Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.017
1.
PLoS One ; 18(4): e0284096, 2023.
Article En | MEDLINE | ID: mdl-37023080

Neurodevelopmental impairment is a significant complication among survivors of preterm birth. To improve outcomes, reliable biomarkers for early detection of brain injury and prognostic assessment are required. Secretoneurin is a promising early biomarker of brain injury in adults and full-term neonates suffering from perinatal asphyxia. Data on preterm infants is currently lacking. The aim of this pilot study was to determine secretoneurin concentrations in preterm infants in the neonatal period, and to assess secretoneurin's potential as a biomarker of preterm brain injury. We included 38 very preterm infants (VPI) born at <32 weeks' gestation in the study. Secretoneurin concentrations were measured in serum samples obtained from the umbilical cord, at 48 hours and 3 weeks of life. Outcome measures included repeated cerebral ultrasonography, magnetic resonance imaging at term-equivalent age, general movements assessment, and neurodevelopmental assessment at a corrected age of 2 years by the Bayley Scales of Infant and Toddler Development, third edition (Bayley-III). In comparison to a term-born reference population, VPI had lower secretoneurin serum concentrations in umbilical cord blood and blood collected at 48 hours of life. When measured at 3 weeks of life, concentrations correlated with gestational age at birth. Secretoneurin concentrations did not differ between VPI with an imaging-based diagnosis of brain injury and those without, but when measured in umbilical cord blood and at 3 weeks of life correlated with and were predictive of Bayley-III motor and cognitive scale scores. Secretoneurin levels in VPI differ from term-born neonates. Secretoneurin seems unsuitable as a diagnostic biomarker of preterm brain injury, but bears some prognostic potential and is worthy of further investigation as a blood-based biomarker of preterm brain injury.


Brain Injuries , Infant, Premature, Diseases , Premature Birth , Infant , Pregnancy , Female , Humans , Infant, Newborn , Child, Preschool , Infant, Premature , Pilot Projects , Premature Birth/pathology , Brain Injuries/diagnosis , Brain Injuries/pathology , Gestational Age , Biomarkers , Infant, Premature, Diseases/pathology , Brain/diagnostic imaging , Brain/pathology
2.
Pediatr Res ; 94(3): 979-986, 2023 09.
Article En | MEDLINE | ID: mdl-36934213

BACKGROUND: Preterm birth adversely impacts brain development and contributes to neurodevelopmental impairment; the temporal lobe may be particularly vulnerable to the impact of very preterm (VP) birth. Yet, no prior magnetic resonance imaging (MRI) scoring system incorporated a method to quantify temporal lobe size in VP infants. METHODS: We developed and applied three metrics (temporal lobe length, extra-axial space, and temporal horn width) to quantify temporal lobe structure on term-equivalent brain MRIs obtained from 74 VP and 16 term infants. We compared metrics between VP and term infants and explored associations of each metric with perinatal risk factors. RESULTS: All metrics had excellent reliability (intra-class correlation coefficient 0.62-0.98). VP infants had lower mean temporal lobe length (76.8 mm versus 79.2 mm, p = 0.02); however, the difference attenuated after correction for postmenstrual age. VP infants had larger temporal horn widths compared with term infants (2.6 mm versus 1.8 mm, p < 0.001). Temporal lobe length was positively associated with gestational age, birth weight, and male sex, and negatively associated with the duration of parenteral nutrition. CONCLUSIONS: The proposed metrics are reliable and sensitive in distinguishing differences in temporal lobe development between VP and full-term infants. IMPACT: We developed a novel method for quantifying temporal lobe size among very preterm infants at term equivalent using simple metrics performed on brain MRI. Temporal lobe metrics were reliable, correlated with brain volume from volumetric analysis, and were sensitive in identifying differences in temporal lobe development among preterm compared with term infants, specifically larger temporal horn size in preterm infants. This temporal lobe metric system will enable future work to delineate the perinatal and postnatal factors that impact temporal lobe growth, and better understand the relationship between temporal lobe disturbance and neurodevelopment in very preterm infants.


Infant, Premature, Diseases , Premature Birth , Infant , Pregnancy , Female , Humans , Infant, Newborn , Male , Infant, Premature , Reproducibility of Results , Benchmarking , Brain , Magnetic Resonance Imaging/methods , Gestational Age , Fetal Growth Retardation/pathology , Infant, Premature, Diseases/pathology , Temporal Lobe/diagnostic imaging
3.
Oxid Med Cell Longev ; 2022: 9010354, 2022.
Article En | MEDLINE | ID: mdl-36193058

Background: Bronchopulmonary dysplasia (BPD) is one of the most serious complications in premature infants. Myeloid-derived suppressor cells (MDSCs) have been indicated to promote immune tolerance and induce anti-inflammatory responses during the neonatal stage. However, the role of MDSCs in BPD has not been completely expounded. Methods: 130 cases of newborns were collected from six tertiary hospitals in Guangzhou from August 2019 to June 2022. They were divided into BPD group, non-BPD preterm infants group, and term infants group according to gestational age and presence of BPD. The peripheral blood was collected and used to analyze the proportion, phenotypic, and function of MDSCs at 3 to 7 days and 8 to 14 days after birth, respectively. Results: We indicated that the number of both MDSCs in premature infants is reduced, and the number of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in peripheral blood of BPD infants was significantly lower than that of non-BPD infants under 34 weeks of gestational age (P < 0.05). Furthermore, PMN-MDSCs from peripheral blood of patients presented inhibitory effect on proliferation of CD4+T and CD8+T cells in each group. However, PMN-MDSCs from BPD group had obviously weaker inhibitory effect on proliferation of CD4+T and CD8+T cells than that from non-BPD preterm infants group. In addition, we demonstrated that the expression of NADPH oxidase (Nox2) and reactive oxygen species (ROS) in PMN-MDSCs of BPD children was significantly lower than that in non-BPD preterm infants, suggesting that ROS pathway was affected in BPD in premature infants. Conclusion: This study preliminarily revealed the role of PMN-MDSCs in the pathogenesis of BPD in premature infants. The specific immune regulation mechanism of PMN-MDSCs in BPD will provide new ideas and strategies for clinical prevention and treatment of BPD in premature infants.


Bronchopulmonary Dysplasia , Infant, Premature, Diseases , Myeloid-Derived Suppressor Cells , Anti-Inflammatory Agents/metabolism , Bronchopulmonary Dysplasia/metabolism , Child , Humans , Infant , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/pathology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism
4.
Glia ; 70(9): 1699-1719, 2022 09.
Article En | MEDLINE | ID: mdl-35579329

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.


Cerebellar Diseases , Infant, Premature, Diseases , Inflammation , Interferon Type I , Interleukin-1beta , Microglia , Animals , Brain Diseases/chemically induced , Brain Diseases/immunology , Brain Diseases/pathology , Cerebellar Diseases/chemically induced , Cerebellar Diseases/immunology , Cerebellar Diseases/pathology , Cerebellum/drug effects , Cerebellum/immunology , Cerebellum/pathology , Disease Models, Animal , Female , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/chemically induced , Infant, Premature, Diseases/immunology , Infant, Premature, Diseases/pathology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interferon Type I/immunology , Interleukin-1beta/adverse effects , Interleukin-1beta/pharmacology , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Pregnancy
5.
Sci Rep ; 12(1): 4498, 2022 03 16.
Article En | MEDLINE | ID: mdl-35296792

To investigate the relationship between morphine exposure in the first week of life and brain injury on term-equivalent age magnetic resonance imaging (MRI) in very preterm infants. A retrospective study included 106 infants with a birth weight of < 1500 g who were born at King Saud Medical City at ≤ 32 gestational weeks, were admitted to the neonatal intensive care unit, and underwent term-equivalent age or pre-discharge brain MRI. A univariate analysis in addition to modified log-Poisson regression with a robust variance estimator was applied, and the effect of early morphine exposure and cumulative dose in the first seven days on brain morphology and growth at term-equivalent age was determined using the Kidokoro score. Sixty-eight (64.2%) infants had received morphine in the first week of life (median cumulative dose: 1.68 mg/kg, interquartile range 0.48-2.52 mg/kg). Early initiation of morphine administration was significantly associated with high total white matter (adjusted relative risk [aRR] 1.32, 95% confidence interval [CI] 1.01-1.72) and cerebellum (aRR 1.36, 95% CI 1.03-1.81) scores and a small cerebellar volume (aRR 1.28, 95% CI 1.02-1.61). Morphine exposure in the first week of life was independently associated with white matter and cerebellar injury on term-equivalent age brain MRI in very preterm infants.


Infant, Premature, Diseases , Morphine , Brain/diagnostic imaging , Brain/pathology , Female , Fetal Growth Retardation/pathology , Gestational Age , Humans , Infant , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/pathology , Magnetic Resonance Imaging/methods , Morphine/adverse effects , Retrospective Studies
6.
Pediatr Res ; 92(1): 299-306, 2022 07.
Article En | MEDLINE | ID: mdl-33654289

BACKGROUND: Noninvasive advanced neuroimaging and neurochemical assessment can identify subtle abnormalities and predict neurodevelopmental impairments. Our objective was to quantify white matter metabolite levels and evaluate their relationship with neurodevelopmental outcomes at age 3 years. METHODS: Our study evaluated a longitudinal prospective cohort of very premature infants (<32 weeks gestational age) with single-voxel proton magnetic resonance spectroscopy from the centrum semiovale performed at term-equivalent age and standardized cognitive, verbal, and motor assessments at 3 years corrected age. We separately examined metabolite ratios in the left and right centrum semiovale. We also conducted an exploratory interaction analysis for high/low socioeconomic status (SES) to evaluate the relationship between metabolites and neurodevelopmental outcomes, after adjusting for confounders. RESULTS: We found significant relationships between choline/creatine levels in the left and right centrum semiovale and motor development scores. Exploratory interaction analyses revealed that, for infants with low SES, there was a negative association between choline/creatine in the left centrum semiovale and motor assessment scores at age 3 years. CONCLUSIONS: Brain metabolites from the centrum semiovale at term-equivalent age were associated with motor outcomes for very preterm infants at 3 years corrected age. This effect may be most pronounced for infants with low SES. IMPACT: Motor development at 3 years corrected age for very preterm infants is inversely associated with choline neurochemistry within the centrum semiovale on magnetic resonance spectroscopy at term-equivalent age, especially in infants with low socioeconomic status. No prior studies have studied metabolites in the centrum semiovale to predict neurodevelopmental outcomes at 3 years corrected age based on high/low socioeconomic status. For very preterm infants with lower socioeconomic status, higher choline-to-creatine ratio in central white matter is associated with worse neurodevelopmental outcomes.


Creatine , Infant, Premature, Diseases , Brain , Child, Preschool , Choline , Creatine/metabolism , Female , Fetal Growth Retardation/metabolism , Humans , Infant , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/pathology , Magnetic Resonance Spectroscopy , Prospective Studies
7.
J Neurosurg Pediatr ; 29(1): 10-20, 2022 Jan 01.
Article En | MEDLINE | ID: mdl-34653990

OBJECTIVE: Posthemorrhagic hydrocephalus (PHH) is associated with significant morbidity, smaller hippocampal volumes, and impaired neurodevelopment in preterm infants. The timing of temporary CSF (tCSF) diversion has been studied; however, the optimal time for permanent CSF (pCSF) diversion is unknown. The objective of this study was to determine whether cumulative ventricle size or timing of pCSF diversion is associated with neurodevelopmental outcome and hippocampal size in preterm infants with PHH. METHODS: Twenty-five very preterm neonates (born at ≤ 32 weeks' gestational age) with high-grade intraventricular hemorrhage (IVH), subsequent PHH, and pCSF diversion with a ventriculoperitoneal shunt (n = 20) or endoscopic third ventriculostomy (n = 5) were followed until 2 years of age. Infants underwent serial cranial ultrasounds from birth until 1 year after pCSF diversion, brain MRI at term-equivalent age, and assessment based on the Bayley Scales of Infant and Toddler Development, Third Edition, at 2 years of age. Frontooccipital horn ratio (FOHR) measurements were derived from cranial ultrasounds and term-equivalent brain MRI. Hippocampal volumes were segmented and calculated from term-equivalent brain MRI. Cumulative ventricle size until the time of pCSF diversion was estimated using FOHR measurements from each cranial ultrasound performed prior to permanent intervention. RESULTS: The average gestational ages at tCSF and pCSF diversion were 28.9 and 39.0 weeks, respectively. An earlier chronological age at the time of pCSF diversion was associated with larger right hippocampal volumes on term-equivalent MRI (Pearson's r = -0.403, p = 0.046) and improved cognitive (r = -0.554, p = 0.047), motor (r = -0.487, p = 0.048), and language (r = -0.414, p = 0.021) outcomes at 2 years of age. Additionally, a smaller cumulative ventricle size from birth to pCSF diversion was associated with larger right hippocampal volumes (r = -0.483, p = 0.014) and improved cognitive (r = -0.711, p = 0.001), motor (r = -0.675, p = 0.003), and language (r = -0.618, p = 0.011) outcomes. There was no relationship between time to tCSF diversion or cumulative ventricle size prior to tCSF diversion and neurodevelopmental outcome or hippocampal size. Finally, a smaller cumulative ventricular size prior to either tCSF diversion or pCSF diversion was associated with a smaller ventricular size 1 year after pCSF diversion (r = 0.422, p = 0.040, R2 = 0.178 and r = 0.519, p = 0.009, R2 = 0.269, respectively). CONCLUSIONS: In infants with PHH, a smaller cumulative ventricular size and shorter time to pCSF diversion were associated with larger right hippocampal volumes, improved neurocognitive outcomes, and reduced long-term ventriculomegaly. Future prospective randomized studies are needed to confirm these findings.


Cerebral Intraventricular Hemorrhage/complications , Cerebral Ventricles/pathology , Hydrocephalus/pathology , Hydrocephalus/surgery , Time-to-Treatment , Child Development , Hippocampus/pathology , Humans , Hydrocephalus/etiology , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/pathology , Infant, Premature, Diseases/surgery , Longitudinal Studies , Neuroendoscopy/methods , Ventriculoperitoneal Shunt/methods , Ventriculostomy/methods
8.
Pediatr Nephrol ; 37(6): 1277-1284, 2022 06.
Article En | MEDLINE | ID: mdl-34761299

Advances in the care of neonates to the extreme limits of viability have increased the risk of severe comorbidities in surviving preemies. The respiratory and the neurodevelopmental consequences of premature birth and/or intra-uterine growth retardation have been well described. Because of the usual clinical silence of the kidney, the long-term renal consequences of low birth weight have not been as well studied. A case report illustrates the risk factors associated with low birth weight and prematurity and discusses the pathogenesis of the late consequences of the congenital nephron deficit associated with a low birth weight. Practical recommendations are given for a tight follow-up of these newly born preemies.


Infant, Newborn, Diseases , Infant, Premature, Diseases , Neonatology , Female , Humans , Infant, Low Birth Weight , Infant, Newborn , Infant, Premature, Diseases/pathology , Intensive Care Units, Neonatal , Nephrons/pathology , Pregnancy
9.
Cells ; 10(8)2021 07 28.
Article En | MEDLINE | ID: mdl-34440681

Globally, approximately 11% of all infants are born preterm, prior to 37 weeks' gestation. In these high-risk neonates, encephalopathy of prematurity (EoP) is a major cause of both morbidity and mortality, especially for neonates who are born very preterm (<32 weeks gestation). EoP encompasses numerous types of preterm birth-related brain abnormalities and injuries, and can culminate in a diverse array of neurodevelopmental impairments. Of note, posthemorrhagic hydrocephalus of prematurity (PHHP) can be conceptualized as a severe manifestation of EoP. PHHP impacts the immature neonatal brain at a crucial timepoint during neurodevelopment, and can result in permanent, detrimental consequences to not only cerebrospinal fluid (CSF) dynamics, but also to white and gray matter development. In this review, the relevant literature related to the diverse mechanisms of cell death in the setting of PHHP will be thoroughly discussed. Loss of the epithelial cells of the choroid plexus, ependymal cells and their motile cilia, and cellular structures within the glymphatic system are of particular interest. Greater insights into the injuries, initiating targets, and downstream signaling pathways involved in excess cell death shed light on promising areas for therapeutic intervention. This will bolster current efforts to prevent, mitigate, and reverse the consequential brain remodeling that occurs as a result of hydrocephalus and other components of EoP.


Cell Death , Hydrocephalus/pathology , Infant, Premature, Diseases/pathology , Brain/growth & development , Brain/metabolism , Brain/pathology , Choroid Plexus/cytology , Choroid Plexus/metabolism , Cilia/metabolism , Ependyma/cytology , Ependyma/metabolism , Humans , Hydrocephalus/cerebrospinal fluid , Hydrocephalus/genetics , Infant, Premature, Diseases/cerebrospinal fluid , Infant, Premature, Diseases/genetics , Premature Birth , Signal Transduction
10.
Sci Rep ; 11(1): 13777, 2021 07 02.
Article En | MEDLINE | ID: mdl-34215818

Necrotizing enterocolitis (NEC) and focal intestinal perforation (FIP) are two of the most common emergencies of the gastrointestinal tract in preterm infants with very low birth weight (VLBW, birth weight < 1500 g). Identification of risk factors among these children is crucial for earlier diagnosis and prompt intervention. In this study, we investigated a relationship between ABO blood groups and the risk for surgical NEC/FIP. We genotyped the ABO locus (rs8176746 and rs8176719) in VLBW infants enrolled in a prospective, population-based cohort study of the German Neonatal Network (GNN). Of the 10,257 VLBW infants, 441 (4.3%) had surgical NEC/FIP. In univariate analyses, the blood group AB was more prevalent in VLBW infants with surgical NEC/FIP compared to non-AB blood groups (OR 1.51, 95% CI 1.07-2.13, p = 0.017; absolute risk difference 2.01%, 95% CI 0.06-3.96%). The association between blood group AB and surgical NEC/FIP was observed in a multivariable logistic regression model (OR of 1.58, 95% CI 1.10-2.26, p = 0.013) as well. In summary, our study suggests that the risk of surgical NEC and FIP is higher in patients with blood group AB and lower in those having non-AB blood groups.


ABO Blood-Group System/blood , Enterocolitis, Necrotizing/epidemiology , Infant, Newborn, Diseases/epidemiology , Infant, Premature, Diseases/epidemiology , Intestinal Perforation/epidemiology , Child, Preschool , Enterocolitis, Necrotizing/blood , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/surgery , Female , Fetal Diseases/blood , Fetal Diseases/pathology , Fetal Diseases/surgery , Gestational Age , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases/blood , Infant, Newborn, Diseases/pathology , Infant, Newborn, Diseases/surgery , Infant, Premature/blood , Infant, Premature, Diseases/pathology , Infant, Premature, Diseases/surgery , Infant, Very Low Birth Weight , Intestinal Perforation/blood , Intestinal Perforation/pathology , Intestinal Perforation/surgery , Male , Risk Factors
11.
Neurosci Lett ; 758: 136013, 2021 07 27.
Article En | MEDLINE | ID: mdl-34111510

Preterm infants often suffer from impaired postnatal brain development, and glutamate excitotoxicity is identified as a pivotal mechanism of hyperoxia-induced neurological abnormality. We aimed to investigate the effect of short time hyperoxia on glutamate homeostasis and glutamate transporters expressions in immature brain. Six-day-old (P6) rat pups were exposed to 80% oxygen for 24 h (the hyperoxia group) or placed in atmospheric air (the control group). The concentrations of glutamate and γ-aminobutyric acid (GABA) in immature cerebrum and cerebellum at P7, P14 and P21 were determined by ELISA. The mRNA levels of glutamate transporters including excitatory amino acid transporter 1 (EAAT1), EAAT2, EAAT3, vesicular glutamate transporter 1 (VGLUT1) and VGLUT2 in brain were determined by qPCR. Glutamate accumulation was induced by hyperoxia both in immature cerebrum and cerebellum at P7 but got gradually attenuated at P14 and P21, as evidenced by the changes of glutamate and GABA concentrations. Hyperoxia also induced sustained glutamatic oxidative stress in both cerebrum and cerebellum, as GSH (reduced glutathione) levels in the hyperoxia group were constantly higher than the control group at three examined time-points. Furthermore, at P7, the expressions of all glutamate transporters decreased in both cerebrum and cerebellum except that of EAAT1. At P21, VGLUT2 in cerebrum and EAAT1, EAAT3 and VGLUT2 in cerebellum still displayed significant decrease in expression levels upon hyperoxia stimulation. Taken together, our results indicate that hyperoxia induces glutamate accumulation in brain of rat pups, which is associated with increased oxidative stress and decreased expressions of glutamate transporters.


Cerebellum/metabolism , Cerebrum/metabolism , Hyperoxia/pathology , Infant, Premature, Diseases/pathology , Animals , Animals, Newborn , Cerebellum/growth & development , Cerebellum/pathology , Cerebrum/growth & development , Cerebrum/pathology , Disease Models, Animal , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamic Acid/metabolism , Humans , Hyperoxia/etiology , Infant, Newborn , Infant, Premature/growth & development , Infant, Premature/metabolism , Infant, Premature, Diseases/etiology , Male , Oxidative Stress , Oxygen/administration & dosage , Oxygen/adverse effects , Rats , Time Factors , Vesicular Glutamate Transport Proteins/metabolism
12.
Sci Rep ; 11(1): 8471, 2021 04 19.
Article En | MEDLINE | ID: mdl-33875758

Despite wide application of high frequency oscillatory ventilation (HFOV) in neonates with respiratory distress, little has been reported about its rescue use in preterm infants. We aimed to evaluate the therapeutic effects of HFOV in preterm neonates with refractory respiratory failure and investigate the independent risk factors of in-hospital mortality. We retrospectively analyzed data collected prospectively (January 2011-December 2018) in four neonatal intensive care units of two tertiary-level medical centers in Taiwan. All premature infants (gestational age 24-34 weeks) receiving HFOV as rescue therapy for refractory respiratory failure were included. A total of 668 preterm neonates with refractory respiratory failure were enrolled. The median (IQR) gestational age and birth weight were 27.3 (25.3-31.0) weeks and 915.0 (710.0-1380.0) g, respectively. Pre-HFOV use of cardiac inotropic agents and inhaled nitric oxide were 70.5% and 23.4%, respectively. The oxygenation index (OI), FiO2, and AaDO2 were markedly increased after HFOV initiation (all p < 0.001), and can be decreased within 24-48 h (all p < 0.001) after use of HFOV. 375 (56.1%) patients had a good response to HFOV within 3 days. The final in-hospital mortality rate was 34.7%. No association was found between specific primary pulmonary disease and survival in multivariate analysis. We found preterm neonates with gestational age < 28 weeks, occurrences of sepsis, severe hypotension, multiple organ dysfunctions, initial higher severity of respiratory failure and response to HFOV within the first 72 h were independently associated with final in-hospital mortality. The mortality rate of preterm neonates with severe respiratory failure remains high after rescue HFOV treatment. Aggressive therapeutic interventions to treat sepsis and prevent organ dysfunctions are the suggested strategies to optimize outcomes.


High-Frequency Ventilation/methods , Infant, Premature, Diseases/therapy , Infant, Premature/physiology , Lung Diseases/prevention & control , Respiratory Distress Syndrome, Newborn/therapy , Birth Weight , Female , Follow-Up Studies , Gestational Age , Humans , Infant, Newborn , Infant, Premature, Diseases/epidemiology , Infant, Premature, Diseases/pathology , Longitudinal Studies , Male , Prognosis , Respiratory Distress Syndrome, Newborn/epidemiology , Respiratory Distress Syndrome, Newborn/pathology , Retrospective Studies , Survival Rate , Taiwan/epidemiology
13.
BMJ Case Rep ; 14(1)2021 Jan 08.
Article En | MEDLINE | ID: mdl-33419746

A 24+5-week preterm neonate with a severe scalp lesion was admitted to the neonatal intensive care unit (NICU) after caesarean section due to maternal chorioamnionitis (MC). An Arabin pessary had been inserted in addition to a previous cervical cerclage due to cervix insufficiency at 21+5 weeks of pregnancy (wp). At 23+5 wp, preterm rupture of membranes was evidenced. Both devices were kept to provide fetal viability. On 24+4 wp, she developed MC. Urgent caesarean section was performed. Transvaginal manual manipulation was required during the procedure. On NICU, she presented severe shock which required high-dose vasopressors and blood products. Following surgical repair, a bilateral grade IV intracranial haemorrhage was evidenced. Subsequently, it was agreed to withdraw life support. We hypothesise that MC and local infection could have acted as predisposing factors, with the presence of a pessary in the setting causing uterine contractions and its manipulation acting as a precipitating factor.


Gangrene/etiology , Infant, Premature, Diseases/etiology , Ischemia/etiology , Scalp/blood supply , Fatal Outcome , Female , Gangrene/pathology , Gangrene/surgery , Humans , Infant, Extremely Premature , Infant, Newborn , Infant, Premature, Diseases/pathology , Infant, Premature, Diseases/surgery , Ischemia/pathology , Ischemia/surgery
14.
Sci Rep ; 11(1): 1729, 2021 01 18.
Article En | MEDLINE | ID: mdl-33462302

To evaluate the impact of maternal hypertensive disorders of pregnancy (HDP) on mortality and neurological outcomes in extremely and very preterm infants using a nationwide neonatal database in Japan. This population-based retrospective study was based on an analysis of data collected by the Neonatal Research Network of Japan from 2003 to 2015 of neonates weighing 1,500 g or less at birth, between 22 and 31 weeks' gestation. A total of 21,659 infants were randomly divided into two groups, HDP (n = 4,584) and non-HDP (n = 4,584), at a ratio of 1:1 after stratification by four factors including maternal age, parity, weeks of gestation, and year of delivery. Short-term (neonatal period) and medium-term (3 years of age) mortality and neurological outcomes were compared between the two groups by logistic regression analyses. In univariate analysis, HDP was associated with an increased risk for in-hospital death (crude odds ratio [OR], 1.31; 95% confidence interval, 1.04-1.63) and a decreased risk for severe intraventricular haemorrhage (0.68; 0.53-0.87) and periventricular leukomalacia (0.60; 0.48-0.77). In multivariate analysis, HDP was significantly associated with a lower risk for in-hospital death (adjusted OR, 0.61; 0.47-0.80), severe intraventricular haemorrhage (0.47; 0.35-0.63), periventricular leukomalacia (0.59; 0.45-0.78), neonatal seizures (0.40; 0.28-0.57) and cerebral palsy (0.70; 0.52-0.95) at 3 years after adjustment for covariates including birth weight. These results were consistent with those of additional analyses, which excluded cases with histological chorioamnionitis and which divided the infants into two subgroups (22-27 gestational weeks and 28-31 gestational weeks). Maternal HDP was associated with an increased risk for in-hospital death without adjusting for covariates, but it was also associated with a lower risk for mortality and adverse neurological outcomes in extremely and very preterm infants if all covariates except HDP were identical.


Birth Weight , Hypertension, Pregnancy-Induced/mortality , Infant, Extremely Premature , Infant, Premature, Diseases/mortality , Leukomalacia, Periventricular/pathology , Nervous System Diseases/mortality , Adult , Databases, Factual , Female , Gestational Age , Hospital Mortality/trends , Humans , Hypertension, Pregnancy-Induced/etiology , Hypertension, Pregnancy-Induced/pathology , Infant, Premature, Diseases/etiology , Infant, Premature, Diseases/pathology , Japan/epidemiology , Maternal Age , Nervous System Diseases/etiology , Nervous System Diseases/pathology , Pregnancy , Retrospective Studies , Survival Rate
18.
Am J Med Genet A ; 185(3): 866-870, 2021 03.
Article En | MEDLINE | ID: mdl-33300650

Infantile liver failure syndrome type 1 (ILFS1) is a recently recognized autosomal recessive disorder caused by deleterious mutations in the leucyl-tRNA synthetase 1 gene (LARS1). The LARS1 enzyme is responsible for incorporation of the amino acid leucine during protein polypeptide synthesis. Individuals with LARS1 mutations typically show liver failure from infancy to early childhood during periods of illness or other physiological stress. While 25 patients from 15 families with ILFS1 have been reported in the literature, histological reports from autopsy findings are limited. We report here a premature male neonate who presented with severe intrauterine growth retardation, microcytic anemia, and fulminant liver failure, and who was a compound heterozygote for two novel deleterious mutations in LARS1. An autopsy showed fulminant hepatitis-like hepatocellular injury and fibrogenesis in the liver and a lack of uniformity in skeletal muscle, accompanied by the disruption of striated muscle fibers. Striking dysgenesis in skeletal muscle detected in the present case indicates the effect of LARS1 functional deficiency on the musculature. Whole-exome sequencing may be useful for neonates with unexplained early liver failure if extensive genetic and metabolic testing is inconclusive.


Infant, Premature, Diseases/genetics , Leucine-tRNA Ligase/genetics , Liver Failure/genetics , Musculoskeletal Abnormalities/genetics , Mutation, Missense , Point Mutation , RNA Splice Sites/genetics , Amino Acid Substitution , Anemia, Neonatal/genetics , Exons/genetics , Fatal Outcome , Fetal Growth Retardation/genetics , Genes, Recessive , Heterozygote , Humans , Hyperbilirubinemia, Neonatal/genetics , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/pathology , Introns/genetics , Leucine-tRNA Ligase/deficiency , Liver Cirrhosis/etiology , Liver Failure/pathology , Liver Failure, Acute/etiology , Liver Failure, Acute/pathology , Male , Multiple Organ Failure/etiology , Muscle, Skeletal/pathology , Musculoskeletal Abnormalities/pathology , Sequence Alignment , Syndrome , Exome Sequencing
20.
Surg Pathol Clin ; 13(4): 657-682, 2020 Dec.
Article En | MEDLINE | ID: mdl-33183726

Neonatal lung biopsy guides important medical decisions when the diagnosis is not clear from prior clinical assessment, imaging, or genetic testing. Common scenarios that lead to biopsy include severe acute respiratory distress in a term neonate, pulmonary hypertension disproportionate to that expected for gestational age or known cardiac anomalies, and assessment of suspected genetic disorder based on clinical features or genetic variant of unknown significance. The differential diagnosis includes genetic developmental disorders, genetic surfactant disorders, vascular disorders, acquired infection, and meconium aspiration. This article describes pathologic patterns in the neonatal lung and correlation with molecular abnormalities, where appropriate.


Biopsy , Lung Diseases/congenital , Lung Diseases/pathology , Lung/abnormalities , Lung/pathology , Diagnosis, Differential , Down Syndrome/complications , Heart Defects, Congenital/complications , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/etiology , Infant, Premature, Diseases/pathology , Lung Diseases/etiology , Prognosis
...