1
|
Murillo C, Eixarch E, Rueda C, Larroya M, Boada D, Grau L, Ponce J, Aldecoa V, Monterde E, Ferrero S, Andreu-Fernández V, Arca G, Oleaga L, Ros O, Hernández MP, Gratacós E, Palacio M, Cobo T. Evidence of brain injury in fetuses of mothers with preterm labor with intact membranes and preterm premature rupture of membranes. Am J Obstet Gynecol 2025; 232:114.e1-114.e24. [PMID: 38685550 DOI: 10.1016/j.ajog.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Brain injury and poor neurodevelopment have been consistently reported in infants and adults born before term. These changes occur, at least in part, prenatally and are associated with intra-amniotic inflammation. The pattern of brain changes has been partially documented by magnetic resonance imaging but not by neurosonography along with amniotic fluid brain injury biomarkers. OBJECTIVE This study aimed to evaluate the prenatal features of brain remodeling and injury in fetuses from patients with preterm labor with intact membranes or preterm premature rupture of membranes and to investigate the potential influence of intra-amniotic inflammation as a risk mediator. STUDY DESIGN In this prospective cohort study, fetal brain remodeling and injury were evaluated using neurosonography and amniocentesis in singleton pregnant patients with preterm labor with intact membranes or preterm premature rupture of membranes between 24.0 and 34.0 weeks of gestation, with (n=41) and without (n=54) intra-amniotic inflammation. The controls for neurosonography were outpatient pregnant patients without preterm labor or preterm premature rupture of membranes matched 2:1 by gestational age at ultrasound. Amniotic fluid controls were patients with an amniocentesis performed for indications other than preterm labor or preterm premature rupture of membranes without brain or genetic defects whose amniotic fluid was collected in our biobank for research purposes matched by gestational age at amniocentesis. The group with intra-amniotic inflammation included those with intra-amniotic infection (microbial invasion of the amniotic cavity and intra-amniotic inflammation) and those with sterile inflammation. Microbial invasion of the amniotic cavity was defined as a positive amniotic fluid culture and/or positive 16S ribosomal RNA gene. Inflammation was defined by amniotic fluid interleukin 6 concentrations of >13.4 ng/mL in preterm labor and >1.43 ng/mL in preterm premature rupture of membranes. Neurosonography included the evaluation of brain structure biometric parameters and cortical development. Neuron-specific enolase, protein S100B, and glial fibrillary acidic protein were selected as amniotic fluid brain injury biomarkers. Data were adjusted for cephalic biometrics, fetal growth percentile, fetal sex, noncephalic presentation, and preterm premature rupture of membranes at admission. RESULTS Fetuses from mothers with preterm labor with intact membranes or preterm premature rupture of membranes showed signs of brain remodeling and injury. First, they had a smaller cerebellum. Thus, in the intra-amniotic inflammation, non-intra-amniotic inflammation, and control groups, the transcerebellar diameter measurements were 32.7 mm (interquartile range, 29.8-37.6), 35.3 mm (interquartile range, 31.2-39.6), and 35.0 mm (interquartile range, 31.3-38.3), respectively (P=.019), and the vermian height measurements were 16.9 mm (interquartile range, 15.5-19.6), 17.2 mm (interquartile range, 16.0-18.9), and 17.1 mm (interquartile range, 15.7-19.0), respectively (P=.041). Second, they presented a lower corpus callosum area (0.72 mm2 [interquartile range, 0.59-0.81], 0.71 mm2 [interquartile range, 0.63-0.82], and 0.78 mm2 [interquartile range, 0.71-0.91], respectively; P=.006). Third, they showed delayed cortical maturation (the Sylvian fissure depth-to-biparietal diameter ratios were 0.14 [interquartile range, 0.12-0.16], 0.14 [interquartile range, 0.13-0.16], and 0.16 [interquartile range, 0.15-0.17], respectively [P<.001], and the right parieto-occipital sulci depth ratios were 0.09 [interquartile range, 0.07-0.12], 0.11 [interquartile range, 0.09-0.14], and 0.11 [interquartile range, 0.09-0.14], respectively [P=.012]). Finally, regarding amniotic fluid brain injury biomarkers, fetuses from mothers with preterm labor with intact membranes or preterm premature rupture of membranes had higher concentrations of neuron-specific enolase (11,804.6 pg/mL [interquartile range, 6213.4-21,098.8], 8397.7 pg/mL [interquartile range, 3682.1-17,398.3], and 2393.7 pg/mL [interquartile range, 1717.1-3209.3], respectively; P<.001), protein S100B (2030.6 pg/mL [interquartile range, 993.0-4883.5], 1070.3 pg/mL [interquartile range, 365.1-1463.2], and 74.8 pg/mL [interquartile range, 44.7-93.7], respectively; P<.001), and glial fibrillary acidic protein (1.01 ng/mL [interquartile range, 0.54-3.88], 0.965 ng/mL [interquartile range, 0.59-2.07], and 0.24 mg/mL [interquartile range, 0.20-0.28], respectively; P=.002). CONCLUSION Fetuses with preterm labor with intact membranes or preterm premature rupture of membranes had prenatal signs of brain remodeling and injury at the time of clinical presentation. These changes were more pronounced in fetuses with intra-amniotic inflammation.
Collapse
Affiliation(s)
- Clara Murillo
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Elisenda Eixarch
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain; Center for Biomedical Research on Rare Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Claudia Rueda
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Marta Larroya
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - David Boada
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain
| | - Laia Grau
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain
| | - Júlia Ponce
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain
| | - Victoria Aldecoa
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain
| | - Elena Monterde
- Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Silvia Ferrero
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain
| | - Vicente Andreu-Fernández
- Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain; Biosanitary Research Institute, Valencian International University, Valencia, Spain
| | - Gemma Arca
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Laura Oleaga
- Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain; Department of Radiology, Clinical Diagnostic Imaging Centre, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Olga Ros
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain
| | - Maria Pilar Hernández
- Department of Radiology, Clinical Diagnostic Imaging Centre, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Eduard Gratacós
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain; Center for Biomedical Research on Rare Diseases, Institute of Health Carlos III, Madrid, Spain.
| | - Montse Palacio
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain; Center for Biomedical Research on Rare Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Teresa Cobo
- BCNatal Fetal Medicine Research Center, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic de Barcelona and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetrícia i Neonatología, Barcelona, Spain; Fundació de Recerca Clínica Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer, Universitat de Barcelona, Barcelona, Spain; Center for Biomedical Research on Rare Diseases, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Prasad J, Van Steenwinckel J, Gunn AJ, Bennet L, Korzeniewski SJ, Gressens P, Dean JM. Chronic Inflammation Offers Hints About Viable Therapeutic Targets for Preeclampsia and Potentially Related Offspring Sequelae. Int J Mol Sci 2024; 25:12999. [PMID: 39684715 DOI: 10.3390/ijms252312999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The combination of hypertension with systemic inflammation during pregnancy is a hallmark of preeclampsia, but both processes also convey dynamic information about its antecedents and correlates (e.g., fetal growth restriction) and potentially related offspring sequelae. Causal inferences are further complicated by the increasingly frequent overlap of preeclampsia, fetal growth restriction, and multiple indicators of acute and chronic inflammation, with decreased gestational length and its correlates (e.g., social vulnerability). This complexity prompted our group to summarize information from mechanistic studies, integrated with key clinical evidence, to discuss the possibility that sustained or intermittent systemic inflammation-related phenomena offer hints about viable therapeutic targets, not only for the prevention of preeclampsia, but also the neurobehavioral and other developmental deficits that appear to be overrepresented in surviving offspring. Importantly, we feel that carefully designed hypothesis-driven observational studies are necessary if we are to translate the mechanistic evidence into child health benefits, namely because multiple pregnancy disorders might contribute to heightened risks of neuroinflammation, arrested brain development, or dysconnectivity in survivors who exhibit developmental problems later in life.
Collapse
Affiliation(s)
- Jaya Prasad
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | | | - Alistair J Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Steven J Korzeniewski
- C.S. Mott Center for Human Growth and Development, Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Pierre Gressens
- Inserm, Neurodiderot, Université de Paris, 75019 Paris, France
- Centre for the Developing Brain, Division of Imaging Sciences and Department of Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, UK
| | - Justin M Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
3
|
Lin J, Zhao X, Qi X, Zhao W, Teng S, Mo T, Xiao X, Li P, Chen T, Yun G, Zeng H. Structural covariance alterations reveal motor damage in periventricular leukomalacia. Brain Commun 2024; 6:fcae405. [PMID: 39600524 PMCID: PMC11589463 DOI: 10.1093/braincomms/fcae405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 08/15/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Periventricular leukomalacia is a common neuroimaging finding in patients with spastic cerebral palsy. Myelin damage disrupts neuronal connectivity. However, specific alterations in the grey matter structure and their impact on the whole brain remain unclear, particularly when differentiating between preterm and full-term periventricular leukomalacia. This study investigated the grey matter network alterations following early white matter injury in infants and young children. High-resolution T1-weighted 3 T brain magnetic resonance imaging, clinical data and motor function scores were collected from 42 children with periventricular leukomalacia and 38 age- and sex-matched healthy controls. Based on gestational age, the periventricular leukomalacia group was stratified into preterm (n = 27) and full-term (n = 15) groups. Voxel-based morphometry was used to analyse whole-brain structural metrics, and motor-related regions were selected as nodes for network construction. Structural covariance analysis was used to quantify the strength of the structural connections between grey matter regions, and graph theory metrics were used to assess network properties. Motor assessments included gross and fine motor skills, and their associations with brain regions were analysed. Both preterm and full-term periventricular leukomalacia groups exhibited abnormal motor networks. Preterm periventricular leukomalacia showed more extensive central grey matter nuclei atrophy, whereas full-term periventricular leukomalacia was predominantly localized to the motor cortex. Children with periventricular leukomalacia displayed decreased connectivity between the central grey matter nuclei and other regions, coupled with increased connectivity between the motor cortex and cerebellar hemispheres. Thalamic volume correlated with gross motor scores in preterm infants. These findings suggest that ischaemic-hypoxic injury disrupts motor grey matter networks, with preterm infants being more severely affected. This study highlights the potential of structural covariance patterns for monitoring brain development and advancing our understanding of aberrant brain development in children with periventricular leukomalacia.
Collapse
Affiliation(s)
- Jieqiong Lin
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen 518038, Guangdong Province, China
| | - Xin Zhao
- China Medical University, Shenyang 110122, Liaoning Province, China
| | - Xinxin Qi
- China Medical University, Shenyang 110122, Liaoning Province, China
| | - Wen Zhao
- Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Songyu Teng
- China Medical University, Shenyang 110122, Liaoning Province, China
| | - Tong Mo
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen 518038, Guangdong Province, China
| | - Xin Xiao
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen 518038, Guangdong Province, China
| | - Peng Li
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen 518038, Guangdong Province, China
| | - Turong Chen
- Department of Rehabilitation, Shenzhen Children’s Hospital, Shenzhen 518038, Guangdong Province, China
| | - Guojun Yun
- Department of Rehabilitation, Shenzhen Children’s Hospital, Shenzhen 518038, Guangdong Province, China
| | - Hongwu Zeng
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen 518038, Guangdong Province, China
- Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
4
|
Lear BA, Zhou KQ, Dhillon SK, Lear CA, Bennet L, Gunn AJ. Preventive, rescue and reparative neuroprotective strategies for the fetus and neonate. Semin Fetal Neonatal Med 2024; 29:101542. [PMID: 39472238 DOI: 10.1016/j.siny.2024.101542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Neonatal encephalopathy remains a major contributor to death and disability around the world. Acute hypoxia-ischaemia before, during or after birth creates a series of events that can lead to neonatal brain injury. Understanding the evolution of injury underpinned the development of therapeutic hypothermia. This review discusses the determinants of injury, including maturity, the pattern of exposure to HI, impaired placental function, often associated with fetal growth restriction and in the long-term, socio-economic deprivation. Chorioamnionitis has been associated with the presence of NE, but it is important to note that experimentally, inflammation can either sensitize to greater neural injury after HI or alleviate injury, depending on its precise timing. As fetal surveillance tools improve it is likely that improved detection of specific pathways will offer future opportunities for preventive and reparative interventions in utero and after birth.
Collapse
Affiliation(s)
- Benjamin A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Kelly Q Zhou
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Simerdeep K Dhillon
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
5
|
Leach JL, Derinkuyu BE, Taylor JM, Vadivelu S. Imaging of Hemorrhagic Stroke in Children. Neuroimaging Clin N Am 2024; 34:615-636. [PMID: 39461768 DOI: 10.1016/j.nic.2024.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Hemorrhagic stroke (HS) is an important cause of neurologic morbidity and mortality in children and is more common than ischemic stroke between the ages of 1 and 14 years, a notable contradistinction relative to adult stroke epidemiology. Rapid neuroimaging is of the utmost importance in making the diagnosis of HS, identifying a likely etiology, and directing acute care. Computed tomography and MR imaging with flow-sensitive MR imaging and other noninvasive vascular imaging studies play a primary role in the initial diagnostic evaluation. Catheter-directed digital subtraction angiography is critical for definitive diagnosis and treatment planning.
Collapse
Affiliation(s)
- James L Leach
- Division of Pediatric Neuroradiology, Department of Radiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Betul E Derinkuyu
- Division of Pediatric Neuroradiology, Department of Radiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John Michael Taylor
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sudhakar Vadivelu
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
6
|
Buchmayer J, Fuiko R, Kienast P, Stummer S, Kasprian G, Berger A, Goeral K. Cerebellar haemorrhage and atrophy in infants born extremely preterm with intraventricular haemorrhage. Dev Med Child Neurol 2024. [PMID: 39428664 DOI: 10.1111/dmcn.16123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024]
Abstract
AIM To investigate the impact of cerebellar haemorrhage (CBH) and atrophy in infants born extremely preterm with intraventricular haemorrhage (IVH) on neurodevelopment at 2 years of age. METHOD This retrospective case-control study included infants born at less than 28 weeks' gestation with IVH over a 10-year period. CBH, along with the assessment of cerebellar size, using magnetic resonance imaging, were studied. The impact of injuries on neurodevelopmental outcome at 2 years' corrected age was conducted, using multivariable regression analysis for comprehensive evaluation. RESULTS In a cohort of 103 patients, 69 (67.0%) showed CBH with a median grade of 1 (interquartile range = 0-3). At the corrected age of 2 years, CBH was significantly associated with impaired cognitive and motor outcome. CBH emerged as an independent predictor of poor cognitive and motor development, as well as cerebral palsy. Cerebellar atrophy, affecting 30 (29.1%) infants, was linked to a significantly worse outcome across all domains. Conversely, an increase in cerebellar size was correlated with improved motor development. INTERPRETATION Infants born extremely preterm with IVH and concomitant CBH exhibited significant cognitive and motor impairment. The severity of developmental delay correlated with the grade of CBH. These findings hold potential to support the prediction of long-term outcome and parental counselling.
Collapse
Affiliation(s)
- Julia Buchmayer
- Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna, Vienna, Austria
| | - Renate Fuiko
- Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna, Vienna, Austria
| | - Patric Kienast
- Department of Radiology, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Vienna, Austria
| | - Sophie Stummer
- Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna, Vienna, Austria
| | - Gregor Kasprian
- Department of Radiology, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna, Vienna, Austria
| | - Katharina Goeral
- Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Jafrasteh B, Lubián-López SP, Trimarco E, Ruiz MR, Barrios CR, Almagro YM, Benavente-Fernández I. MGA-Net: A novel mask-guided attention neural network for precision neonatal brain imaging. Neuroimage 2024; 300:120872. [PMID: 39349149 DOI: 10.1016/j.neuroimage.2024.120872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024] Open
Abstract
In this study, we introduce MGA-Net, a novel mask-guided attention neural network, which extends the U-net model for precision neonatal brain imaging. MGA-Net is designed to extract the brain from other structures and reconstruct high-quality brain images. The network employs a common encoder and two decoders: one for brain mask extraction and the other for brain region reconstruction. A key feature of MGA-Net is its high-level mask-guided attention module, which leverages features from the brain mask decoder to enhance image reconstruction. To enable the same encoder and decoder to process both MRI and ultrasound (US) images, MGA-Net integrates sinusoidal positional encoding. This encoding assigns distinct positional values to MRI and US images, allowing the model to effectively learn from both modalities. Consequently, features learned from a single modality can aid in learning a modality with less available data, such as US. We extensively validated the proposed MGA-Net on diverse and independent datasets from varied clinical settings and neonatal age groups. The metrics used for assessment included the DICE similarity coefficient, recall, and accuracy for image segmentation; structural similarity for image reconstruction; and root mean squared error for total brain volume estimation from 3D ultrasound images. Our results demonstrate that MGA-Net significantly outperforms traditional methods, offering superior performance in brain extraction and segmentation while achieving high precision in image reconstruction and volumetric analysis. Thus, MGA-Net represents a robust and effective preprocessing tool for MRI and 3D ultrasound images, marking a significant advance in neuroimaging that enhances both research and clinical diagnostics in the neonatal period and beyond.
Collapse
Affiliation(s)
- Bahram Jafrasteh
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University, Cádiz, Spain.
| | - Simón Pedro Lubián-López
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University, Cádiz, Spain; Division of Neonatology, Department of Pediatrics, Puerta del Mar University Hospital, Cádiz, Spain.
| | - Emiliano Trimarco
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University, Cádiz, Spain
| | - Macarena Román Ruiz
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University, Cádiz, Spain
| | - Carmen Rodríguez Barrios
- Division of Neonatology, Department of Pediatrics, Puerta del Mar University Hospital, Cádiz, Spain
| | - Yolanda Marín Almagro
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University, Cádiz, Spain
| | - Isabel Benavente-Fernández
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University, Cádiz, Spain; Division of Neonatology, Department of Pediatrics, Puerta del Mar University Hospital, Cádiz, Spain; Area of Pediatrics, Department of Child and Mother Health and Radiology, Medical School, University of Cádiz, Cádiz, Spain.
| |
Collapse
|
8
|
Brandt M, Kosmeijer C, Achterberg E, de Theije C, Nijboer C. Timed fetal inflammation and postnatal hypoxia cause cortical white matter injury, interneuron imbalances, and behavioral deficits in a double-hit rat model of encephalopathy of prematurity. Brain Behav Immun Health 2024; 40:100817. [PMID: 39188404 PMCID: PMC11345510 DOI: 10.1016/j.bbih.2024.100817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 08/28/2024] Open
Abstract
Extreme preterm birth-associated adversities are a major risk factor for aberrant brain development, known as encephalopathy of prematurity (EoP), which can lead to long-term neurodevelopmental impairments. Although progress in clinical care for preterm infants has markedly improved perinatal outcomes, there are currently no curative treatment options available to combat EoP. EoP has a multifactorial etiology, including but not limited to pre- or postnatal immune activation and oxygen fluctuations. Elucidating the underlying mechanisms of EoP and determining the efficacy of potential therapies relies on valid, clinically translatable experimental models that reflect the neurodevelopmental and pathophysiological hallmarks of EoP. Here, we expand on our double-hit rat model that can be used to study EoP disease mechanisms and therapeutic options in a preclinical setting. Pregnant Wistar dams were intraperitoneally injected with 10 μg/kg LPS on embryonic day (E)20 and offspring was subjected to hypoxia (140 min, 8% O2) at postnatal day 4. Rats exposed to fetal inflammation and postnatal hypoxia (FIPH) showed neurodevelopmental impairments, such as reduced nest-seeking ability, ultrasonic vocalizations, social engagement, and working memory, and increased anxiety and sensitivity. Impairments in myelination, oligodendrocyte maturation and interneuron development were examined as hallmarks for EoP, in different layers and coordinates of the cortex using histological and molecular techniques. Myelin density and complexity was decreased in the cortex, which partially coincided with a decrease in mature oligodendrocytes. Furthermore, interneuron populations (GAD67+ and PVALB+) were affected. To determine if the timing of inducing fetal inflammation affected the severity of EoP hallmarks in the cortex, multiple timepoints of fetal inflammation were compared. Inflammation at E20 combined with postnatal hypoxia gave the most severe EoP phenotype in the cortex. In conclusion, we present a double-hit rat model which displays various behavioral, anatomical and molecular hallmarks of EoP, including diffuse white matter injury. This double-hit model can be used to investigate pathophysiological mechanisms and potential therapies for EoP.
Collapse
Affiliation(s)
- M.J.V. Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| | - C.M. Kosmeijer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| | - E.J.M. Achterberg
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, the Netherlands
| | - C.G.M. de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| | - C.H. Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| |
Collapse
|
9
|
Fuma K, Kotani T, Tsuda H, Oshiro M, Tano S, Ushida T, Imai K, Sato Y, Kajiyama H. Impact of antenatal corticosteroids-to-delivery interval on very preterm neonatal outcomes: a retrospective study in two tertiary centers in Japan. BMC Pregnancy Childbirth 2024; 24:607. [PMID: 39294574 PMCID: PMC11411863 DOI: 10.1186/s12884-024-06790-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Antenatal corticosteroids (ACS) are administered to prevent neonatal complications and death in women at risk of imminent preterm birth. Internationally, the optimal interval from ACS to delivery (ACS-to-delivery interval) is within seven days; however, evidence in Asian populations specifically is limited. This study aimed to investigate the association between ACS-to-delivery interval and the incidence of neonatal complications in Japan. METHODS This retrospective observational study enrolled singleton neonates born preterm at < 32 weeks of gestational age between 2012 and 2020 at two tertiary centers. A total of 625 neonates were divided into the following four groups according to the timing of ACS (measured in days): no ACS (n = 145), partial ACS (n = 85), ACS 1-7 (n = 307), and ACS ≥ 8 (n = 88). The following outcomes were compared between the groups: treated respiratory distress syndrome (RDS), severe intraventricular hemorrhage (IVH), treated patent ductus arteriosus (PDA), necrotizing enterocolitis, sepsis, bronchopulmonary dysplasia (BPD), treated retinopathy of prematurity (ROP), periventricular leukomalacia, and death discharge. RESULTS The ACS 1-7 group had significantly decreased adjusted odds ratios (ORs) for treated RDS (0.37 [95% confidence interval: 0.23, 0.57]), severe IVH (0.21 [0.07, 0.63]), treated PDA (0.47 [0.29, 0.75]), and treated ROP (0.50 [0.25, 0.99]) compared with the no ACS group. The ACS ≥ 8 group also showed significantly reduced adjusted ORs for RDS (0.37 [0.20, 0.66]) and treated PDA (0.48 [0.25, 0.91]) compared with the no ACS group. However, the adjusted ORs for BPD significantly increased in both the ACS 1-7 (1.86 [1.06, 3.28]) and ACS ≥ 8 groups (2.94 [1.43, 6.05]) compared to the no ACS group. CONCLUSIONS An ACS-to-delivery interval of 1-7 days achieved the lowest incidence of several complications in preterm neonates born at < 32 weeks of gestational age. Some of the favorable effects of ACS seem to continue even beyond ≥ 8 days from administration. In contrast, ACS might be associated with an increased incidence of BPD, which was most likely to be prominent in neonates delivered ≥ 8 days after receiving ACS. Based on these findings, the duration of the effect of ACS on neonatal complications should be studied further.
Collapse
MESH Headings
- Humans
- Retrospective Studies
- Female
- Infant, Newborn
- Japan/epidemiology
- Pregnancy
- Tertiary Care Centers
- Adrenal Cortex Hormones/administration & dosage
- Male
- Gestational Age
- Infant, Extremely Premature
- Respiratory Distress Syndrome, Newborn/prevention & control
- Respiratory Distress Syndrome, Newborn/epidemiology
- Premature Birth/epidemiology
- Premature Birth/prevention & control
- Adult
- Infant, Premature, Diseases/prevention & control
- Infant, Premature, Diseases/epidemiology
- Time Factors
- Prenatal Care/methods
- Delivery, Obstetric/methods
- Delivery, Obstetric/statistics & numerical data
- Infant, Premature
Collapse
Affiliation(s)
- Kazuya Fuma
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
- Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan.
| | - Hiroyuki Tsuda
- Department of Obstetrics and Gynecology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, 453-8511, Japan
| | - Makoto Oshiro
- Department of Pediatrics, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, 453-8511, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| |
Collapse
|
10
|
Drommelschmidt K, Mayrhofer T, Hüning B, Stein A, Foldyna B, Schweiger B, Felderhoff-Müser U, Sirin S. Incidence of brain injuries in a large cohort of very preterm and extremely preterm infants at term-equivalent age: results of a single tertiary neonatal care center over 10 years. Eur Radiol 2024; 34:5239-5249. [PMID: 38279057 PMCID: PMC11255071 DOI: 10.1007/s00330-024-10592-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/30/2023] [Accepted: 12/22/2023] [Indexed: 01/28/2024]
Abstract
OBJECTIVES Cerebral magnetic resonance imaging (cMRI) at term-equivalent age (TEA) can detect brain injury (BI) associated with adverse neurological outcomes in preterm infants. This study aimed to assess BI incidences in a large, consecutive cohort of preterm infants born < 32 weeks of gestation, the comparison between very (VPT, ≥ 28 + 0 to < 32 + 0 weeks of gestation) and extremely preterm infants (EPT, < 28 + 0 weeks of gestation) and across weeks of gestation. METHODS We retrospectively analyzed cMRIs at TEA of VPT and EPT infants born at a large tertiary center (2009-2018). We recorded and compared the incidences of BI, severe BI, intraventricular hemorrhage (IVH), periventricular hemorrhagic infarction (PVHI), cerebellar hemorrhage (CBH), cystic periventricular leukomalacia (cPVL), and punctate white matter lesions (PWML) between VPTs, EPTs, and across weeks of gestation. RESULTS We included 507 preterm infants (VPT, 335/507 (66.1%); EPT, 172/507 (33.9%); mean gestational age (GA), 28 + 2 weeks (SD 2 + 2 weeks); male, 52.1%). BIs were found in 48.3% of the preterm infants (severe BI, 12.0%) and increased with decreasing GA. IVH, PVHI, CBH, cPVL, and PWML were seen in 16.8%, 0.8%, 10.5%, 3.4%, and 18.1%, respectively. EPT vs. VPT infants suffered more frequently from BI (59.3% vs. 42.7%, p < 0.001), severe BI (18.6% vs. 8.7%, p = 0.001), IVH (31.9% vs. 9.0%, p < 0.001), and CBH (18.0% vs. 6.6%, p < 0.001). CONCLUSION Brain injuries are common cMRI findings among preterm infants with a higher incidence of EPT compared to VPT infants. These results may serve as reference values for clinical management and research. CLINICAL RELEVANCE STATEMENT Our results with regard to gestational age might provide valuable clinical insights, serving as a key reference for parental advice, structured follow-up planning, and enhancing research and management within the Neonatal Intensive Care Unit. KEY POINTS • Brain injury is a common cMRI finding in preterm infants seen in 48.3% individuals. • Extremely preterm compared to very preterm infants have higher brain injury incidences driven by brain injuries such as intraventricular and cerebellar hemorrhage. • Reference incidence values are crucial for parental advice and structured follow-up planning.
Collapse
Affiliation(s)
- Karla Drommelschmidt
- Department of Pediatrics I, Neonatology, Pediatric Intensive Care, and Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (cTNBS), University Medicine Essen, Essen, Germany
| | - Thomas Mayrhofer
- School of Business Studies, Stralsund, University of Applied Sciences, Stralsund, Germany
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital - Harvard Medical School, Boston, MA, USA
| | - Britta Hüning
- Department of Pediatrics I, Neonatology, Pediatric Intensive Care, and Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (cTNBS), University Medicine Essen, Essen, Germany
| | - Anja Stein
- Department of Pediatrics I, Neonatology, Pediatric Intensive Care, and Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (cTNBS), University Medicine Essen, Essen, Germany
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital - Harvard Medical School, Boston, MA, USA
| | - Bernd Schweiger
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Müser
- Department of Pediatrics I, Neonatology, Pediatric Intensive Care, and Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (cTNBS), University Medicine Essen, Essen, Germany
| | - Selma Sirin
- Department of Diagnostic Imaging, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
11
|
Vanderhasselt T, Naeyaert M, Buls N, Allemeersch GJ, Raeymaeckers S, Raeymaekers H, Smeets N, Cools F, de Mey J, Dudink J. Synthetic magnetic resonance-based relaxometry and brain volume: cutoff values for predicting neurocognitive outcomes in very preterm infants. Pediatr Radiol 2024; 54:1523-1531. [PMID: 38980354 PMCID: PMC11324712 DOI: 10.1007/s00247-024-05981-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Early neurorehabilitation can enhance neurocognitive outcomes in very preterm infants (<32 weeks), and conventional magnetic resonance imaging (MRI) is commonly used to assess neonatal brain injury; however, the predictive value for neurodevelopmental delay is limited. Timely predictive quantitative biomarkers are needed to improve early identification and management of infants at risk of neurodevelopmental delay. OBJECTIVE To evaluate the potential of quantitative synthetic MRI measurements at term-equivalent age as predictive biomarkers of neurodevelopmental impairment and establish practical cutoff values to guide clinical decision-making. MATERIALS AND METHODS This retrospective study included 93 very preterm infants who underwent synthetic MRI at term-equivalent age between January 2017 and September 2020. Clinical outcomes were assessed using the Bayley-III scale of infant development (mean age 2.1 years). The predictive value for impaired development was analyzed using receiver operating characteristic curves for synthetic MRI-based volumetry and T1 and T2 relaxation measurements. RESULTS The T1 relaxation time in the posterior limb of the internal capsule was a potent predictor of severe (sensitivity, 92%; specificity, 80%; area under the curve (AUC), 0.91) and mild or severe (AUC, 0.75) developmental impairment. T2 relaxation time in the posterior limb of the internal capsule was a significant predictor of severe impairment (AUC, 0.76), whereas the brain parenchymal volume was a significant predictor of severe (AUC, 0.72) and mild or severe impairment (AUC, 0.71) outperforming the reported qualitative MRI scores (AUC, 0.66). CONCLUSION The proposed cutoff values for T1 relaxation time in the posterior limb of the internal capsule and for total brain volume measurements, derived from synthetic MRI, show promise as predictors of both mild and severe neurodevelopmental impairment in very preterm infants.
Collapse
Affiliation(s)
- Tim Vanderhasselt
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium.
| | - Maarten Naeyaert
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Nico Buls
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Gert-Jan Allemeersch
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steven Raeymaeckers
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Hubert Raeymaekers
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Nathalie Smeets
- Department of Pediatric Neurology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Filip Cools
- Department of Neonatology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Johan de Mey
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
12
|
Kienast P, Schmidbauer V, Yildirim MS, Seeliger S, Stuempflen M, Elis J, Giordano V, Fuiko R, Olischar M, Vierlinger K, Noehammer C, Berger A, Prayer D, Kasprian G, Goeral K. Neurodevelopmental outcome in preterm infants with intraventricular hemorrhages: the potential of quantitative brainstem MRI. Cereb Cortex 2024; 34:bhae189. [PMID: 38715405 PMCID: PMC11077078 DOI: 10.1093/cercor/bhae189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
OBJECTIVES This retrospective study aimed to identify quantitative magnetic resonance imaging markers in the brainstem of preterm neonates with intraventricular hemorrhages. It delves into the intricate associations between quantitative brainstem magnetic resonance imaging metrics and neurodevelopmental outcomes in preterm infants with intraventricular hemorrhage, aiming to elucidate potential relationships and their clinical implications. MATERIALS AND METHODS Neuroimaging was performed on preterm neonates with intraventricular hemorrhage using a multi-dynamic multi-echo sequence to determine T1 relaxation time, T2 relaxation time, and proton density in specific brainstem regions. Neonatal outcome scores were collected using the Bayley Scales of Infant and Toddler Development. Statistical analysis aimed to explore potential correlations between magnetic resonance imaging metrics and neurodevelopmental outcomes. RESULTS Sixty preterm neonates (mean gestational age at birth 26.26 ± 2.69 wk; n = 24 [40%] females) were included. The T2 relaxation time of the midbrain exhibited significant positive correlations with cognitive (r = 0.538, P < 0.0001, Pearson's correlation), motor (r = 0.530, P < 0.0001), and language (r = 0.449, P = 0.0008) composite scores at 1 yr of age. CONCLUSION Quantitative magnetic resonance imaging can provide valuable insights into neurodevelopmental outcomes after intraventricular hemorrhage, potentially aiding in identifying at-risk neonates. Multi-dynamic multi-echo sequence sequences hold promise as an adjunct to conventional sequences, enhancing the sensitivity of neonatal magnetic resonance neuroimaging and supporting clinical decision-making for these vulnerable patients.
Collapse
Affiliation(s)
- Patric Kienast
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Victor Schmidbauer
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Mehmet Salih Yildirim
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Selina Seeliger
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Marlene Stuempflen
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Julia Elis
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Vito Giordano
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Renate Fuiko
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Monika Olischar
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Klemens Vierlinger
- Center for Health and Bioresources, Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria
| | - Christa Noehammer
- Center for Health and Bioresources, Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Daniela Prayer
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Gregor Kasprian
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Katharina Goeral
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| |
Collapse
|
13
|
Zhu T, Zhang S, Jiang W, Chai D, Mao J, Wei Y, Xiong J. A Multiplanar Radiomics Model Based on Cranial Ultrasound to Predict the White Matter Injury in Premature Infants and an Analysis of its Correlation With Neurodevelopment. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:899-911. [PMID: 38269595 DOI: 10.1002/jum.16419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/14/2023] [Accepted: 01/07/2024] [Indexed: 01/26/2024]
Abstract
OBJECTIVES To develop and evaluate a multiplanar radiomics model based on cranial ultrasound (CUS) to predict white matter injury (WMI) in premature infants and explore its correlation with neurodevelopment. METHODS We retrospectively reviewed 267 premature infants. The radiomics features were extracted from five standard sections of CUS. The Spearman's correlation coefficient combined with the least absolute shrinkage and selection operator (LASSO) was applied to select features and build radiomics signature, and a multiplanar radiomics model was constructed based on the radiomics signature of five planes. The performance of the model was evaluated using the area under the receiver operating characteristic curve (AUC). Infants with WMI were re-examined by ultrasound at 2 and 4 weeks after birth, and the recovery degree of WMI was evaluated using multiplanar radiomics. The relationship between WMI and the recovery degree and neurodevelopment was analyzed. RESULTS The AUC of the multiplanar radiomics in the training and validation sets were 0.94 and 0.91, respectively. The neurodevelopmental function scores in infants with WMI were significantly lower than those in healthy preterm infants and full-term newborns (P < .001). There were statistically significant differences in the neurodevelopmental function scores of infants between the 2- and 4-week lesion disappearance and 4-week lesion persistence (P < .001). CONCLUSIONS The multiplanar radiomics model showed a good performance in predicting the WMI of premature infants. It can not only provide objective and accurate results but also dynamically monitor the degree of recovery of WMI to predict the prognosis of premature infants.
Collapse
Affiliation(s)
- Ting Zhu
- Department of Ultrasound, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Shuang Zhang
- Educational Technology and Information, Shenzhen Polytechnic University, Shenzhen, China
| | - Wei Jiang
- Department of Ultrasound, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Dan Chai
- Department of Obstetrics, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Jiaoyu Mao
- Department of Neonatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Yuya Wei
- Department of Ultrasound, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Jiayu Xiong
- Department of Ultrasound, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
14
|
Metallinou D, Karampas G, Pavlou ML, Louma MI, Mantzou A, Sarantaki A, Nanou C, Gourounti K, Tzeli M, Pantelaki N, Tzamakos E, Boutsikou T, Lykeridou A, Iacovidou N. Serum Neuron-Specific Enolase as a Biomarker of Neonatal Brain Injury-New Perspectives for the Identification of Preterm Neonates at High Risk for Severe Intraventricular Hemorrhage. Biomolecules 2024; 14:434. [PMID: 38672451 PMCID: PMC11048112 DOI: 10.3390/biom14040434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Neonatal brain injury (NBI) is a critical condition for preterm neonates with potential long-term adverse neurodevelopmental outcomes. This prospective longitudinal case-control study aimed at investigating the levels and prognostic value of serum neuron-specific enolase (NSE) during the first 3 days of life in preterm neonates (<34 weeks) that later developed brain injury in the form of either periventricular leukomalacia (PVL) or intraventricular hemorrhage (IVH) during their hospitalization. Participants were recruited from one neonatal intensive care unit, and on the basis of birth weight and gestational age, we matched each case (n = 29) with a neonate who had a normal head ultrasound scan (n = 29). We report that serum NSE levels during the first three days of life do not differ significantly between control and preterm neonates with NBI. Nevertheless, subgroup analysis revealed that neonates with IVH had significantly higher concentrations of serum NSE in comparison to controls and neonates with PVL on the third day of life (p = 0.014 and p = 0.033, respectively). The same pattern on the levels of NSE on the third day of life was also observed between (a) neonates with IVH and all other neonates (PVL and control; p = 0.003), (b) neonates with II-IV degree IVH and all other neonates (p = 0.003), and (c) between control and the five (n = 5) neonates that died from the case group (p = 0.023). We conclude that NSE could be an effective and useful biomarker on the third day of life for the identification of preterm neonates at high risk of developing severe forms of IVH.
Collapse
Affiliation(s)
- Dimitra Metallinou
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Grigorios Karampas
- Second Department of Obstetrics and Gynaecology, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Maria-Loukia Pavlou
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Maria-Ioanna Louma
- Department of Biochemistry & Biotechnology, University of Thessaly, 41500 Larissa, Greece;
| | - Aimilia Mantzou
- Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, Medical School, Aghia Sophia Children’s Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Antigoni Sarantaki
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Christina Nanou
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Kleanthi Gourounti
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Maria Tzeli
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Nikoletta Pantelaki
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Evangelos Tzamakos
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Theodora Boutsikou
- Department of Neonatology, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (T.B.); (N.I.)
| | - Aikaterini Lykeridou
- Department of Midwifery, University of West Attica, 12243 Athens, Greece; (M.-L.P.); (A.S.); (C.N.); (K.G.); (M.T.); (N.P.); (E.T.); (A.L.)
| | - Nicoletta Iacovidou
- Department of Neonatology, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (T.B.); (N.I.)
| |
Collapse
|
15
|
Feng Y, Wang Y, Li X, Dai L, Zhang J. Differences in the amplitude of low-frequency fluctuations of spontaneous brain activity between preterm and term infants. Front Neurol 2024; 15:1346632. [PMID: 38497040 PMCID: PMC10941683 DOI: 10.3389/fneur.2024.1346632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/22/2024] [Indexed: 03/19/2024] Open
Abstract
Objectives To date, the majority of research on resting-state functional magnetic resonance imaging (rs-fMRI) in the developing brain has primarily centered on adolescents and adults, leaving a gap in understanding variations in spontaneous brain activity at rest in preterm infants. This study aimed to uncover and comprehend the distinctions in spontaneous brain activity between preterm and term infants, with the goal of establishing a foundation for assessing the condition of preterm infants. Methods In this study, 14 term infants and 15 preterm infants with equivalent gestational age were carefully chosen from the neonatal unit of Anhui Provincial Children's Hospital. The amplitude of low-frequency fluctuations (ALFF) intensity was assessed using resting-state functional magnetic resonance imaging (rs-fMRI) to examine brain activity in both groups. Subsequently, the differences between the term and preterm infants were statistically analyzed using a two-sample t-test. A p-value of <0.05, corrected for the REST Gaussian Random Fields, was deemed to be statistically significant. Results In comparison to the term infant group, the preterm infant group exhibited a significant increase in the ALFF value in the left precuneus, left frontal superior orbital gyrus, and left calcarine cortex. Conclusion Significant variances in spontaneous brain activity have been observed in various regions between term infants and preterm infants of equivalent gestational age. These variations could potentially impact the emotional and cognitive development of preterm infants in the long term.
Collapse
Affiliation(s)
- Ye Feng
- Department of Neonatology, Anhui Provincial Children’s Hospital, Hefei, China
| | - Yuanchong Wang
- Department of Neonatology, Anhui Provincial Children’s Hospital, Hefei, China
- Department of Pediatric Medicine, Anhui Provincial Children’s Hospital, Hefei, China
| | - Xu Li
- Department of Imaging, Anhui Provincial Children’s Hospital, Hefei, China
| | - Liying Dai
- Neonate Follow-up Center, Anhui Provincial Children’s Hospital, Hefei, China
| | - Jian Zhang
- Department of Neonatology, Anhui Provincial Children’s Hospital, Hefei, China
- Neonate Follow-up Center, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
16
|
Griesmaier E, Schreiner C, Winkler I, Posod A, Sappler M, Kiechl-Kohlendorfer U, Neubauer V. Association of aEEG and brain injury severity on MRI at term-equivalent age in preterm infants. Acta Paediatr 2024; 113:229-238. [PMID: 37897122 DOI: 10.1111/apa.17017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
AIM Measures to detect and monitor brain injury in preterm infants are amplitude-integrated electroencephalography (aEEG) and magnetic resonance imaging (MRI). To investigate the association between aEEG and MRI in a large cohort of preterm infants. Five hundred and twenty-three preterm infants were included in the study. METHODS AEEG was interpreted for the total maturation score (TMS) according to Burdjalov. Cerebral MRI was evaluated using a validated scoring system by Kidokoro. RESULTS One hundred and forty-six infants (27.9%) showed some form of brain injury, with 111 infants (21.2%) showing mild injury and 35 (6.7%) showing severe injury. TMS were significantly higher in infants without injury compared to severe injury. When comparing infants with isolated intraventricular haemorrhage to infants without brain injury, TMS were significantly lower. CONCLUSION Prediction of adverse outcome is an important aspect of neonatal care. The combination of diagnostic measures evaluating brain injury might enhance our abilities in neonatal care to provide accurate information about later outcome. Early aEEG is predictive for the severity of brain injury detected by MRI at term-equivalent age. Whether aEEG is also predictive for neurodevelopmental outcome needs to be further investigated in relation to the various patterns of preterm brain injury.
Collapse
Affiliation(s)
- Elke Griesmaier
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Schreiner
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Ira Winkler
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Posod
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Maria Sappler
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | | | - Vera Neubauer
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Parillo M, Vaccarino F, Di Gennaro G, Kumar S, Van Goethem J, Beomonte Zobel B, Quattrocchi CC, Parizel PM, Mallio CA. Overview of the Current Knowledge and Conventional MRI Characteristics of Peri- and Para-Vascular Spaces. Brain Sci 2024; 14:138. [PMID: 38391713 PMCID: PMC10886993 DOI: 10.3390/brainsci14020138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/10/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Brain spaces around (perivascular spaces) and alongside (paravascular or Virchow-Robin spaces) vessels have gained significant attention in recent years due to the advancements of in vivo imaging tools and to their crucial role in maintaining brain health, contributing to the anatomic foundation of the glymphatic system. In fact, it is widely accepted that peri- and para-vascular spaces function as waste clearance pathways for the brain for materials such as ß-amyloid by allowing exchange between cerebrospinal fluid and interstitial fluid. Visible brain spaces on magnetic resonance imaging are often a normal finding, but they have also been associated with a wide range of neurological and systemic conditions, suggesting their potential as early indicators of intracranial pressure and neurofluid imbalance. Nonetheless, several aspects of these spaces are still controversial. This article offers an overview of the current knowledge and magnetic resonance imaging characteristics of peri- and para-vascular spaces, which can help in daily clinical practice image description and interpretation. This paper is organized into different sections, including the microscopic anatomy of peri- and para-vascular spaces, their associations with pathological and physiological events, and their differential diagnosis.
Collapse
Affiliation(s)
- Marco Parillo
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Research Unit of Diagnostic Imaging and Interventional Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Federica Vaccarino
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Research Unit of Diagnostic Imaging and Interventional Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Gianfranco Di Gennaro
- Department of Health Sciences, Chair of Medical Statistics, University of Catanzaro "Magna Græcia", 88100 Catanzaro, Italy
| | - Sumeet Kumar
- Department of Neuroradiology, National Neuroscience Institute, Singapore 308433, Singapore
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Johan Van Goethem
- Department of Radiology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Bruno Beomonte Zobel
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Research Unit of Diagnostic Imaging and Interventional Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Carlo Cosimo Quattrocchi
- Centre for Medical Sciences-CISMed, University of Trento, Via S. Maria Maddalena 1, 38122 Trento, Italy
| | - Paul M Parizel
- Royal Perth Hospital & University of Western Australia, Perth, WA 6000, Australia
- Medical School, University of Western Australia, Perth, WA 6009, Australia
| | - Carlo Augusto Mallio
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Research Unit of Diagnostic Imaging and Interventional Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
18
|
Pande GS, Vagha JD. A Review of the Occurrence of Intraventricular Hemorrhage in Preterm Newborns and its Future Neurodevelopmental Consequences. Cureus 2023; 15:e48968. [PMID: 38111458 PMCID: PMC10726079 DOI: 10.7759/cureus.48968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Intraventricular hemorrhage (IVH) is a type of bleeding that occurs through the germinal matrix and comes through the ependymal cells into the ventricular cavity. It is mostly seen in preterm neonates but can also be seen sometimes in term neonates. Various factors predispose to preterm delivery; it can be spontaneous or medically induced. Spontaneous IVH occurs in cases of intrauterine infections in the mother, and it can be induced in cases of medical emergencies such as preeclampsia and eclampsia. The brain of a preterm newborn is not fully developed as it does not have pericytes and proteins, so it can bleed very quickly, which can cause IVH. Also, the vessels supplying the germinal matrix are immature and highly vascularized. IVH has four grades based on findings detected on cranial ultrasound and MRI. Management includes medical and surgical management; medical management includes phenobarbitone used for seizures and prophylaxis. Surgical management includes drainage, irrigation, and fibrinolytic therapy (DRIFT), and neuro-endoscopic lavage. IVH causes various short-term and long-term neurodevelopmental consequences. Long-term complications include cerebral palsy and intellectual disability, which hamper the life of the child. It mainly presents with seizures, flaccidity, decerebrate posture, etc. Various preventive measures can be taken to tackle IVH in newborns. First of all, preterm delivery should be avoided, and intrauterine infections in mothers should be treated. The administration of corticosteroids should be done for all preterm deliveries as it helps in the maturation of organs. The administration of magnesium sulfate should be done as it is neuroprotective and reduces cerebral palsy in the future. Delayed cord clamping is to be done to reduce recurrent blood transfusions and decrease the risk of IVH. This article explains the pathogenesis, management, prevention, and future outcomes of IVH.
Collapse
Affiliation(s)
- Gauri S Pande
- Department of Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Jayant D Vagha
- Department of Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
19
|
Pascal A, de Bruyn N, Naulaers G, Ortibus E, Hanssen B, Oostra A, de Coen K, Sonnaert M, Cloet E, Casaer A, D'Haese J, Laroche S, Jonckheere A, Plaskie K, van Mol C, Bruneel E, van Hoestenberghe MR, Samijn B, Govaert P, Van den Broeck C. The Impact of Intraventricular Hemorrhage and Periventricular Leukomalacia on Mortality and Neurodevelopmental Outcome in Very Preterm and Very Low Birthweight Infants: A Prospective Population-based Cohort Study. J Pediatr 2023; 262:113600. [PMID: 37402440 DOI: 10.1016/j.jpeds.2023.113600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/09/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023]
Abstract
OBJECTIVE To survey the incidence of intraventricular hemorrhage (IVH) and periventricular leukomalacia (PVL) by gestational age and to report the impact on mortality and neurodevelopmental outcome in very preterm/very low birthweight infants. STUDY DESIGN This was a population-based cohort study of 1927 very preterm/very low birthweight infants born in 2014-2016 and admitted to Flemish neonatal intensive care units. Infants underwent standard follow-up assessment until 2 years corrected age with the Bayley Scales of Infant and Toddler Development and neurological assessments. RESULTS No brain lesion was present in 31% of infants born at <26 weeks of gestation and 75.8% in infants born at 29-32 weeks of gestation. The prevalence of low-grade IVH/PVL (grades I and II) was 16.8% and 12.7%, respectively. Low-grade IVH/PVL was not related significantly to an increased likelihood of mortality, motor delay, or cognitive delay, except for PVL grade II, which was associated with a 4-fold increase in developing cerebral palsy (OR, 4.1; 95% CI, 1.2-14.6). High-grade lesions (III-IV) were present in 22.0% of the infants born at <26 weeks of gestational and 3.1% at 29-32 weeks of gestation, and the odds of death were ≥14.0 (IVH: OR, 14.0; 95% CI, 9.0-21.9; PVL: OR, 14.1; 95% CI, 6.6-29.9). PVL grades III-IV showed an increased odds of 17.2 for motor delay and 12.3 for cerebral palsy, but were not found to be associated significantly with cognitive delay (OR, 2.9; 95% CI, 0.5-17.5; P = .24). CONCLUSIONS Both the prevalence and severity of IVH/PVL decreased significantly with advancing gestational age. More than 75% of all infants with low grades of IVH/PVL showed normal motor and cognitive outcome at 2 years corrected age. High-grade PVL/IVH has become less common and is associated with adverse outcomes.
Collapse
Affiliation(s)
- Aurelie Pascal
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium; Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Nele de Bruyn
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
| | - Gunnar Naulaers
- Department of Neonatology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Els Ortibus
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium; Center for Developmental Disabilities, University Hospital Gasthuisberg, Leuven, Belgium
| | - Britta Hanssen
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium; Department of Rehabilitation Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ann Oostra
- Center for Developmental Disorders, University Hospital Ghent, Ghent, Belgium
| | - Kris de Coen
- Department of Neonatology, University Hospital Ghent, Ghent, Belgium
| | - Michel Sonnaert
- Department of Neonatology, University Hospital Brussels, Brussels, Belgium
| | - Eva Cloet
- Department of Pediatric Neurology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Alexandra Casaer
- Center for Developmental Disorders, University Hospital Ghent, Ghent, Belgium; Department of Neonatology, AZ Sint-Jan, Brugge, Brugge, Belgium
| | - James D'Haese
- Department of Neonatology, AZ Sint-Jan, Brugge, Brugge, Belgium
| | - Sabine Laroche
- Department of Neonatology, University Hospital Antwerp, Antwerp, Belgium; Center for Developmental Disorders, University Hospital Antwerp, Antwerp, Belgium
| | - An Jonckheere
- Center for Developmental Disorders, University Hospital Antwerp, Antwerp, Belgium
| | - Katleen Plaskie
- Department of Neonatology, GasthuisZusters Antwerpen, Antwerp, Belgium
| | - Christine van Mol
- Department of Neonatology, GasthuisZusters Antwerpen, Antwerp, Belgium
| | - Els Bruneel
- Department of Neonatology, Algemeen Ziekenhuis Oost-Limburg, Genk, Belgium
| | | | - Bieke Samijn
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
| | - Paul Govaert
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Ghent, Belgium
| | | |
Collapse
|
20
|
Aalten M, Tataranno ML, Dudink J, Lemmers PMA, Lindeboom MYA, Benders MJNL. Brain injury and long-term outcome after neonatal surgery for non-cardiac congenital anomalies. Pediatr Res 2023; 94:1265-1272. [PMID: 37217607 DOI: 10.1038/s41390-023-02629-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND There is growing evidence that neonatal surgery for non-cardiac congenital anomalies (NCCAs) in the neonatal period adversely affects long-term neurodevelopmental outcome. However, less is known about acquired brain injury after surgery for NCCA and abnormal brain maturation leading to these impairments. METHODS A systematic search was performed in PubMed, Embase, and The Cochrane Library on May 6, 2022 on brain injury and maturation abnormalities seen on magnetic resonance imaging (MRI) and its associations with neurodevelopment in neonates undergoing NCCA surgery the first month postpartum. Rayyan was used for article screening and ROBINS-I for risk of bias assessment. Data on the studies, infants, surgery, MRI, and outcome were extracted. RESULTS Three eligible studies were included, reporting 197 infants. Brain injury was found in n = 120 (50%) patients after NCCA surgery. Sixty (30%) were diagnosed with white matter injury. Cortical folding was delayed in the majority of cases. Brain injury and delayed brain maturation was associated with a decrease in neurodevelopmental outcome at 2 years of age. CONCLUSIONS Surgery for NCCA was associated with high risk of brain injury and delay in maturation leading to delay in neurocognitive and motor development. However, more research is recommended for strong conclusions in this group of patients. IMPACT Brain injury was found in 50% of neonates who underwent NCCA surgery. NCCA surgery is associated with a delay in cortical folding. There is an important research gap regarding perioperative brain injury and NCCA surgery.
Collapse
Affiliation(s)
- Mark Aalten
- Department of Neonatology, University Medical Center, Utrecht Brain Center and Wilhelmina Children's Hospital, University Utrecht, Utrecht, Netherlands
| | - Maria Luisa Tataranno
- Department of Neonatology, University Medical Center, Utrecht Brain Center and Wilhelmina Children's Hospital, University Utrecht, Utrecht, Netherlands
| | - Jeroen Dudink
- Department of Neonatology, University Medical Center, Utrecht Brain Center and Wilhelmina Children's Hospital, University Utrecht, Utrecht, Netherlands
| | - Petra M A Lemmers
- Department of Neonatology, University Medical Center, Utrecht Brain Center and Wilhelmina Children's Hospital, University Utrecht, Utrecht, Netherlands
| | - Maud Y A Lindeboom
- Department of Pediatric Surgery, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center, Utrecht Brain Center and Wilhelmina Children's Hospital, University Utrecht, Utrecht, Netherlands.
| |
Collapse
|
21
|
Larson AC, Sridharan A, Moon JK, Agarwal D, Chang J, Wallace KD, Forsberg F, Didier RA. Contrast-enhanced subharmonic aided pressure estimation for assessment of intracranial pressure in vivo. Pediatr Radiol 2023; 53:1640-1647. [PMID: 37062765 DOI: 10.1007/s00247-023-05637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND Intracranial pressure (ICP) monitoring in children currently requires invasive techniques. Subharmonic aided pressure estimation (SHAPE) uses contrast-enhanced ultrasound (CEUS) to measure intravascular and interstitial pressure, but utility in ICP measurements has yet to be explored. OBJECTIVE The objective of this study was to investigate SHAPE as a novel tool for noninvasive ICP measurements in fetal lambs. MATERIALS AND METHODS Eighteen fetal lambs at 107-139 days gestational age (term = 145 days) underwent subdural ICP catheter placement. The brain was imaged in the coronal plane in CEUS mode optimized for SHAPE, while infusing an US contrast agent into the fetal circulation. After SHAPE calibration, saline was infused via the subdural catheter to increase ICP. Five-second SHAPE cine clips were obtained at various ICPs. Subharmonic intensity values of the whole brain and thalami were correlated with ICP values using mixed effects linear regression analyses and the strength of the relationship was evaluated by Spearman's rank-order correlation. RESULTS Forty-nine experiments produced 723 datapoints, including SHAPE intensity values and mean ICP measurements. There was a statistically significant inverse relationship between SHAPE intensity values and ICP measurements in the whole brain and thalami (median rho value - 0.58 and - 0.56, respectively). CONCLUSION SHAPE intensity values of the brain demonstrate an inverse and statistically significant correlation with in vivo ICP measurements in an animal model.
Collapse
Affiliation(s)
- Abby C Larson
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anush Sridharan
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - James K Moon
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Divyansh Agarwal
- Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan Chang
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryne A Didier
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA.
- Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Gonzalez-Moreira E, Harmony T, Hinojosa-Rodríguez M, Carrillo-Prado C, Juárez-Colín ME, Gutiérrez-Hernández CC, Carlier MEM, Cubero-Rego L, Castro-Chavira SA, Fernández T. Prevention of Neurological Sequelae in Preterm Infants. Brain Sci 2023; 13:brainsci13050753. [PMID: 37239225 DOI: 10.3390/brainsci13050753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Preterm birth is one of the world's critical health problems, with an incidence of 5% to 18% of living newborns according to various countries. White matter injuries due to preoligodendrocytes deficits cause hypomyelination in children born preterm. Preterm infants also have multiple neurodevelopmental sequelae due to prenatal and perinatal risk factors for brain damage. The purpose of this work was to explore the effects of the brain risk factors and MRI volumes and abnormalities on the posterior motor and cognitive development at 3 years of age. METHODS A total of 166 preterm infants were examined before 4 months and clinical and MRI evaluations were performed. MRI showed abnormal findings in 89% of the infants. Parents of all infants were invited to receive the Katona neurohabilitation treatment. The parents of 128 infants accepted and received Katona's neurohabilitation treatment. The remaining 38 infants did not receive treatment for a variety of reasons. At the three-year follow-up, Bayley's II Mental Developmental Index (MDI) and the Psychomotor Developmental Index (PDI) were compared between treated and untreated subjects. RESULTS The treated children had higher values of both indices than the untreated. Linear regression showed that the antecedents of placenta disorders and sepsis as well as volumes of the corpus callosum and of the left lateral ventricle significantly predicted both MDI and PDI, while Apgar < 7 and volume of the right lateral ventricle predicted the PDI. CONCLUSIONS (1) The results indicate that preterm infants who received Katona's neurohabilitation procedure exhibited significantly better outcomes at 3 years of age compared to those who did not receive the treatment. (2) The presence of sepsis and the volumes of the corpus callosum and lateral ventricles at 3-4 months were significant predictors of the outcome at 3 years of age.
Collapse
Affiliation(s)
- Eduardo Gonzalez-Moreira
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Thalía Harmony
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - Manuel Hinojosa-Rodríguez
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - Cristina Carrillo-Prado
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - María Elena Juárez-Colín
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - Claudia Calipso Gutiérrez-Hernández
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - María Elizabeth Mónica Carlier
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - Lourdes Cubero-Rego
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - Susana A Castro-Chavira
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Juriquilla, Querétaro 76230, Mexico
| | - Thalía Fernández
- Laboratorio de Psicofisiología, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| |
Collapse
|
23
|
Bokobza C, Jacquens A, Guenoun D, Bianco B, Galland A, Pispisa M, Cruz A, Zinni M, Faivre V, Roumier A, Lebon S, Vitalis T, Csaba Z, Le Charpentier T, Schwendimann L, Young-Ten P, Degos V, Monteiro P, Dournaud P, Gressens P, Van Steenwinckel J. Targeting the brain 5-HT7 receptor to prevent hypomyelination in a rodent model of perinatal white matter injuries. J Neural Transm (Vienna) 2023; 130:281-297. [PMID: 36335540 PMCID: PMC10033587 DOI: 10.1007/s00702-022-02556-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Abstract
Approximately 15 million babies are born prematurely every year and many will face lifetime motor and/or cognitive deficits. Children born prematurely are at higher risk of developing perinatal brain lesions, especially white matter injuries (WMI). Evidence in humans and rodents demonstrates that systemic inflammation-induced neuroinflammation, including microglial and astrocyte reactivity, is the prominent processes of WMI associated with preterm birth. Thus, a new challenge in the field of perinatal brain injuries is to develop new neuroprotective strategies to target neuroinflammation to prevent WMI. Serotonin (5-HT) and its receptors play an important role in inflammation, and emerging evidence indicates that 5-HT may regulate brain inflammation by the modulation of microglial reactivity and astrocyte functions. The present study is based on a mouse model of WMI induced by intraperitoneal (i.p.) injections of IL-1β during the first 5 days of life. In this model, certain key lesions of preterm brain injuries can be summarized by (i) systemic inflammation, (ii) pro-inflammatory microglial and astrocyte activation, and (iii) inhibition of oligodendrocyte maturation, leading to hypomyelination. We demonstrate that Htr7 mRNA (coding for the HTR7/5-HT7 receptor) is significantly overexpressed in the anterior cortex of IL-1β-exposed animals, suggesting it as a potential therapeutic target. LP-211 is a specific high-affinity HTR7 agonist that crosses the blood-brain barrier (BBB). When co-injected with IL-1β, LP-211 treatment prevented glial reactivity, the down-regulation of myelin-associated proteins, and the apparition of anxiety-like phenotypes. Thus, HTR7 may represent an innovative therapeutic target to protect the developing brain from preterm brain injuries.
Collapse
Affiliation(s)
- Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France.
| | - Alice Jacquens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Anesthesia and Critical Care, APHP-Sorbonne University, Hôpital La Pitié- Salpêtrière, Paris, France
| | - David Guenoun
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Pharmacy, APHP, Hôpital Robert Debré, Université de Paris, Paris, France
| | - Blandine Bianco
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Anne Galland
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Maxime Pispisa
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Alexandra Cruz
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Manuela Zinni
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Valérie Faivre
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Anne Roumier
- Sorbonne Université, Inserm, UMR-S 1270, Paris, France
| | - Sophie Lebon
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Tania Vitalis
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Zsolt Csaba
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | | | | | | | - Vincent Degos
- Department of Anesthesia and Critical Care, APHP-Sorbonne University, Hôpital La Pitié- Salpêtrière, Paris, France
| | - Patricia Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Pascal Dournaud
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | | |
Collapse
|
24
|
Ruiz-González E, Benavente-Fernández I, Lubián-Gutiérrez M, Segado-Arenas A, Zafra-Rodríguez P, Méndez-Abad P, Lubián-López SP. Ultrasonographic evaluation of the early brain growth pattern in very low birth weight infants. Pediatr Res 2023:10.1038/s41390-022-02425-w. [PMID: 36624287 DOI: 10.1038/s41390-022-02425-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Preterm infants develop smaller brain volumes compared to term newborns. Our aim is to study early brain growth related to perinatal factors in very low birth weight infants (VLBWI). METHODS Manual segmentation of total brain volume (TBV) was performed in weekly 3D-ultrasonographies in our cohort of VLBWI. We studied the brain growth pattern related to term magnetic resonance image (term-MRI). RESULTS We found different brain growth trajectories, with smaller brain volumes and a decrease in brain growth rate in those VLBWI who would later have an abnormal term-MRI (mean TBV 190.68 vs. 213.9 cm3; P = 0.0001 and mean TBV growth rate 14.35 (±1.27) vs. 16.94 (±2.29) cm3/week; P = 0.0001). TBV in those with normal term-MRI was related to gestational age (GA), being small for gestational age (SGA), sex, and duration of parenteral nutrition (TPN) while in those with abnormal term-MRI findings it was related to GA, SGA, TPN, and comorbidities. We found a deceleration in brain growth rate in those with ≥3 comorbidities. CONCLUSIONS An altered brain growth pattern in VLBWI who subsequently present worst scores on term-MRI is related to GA, being SGA and comorbidities. Early ultrasonographic monitoring of TBV could be useful to detect deviated patterns of brain growth. IMPACT STATEMENT We describe the brain growth pattern in very low birth weight infants during their first postnatal weeks. Brain growth may be affected in the presence of certain perinatal factors and comorbidities, conditioning a deviation of the normal growth pattern. The serial ultrasound follow-up of these at-risk patients allows identifying these brain growth patterns early, which offers a window of opportunity for implementing earlier interventions.
Collapse
Affiliation(s)
- Estefanía Ruiz-González
- Division of Neonatology, Department of Paediatrics, Puerta del Mar University Hospital, Cádiz, Spain.,Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
| | - Isabel Benavente-Fernández
- Division of Neonatology, Department of Paediatrics, Puerta del Mar University Hospital, Cádiz, Spain. .,Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain. .,Area of Paediatrics, Department of Child and Mother Health and Radiology, Medical School, University of Cádiz, C/Doctor Marañon, 3, Cádiz, Spain.
| | - Manuel Lubián-Gutiérrez
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain.,Division of Neurology, Department of Paediatrics, Puerta del Mar University Hospital, Cádiz, Spain
| | - Antonio Segado-Arenas
- Division of Neonatology, Department of Paediatrics, Puerta del Mar University Hospital, Cádiz, Spain.,Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
| | - Pamela Zafra-Rodríguez
- Division of Neonatology, Department of Paediatrics, Puerta del Mar University Hospital, Cádiz, Spain.,Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
| | - Paula Méndez-Abad
- Division of Neonatology, Department of Paediatrics, Puerta del Mar University Hospital, Cádiz, Spain.,Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
| | - Simón P Lubián-López
- Division of Neonatology, Department of Paediatrics, Puerta del Mar University Hospital, Cádiz, Spain.,Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
| |
Collapse
|
25
|
Xie Y, Yang Y, Yuan T. Brain Damage in the Preterm Infant: Clinical Aspects and Recent Progress in the Prevention and Treatment. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:27-40. [PMID: 35209835 DOI: 10.2174/1871527321666220223092905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/16/2022] [Accepted: 01/16/2022] [Indexed: 12/16/2022]
Abstract
Although the prevalence of brain injury and related neurodevelopmental disabilities resulting from preterm birth are major public health concerns, there are no definite neuroprotective strategies to prevent or reduce brain injury. The pattern of brain injury seen in preterm infants has evolved into more subtle lesions that are still essential to diagnose regarding neurodevelopmental outcomes. There is no specific effective method for the treatment of premature infant brain injury, and the focus of clinical treatment is still on prevention. Prevention of this injury requires insight into the pathogenesis, but many gaps exist in our understanding of how neonatal treatment procedures and medications impact cerebral hemodynamics and preterm brain injury. Many studies provide evidence about the prevention of premature infant brain injury, which is related to some drugs (such as erythropoietin, melatonin, mesenchymal stem cells, etc.). However, there are still some controversies about the quality of research and the effectiveness of therapy. This review aims to recapitulate the results of preclinical studies and provide an update on the latest developments around etiological pathways, prevention, and treatment.
Collapse
Affiliation(s)
- Yixuan Xie
- Department of Neonatology, Children\'s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, P.R. China
| | - Yue Yang
- Department of Neonatology, Children\'s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, P.R. China
| | - Tianming Yuan
- Department of Neonatology, Children\'s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, P.R. China
| |
Collapse
|
26
|
Yuan M, Jin X, Qin F, Zhang X, Wang X, Yuan E, Shi Y, Xu F. The association of γδT lymphocytes with cystic leukomalacia in premature infants. Front Neurol 2022; 13:1043142. [PMID: 36530609 PMCID: PMC9755680 DOI: 10.3389/fneur.2022.1043142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/09/2022] [Indexed: 09/19/2023] Open
Abstract
Background Periventricular leukomalacia (PVL) is an essential cause of cerebral palsy in preterm infants, and cystic PVL (cPVL) is the most severe form of the disease. The pathogenesis of cPVL is complex, and immune imbalances and inflammatory responses may play an essential role in it. Objective This study aimed to investigate the correlation between peripheral blood lymphocyte subsets, especially γδT cells with the pathogenesis of cPVL in preterm infants. Methods Peripheral blood from preterm infants with GA < 32 weeks and BW < 1,500 g was used in this study and was collected at 34 weeks corrected gestational age and within 24 h after the diagnosis with cranial MRI or cranial ultrasound. The infants were divided into cPVL groups and control groups. Flow cytometry was used to detect peripheral blood γδT, CD3+, CD4+, CD8+, and the proportion of total lymphocytes. Multiplex cell assays were used to detect the concentration of extracellular serum cytokines IL-6, IL-2, IL-8, IL-17A, IL-10, IL-1RA, eotaxin (CCL11), MCP-1 (CCL2), CXCL1, G-CSF, and IFNγ. A follow-up visit was carried out when the patient was 3 years old. Results After correcting for confounding factors, the proportion of peripheral blood γδT in the cPVL group was significantly lower than that in the control group (β: 0.216; 95% CI: 0.058-0.800, P < 0.022). Peripheral blood γδT (AUC: 0.722, P=0.006) and multivariate binary regression model (AUC: 0.865, P < 0.000) have good diagnostic values for cPVL. Peripheral blood γδT has some predictive power for neurodevelopmental outcomes in preterm infants (AUC: 0.743, P = 0.002). Conclusion It seems that peripheral blood γδT cells are inversely correlated with cPVL, which is not only a risk factor for cPVL disease but also neurodevelopmental outcomes in preterm infants. However, the causality of cPVL and various lymphocytes is unclear and needs further study.
Collapse
Affiliation(s)
- Mengjie Yuan
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Xinyun Jin
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Fanyue Qin
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Enwu Yuan
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Shi
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Falin Xu
- Department of Neonatology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
- Advanced Medical Research Center of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
27
|
Hüls A, Wedderburn CJ, Groenewold NA, Gladish N, Jones MJ, Koen N, MacIsaac JL, Lin DTS, Ramadori KE, Epstein MP, Donald KA, Kobor MS, Zar HJ, Stein DJ. Newborn differential DNA methylation and subcortical brain volumes as early signs of severe neurodevelopmental delay in a South African Birth Cohort Study. World J Biol Psychiatry 2022; 23:601-612. [PMID: 34895032 PMCID: PMC9273810 DOI: 10.1080/15622975.2021.2016955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 01/25/2023]
Abstract
OBJECTIVES Early detection of neurodevelopmental delay is crucial for intervention and treatment strategies. We analysed associations between newborn DNA methylation (DNAm), neonatal magnetic resonance imaging (MRI) neuroimaging data, and neurodevelopment. METHODS Neurodevelopment was assessed in 161 children from the South African Drakenstein Child Health Study at 2 years of age using the Bayley Scales of Infant and Toddler Development III. We performed an epigenome-wide association study of neurodevelopmental delay using DNAm from cord blood. Subsequently, we analysed if associations between DNAm and neurodevelopmental delay were mediated by altered neonatal brain volumes (subset of 51 children). RESULTS Differential DNAm at SPTBN4 (cg26971411, Δbeta = -0.024, p-value = 3.28 × 10-08), and two intergenic regions (chromosome 11: cg00490349, Δbeta = -0.036, p-value = 3.02 × 10-08; chromosome 17: cg15660740, Δbeta = -0.078, p-value = 6.49 × 10-08) were significantly associated with severe neurodevelopmental delay. While these associations were not mediated by neonatal brain volume, neonatal caudate volumes were independently associated with neurodevelopmental delay, particularly in language (Δcaudate volume = 165.30 mm3, p = 0.0443) and motor (Δcaudate volume = 365.36 mm3, p-value = 0.0082) domains. CONCLUSIONS Differential DNAm from cord blood and increased neonatal caudate volumes were independently associated with severe neurodevelopmental delay at 2 years of age. These findings suggest that neurobiological signals for severe developmental delay may be detectable in very early life.
Collapse
Affiliation(s)
- Anke Hüls
- Department of Epidemiology and Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Catherine J Wedderburn
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Nynke A Groenewold
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Nicole Gladish
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, and Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Nastassja Koen
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| | - Julia L MacIsaac
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - David T S Lin
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Katia E Ramadori
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Michael P Epstein
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Kirsten A Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Michael S Kobor
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
28
|
Huang J, Zheng Z, Zhao X, Huang L, Wang L, Zhang X, Lin X. Short-term effects of fresh mother's own milk in very preterm infants. MATERNAL & CHILD NUTRITION 2022; 19:e13430. [PMID: 36098334 PMCID: PMC9749607 DOI: 10.1111/mcn.13430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 12/15/2022]
Abstract
Fresh mother's own milk (MOM) can protect preterm infants from many complications. Often MOM is pasteurized for safety, which can deactivate cellular and bioactive components with protective benefits. Questions remain regarding whether pasteurized MOM provides the same benefits as fresh MOM. The aim of this study was to evaluate the association and feasibility of feeding very preterm infants with fresh MOM. This prospective cohort study included 157 very preterm infants born before 32 weeks' gestational age and with a birthweight below 1500 g. Of these, 82 infants were included in the fresh MOM without any processing group and 75 infants were included in the pasteurized never-frozen MOM (PNFMOM) group. The mortality rate, survival rate without severe complication, incidence of complications, feeding indexes and growth velocities were compared to assess the association and feasibility of feeding fresh MOM. Compared with the PNFMOM group, the fresh MOM group had a higher survival rate without severe complications (p = 0.014) and a lower incidence of bronchopulmonary dysplasia (p = 0.010) after adjustment for confounders. The fresh MOM group regained birthweight earlier (p = 0.021), reached total enteral feeding earlier (p = 0.024), and received total parenteral nutrition for less time (p = 0.045). No adverse events associated with fresh MOM feeding were recorded. Feeding fresh MOM may reduce the incidence of complications in very premature infants. Fresh MOM was shown to be a feasible feeding strategy to improve preterm infants' outcomes.
Collapse
Affiliation(s)
- Jing Huang
- Department of Neonatology, Xiamen Maternal and Child Care Hospital, Women and Children's Hospital, School of MedicineXiamen UniversityXiamenChina
| | - Zhi Zheng
- Department of Neonatology, Xiamen Maternal and Child Care Hospital, Women and Children's Hospital, School of MedicineXiamen UniversityXiamenChina
| | - Xiao‐yan Zhao
- Department of Neonatology, Xiamen Maternal and Child Care Hospital, Women and Children's Hospital, School of MedicineXiamen UniversityXiamenChina
| | - Li‐han Huang
- Department of Neonatology, Xiamen Maternal and Child Care Hospital, Women and Children's Hospital, School of MedicineXiamen UniversityXiamenChina
| | - Lian Wang
- Department of Neonatology, Xiamen Maternal and Child Care Hospital, Women and Children's Hospital, School of MedicineXiamen UniversityXiamenChina
| | - Xiao‐lan Zhang
- Department of NeonatologyXiamen Humanity HospitalXiamenChina
| | - Xin‐zhu Lin
- Department of Neonatology, Xiamen Maternal and Child Care Hospital, Women and Children's Hospital, School of MedicineXiamen UniversityXiamenChina
| |
Collapse
|
29
|
Abstract
According to the World Health Organization (WHO), 15 million babies are born preterm each year. Preterm infants are those born at less than 37 weeks, while extremely and very preterm neonates include those born at 22 to less than 32 weeks gestational age. Infants that fail to make it to term are missing a key part in neurodevelopment, as weeks 24 to 40 are a critical period of brain development. Neonatal brain injury is a crucial predictor for mortality and morbidity in premature and low birth weight (<1500 g) infants. Although the complications associated with preterm birth continue to be the number one cause of death in children under 5, the survival rates are increasing (Volpe, 2019). Despite this, the incidence of comorbidities, such as learning disabilities and visual and hearing problems, is still high. The functional deficits seen in these infants can be contributed to the white matter abnormalities (WMA) that have been found in 50% to 80% of extremely and very preterm neonates. While numerous, the etiology of the neonatal brain injury is essential for determining the mortality and morbidities of the infant, as there is an increased risk for both intraventricular hemorrhage (IVH) and periventricular leukomalacia (PVL), which can be attributed to their lack of cerebrovascular autoregulation and hypoxic events. Neuroimaging plays a key role in detecting and assessing these neurologic injuries that preterm infants are at risk for. It is essential to diagnose these events early on to assess neurologic damage, minimize disease progression, and provide supportive care. Brain MRI and cranial ultrasound (CUS) are both extensively used neuroimaging techniques to assess WMA, and it has become ever more important to determine the best imaging techniques and modalities with the increasing survival rates and high incidence of comorbidities among these infants.
Collapse
Affiliation(s)
- Abigail Locke
- Radiology Research, Division of Neuroradiology, Penn State Health, Penn State College of Medicine, Mail Code H066 500 University Drive, Hershey, PA 17033, USA
| | - Sangam Kanekar
- Radiology Research, Division of Neuroradiology, Penn State Health, Penn State College of Medicine, Mail Code H066 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
30
|
Favrais G, Bokobza C, Saliba E, Chalon S, Gressens P. Alteration of the Oligodendrocyte Lineage Varies According to the Systemic Inflammatory Stimulus in Animal Models That Mimic the Encephalopathy of Prematurity. Front Physiol 2022; 13:881674. [PMID: 35928559 PMCID: PMC9343871 DOI: 10.3389/fphys.2022.881674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Preterm birth before the gestational age of 32 weeks is associated with the occurrence of specific white matter damage (WMD) that can compromise the neurological outcome. These white matter abnormalities are embedded in more global brain damage defining the encephalopathy of prematurity (EoP). A global reduction in white matter volume that corresponds to chronic diffuse WMD is the most frequent form in contemporary cohorts of very preterm infants. This WMD partly results from alterations of the oligodendrocyte (OL) lineage during the vulnerability window preceding the beginning of brain myelination. The occurrence of prenatal, perinatal and postnatal events in addition to preterm birth is related to the intensity of WMD. Systemic inflammation is widely recognised as a risk factor of WMD in humans and in animal models. This review reports the OL lineage alterations associated with the WMD observed in infants suffering from EoP and emphasizes the role of systemic inflammation in inducing these alterations. This issue is addressed through data on human tissue and imaging, and through neonatal animal models that use systemic inflammation to induce WMD. Interestingly, the OL lineage damage varies according to the inflammatory stimulus, i.e., the liposaccharide portion of the E.Coli membrane (LPS) or the proinflammatory cytokine Interleukin-1β (IL-1β). This discrepancy reveals multiple cellular pathways inducible by inflammation that result in EoP. Variable long-term consequences on the white matter morphology and functioning may be speculated upon according to the intensity of the inflammatory challenge. This hypothesis emerges from this review and requires further exploration.
Collapse
Affiliation(s)
- Geraldine Favrais
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
- Neonatology Unit, CHRU de Tours, Tours, France
- *Correspondence: Geraldine Favrais,
| | - Cindy Bokobza
- Inserm, NeuroDiderot, Université Paris Cité, Paris, France
| | - Elie Saliba
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Sylvie Chalon
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | | |
Collapse
|
31
|
Haffner DN, Sankovic A. A Neurologist's Guide to Neonatal Neurodevelopmental Assessments and Preterm Brain Injury. Semin Pediatr Neurol 2022; 42:100974. [PMID: 35868724 DOI: 10.1016/j.spen.2022.100974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 11/19/2022]
Abstract
Despite advances in medical care and improved survival of extremely preterm infants, rates of neurodevelopmental impairment remain high. Outcomes are significantly improved with early intervention, but infants must be appropriately identified to facilitate services. Neuroimaging provides important information regarding neurodevelopmental outcomes but prognosticating and communicating risk remains challenging. Standardized neonatal neurodevelopmental assessments provide supplemental information to aid in the identification of high-risk infants and counseling for their families.
Collapse
Affiliation(s)
- Darrah N Haffner
- Division of Pediatric Neurology, Nationwide Children's Hospital and the Ohio State University, 700 Children's Dr Columbus, 43205 OH, United States.
| | - Alexandra Sankovic
- Division of Pediatric Neurology, Nationwide Children's Hospital and the Ohio State University, 700 Children's Dr Columbus, 43205 OH, United States
| |
Collapse
|
32
|
van 't Westende C, Steggerda SJ, Jansen L, van den Berg-Huysmans AA, van de Pol LA, Wiggers-de Bruine FT, Stam CJ, Peeters-Scholte CMPCD. Combining advanced MRI and EEG techniques better explains long-term motor outcome after very preterm birth. Pediatr Res 2022; 91:1874-1881. [PMID: 34031571 DOI: 10.1038/s41390-021-01571-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/20/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Preterm born children are at high risk for adverse motor neurodevelopment. The aim of this study was to establish the relationship between motor outcome and advanced magnetic resonance imaging (MRI) and electroencephalography (EEG) measures. METHODS In a prospective cohort study of 64 very preterm born children, the motor outcome was assessed at 9.83 (SD 0.70) years. Volumetric MRI, diffusion tensor imaging (DTI), and EEG were acquired at 10.85 (SD 0.49) years. We investigated associations between motor outcome and brain volumes (white matter, deep gray matter, cerebellum, and ventricles), white matter integrity (fractional anisotropy and mean, axial and radial diffusivity), and brain activity (upper alpha (A2) functional connectivity and relative A2 power). The independence of associations with motor outcome was investigated with a final model. For each technique, the measure with the strongest association was selected to avoid multicollinearity. RESULTS Ventricular volume, radial diffusivity, mean diffusivity, relative A2 power, and A2 functional connectivity were significantly correlated to motor outcome. The final model showed that ventricular volume and relative A2 power were independently associated with motor outcome (B = -9.42 × 10-5, p = 0.027 and B = 28.9, p = 0.007, respectively). CONCLUSIONS This study suggests that a lasting interplay exists between brain structure and function that might underlie motor outcome at school age. IMPACT This is the first study that investigates the relationships between motor outcome and brain volumes, DTI, and brain function in preterm born children at school age. Ventricular volume and relative upper alpha power on EEG have an independent relation with motor outcome in preterm born children at school age. This suggests that there is a lasting interplay between structure and function that underlies adverse motor outcome.
Collapse
Affiliation(s)
- Charlotte van 't Westende
- Department of Child Neurology, Amsterdam University Medical Centers, AMC Site, Amsterdam, The Netherlands. .,Department of Neonatology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Sylke J Steggerda
- Department of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisette Jansen
- Department of Psychology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Laura A van de Pol
- Department of Child Neurology, Amsterdam University Medical Centers, AMC Site, Amsterdam, The Netherlands
| | | | - Cornelis J Stam
- Department of Clinical Neurophysiology, Amsterdam University Medical Centers, VUmc Site, Amsterdam, The Netherlands
| | | |
Collapse
|
33
|
D’Andrea CB, Kenley JK, Montez DF, Mirro AE, Miller RL, Earl EA, Koller JM, Sung S, Yacoub E, Elison JT, Fair DA, Dosenbach NU, Rogers CE, Smyser CD, Greene DJ. Real-time motion monitoring improves functional MRI data quality in infants. Dev Cogn Neurosci 2022; 55:101116. [PMID: 35636344 PMCID: PMC9157440 DOI: 10.1016/j.dcn.2022.101116] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/24/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Imaging the infant brain with MRI has improved our understanding of early neurodevelopment. However, head motion during MRI acquisition is detrimental to both functional and structural MRI scan quality. Though infants are typically scanned while asleep, they commonly exhibit motion during scanning causing data loss. Our group has shown that providing MRI technicians with real-time motion estimates via Framewise Integrated Real-Time MRI Monitoring (FIRMM) software helps obtain high-quality, low motion fMRI data. By estimating head motion in real time and displaying motion metrics to the MR technician during an fMRI scan, FIRMM can improve scanning efficiency. Here, we compared average framewise displacement (FD), a proxy for head motion, and the amount of usable fMRI data (FD ≤ 0.2 mm) in infants scanned with (n = 407) and without FIRMM (n = 295). Using a mixed-effects model, we found that the addition of FIRMM to current state-of-the-art infant scanning protocols significantly increased the amount of usable fMRI data acquired per infant, demonstrating its value for research and clinical infant neuroimaging. MRI studies of the infant brain are critical for studying early neurodevelopment. Head motion diminishes MRI data quality, which can adversely affect infant imaging. We show that real-time head motion monitoring improves fMRI scan quality in infants. Being able to monitor motion during fMRI acquisition improves scanning efficiency.
Collapse
|
34
|
Wang Y, Song J, Zhang X, Kang W, Li W, Yue Y, Zhang S, Xu F, Wang X, Zhu C. The Impact of Different Degrees of Intraventricular Hemorrhage on Mortality and Neurological Outcomes in Very Preterm Infants: A Prospective Cohort Study. Front Neurol 2022; 13:853417. [PMID: 35386416 PMCID: PMC8978798 DOI: 10.3389/fneur.2022.853417] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveIntraventricular hemorrhage (IVH) is a common complication in preterm infants and is related to neurodevelopmental outcomes. Infants with severe IVH are at higher risk of adverse neurological outcomes and death, but the effect of low-grade IVH remains controversial. The purpose of this study was to evaluate the impact of different degrees of IVH on mortality and neurodevelopmental outcomes in very preterm infants.MethodsPreterm infants with a gestational age of <30 weeks admitted to neonatal intensive care units were included. Cerebral ultrasound was examined repeatedly until discharge or death. All infants were followed up to 18–24 months of corrected age. The impact of different grades of IVH on death and neurodevelopmental disability was assessed by multiple logistic regression.ResultsA total of 1,079 preterm infants were included, and 380 (35.2%) infants had grade I-II IVH, 74 (6.9%) infants had grade III-IV IVH, and 625 (57.9%) infants did not have IVH. The mortality in the non-IVH, I-II IVH, and III-IV IVH groups was 20.1, 19.7, and 55.2%, respectively (p < 0.05), and the incidence of neurodevelopmental disabilities was 13.9, 16.1, and 43.3%, respectively (p < 0.05), at 18–24 months of corrected age. After adjusting for confounding factors, preterm infants with III-IV IVH had higher rates of cerebral palsy [26.7 vs. 2.4%, OR = 6.10, 95% CI (1.840–20.231), p = 0.003], disability [43.3 vs. 13.9%, OR = 2.49, 95% CI (1.059–5.873), p = 0.037], death [55.2 vs. 20.1%, OR = 3.84, 95% CI (2.090–7.067), p < 0.001], and disability + death [73.7 vs. 28.7%, OR = 4.77, 95% CI (2.518–9.021), p < 0.001] compared to those without IVH. However, the mortality and the incidence of neurodevelopmental disability in infants with I-II IVH were similar to those without IVH (p > 0.05).ConclusionsSevere IVH but not mild IVH increased the risk of mortality and neurodevelopmental disability in very preterm infants.
Collapse
Affiliation(s)
- Yong Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juan Song
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenqing Kang
- Department of Neonatology, Children's Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenhua Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuyang Yue
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Falin Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden
- *Correspondence: Changlian Zhu ;
| |
Collapse
|
35
|
Yuan S, Liu M, Kim S, Yang J, Barkovich AJ, Xu D, Kim H. Cyto/myeloarchitecture of cortical gray matter and superficial white matter in early neurodevelopment: multimodal MRI study in preterm neonates. Cereb Cortex 2022; 33:357-373. [PMID: 35235643 PMCID: PMC9837610 DOI: 10.1093/cercor/bhac071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 01/19/2023] Open
Abstract
The cerebral cortex undergoes rapid microstructural changes throughout the third trimester. Recently, there has been growing interest on imaging features that represent cyto/myeloarchitecture underlying intracortical myelination, cortical gray matter (GM), and its adjacent superficial whitematter (sWM). Using 92 magnetic resonance imaging scans from 78 preterm neonates, the current study used combined T1-weighted/T2-weighted (T1w/T2w) intensity ratio and diffusion tensor imaging (DTI) measurements, including fractional anisotropy (FA) and mean diffusivity (MD), to characterize the developing cyto/myeloarchitectural architecture. DTI metrics showed a linear trajectory: FA decreased in GM but increased in sWM with time; and MD decreased in both GM and sWM. Conversely, T1w/T2w measurements showed a distinctive parabolic trajectory, revealing additional cyto/myeloarchitectural signature inferred. Furthermore, the spatiotemporal courses were regionally heterogeneous: central, ventral, and temporal regions of GM and sWM exhibited faster T1w/T2w changes; anterior sWM areas exhibited faster FA increases; and central and cingulate areas in GM and sWM exhibited faster MD decreases. These results may explain cyto/myeloarchitectural processes, including dendritic arborization, synaptogenesis, glial proliferation, and radial glial cell organization and apoptosis. Finally, T1w/T2w values were significantly associated with 1-year language and cognitive outcome scores, while MD significantly decreased with intraventricular hemorrhage.
Collapse
Affiliation(s)
| | | | | | - Jingda Yang
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Anthony James Barkovich
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Duan Xu
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hosung Kim
- Corresponding author: 2025 Zonal Ave, Los Angeles, CA 90033, USA.
| |
Collapse
|
36
|
Nosko D, Strindberg M, Svoboda J, Kvanta H, Broström L, Padilla N, Mårtensson G, Örtqvist M, Moreira NC, Ådén U. Discrete white matter abnormalities at age 8-11 years in children born extremely preterm are not associated with adverse cognitive or motor outcomes. Acta Paediatr 2022; 111:566-575. [PMID: 34665877 DOI: 10.1111/apa.16158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 11/27/2022]
Abstract
AIM Little is known about the prevalence of discrete white matter abnormalities (WMA) beyond the first years in children born extremely preterm (EPT) and the relation to neurodevelopmental outcomes. Our aim was to investigate the prevalence of discrete WMA in children born EPT and the relationship to neonatal white matter injuries (WMI), white matter (WM) volume, WM diffusivity and neurodevelopment. METHODS The study was a part of a longitudinal follow-up study of EPT neonates. All children were scanned at Karolinska University hospital 2004-2007 (neonates) and 2014-2015 (children at 8-11 years). WMA was qualitatively assessed by visual inspection. Developmental assessment was conducted at 12 years. RESULTS In total, 112 children (median age 10.3 years, 56 girls) underwent MRI of the brain (68 EPT, 45 controls). In the EPT group, a subset had MRI around term equivalent age (n = 61). In the EPT group, the prevalence of discrete WMA at 8-11 years was 52%. There was a positive association between WMI at TEA and 8-11 years. There was no association between WMI and WM volumes or diffusivity at 8-11 years. Discrete WMA was not related to neurodevelopmental outcomes. CONCLUSION Discrete WMA was prevalent in children born EPT at 8-11 years but were not related to neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Daniela Nosko
- Department of Paediatrics Örebro University Hospital Örebro Sweden
| | - Marika Strindberg
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Jan Svoboda
- Department of Neuroradiology Karolinska University Hospital Stockholm Sweden
| | - Hedvig Kvanta
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Lina Broström
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Nelly Padilla
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Gustaf Mårtensson
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Maria Örtqvist
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Nuno Canto Moreira
- Department of Neuroradiology Karolinska University Hospital Stockholm Sweden
| | - Ulrika Ådén
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
- Department of Neonatal Medicine Karolinska University Hospital Stockholm Sweden
| |
Collapse
|
37
|
Julien-Marsollier F, Cholet C, Coeffic A, Dupont T, Gauthier T, Loiselle M, Brouns K, Bonnard A, Biran V, Brasher C, Dahmani S. Intraoperative cerebral oxygen saturation and neurological outcomes following surgical management of necrotizing enterocolitis: Predictive factors of neurological complications following neonatal necrotizing enterocolitis: Predictive factors of neurological complications following neonatal necrotizing enterocolitis. Paediatr Anaesth 2022; 32:421-428. [PMID: 34984774 DOI: 10.1111/pan.14392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/19/2021] [Accepted: 12/28/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND The goal of the present study was to investigate intraoperative factors associated with major neurological complications at 1 year following surgery for necrotizing enterocolitis. MATERIAL AND METHODS The study consisted of a retrospective review of medical charts of patients operated for over one calendar year in one institution. Data collected included demographic data, cardiac resuscitation at birth, Bell classification, antibiotics usage, time of day of surgery, surgical technique, surgical duration, type of ventilation, intraoperative vasoactive agents, and albumin use, nadir cerebral saturation, the decrease in cerebral saturation from baseline, the time period when cerebral saturation was at least 20% below baseline, and the mean arterial pressure at nadir cerebral saturation. Reported follow-up complications were assessed during formal neonatologist consultation and additional imaging exploration as needed. Analyses included descriptive statistics, and univariable and multivariable statistics. RESULTS The study included 32 patients with no prior clinical neurological complications, of which 25 had normal cerebral imaging. Severe neurological complications occurred in nine patients at 1 year: Intraventricular hemorrhage (N = 2) and Periventricular leukomalacia (N = 7). However, preoperative cerebral imaging was lacking in seven patients. Consequently, the observed neurological complications at 1 year might be present before the surgery. Multivariable analysis found the decrease in cerebral saturation ≥36% from baseline as the only factor associated with the occurrence of those complications. CONCLUSION Intraoperative decrease of cerebral oxygen saturation below ≥36% from baseline is associated with severe neurological complications in neonates undergoing surgery for necrotizing enterocolitis.
Collapse
Affiliation(s)
- Florence Julien-Marsollier
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| | - Clementine Cholet
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| | - Adrien Coeffic
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| | - Thibault Dupont
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| | - Thibault Gauthier
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| | - Maud Loiselle
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| | - Kelly Brouns
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| | - Arnaud Bonnard
- Department of general and urological surgery, Robert Debré University Hospital, Paris, France
| | - Valerie Biran
- Université de Paris, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France.,Department of Neonatology, Robert Debré University Hospital, Paris, France
| | - Christopher Brasher
- Department of Anesthesia & Pain Management, Royal Children's Hospital, Melbourne, Australia.,Anesthesia and Pain Management Research Group, Murdoch Children's Research Institute, Melbourne, Australia.,Centre for Integrated Critical Care, University of Melbourne, Australia
| | - Souhayl Dahmani
- Université de Paris, Paris, France.,Department of Anesthesia and Intensive care, Robert Debré University Hospital, Paris, France.,University Hospital Federation I2-D2, INSERM U1141, Robert Debré University Hospital, Paris, France
| |
Collapse
|
38
|
Savchenko O, Pavlinova E. Long-term neurological outcomes in premature children with excitotoxic brain damage. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:37-41. [DOI: 10.17116/jnevro202212209237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
39
|
Harmony T, Gutiérrez-Hernández CC, Carlier M, Hinojosa-Rodríguez M, Carrillo C. Early detection and treatment of attention deficits in preterm and at term infants with risk factors for brain damage. Int J Psychophysiol 2021; 172:17-23. [PMID: 34921894 DOI: 10.1016/j.ijpsycho.2021.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/25/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022]
Abstract
Cognitive deficits in infants born preterm and infants at term with risk factors for brain damage are a common outcome. Attention deficits in preterm infants are related to the development of attention-deficit/hyperactivity disorder (ADHD), and therefore, there is a need for earlier evaluations and treatment procedures that are implemented before the presence of signs of ADHD. METHODS We studied preterm (74%) and term infants with the Infant Scale of Selective Attention (ISSA, Escala de Evaluación de la Atención Selectiva (EEAS), in Spanish). This scale evaluates both visual- and auditory-orienting attention. Two groups participated, one with attention deficits (n = 26) and another with regular performance (n = 36). An early attention-stimulation program (EASP) was implemented in the infant group with attention deficits from three to eight months of age. All infants underwent magnetic resonance imaging (MRI), and visual and auditory evoked responses were assessed. RESULTS All infants had prenatal and perinatal risk factors for brain damage and abnormal MRI findings, and the majority had abnormalities compatible with white matter injury. However, there were four infants with porencephalic cysts; 3 of them were in the treated group. At the beginning of the treatment, ISSA values showed differences between groups. These differences persisted for five months in the visual test and up to the sixth month in the auditory evaluation. Afterward, there were no significant differences, indicating that infants with attention deficits had satisfactorily responded to the treatment. CONCLUSIONS The ISSA is helpful for the early evaluation of visual and auditory attention. Infants with attention deficits react well enough after six months of EASP.
Collapse
Affiliation(s)
- Thalía Harmony
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva del Instituto de Neurobiología de la Universidad Nacional Autónoma de México Campus Juriquilla, Mexico.
| | - Claudia Calipso Gutiérrez-Hernández
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva del Instituto de Neurobiología de la Universidad Nacional Autónoma de México Campus Juriquilla, Mexico
| | - Mónica Carlier
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva del Instituto de Neurobiología de la Universidad Nacional Autónoma de México Campus Juriquilla, Mexico
| | - Manuel Hinojosa-Rodríguez
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva del Instituto de Neurobiología de la Universidad Nacional Autónoma de México Campus Juriquilla, Mexico
| | - Cristina Carrillo
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva del Instituto de Neurobiología de la Universidad Nacional Autónoma de México Campus Juriquilla, Mexico
| |
Collapse
|
40
|
Relationship between the Quantitative Indicators of Cranial MRI and the Early Neurodevelopment of Preterm Infants. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6486452. [PMID: 34840597 PMCID: PMC8626187 DOI: 10.1155/2021/6486452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/07/2021] [Indexed: 11/17/2022]
Abstract
Aim To explore the relationship between the quantitative indicators (biparietal width, interhemispheric distance) of the cranial MRI for preterm infants at 37 weeks of postmenstrual age (PMA) and neurodevelopment at 6 months of corrected age. Methods A total of 113 preterm infants (gestational age < 37 weeks) delivered in the Obstetrics Department of the First People's Hospital of Lianyungang from September 2018 to February 2020 and directly transferred to the Neonatology Department for treatment were enrolled in this study. Based on their development quotient (DQ), the patients were divided into the normal (DQ ≥ 85, n = 76) group and the abnormal (DQ < 85, n = 37) group. Routine cranial MRI (cMRI) was performed at 37 weeks of PMA to measure the biparietal width (BPW) and interhemispheric distance (IHD). At the corrected age of 6 months, Development Screening Test (for children under six) was used to assess the participants' neurodevelopment. Results Univariate analysis showed statistically significant differences in BPW, IHD, and the incidence of bronchopulmonary dysplasia between the normal and the abnormal groups (P < 0.05), while no statistically significant differences were found in maternal complications and other clinical conditions between the two groups (P > 0.05). Binary logistic regression analysis demonstrated statistically significant differences in IHD and BPW between the normal and the abnormal groups (95% CI: 1.629-12.651 and 0.570-0.805, respectively; P = 0.004 and P < 0.001, respectively), while no significant differences were found in the incidence of bronchopulmonary dysplasia between the two groups (95% CI: 0.669-77.227, P = 0.104). Receiver operating characteristic curve revealed that the area under the curve of BPW, IHD, and the joint predictor (BPW + IHD) were 0.867, 0.805, and 0.881, respectively (95% CI: 0.800-0.933, 0.710-0.900, and 0.819-0.943, respectively; all P values < 0.001). Conclusion BPW and IHD, the two quantitative indicators acquired by cMRI, could predict the neurodevelopmental outcome of preterm infants at the corrected age of 6 months. The combination of the two indicators showed an even higher predictive value.
Collapse
|
41
|
Timmerman BM, Mooney-Leber SM, Brummelte S. The effects of neonatal procedural pain and maternal isolation on hippocampal cell proliferation and reelin concentration in neonatal and adult male and female rats. Dev Psychobiol 2021; 63:e22212. [PMID: 34813104 DOI: 10.1002/dev.22212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 11/11/2022]
Abstract
Preterm births accounted for over 10% of all U.S. live births in 2019 and the rate is rising. Neonatal stressors, especially procedural pain, experienced by preterm infants in the neonatal intensive care unit (NICU) have been associated with neurodevelopmental impairments. Parental care can alleviate stress during stressful or painful procedures; however, infants in the NICU often receive reduced parental care compared with their peers. Animal studies suggest that decreased maternal care similarly impairs neurodevelopment but also influences the effects of neonatal pain. It is important to mimic both stressors in animal models of neonatal stress exposure. In this study, researchers investigated the individual and combined impact of neonatal pain and maternal isolation on reelin protein levels and cellular proliferation in the hippocampal dentate gyrus of 8 days old and adult rats. Exposure to either stressor individually, but not both, increased reelin levels in the dentate gyrus of adult females without significantly altering reelin levels in adult males. However, cell proliferation levels at either age were unaffected by the early-life stressors. These results suggest that each early-life stressor has a unique effect on markers of brain development and more research is needed to further investigate their distinct influences.
Collapse
Affiliation(s)
- Brian M Timmerman
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
| | - Sean M Mooney-Leber
- Department of Psychology, University of Wisconsin-Stevens Points, Stevens Point, Wisconsin, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, Michigan, USA.,Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
42
|
Abstract
While medical advancements have led to improved survival of extremely premature infants, children remain at risk for brain injury and neurodevelopmental impairment. Brain imaging can offer insight into an infant's acute and long-term outcome; however, counseling parents about the results and implications of brain imaging remains challenging. The purpose of this article is to review the current literature and describe the challenges associated with counseling families of premature infants on neuroimaging findings. We propose a framework to guide clinicians in counseling parents about brain imaging results, informed by best practices in other disciplines: (FIGURE): 1) Formulate a plan 2) Identify parental needs and values 3) Give information 4) Acknowledge Uncertainty 5) Recognize and Respond to emotions 6) Discuss Expectations and Establish follow-up.
Collapse
Affiliation(s)
- Sarah M Bernstein
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | | | - Monica E Lemmon
- Departments of Pediatrics and Population Health Sciences, Duke University Medical Center, Duke-Margolis Center for Health Policy, DUMC 3936, Durham, NC 27710, United States.
| |
Collapse
|
43
|
Williamson M, Poorun R, Hartley C. Apnoea of Prematurity and Neurodevelopmental Outcomes: Current Understanding and Future Prospects for Research. Front Pediatr 2021; 9:755677. [PMID: 34760852 PMCID: PMC8573333 DOI: 10.3389/fped.2021.755677] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Infants who are born prematurely are at significant risk of apnoea. In addition to the short-term consequences such as hypoxia, apnoea of prematurity has been associated with long-term morbidity, including poor neurodevelopmental outcomes. Clinical trials have illustrated the importance of methylxanthine drugs, in particular caffeine, in reducing the risk of long term adverse neurodevelopmental outcomes. However, the extent to which apnoea is causative of this secondary neurodevelopmental delay or is just associated in a background of other sequelae of prematurity remains unclear. In this review, we first discuss the pathophysiology of apnoea of prematurity, previous studies investigating the relationship between apnoea and neurodevelopmental delay, and treatment of apnoea with caffeine therapy. We propose a need for better methods of measuring apnoea, along with improved understanding of the neonatal brain's response to consequent hypoxia. Only then can we start to disentangle the effects of apnoea on neurodevelopment in preterm infants. Moreover, by better identifying those infants who are at risk of apnoea, and neurodevelopmental delay, we can work toward a risk stratification system for these infants that is clinically actionable, for example, with doses of caffeine tailored to the individual. Optimising treatment of apnoea for individual infants will improve neonatal care and long-term outcomes for this population.
Collapse
Affiliation(s)
- Max Williamson
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Ravi Poorun
- Department of Paediatrics, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Caroline Hartley
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
44
|
Predictive value of brain MRI at term-equivalent age in extremely preterm children on neurodevelopmental outcome at school-age. Brain Imaging Behav 2021; 16:878-887. [PMID: 34661873 DOI: 10.1007/s11682-021-00559-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
This study's objective was to correlate the abnormalities in brain MRIs performed at corrected-term age for minor or moderate neurocognitive disorders in children school-age born extremely premature (EPT) and without serious sequelae such as autism, cerebral palsy, mental impairment. Data were issued from a cross-sectional multicenter study (GP-Qol study, number NCT01675726). Clinical examination and psychometric assessments were performed when the children were between 7 and 10 years old during a day-long evaluation. Term-equivalent age brain MRIs on EPT were analyzed with a standardized scoring system. There were 114 children included in the study. The mean age at the time of evaluation, was 8.47 years old (± 0.70). 59% of children with at least one cognitive impairment and 53% who had a dysexecutive disorder. Only ten EPT (8.7%) presented moderate to severe white and grey matter abnormalities. These moderate to severe grey matter abnormalities were associated with at least two abnormal executive functions [OR 3.08 (95% CI 1.04-8.79), p = 0.04] and language delay [OR 3.25 (95% CI 1.03-9.80), p = 0.04]. These results remained significant in the multivariate analysis. Moderate to severe ventricular dilatation abnormalities (15%, n = 17) were associated with ideomotor dyspraxia [OR 7.49 (95% CI 1.48-35.95), p = 0.02] and remained significant in multivariate analysis [OR 11.2 (95% CI 1.45-131.4), p = 0.02]. Biparietal corrected diameters were moderate abnormal in 20% of cases (n = 23) and were associated to visuo spatial integration delay [OR 4.13 (95% CI 1.23-13.63), p = 0.02]. Cerebral MRI at term-equivalent age with scoring system analysis can provide information on long-term neuropsychological outcomes at school-age in EPTs children having no severe disability.
Collapse
|
45
|
Metallinou D, Karampas G, Lazarou E, Iacovidou N, Pervanidou P, Lykeridou K, Mastorakos G, Rizos D. Serum Activin A as Brain Injury Biomarker in the First Three Days of Life. A Prospective Case-Control Longitudinal Study in Human Premature Neonates. Brain Sci 2021; 11:brainsci11091243. [PMID: 34573263 PMCID: PMC8468004 DOI: 10.3390/brainsci11091243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
Disruption of normal intrauterine brain development is a significant consequence of premature birth and may lead to serious complications, such as neonatal brain injury (NBI). This prospective case-control longitudinal study aimed at determining the levels and prognostic value of serum activin A during the first three days of life in human premature neonates which later developed NBI. It was conducted in a single tertiary hospital and eligible participants were live-born premature (<34 weeks) neonates. Each case (n = 29) developed NBI in the form of an intraventricular haemorrhage, or periventricular leukomalacia, and was matched according to birth weight and gestational age to one neonate with normal head ultrasound scans. Serum activin A levels in both groups showed a stable concentration during the first three days of life as no difference was observed within the two groups from the first to the third day. Neonates diagnosed with NBI had significantly higher activin A levels during the first two days of life compared to control neonates and its levels correlated to the severity of NBI during the second and third day of life. Although serum activin A on the second day was the best predictor for neonates at risk to develop NBI, the overall predictive value was marginally fair (area under the ROC-curve 69.2%). Activin A, in combination with other biomarkers, may provide the first clinically useful panel for the early detection of premature neonates at high risk of NBI.
Collapse
Affiliation(s)
- Dimitra Metallinou
- Department of Midwifery, University of West Attica, Ag. Spyridonos Street, 12243 Egaleo, Greece;
- Correspondence:
| | - Grigorios Karampas
- 2nd Department of Obstetrics and Gynecology, Aretaieio University Hospital, 46 Vasilissis Sofias Avenue, 11528 Athens, Greece;
| | - Eleftheria Lazarou
- Department of Obstetrics, Iasis Private Hospital Paphos, 8 Voriou Ipirou Street, 8036 Paphos, Cyprus;
| | - Nikoletta Iacovidou
- Neonatal Department, Aretaieio University Hospital, 46 Vasilissis Sofias Avenue, 11528 Athens, Greece;
| | - Panagiota Pervanidou
- Unit of Developmental and Behavioral Pediatrics, 1st Department of Pediatrics, National and Kapodistrian University of Athens, 5 Mikras Asias Street, 11527 Athens, Greece;
| | - Katerina Lykeridou
- Department of Midwifery, University of West Attica, Ag. Spyridonos Street, 12243 Egaleo, Greece;
| | - George Mastorakos
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieio University Hospital, 46 Vasilissis Sofias Avenue, 11528 Athens, Greece;
| | - Demetrios Rizos
- Hormone Laboratory, Aretaieio University Hospital, 46 Vasilissis Sofias Avenue, 11528 Athens, Greece;
| |
Collapse
|
46
|
Li L, Wang L, Niu C, Liu C, Lv T, Ji F, Yu L, Yan W, Dou YL, Wang Y, Cao Y, Huang G, Hu X. Early skin contact combined with mother's breastfeeding to shorten the process of premature infants ≤ 30 weeks of gestation to achieve full oral feeding: the study protocol of a randomized controlled trial. Trials 2021; 22:637. [PMID: 34535164 PMCID: PMC8447630 DOI: 10.1186/s13063-021-05605-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Most hospitalized preterm infants experience difficulties in transitioning from tube feeding to full oral feeding. Interventions to promote full oral feeding in preterm infants in the neonatal intensive care unit (NICU) are limited to pacifier use or bottle-feeding exercises. Skin contact has been shown to be beneficial to start and maintain lactation and provide preterm infants with the opportunity to suck on the mother's breast, which may promote further development of the preterm infant's suckling patterns. The objective of this study is to compare and evaluate the effects of skin contact combined with breastfeeding (suck on the mother's empty breast) as compared to the routine pacifier suckling training model in achieving full oral feeding for infants whose gestational age are ≤ 30 weeks. METHODS This is a single-center, randomized controlled clinical trial conducted in the NICU and designed according to the SPIRIT Statement. The subjects included in the study are premature infants born between April 2020 and July 2021 with a gestational age of ≤30 weeks, birth weight of <1500 g, admission age of <72 h, and absence of congenital malformations. Those with oxygenation indices of >40 and those born to mothers with poor verbal communication skills will be excluded. A sample of 148 infants is needed. The infants will be randomized to the intervention (skin contact combined with mother's breastfeeding model) or control group (routine pacifier sucking training model). The primary outcome is the time required to achieve full oral feeding. The secondary outcomes are the breastfeeding abilities of preterm infants as assessed by the Preterm Infant Breastfeeding Behavior Scale (PIBBS), breastfeeding rates at 3 and 6 months corrected gestational age, complication rates, duration of oxygen requirement, days of hospital stay, and satisfaction of parents. DISCUSSION This paper describes the first single-center, open-label, randomized clinical trial on this topic and will provide crucial information to support the implementation of skin contact combined with the breastfeeding model in the NICU setting. TRIAL REGISTRATION ClinicalTrials.gov NCT04283682. Registered on 8 February 2020.
Collapse
Affiliation(s)
- Liling Li
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Li Wang
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Conway Niu
- King Edward Memorial Hospital, Western Australia, Subiaco, Australia
| | - Chan Liu
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Tianchan Lv
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Futing Ji
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Ling Yu
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Weili Yan
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Ya Lan Dou
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Yin Wang
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Yun Cao
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Guoying Huang
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Xiaojing Hu
- Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China.
| |
Collapse
|
47
|
Ottolini KM, Andescavage N, Limperopoulos C. Lipid Intake and Neurodevelopment in Preterm Infants. Neoreviews 2021; 22:e370-e381. [PMID: 34074642 DOI: 10.1542/neo.22-6-e370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Preterm infants are born before the critical period of lipid accretion and brain development that occurs during the third trimester of pregnancy. Dietary lipids serve as an important source of energy and are involved in complex processes that are essential for normal central nervous system development. In addition to traditional neurodevelopmental testing, novel quantitative magnetic resonance imaging (MRI) techniques are now available to evaluate the impact of nutritional interventions on early preterm brain development. Trials of long-chain polyunsaturated fatty acid supplementation have yielded inconsistent effects on neurodevelopmental outcomes and quantitative MRI findings. Recent studies using quantitative MRI suggest a positive impact of early lipid intake on brain volumes and white matter microstructural organization by term-equivalent age.
Collapse
Affiliation(s)
- Katherine M Ottolini
- Department of Pediatrics, Division of Neonatology, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Nickie Andescavage
- Department of Neonatology and.,Developing Brain Research Laboratory, Children's National Hospital, Washington, DC
| | - Catherine Limperopoulos
- Developing Brain Research Laboratory, Children's National Hospital, Washington, DC.,Departments of Pediatrics and Radiology, George Washington University School of Medicine, Washington, DC
| |
Collapse
|
48
|
Tataranno ML, Vijlbrief DC, Dudink J, Benders MJNL. Precision Medicine in Neonates: A Tailored Approach to Neonatal Brain Injury. Front Pediatr 2021; 9:634092. [PMID: 34095022 PMCID: PMC8171663 DOI: 10.3389/fped.2021.634092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/14/2021] [Indexed: 11/27/2022] Open
Abstract
Despite advances in neonatal care to prevent neonatal brain injury and neurodevelopmental impairment, predicting long-term outcome in neonates at risk for brain injury remains difficult. Early prognosis is currently based on cranial ultrasound (CUS), MRI, EEG, NIRS, and/or general movements assessed at specific ages, and predicting outcome in an individual (precision medicine) is not yet possible. New algorithms based on large databases and machine learning applied to clinical, neuromonitoring, and neuroimaging data and genetic analysis and assays measuring multiple biomarkers (omics) can fulfill the needs of modern neonatology. A synergy of all these techniques and the use of automatic quantitative analysis might give clinicians the possibility to provide patient-targeted decision-making for individualized diagnosis, therapy, and outcome prediction. This review will first focus on common neonatal neurological diseases, associated risk factors, and most common treatments. After that, we will discuss how precision medicine and machine learning (ML) approaches could change the future of prediction and prognosis in this field.
Collapse
Affiliation(s)
| | | | | | - Manon J. N. L. Benders
- Department of Neonatology, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
49
|
Epstein S, Bauer S, Levkovitz Stern O, Litmanovitz I, Elefant C, Yakobson D, Arnon S. Preterm infants with severe brain injury demonstrate unstable physiological responses during maternal singing with music therapy: a randomized controlled study. Eur J Pediatr 2021; 180:1403-1412. [PMID: 33244709 DOI: 10.1007/s00431-020-03890-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022]
Abstract
Preterm infants with severe brain injury are at high risk for poor outcomes and, therefore, may benefit from developmental care modalities such as music therapy (MT). In this prospective, randomized intervention, preterm infants with severe brain injury (grade 3 or 4 intraventricular hemorrhage or periventricular leukomalacia) who underwent skin-to-skin contact (SSC) with or without maternal singing during MT were evaluated for physiological responses, including autonomic nervous system stability (low frequency (LF)/high frequency (HF) power), heart rate, respiratory rate, oxygen saturation, and behavioral state. Maternal anxiety state and physiological data were also evaluated. A total of 35 preterm infants with severe brain injuries were included in the study analysis. Higher mean ± standard deviation (SD) LF/HF ratio (1.8 ± 0.7 vs. 1.1 ± 0.25, p = 0.01), higher mean ± SD heart rate (145 ± 15 vs. 132 ± 12 beats per minute, p = 0.04), higher median (interquartile range) infant behavioral state (NIDCAP manual for naturalistic observation and the Brazelton Neonatal Behavioral Assessment) score (3 (2-5) vs. 1 (1-3), p = 0.03), and higher mean ± SD maternal anxiety (state-trait anxiety inventory) score (39.1 ± 10.4 vs. 31.5 ± 7.3, p = 0.04) were documented in SSC combined with maternal singing during MT, as compared to SSC alone.Conclusion: Maternal singing during MT for preterm infants with severe brain injury induces physiological and behavioral instability and increases maternal anxiety during NICU hospitalization. A unique MT intervention should be designed for preterm infants with severe brain injury and their mothers. What is Known: • Preterm infants with severe brain injury are at high risk for poor outcomes. • Music therapy benefits brain development of preterm infants without severe brain injury, however it is unknown whether maternal singing during music therapy for preterm infants with severe brain injury is beneficial. What is New: • Maternal singing during music therapy for preterm infants with severe brain injury induces physiological and behavioral instability and increases maternal anxiety during NICU hospitalization. • A unique music therapy intervention should be designed for preterm infants with severe brain injury and their mothers.
Collapse
Affiliation(s)
- Shulamit Epstein
- School of Creative Arts Therapies, University of Haifa, Haifa, Israel
| | - Sofia Bauer
- Department of Neonatology, Meir Medical Center, 59 Tchernichovsky St., 44281, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orly Levkovitz Stern
- Department of Neonatology, Meir Medical Center, 59 Tchernichovsky St., 44281, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ita Litmanovitz
- Department of Neonatology, Meir Medical Center, 59 Tchernichovsky St., 44281, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Cochavit Elefant
- School of Creative Arts Therapies, University of Haifa, Haifa, Israel
| | - Dana Yakobson
- Department of Neonatology, Meir Medical Center, 59 Tchernichovsky St., 44281, Kfar Saba, Israel.,Doctoral program in Music Therapy, Aalborg University, Aalborg, Denmark
| | - Shmuel Arnon
- Department of Neonatology, Meir Medical Center, 59 Tchernichovsky St., 44281, Kfar Saba, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
50
|
Story L, Davidson A, Patkee P, Fleiss B, Kyriakopoulou V, Colford K, Sankaran S, Seed P, Jones A, Hutter J, Shennan A, Rutherford M. Brain volumetry in fetuses that deliver very preterm: An MRI pilot study. NEUROIMAGE-CLINICAL 2021; 30:102650. [PMID: 33838546 PMCID: PMC8045030 DOI: 10.1016/j.nicl.2021.102650] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/10/2021] [Accepted: 03/26/2021] [Indexed: 11/17/2022]
Abstract
Fetuses that subsequently deliver very preterm have a reduction in cortical and extra cerebrospinal fluid volumes. If such alterations commence antenatally this suggests a role for earlier administration of neuroprotective agents.
Background Infants born preterm are at increased risk of neurological complications resulting in significant morbidity and mortality. The exact mechanism and the impact of antenatal factors has not been fully elucidated, although antenatal infection/inflammation has been implicated in both the aetiology of preterm birth and subsequent neurological sequelae. It is therefore hypothesized that processes driving preterm birth are affecting brain development in utero. This study aims to compare MRI derived regional brain volumes in fetuses that deliver < 32 weeks with fetuses that subsequently deliver at term. Methods Women at high risk of preterm birth, with gestation 19.4–32 weeks were recruited prospectively. A control group was obtained from existing study datasets. Fetal MRI was performed on a 1.5 T or 3 T MRI scanner: T2-weighted images were obtained of the fetal brain. 3D brain volumetric datsets were produced using slice to volume reconstruction and regional segmentations were produced using multi-atlas approaches for supratentorial brain tissue, lateral ventricles, cerebellum cerebral cortex and extra-cerebrospinal fluid (eCSF). Statistical comparison of control and high-risk for preterm delivery fetuses was performed by creating normal ranges for each parameter from the control datasets and then calculating gestation adjusted z scores. Groups were compared using t-tests. Results Fetal image datasets from 24 pregnancies with delivery < 32 weeks and 87 control pregnancies that delivered > 37 weeks were included. Median gestation at MRI of the preterm group was 26.8 weeks (range 19.4–31.4) and control group 26.2 weeks (range 21.7–31.9). No difference was found in supra-tentorial brain volume, ventricular volume or cerebellar volume but the eCSF and cerebral cortex volumes were smaller in fetuses that delivered preterm (p < 0.001 in both cases). Conclusion Fetuses that deliver preterm have a reduction in cortical and eCSF volumes. This is a novel finding and needs further investigation. If alterations in brain development are commencing antenatally in fetuses that subsequently deliver preterm, this may present a window for in utero therapy in the future.
Collapse
Affiliation(s)
- Lisa Story
- Department of Women and Children's Health, King's College London, UK.
| | - Alice Davidson
- Centre for the Developing Brain, King's College London, London, UK
| | - Prachi Patkee
- Centre for the Developing Brain, King's College London, London, UK
| | - Bobbi Fleiss
- Centre for the Developing Brain, King's College London, London, UK; School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, VIC, Australia; Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | | | - Kathleen Colford
- Centre for the Developing Brain, King's College London, London, UK; Centre for Medical Engineering, King's College London, London, UK
| | | | - Paul Seed
- Department of Women and Children's Health, King's College London, UK
| | - Alice Jones
- Centre for the Developing Brain, King's College London, London, UK; Queen Mary University Medical School, UK
| | - Jana Hutter
- Centre for the Developing Brain, King's College London, London, UK; School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, VIC, Australia
| | - Andrew Shennan
- Department of Women and Children's Health, King's College London, UK
| | - Mary Rutherford
- Centre for the Developing Brain, King's College London, London, UK
| |
Collapse
|