Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Dev Neurosci ; 46(2): 84-97, 2024.
Article in English | MEDLINE | ID: mdl-37231871

ABSTRACT

Fetal growth restriction (FGR) and small for gestational age (SGA) infants have increased risk of mortality and morbidity. Although both FGR and SGA infants have low birthweights for gestational age, a diagnosis of FGR also requires assessments of umbilical artery Doppler, physiological determinants, neonatal features of malnutrition, and in utero growth retardation. Both FGR and SGA are associated with adverse neurodevelopmental outcomes ranging from learning and behavioral difficulties to cerebral palsy. Up to 50% of FGR, newborns are not diagnosed until around the time of birth, yet this diagnosis lacks further indication of the risk of brain injury or adverse neurodevelopmental outcomes. Blood biomarkers may be a promising tool. Defining blood biomarkers indicating an infant's risk of brain injury would provide the opportunity for early detection and therefore earlier support. The aim of this review was to summarize the current literature to assist in guiding the future direction for the early detection of adverse brain outcomes in FGR and SGA neonates. The studies investigated potential diagnostic blood biomarkers from cord and neonatal blood or serum from FGR and SGA human neonates. Results were often conflicting with heterogeneity common in the biomarkers examined, timepoints, gestational age, and definitions of FGR and SGA used. Due to these variations, it was difficult to draw strong conclusions from the results. The search for blood biomarkers of brain injury in FGR and SGA neonates should continue as early detection and intervention is critical to improve outcomes for these neonates.


Subject(s)
Brain Injuries , Fetal Growth Retardation , Female , Infant, Newborn , Humans , Fetal Growth Retardation/diagnosis , Gestational Age , Infant, Small for Gestational Age , Brain Injuries/diagnosis , Biomarkers
2.
Pediatr Res ; 95(1): 59-69, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37674023

ABSTRACT

The neurovascular unit (NVU) within the brain is a multicellular unit that synergistically acts to maintain blood-brain barrier function and meet cerebral metabolic demand. Recent studies have indicated disruption to the NVU is associated with neuropathology in the perinatal brain. Infants with fetal growth restriction (FGR) are known to be at increased risk of neurodevelopmental conditions including motor, learning, and behavioural deficits. There are currently no neuroprotective treatments for these conditions. In this review, we analyse large animal studies examining the effects of FGR on the perinatal NVU. These studies show altered vascularity in the FGR brain as well as blood-brain barrier dysfunction due to underlying cellular changes, mediated by neuroinflammation. Neuroinflammation is a key mechanism associated with pathological effects in the FGR brain. Hence, targeting inflammation may be key to preserving the multicellular NVU and providing neuroprotection in FGR. A number of maternal and postnatal therapies with anti-inflammatory components have been investigated in FGR animal models examining targets for amelioration of NVU disruption. Each therapy showed promise by uniquely ameliorating the adverse effects of FGR on multiple aspects of the NVU. The successful implementation of a clinically viable neuroprotective treatment has the potential to improve outcomes for neonates affected by FGR. IMPACT: Disruption to the neurovascular unit is associated with neuropathology in fetal growth restriction. Inflammation is a key mechanism associated with neurovascular unit disruption in the growth-restricted brain. Anti-inflammatory treatments ameliorate adverse effects on the neurovascular unit and may provide neuroprotection.


Subject(s)
Fetal Growth Retardation , Neuroinflammatory Diseases , Pregnancy , Animals , Infant, Newborn , Infant , Female , Humans , Brain/metabolism , Blood-Brain Barrier , Anti-Inflammatory Agents/therapeutic use
3.
Dev Med Child Neurol ; 65(3): 346-357, 2023 03.
Article in English | MEDLINE | ID: mdl-37017185

ABSTRACT

AIM: This study aimed to identify early clinical biomarkers from birth to 16 weeks corrected age to predict typical outcome and developmental delay in infants born very preterm or with very low birthweight. METHOD: A prospective cohort of infants on the Sunshine Coast, Australia, was assessed using the Premie-Neuro Examination, the General Movement Assessment (GMA), the Alberta Infant Motor Scale, and the Infant Sensory Profile 2. At 24 months corrected age, delay was identified using the Bayley Scales of Infant and Toddler Development, Third Edition (Bayley-III) and Neurosensory Motor Developmental Assessment (NSMDA). RESULTS: One hundred and four infants were recruited; 79 completed outcome assessments (43 females, 36 males; mean gestational age 30 weeks [SD 1 week 6 days], mean birthweight 1346 g [SD 323]). The incidence of developmental delay (motor or cognitive) was 6.3%. Suboptimal quality of fidgety general movements (temporal organization) at 16 weeks corrected age demonstrated the best predictive accuracy (Bayley-III motor: sensitivity 100% [95% confidence interval {CI} 3-100], specificity 75% [95% CI 63-84], area under the curve [AUC] 0.87); Bayley-III cognitive: sensitivity 100% [95% CI 3-100], specificity 75% [95% CI 64-84], AUC 0.88); NSMDA motor: sensitivity 100% [95% CI 40-100], specificity 81% [95% CI 70-90], AUC 0.91 [95% CI 0.86-0.95]). GMA trajectories that combined abnormal writhing general movements at 4 to 5 weeks corrected age with suboptimal quality of fidgety movement at 16 weeks corrected age were strongly predictive of developmental delay, superior to all other clinical tools, and perinatal and demographic variables investigated (p = 0.01, Akaike information criterion method 18.79 [score corrected for small sample size], accounting for 93% of the cumulative weight). INTERPRETATION: Only the GMA had sufficient predictive validity to act as a biomarker for both conditions: typical outcome and developmental delay (motor or cognitive). GMA trajectories that assessed both writhing general movements at 4 to 5 weeks corrected age and quality of fidgety movement at 16 weeks corrected age predicted adverse neurodevelopmental outcome, accurately differentiating between infants with typical outcomes and those at increased risk for motor or cognitive delay.


Subject(s)
Developmental Disabilities , Infant, Extremely Premature , Infant, Newborn , Male , Female , Pregnancy , Child , Infant , Humans , Developmental Disabilities/diagnosis , Child Development , Prospective Studies , Infant, Very Low Birth Weight
4.
Dev Med Child Neurol ; 65(8): 1061-1072, 2023 08.
Article in English | MEDLINE | ID: mdl-36683126

ABSTRACT

AIM: To evaluate the predictive validity of the Hammersmith Neonatal Neurological Examination (HNNE) performed early (at 32 weeks postmenstrual age) and at term-equivalent age (TEA) for 12-month motor outcomes in infants born very preterm. METHOD: This was a diagnostic study using data from a prospective birth cohort. A total of 104 infants born preterm at less than 31 weeks gestational age (males n = 61; mean = 28 weeks 1 day [SD 1 week 6 days], range 23 weeks 1 day-30 weeks 6 days) underwent HNNE early and at TEA, which were scored by comparison with term data. Motor outcomes at 12 months corrected age were determined using the Bayley Scales of Infant and Toddler Development, Third Edition (scores ≤85). Cut-off points were determined using receiver operating characteristic curves. RESULTS: Sixteen (15%) infants born preterm had motor impairment at 12 months corrected age. The HNNE total score cut-off points with the best combination of sensitivity and specificity at early and TEA assessments were 15.2 or lower (sensitivity 77%, 95% confidence interval [CI] = 46%-95%; specificity 74%, 95% CI = 63%-83%) and 23.5 or lower (sensitivity 67%, 95% CI = 38%-88%; specificity 66%, 95% CI = 54%-76%) respectively. The most predictive subscale at the early assessment was reflexes (sensitivity 86%, 95% CI = 57%-98%; specificity 62%, 95% CI = 51%-72%; cut-off point ≤3); at TEA, it was spontaneous movements (sensitivity 73%, 95% CI = 45%-92%; specificity 60%, 95% CI = 48%-70%; cut-off point ≤2). INTERPRETATION: The HNNE provides moderate predictive accuracy for motor outcome at 12 months corrected age in infants born very preterm. Although modest at both time points, early assessment had stronger predictive ability for motor outcomes than TEA when scored using term data, highlighting the value of performing the HNNE earlier in the neonatal period. Performing HNNE earlier may assist risk stratification when planning follow-up services.


Subject(s)
Child Development , Infant, Extremely Premature , Infant, Newborn , Male , Humans , Infant , Female , Prospective Studies , Neurologic Examination , Gestational Age
5.
Sci Rep ; 13(1): 282, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36609414

ABSTRACT

Neuroinflammation is a hallmark of hypoxic-ischemic injury and can be characterized by the activation of glial cells and the expression of inflammatory cytokines and chemokines. Interleukin (IL)-1ß and tumor necrosis factor (TNF)α are among the best-characterized early response cytokines and are often expressed concurrently. Several types of central nervous system cells secrete IL-1ß and TNFα, including microglia, astrocytes, and neurons, and these cytokines convey potent pro-inflammatory actions. Chemokines also play a central role in neuroinflammation by controlling inflammatory cell trafficking. Our aim was to characterise the evolution of early neuroinflammation in the neonatal piglet model of hypoxic-ischemic encephalopathy (HIE). Piglets (< 24 h old) were exposed to HI insult, and recovered to 2, 4, 8, 12 or 24H post-insult. Brain tissue from the frontal cortex and basal ganglia was harvested for assessment of glial cell activation profiles and transcription levels of inflammatory markers in HI piglets with comparison to a control group of newborn piglets. Fluorescence microscopy was used to observe microglia, astrocytes, neurons, degenerating neurons and possibly apoptotic cells, and quantitative polymerase chain reaction was used to measure gene expression of several cytokines and chemokines. HI injury was associated with microglial activation and morphological changes to astrocytes at all time points examined. Gene expression analyses of inflammation-related markers revealed significantly higher expression of pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin 1 beta (IL-1ß), chemokines cxc-chemokine motif ligand (CXCL)8 and CXCL10, and anti-inflammatory cytokine transforming growth factor (TGF)ß in every HI group, with some region-specific differences noted. No significant difference was observed in the level of C-X-C chemokine receptor (CCR)5 over time. This high degree of neuroinflammation was associated with a reduction in the number of neurons in piglets at 12H and 24H in the frontal cortex, and the putamen at 12H. This reduction of neurons was not associated with increased numbers of degenerating neurons or potentially apoptotic cells. HI injury triggered a robust early neuroinflammatory response associated with a reduction in neurons in cortical and subcortical regions in our piglet model of HIE. This neuroinflammatory response may be targeted using novel therapeutics to reduce neuropathology in our piglet model of neonatal HIE.


Subject(s)
Cytokines , Hypoxia-Ischemia, Brain , Animals , Swine , Cytokines/metabolism , Animals, Newborn , Tumor Necrosis Factor-alpha/metabolism , Neuroinflammatory Diseases , Neuroglia/metabolism , Brain/metabolism , Hypoxia/metabolism , Microglia/metabolism , Hypoxia-Ischemia, Brain/pathology , Transforming Growth Factor beta/metabolism , Inflammation/pathology
6.
Neuroimage ; 267: 119815, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36529204

ABSTRACT

Infants born very preterm face a range of neurodevelopmental challenges in cognitive, language, behavioural and/or motor domains. Early accurate identification of those at risk of adverse neurodevelopmental outcomes, through clinical assessment and Magnetic Resonance Imaging (MRI), enables prognostication of outcomes and the initiation of targeted early interventions. This study utilises a prospective cohort of 181 infants born <31 weeks gestation, who had 3T MRIs acquired at 29-35 weeks postmenstrual age and a comprehensive neurodevelopmental evaluation at 2 years corrected age (CA). Cognitive, language and motor outcomes were assessed using the Bayley Scales of Infant and Toddler Development - Third Edition and functional motor outcomes using the Neuro-sensory Motor Developmental Assessment. By leveraging advanced structural MRI pre-processing steps to standardise the data, and the state-of-the-art developing Human Connectome Pipeline, early MRI biomarkers of neurodevelopmental outcomes were identified. Using Least Absolute Shrinkage and Selection Operator (LASSO) regression, significant associations between brain structure on early MRIs with 2-year outcomes were obtained (r = 0.51 and 0.48 for motor and cognitive outcomes respectively) on an independent 25% of the data. Additionally, important brain biomarkers from early MRIs were identified, including cortical grey matter volumes, as well as cortical thickness and sulcal depth across the entire cortex. Adverse outcome on the Bayley-III motor and cognitive composite scores were accurately predicted, with an Area Under the Curve of 0.86 for both scores. These associations between 2-year outcomes and patient prognosis and early neonatal MRI measures demonstrate the utility of imaging prior to term equivalent age for providing earlier commencement of targeted interventions for infants born preterm.


Subject(s)
Brain , Infant, Premature , Infant , Infant, Newborn , Humans , Prospective Studies , Brain/diagnostic imaging , Magnetic Resonance Imaging , Cognition , Biomarkers , Child Development
7.
Cells Tissues Organs ; 212(6): 546-553, 2023.
Article in English | MEDLINE | ID: mdl-36261026

ABSTRACT

Umbilical cord blood cells have therapeutic potential for neurological disorders, through a paracrine mechanism of action. A greater understanding of the safety and immunological effects of allogeneic donor cord blood cells in the context of a healthy recipient immune system, such as in cerebral palsy, is needed. This study aimed to determine how quickly donor cord blood cells were cleared from the circulation in children with cerebral palsy who received a single intravenous infusion of 12/12 human leucocyte antigen (HLA)-matched sibling cord blood cells. Twelve participants with cerebral palsy aged 2-12 years received cord blood cell infusions as part of a phase I trial of umbilical blood infusion for cerebral palsy. Digital droplet PCR analysis of DNA copy number variants specific to donor and recipient was used to assess donor DNA clearance at five timepoints post-infusion, a surrogate measure of cell clearance. Donor cells were cleared by 3 months post-infusion in 11/12 participants. When detected, donor DNA was at a fraction of 0.01-0.31% of total DNA with no signs of graft-versus-host disease in any participant. The donor DNA clearance times provided by this study have important implications for understanding the safety of allogeneic cord blood cell infusion for cerebral palsy and translational tissue engineering or regenerative medicine research in other disorders.


Subject(s)
Cerebral Palsy , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Child , Humans , Cerebral Palsy/therapy , DNA , Fetal Blood
8.
Pediatr Res ; 93(6): 1721-1727, 2023 05.
Article in English | MEDLINE | ID: mdl-36151299

ABSTRACT

BACKGROUND: To determine the diagnostic accuracy of Hammersmith Neonatal Neurological Examination (HNNE) at 30-32 weeks postmenstrual age (PMA, 'Early') and term equivalent age (TEA) in infants born <31 weeks PMA to predict cognitive outcomes at 12 months corrected age (CA). METHODS: Prospective cohort study of 119 infants (73 males; median 28.4 weeks gestational age at birth) who underwent Early and TEA HNNE. At 12 months CA, 104 participants completed Bayley Scales of Infant and Toddler Development, 3rd Edition, (Bayley-III). Optimum cut-off points for each HNNE subscale were determined to establish diagnostic accuracy for predicting adverse cognitive outcomes on the Bayley-III Cognitive Composite Scale (≤85). RESULTS: The best diagnostic accuracy for HNNE total score at 30-32 weeks PMA predicting cognitive impairment occurred at cut-off ≤16.7 (sensitivity (Se) = 71%, specificity (Sp) = 51%). The Abnormal Signs subscale demonstrated the best balance of sensitivity/specificity combination (Se = 71%, Sp = 71%; cut-off ≤1.5). For HNNE at TEA, the total score at cut-off ≤24.5 had Se = 71% and Sp = 47% for predicting cognitive impairment. The Tone Patterns subscale demonstrated the strongest diagnostic accuracy at TEA (Se = 71%, Sp = 63%; cut-off ≤3). CONCLUSIONS: Early and TEA HNNE demonstrated moderate diagnostic accuracy for cognitive outcomes at 12-months CA in infants born <31 weeks gestational age. CLINICAL TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry; Trial Registration Number: ACTRN12613000280707; web address of trial: http://www.ANZCTR.org.au/ACTRN12613000280707.aspx . IMPACT: Early Hammersmith Neonatal Neurological Examination (HNNE) assessment at 30-32 weeks postmenstrual age has moderate diagnostic accuracy for cognitive outcomes at 12 months corrected age in infants born <31 weeks gestation. Early HNNE at 30-32 weeks has stronger predictive validity than HNNE at term equivalent age. Early HNNE may provide an early marker for risk-stratification to optimise the planning of post-discharge support and follow-up services for infants born preterm.


Subject(s)
Aftercare , Infant, Extremely Premature , Infant, Newborn , Male , Female , Humans , Infant , Prospective Studies , Australia , Patient Discharge , Neurologic Examination , Gestational Age , Fetal Growth Retardation , Cognition , Child Development
9.
BMJ Open ; 12(8): e065823, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35977775

ABSTRACT

INTRODUCTION: The increasing prevalence of developmental disorders in early childhood poses a significant global health burden. Early detection of developmental problems is vital to ensure timely access to early intervention, and universal developmental surveillance is recommended best practice for identifying issues. Despite this, there is currently considerable variation in developmental surveillance and screening between Australian states and territories and low rates of developmental screening uptake by parents. This study aims to evaluate an innovative web-based developmental surveillance programme and a sustainable approach to referral and care pathways, linking primary care general practice (GP) services that fall under federal policy responsibility and state government-funded child health services. METHODS AND ANALYSIS: The proposed study describes a longitudinal cluster randomised controlled trial (c-RCT) comparing a 'Watch Me Grow Integrated' (WMG-I) approach for developmental screening, to Surveillance as Usual (SaU) in GPs. Forty practices will be recruited across New South Wales and Queensland, and randomly allocated into either the (1) WMG-I or (2) SaU group. A cohort of 2000 children will be recruited during their 18-month vaccination visit or opportunistic visit to GP. At the end of the c-RCT, a qualitative study using focus groups/interviews will evaluate parent and practitioner views of the WMG-I programme and inform national and state policy recommendations. ETHICS AND DISSEMINATION: The South Western Sydney Local Health District (2020/ETH01625), UNSW Sydney (2020/ETH01625) and University of Queensland (2021/HE000667) Human Research Ethics Committees independently reviewed and approved this study. Findings will be reported to the funding bodies, study institutes and partners; families and peer-reviewed conferences/publications. TRIAL REGISTRATION NUMBER: ANZCTR12621000680864.


Subject(s)
Child Health Services , Mass Screening , Australia , Child , Child, Preschool , Humans , Internet , Primary Health Care , Randomized Controlled Trials as Topic
10.
Early Hum Dev ; 172: 105632, 2022 09.
Article in English | MEDLINE | ID: mdl-35905636

ABSTRACT

BACKGROUND: Very preterm (VPT) infants develop adverse neurological sequelae from early exposure of the immature brain to the extrauterine environment. AIMS: To determine the effects of infant massage on brain maturation in low-risk VPT infants. STUDY DESIGN: A randomised controlled trial of VPT infants, who received standard care or daily massage therapy, administered by the mother, from 34 weeks' to 40 weeks' corrected age (CA). SUBJECTS: VPT infants (born at 28 weeks to 32 + 6 weeks' gestational age, G.A.) and a healthy at term cohort for comparison. OUTCOME MEASURES: At term equivalent age (39 weeks' to 42 weeks' CA), EEG was recorded to calculate global relative power (GRP), using power spectral analysis. RESULTS: Sixty infants were recruited, and EEGs of 25 massage and 20 standard care infants were analysable. There was no difference between groups in primary outcome (beta GRP). There was a significantly higher central alpha relative power measured in the intervention group infants, compared to standard care (SC) group (mean difference = 1.42, 95 % confidence interval (CI): 0.12 to 2.73; p = 0.03). A massage dose effect was shown by a positive correlation between, massage dose and beta, alpha and theta GRP (r = 0.42, 95%CI = 0.12 to 0.64, r = 0.45; 95%CI = 0.16 to 0.66, r = 0.39; 95%CI = 0.10 to 0.62 respectively) and a negative correlation between massage dose and delta GRP (r = -0.41, 95%CI = -0.64 to -0.12), suggesting that a higher dose of massage is associated with more favourable brain maturation. CONCLUSIONS: Central alpha regional relative power was greater in massaged infants compared to SC group infants, suggesting relatively greater brain maturation in this area. A measurable massage dose effect in favour of greater brain maturation, shows promise for verification in a larger clinical trial.


Subject(s)
Infant, Premature, Diseases , Infant, Premature , Brain , Electroencephalography , Female , Fetal Growth Retardation , Gestational Age , Humans , Infant , Infant, Newborn , Massage
11.
Eur Respir J ; 60(5)2022 11.
Article in English | MEDLINE | ID: mdl-35777773

ABSTRACT

INTRODUCTION: Asthma exacerbations in pregnancy are associated with adverse perinatal outcomes. We aimed to determine whether fractional exhaled nitric oxide (F ENO)-based asthma management improves perinatal outcomes compared to usual care. METHODS: The Breathing for Life Trial was a multicentre, parallel-group, randomised controlled trial conducted in six hospital antenatal clinics, which compared asthma management guided by F ENO (adjustment of asthma treatment according to exhaled nitric oxide and symptoms each 6-12 weeks) to usual care (no treatment adjustment as part of the trial). The primary outcome was a composite of adverse perinatal events (preterm birth, small for gestational age (SGA), perinatal mortality or neonatal hospitalisation) assessed using hospital records. Secondary outcomes included maternal asthma exacerbations. Concealed random allocation, stratified by study site and self-reported smoking status was used, with blinded outcome assessment and statistical analysis (intention to treat). RESULTS: Pregnant women with current asthma were recruited; 599 to the control group (608 infants) and 601 to the intervention (615 infants). There were no significant group differences for the primary composite perinatal outcome (152 (25.6%) out of 594 control, 177 (29.4%) out of 603 intervention; OR 1.21, 95% CI 0.94-1.56; p=0.15), preterm birth (OR 1.14, 95% CI 0.78-1.68), SGA (OR 1.06, 95% CI 0.78-1.68), perinatal mortality (OR 3.62, 95% CI 0.80-16.5), neonatal hospitalisation (OR 1.24, 95% CI 0.89-1.72) or maternal asthma exacerbations requiring hospital admission or emergency department presentation (OR 1.19, 95% CI 0.69-2.05). CONCLUSION: F ENO-guided asthma pharmacotherapy delivered by a nurse or midwife in the antenatal clinic setting did not improve perinatal outcomes.


Subject(s)
Asthma , Premature Birth , Infant , Female , Infant, Newborn , Pregnancy , Humans , Nitric Oxide , Exhalation , Asthma/drug therapy , Respiration
12.
Comput Methods Programs Biomed ; 224: 107014, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35849896

ABSTRACT

BACKGROUND AND OBJECTIVE: In newborns, it is often difficult to accurately differentiate between seizure and non-seizure based solely on clinical manifestations. This highlights the importance of electroencephalogram (EEG) in the recognition and management of neonatal seizures. This paper proposes an effective algorithm for the detection of neonatal seizure using multichannel EEG. METHODS: Neonatal EEG changes morphology as it alternates between seizure and non-seizure states. A new signal complexity measure based on matching pursuit (MP) decomposition is proposed and used to detect transitions between these two states. The new measure, referred to as weighted structural complexity (WSC), was used for the detection of seizures in 30 newborn EEG records. Multiple IIR filters and an MP-based filter were designed and used to remove artifacts from the EEG data. Geometrical correlation between the EEG data channels was applied to reduce the number of false detections caused by remnant artifacts. The seizure detector's performance was assessed using several epoch-based (e.g., accuracy) and event-based (GDR = good detection rate and FD/h = false detections per hour) metrics. RESULTS: Compared to the neurologist marking, the proposed detector was able to detect EEG seizures with 94% accuracy, 90.9% GDR, and 0.14 FD/h (95% CI: [0.06, 0.34]). CONCLUSIONS: The high performance of the MP-based detector may have significant implications for the accurate diagnosis of neonatal seizures and the appropriate use of anticonvulsants and ongoing clinical assessment and care of the newborn.


Subject(s)
Electroencephalography , Epilepsy , Algorithms , Artifacts , Humans , Infant, Newborn , Seizures/diagnosis , Signal Processing, Computer-Assisted
13.
J Clin Med ; 11(7)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35407440

ABSTRACT

The Motor Optimality Score, revised (MOS-R) is an extension of the Prechtl General Movements Assessment. This study aims to determine the relationship between MOS-R and 2-year neurodevelopmental outcomes in a cohort of 169 infants born very preterm (<31 weeks' gestational age), and to examine the predictive validity of the MOS-R at 3−4 months' corrected age (CA) above perinatal variables associated with poor outcomes, including Prechtl fidgety movements. Development at 2 years' CA was assessed using Bayley Scales of Infant and Toddler Development, Third edition (Bayley-III) (motor/cognitive impairment: Bayley-III ≤ 85) and Neurological, Sensory, Motor, Developmental Assessment (NSMDA) (neurosensory motor impairment: NSMDA ≥ 12). Cerebral palsy (CP) was classified at 2 years as definite or clinical. The MOS-R was related to 2-year outcomes: Bayley-III motor (BMOS-R = 1.24 95% confidence interval (0.78, 1.70)), cognitive (BMOS-R = 0.91 (0.48, 1.35)), NSMDA scores (BMOS-R = −0.34 (−0.42, −0.25)), definite CP (odds ratio [OR] 0.67 (0.53, 0.86)), clinical CP (OR 0.74 (0.66, 0.83)) for each 1-point increase in MOS-R. MOS-R ≤ 23 predicted motor (sensitivity 78% (60−91%); specificity 63% (54−72%)) and neurosensory motor impairment (sensitivity 86% (64−97%); specificity 59% (51−68%)). The MOS-R is strongly related to CP and motor and cognitive delay at 2 years and is a good predictor of motor and neurosensory motor impairment.

14.
Dev Neurosci ; 44(4-5): 194-204, 2022.
Article in English | MEDLINE | ID: mdl-35263744

ABSTRACT

Fetal growth restriction (FGR) is associated with long-term neurodevelopmental disabilities including learning and behavioral disorders, autism, and cerebral palsy. Persistent changes in brain structure and function that are associated with developmental disabilities are demonstrated in FGR neonates. However, the mechanisms underlying these changes remain to be determined. There are currently no therapeutic interventions available to protect the FGR newborn brain. With the wide range of long-term neurodevelopmental disorders associated with FGR, the use of an animal model appropriate to investigating mechanisms of injury in the FGR newborn is crucial for the development of effective and targeted therapies for babies. Piglets are ideal animals to explore how perinatal insults affect brain structure and function. FGR occurs spontaneously in the piglet, unlike other animal models that require surgical or chemical intervention, allowing brain outcomes to be studied without the confounding impacts of experimental interventions. The FGR piglet mimics many of the human pathophysiological outcomes associated with FGR including asymmetrical growth restriction with brain sparing. This review will discuss the similarities observed in brain outcomes between the FGR human and FGR piglet from a magnetic resonance imaging in the living and a histological perspective. FGR piglet studies provide the opportunity to determine and track mechanisms of brain injury in a clinically relevant animal model of FGR. Findings from these FGR piglet studies may provide critical information to rapidly translate neuroprotective interventions to clinic to improve outcomes for newborn babies.


Subject(s)
Brain Injuries , Cerebral Palsy , Animals , Brain/pathology , Brain Injuries/pathology , Cerebral Palsy/pathology , Disease Models, Animal , Female , Fetal Growth Retardation/pathology , Humans , Infant, Newborn , Magnetic Resonance Imaging , Pregnancy , Swine
15.
Pediatr Res ; 92(6): 1527-1534, 2022 12.
Article in English | MEDLINE | ID: mdl-35197567

ABSTRACT

Foetal growth restriction (FGR) and being born small for gestational age (SGA) are associated with neurodevelopmental delay. Early diagnosis of neurological damage is difficult in FGR and SGA neonates. Electroencephalography (EEG) has the potential as a tool for the assessment of brain development in FGR/SGA neonates. In this review, we analyse the evidence base on the use of EEG for the assessment of neonates with FGR or SGA. We found consistent findings that FGR/SGA is associated with measurable changes in the EEG that present immediately after birth and persist into childhood. Early manifestations of FGR/SGA in the EEG include changes in spectral power, symmetry/synchrony, sleep-wake cycling, and the continuity of EEG amplitude. Later manifestations of FGR/SGA into infancy and early childhood include changes in spectral power, sleep architecture, and EEG amplitude. FGR/SGA infants had poorer neurodevelopmental outcomes than appropriate for gestational age controls. The EEG has the potential to identify FGR/SGA infants and assess the functional correlates of neurological damage. IMPACT: FGR/SGA neonates have significantly different EEG activity compared to AGA neonates. EEG differences persist into childhood and are associated with adverse neurodevelopmental outcomes. EEG has the potential for early identification of brain impairment in FGR/SGA neonates.


Subject(s)
Fetal Growth Retardation , Infant, Small for Gestational Age , Child, Preschool , Infant, Newborn , Pregnancy , Infant , Female , Humans , Fetal Growth Retardation/diagnosis , Birth Weight , Parturition , Gestational Age
16.
J Clin Psychol Med Settings ; 29(2): 391-402, 2022 06.
Article in English | MEDLINE | ID: mdl-35066796

ABSTRACT

Identify predictors of maternal bonding and responsiveness for mothers of very preterm infants (< 32 weeks gestational age) at 6 weeks and 12 months corrected-age (CA). Cross-sectional and longitudinal study containing 39 mothers of very preterm infants. At 6 weeks CA maternal self-efficacy made a significant unique contribution to the variance in self-reported maternal bonding and responsiveness (21% and 26%, respectively). At 12 months CA maternal trauma symptoms, depressive symptoms and self-efficacy made a significant unique contribution to the variance in bonding (14%, 9% and 9%, respectively). Maternal self-efficacy made a significant 31% unique contribution to the variance in responsiveness. The combined effects of maternal trauma symptoms, depressive symptoms and self-efficacy at 6 weeks CA predicted maternal responsiveness at 12 months CA (p = .042). Supporting maternal self-efficacy is key to facilitating bonding and responsiveness up to 12 months CA following a very preterm birth.Trial registration: Australian New Zealand Clinical Trials Registry: ACTRN12612000194864.


Subject(s)
Mothers , Premature Birth , Australia , Cross-Sectional Studies , Female , Humans , Infant , Infant, Newborn , Infant, Premature , Longitudinal Studies , Mother-Child Relations
17.
Pediatr Res ; 92(1): 25-31, 2022 07.
Article in English | MEDLINE | ID: mdl-34482377

ABSTRACT

BACKGROUND: The objective of this study was to systematically review the literature to determine the effect of combined hypothermia (HTH) and mesenchymal stem cell (MSC) therapy (administered during or immediately before or after HTH) compared with HTH alone on brain injury and neurobehavioural outcomes in animal models of neonatal hypoxic-ischaemic encephalopathy. METHODS: Primary outcomes assessed were neuropathological measures and neurobehavioural measures of brain outcome. Secondary outcomes were brain protein proinflammatory cytokine status. Risk of bias (ROB) was assessed with the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) ROB assessment tool. RESULTS: Of 393 studies identified, 3 studies in postnatal day 7 (P7) male Sprague-Dawley rats met the inclusion criteria. Meta-analyses were undertaken for neuropathological measures (apoptotic cells, astrocytes, microglia), neurobehavioral measures (rotarod test and negative geotaxis), and proinflammatory cytokine levels. Two of the three studies scored low or unclear ROB across all measures. Treatment with HTH-MSCs together significantly improved astrocyte optical density by standardised mean difference (SMD) of 0.71 [95% confidence interval (CI) -1.14, -0.28]. No other measures showed significant differences. CONCLUSIONS: There is insufficient preclinical data to confirm the efficacy of combined HTH-MSC therapy over HTH alone. Future studies should utilise a reporting checklist such as in SYRCLE or Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines to improve reporting standards. IMPACT: Very few articles investigating the use of MSCs for the treatment of hypoxic-ischaemic encephalopathy are clinically relevant. Continuing to publish studies in models of hypoxic-ischaemic encephalopathy without the inclusion of HTH therapy does not progress the field towards improved clinical outcomes. This study shows that HTH and MSC therapy improves measures of astrogliosis. More studies are required to establish the efficacy of HTH and MSCs on measures of neuropathology and neurobehavior. The reporting of preclinical data in this space could be improved by using reporting checklists such as the SYRCLE or ARRIVE tools.


Subject(s)
Hypothermia , Hypoxia-Ischemia, Brain , Mesenchymal Stem Cells , Animals , Cytokines , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/therapy , Male , Models, Animal , Rats , Rats, Sprague-Dawley
18.
Mol Neurobiol ; 59(2): 1018-1040, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34825315

ABSTRACT

The developing brain is particularly vulnerable to foetal growth restriction (FGR) and abnormal neurodevelopment is common in the FGR infant ranging from behavioural and learning disorders to cerebral palsy. No treatment exists to protect the FGR newborn brain. Recent evidence suggests inflammation may play a key role in the mechanism responsible for the progression of brain impairment in the FGR newborn, including disruption to the neurovascular unit (NVU). We explored whether ibuprofen, an anti-inflammatory drug, could reduce NVU disruption and brain impairment in the FGR newborn. Using a preclinical FGR piglet model, ibuprofen was orally administered for 3 days from birth. FGR brains demonstrated a proinflammatory state, with changes to glial morphology (astrocytes and microglia), and blood-brain barrier disruption, assessed by IgG and albumin leakage into the brain parenchyma and a decrease in blood vessel density. Loss of interaction between astrocytic end-feet and blood vessels was evident where plasma protein leakage was present, suggestive of structural deficits to the NVU. T-cell infiltration was also evident in the parenchyma of FGR piglet brains. Ibuprofen treatment reduced the pro-inflammatory response in FGR piglets, reducing the number of activated microglia and enhancing astrocyte interaction with blood vessels. Ibuprofen also attenuated plasma protein leakage, regained astrocytic end-feet interaction around vessels, and decreased T-cell infiltration into the FGR brain. These findings suggest postnatal administration of ibuprofen modulates the inflammatory state, allowing for stronger interaction between vasculature and astrocytic end-feet to restore NVU integrity. Modulation of the NVU improves the FGR brain microenvironment and may be key to neuroprotection.


Subject(s)
Brain , Ibuprofen , Animals , Astrocytes/metabolism , Brain/metabolism , Humans , Ibuprofen/pharmacology , Ibuprofen/therapeutic use , Microglia , Neuroglia , Swine
19.
JAMA Netw Open ; 4(12): e2139604, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34919132

ABSTRACT

Importance: Seizures in the neonatal period are associated with increased mortality and morbidity. Bedside amplitude-integrated electroencephalography (aEEG) has facilitated the detection of electrographic seizures; however, whether these seizures should be treated remains uncertain. Objective: To determine if the active management of electrographic and clinical seizures in encephalopathic term or near-term neonates improves survival free of severe disability at 2 years of age compared with only treating clinically detected seizures. Design, Setting, and Participants: This randomized clinical trial was conducted in tertiary newborn intensive care units recruited from 2012 to 2016 and followed up until 2 years of age. Participants included neonates with encephalopathy at 35 weeks' gestation or more and younger than 48 hours old. Data analysis was completed in April 2021. Interventions: Randomization was to an electrographic seizure group (ESG) in which seizures detected on aEEG were treated in addition to clinical seizures or a clinical seizure group (CSG) in which only seizures detected clinically were treated. Main Outcomes and Measures: Primary outcome was death or severe disability at 2 years, defined as scores in any developmental domain more than 2 SD below the Australian mean assessed with Bayley Scales of Neonate and Toddler Development, 3rd ed (BSID-III), or the presence of cerebral palsy, blindness, or deafness. Secondary outcomes included magnetic resonance imaging brain injury score at 5 to 14 days, time to full suck feeds, and individual domain scores on BSID-III at 2 years. Results: Of 212 randomized neonates, the mean (SD) gestational age was 39.2 (1.7) weeks and 122 (58%) were male; 152 (72%) had moderate to severe hypoxic-ischemic encephalopathy (HIE) and 147 (84%) had electrographic seizures. A total of 86 neonates were included in the ESG group and 86 were included in the CSG group. Ten of 86 (9%) neonates in the ESG and 4 of 86 (4%) in the CSG died before the 2-year assessment. The odds of the primary outcome were not significantly different in the ESG group compared with the CSG group (ESG, 38 of 86 [44%] vs CSG, 27 of 86 [31%]; odds ratio [OR], 1.83; 95% CI, 0.96 to 3.49; P = .14). There was also no significant difference in those with HIE (OR, 1.77; 95% CI, 0.84 to 3.73; P = .26). There was evidence that cognitive outcomes were worse in the ESG (mean [SD] scores, ESG: 97.4 [17.7] vs CSG: 103.8 [17.3]; mean difference, -6.5 [95% CI, -1.2 to -11.8]; P = .01). There was little evidence of a difference in secondary outcomes, including time to suck feeds, seizure burden, or brain injury score. Conclusions and Relevance: Treating electrographic and clinical seizures with currently used anticonvulsants did not significantly reduce the rate of death or disability at 2 years in a heterogeneous group of neonates with seizures. Trial Registration: http://anzctr.org.au Identifier: ACTRN12611000327987.


Subject(s)
Anticonvulsants/therapeutic use , Electroencephalography , Hypoxia-Ischemia, Brain/diagnosis , Seizures/diagnosis , Seizures/drug therapy , Australia , Female , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Magnetic Resonance Imaging , Male , Prospective Studies , Seizures/mortality
20.
NPJ Regen Med ; 6(1): 75, 2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34795316

ABSTRACT

The foetal brain is particularly vulnerable to the detrimental effects of foetal growth restriction (FGR) with subsequent abnormal neurodevelopment being common. There are no current treatments to protect the FGR newborn from lifelong neurological disorders. This study examines whether pure foetal mesenchymal stromal cells (MSC) and endothelial colony-forming cells (ECFC) from the human term placenta are neuroprotective through modulating neuroinflammation and supporting the brain vasculature. We determined that one dose of combined MSC-ECFCs (cECFC; 106 ECFC 106 MSC) on the first day of life to the newborn FGR piglet improved damaged vasculature, restored the neurovascular unit, reduced brain inflammation and improved adverse neuronal and white matter changes present in the FGR newborn piglet brain. These findings could not be reproduced using MSCs alone. These results demonstrate cECFC treatment exerts beneficial effects on multiple cellular components in the FGR brain and may act as a neuroprotectant.

SELECTION OF CITATIONS
SEARCH DETAIL
...