, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29382648

RESUMEN

One in 10 newborns will be born before completion of 36 weeks' gestation (premature birth). Infection and sepsis in preterm infants remain a significant clinical problem that represents a substantial financial burden on the healthcare system. Many factors predispose premature infants for having the greatest risk of developing and succumbing to infection as compared with all other age groups across the age spectrum. It is clear that the immune system of preterm infants exhibits distinct, rather than simply deficient, function as compared with more mature and older humans and that the immune function in preterm infants contributes to infection risk. While no single review can cover all aspects of immune function in this population, we will discuss key aspects of preterm neonatal innate and adaptive immune function that place them at high risk for developing infections and sepsis, as well as sepsis-associated morbidity and mortality.


Asunto(s)
Enfermedades del Prematuro/inmunología , Infecciones/inmunología , Sepsis/inmunología , Inmunidad Adaptativa/fisiología , Linfocitos B/inmunología , Humanos , Inmunidad Celular/fisiología , Inmunidad Innata/fisiología , Inmunoglobulinas/inmunología , Recien Nacido Prematuro
15.
J Matern Fetal Neonatal Med ; 31(9): 1227-1233, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-28423971

RESUMEN

Preterm birth accounts for nearly one million deaths among children under five years of age, and although its etiopathogenesis is not fully elucidated, ascending intrauterine infection and fetal inflammatory response seem to be the main triggers. The intense inflammatory response mediated by IL-1ß, TNF-α, PAF, IFN-γ and IL-6, PGE2 and MMP-1 and MMP-9 causes fetal membrane damage and rupture, increased uterine contractions and biochemical and structural changes in the cervix. Furthermore, preterm neonates have deficient innate and adaptive immune responses characterized by reduced levels of IgG, opsonization and phagocytosis, as well as increased activation of Th1 cells in relation to Th2 cells. Therefore, this triad is favors the occurrence of neonatal complications, such as respiratory distress syndrome, necrotizing enterocolitis, retinopathy of prematurity and bronchopulmonary dysplasia. Due to serious maternal and child health complications of intrauterine infection, several studies have tried to identify biomarkers for the early diagnosis of this entity. This literature review aims to discuss the main scientific findings regarding the association between ascending intrauterine infection, immune system and preterm birth.


Asunto(s)
Sistema Inmunológico/inmunología , Infecciones/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Nacimiento Prematuro/inmunología , Enfermedades Uterinas/inmunología , Biomarcadores/análisis , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro/inmunología , Enfermedades del Prematuro/epidemiología , Enfermedades del Prematuro/inmunología , Inflamación/inmunología , Embarazo
18.
Br J Nutr ; 117(7): 994-1000, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28443531

RESUMEN

The aim of the study was to determine whether routine probiotic supplementation (RPS) with Lactobacillus rhamnosus GG (LGG) or Lactobacillus acidophilus +Lactobacillus bifidum is associated with reduced risk of necrotising enterocolitis (NEC)≥Stage II in preterm neonates born at ≤32 weeks' gestation. We conducted a retrospective cohort study on the effect of probiotic supplementation in very low birth weight infants in our neonatal unit by comparing two periods: before and after supplementation. The incidence of NEC≥Stage II, late-onset sepsis and all-cause mortality was compared for an equal period 'before' (Period I) and 'after' (Period II) RPS with LGG or L. acidophillus+L. bifidum. Multivariate logistic regression analysis was conducted to adjust for relevant confounders. The study population was composed of 261 neonates (Period I v. II: 134 v. 127) with comparable gestation duration and birth weights. In <32 weeks, we observed a significant reduction in NEC≥Stage II (11·3 v. 4·8 %), late-onset sepsis (16 v. 10·5 %) and mortality (19·4 v. 2·3 %). The benefits in neonates aged ≤27 weeks did not reach statistical significance. RPS with LGG or L. acidophillus+L. bifidum is associated with a reduced risk of NEC≥Stage II, late-onset sepsis and mortality in preterm neonates born at ≤32 weeks' gestation.


Asunto(s)
Infección Hospitalaria/prevención & control , Enterocolitis Necrotizante/prevención & control , Microbioma Gastrointestinal , Fenómenos Fisiológicos Nutricionales del Lactante , Enfermedades del Prematuro/prevención & control , Nacimiento Prematuro/terapia , Probióticos/uso terapéutico , Estudios de Cohortes , Terapia Combinada , Infección Hospitalaria/epidemiología , Infección Hospitalaria/inmunología , Infección Hospitalaria/microbiología , Enterocolitis Necrotizante/epidemiología , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/microbiología , Microbioma Gastrointestinal/inmunología , Humanos , Incidencia , Lactante , Mortalidad Infantil , Recién Nacido , Enfermedades del Prematuro/epidemiología , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/microbiología , Recién Nacido de muy Bajo Peso , Unidades de Cuidado Intensivo Neonatal , Lactobacillus acidophilus/inmunología , Levilactobacillus brevis/inmunología , Lacticaseibacillus rhamnosus/inmunología , Guías de Práctica Clínica como Asunto , Nacimiento Prematuro/inmunología , Nacimiento Prematuro/microbiología , Nacimiento Prematuro/fisiopatología , Probióticos/efectos adversos , Estudios Retrospectivos , Riesgo , Sepsis/epidemiología , Sepsis/inmunología , Sepsis/microbiología , Sepsis/prevención & control , España/epidemiología
19.
Semin Perinatol ; 41(1): 29-35, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27986328

RESUMEN

Necrotizing enterocolitis (NEC), a disease most commonly seen in preterm infants, often presents without warning and is associated with very high mortality and morbidity. Progress in the prevention and treatment of NEC has been slow. In this article, we will discuss some of the reasons as to why this progress has been slow. We will describe some of the factors that appear to be highly associated and important components in the pathophysiology of NEC. We will discuss the intestinal microbial environment of the fetus as well as the preterm infant and how interaction of dysbiosis with an immature gastrointestinal tract combined with dietary factors play a role in the pathogenesis of NEC. Testable hypotheses are discussed as well as how these may lead to not only a better understanding of the pathophysiology of the disease but also the preventative strategies.


Asunto(s)
Enterocolitis Necrotizante/fisiopatología , Microbioma Gastrointestinal , Enfermedades del Prematuro/fisiopatología , Lactancia Materna , Disbiosis/inmunología , Disbiosis/fisiopatología , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/prevención & control , Microbioma Gastrointestinal/inmunología , Humanos , Inmunidad Innata , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/prevención & control , Intestinos/embriología , Intestinos/microbiología , Intestinos/fisiopatología
20.
Semin Perinatol ; 41(1): 61-69, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27836427

RESUMEN

An underlying genetic predisposition to necrotizing enterocolitis (NEC) is increasingly being recognized. Candidate gene or pathway approaches as well as genome-wide approaches are beginning to identify potential pathogenic variants for NEC in premature infants. However, a majority of these studies have not yielded definitive results because of limited sample size and lack of validation. Despite these challenges, understanding the contribution of genetic variation to NEC is important for providing new insights into the pathogenesis of NEC as well as allowing for targeted care of infants with inherent susceptibility. In this review we provide a summary of published genetic association studies in NEC along with defining the challenges and possible future approaches.


Asunto(s)
Enterocolitis Necrotizante/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Enfermedades del Prematuro/genética , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/terapia , Regulación del Desarrollo de la Expresión Génica/inmunología , Variación Genética , Humanos , Inmunidad Innata , Recien Nacido Extremadamente Prematuro , Recién Nacido , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/prevención & control , Enfermedades del Prematuro/terapia , Inflamación , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiopatología , Terapia Molecular Dirigida/tendencias , Mutación/genética , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...