1
|
Meijer RF, Wang X, van Ooijen IM, van der Velde B, Dudink J, Benders MJNL, Tataranno ML. The relationship between early life EEG and brain MRI in preterm infants: A systematic review. Clin Neurophysiol 2025; 170:168-179. [PMID: 39729686 DOI: 10.1016/j.clinph.2024.12.014] [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: 05/08/2024] [Revised: 10/08/2024] [Accepted: 12/07/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVE To systematically review the literature on the associations between electroencephalogram (EEG) and brain magnetic resonance imaging (MRI) measures in preterm infants (gestational age < 37 weeks). METHODS A comprehensive search was performed in PubMed and EMBASE databases up to February 12th, 2024. Non-relevant studies were eliminated following the PRISMA guidelines. RESULTS Ten out of 991 identified studies were included. Brain MRI metrics used in these studies include volumes, cortical features, microstructural integrity, visual assessments, and cerebral linear measurements. EEG parameters were classified as qualitative (Burdjalov maturity score, seizure burden, and background activity) or quantitative (discontinuity, spectral content, amplitude, and connectivity). Among them, discontinuity and the Burdjalov score were most frequently examined. Higher discontinuity was associated with reduced brain volume, cortical surface, microstructural integrity, and linear measurements. The Burdjalov score related to brain maturation qualitatively assessed on MRI. No other consistent correlations could be established due to the variability across studies. CONCLUSIONS The reviewed studies utilized a variety of EEG and MRI measurements, while discontinuity and the Burdjalov score stood out as significant indicators of structural brain development. SIGNIFICANCE This review, for the first time, provides an extensive overview of EEG-MRI associations in preterm infants, potentially facilitating their clinical application.
Collapse
Affiliation(s)
- Roos F Meijer
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Xiaowan Wang
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Inge M van Ooijen
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Bauke van der Velde
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Manon J N L Benders
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Maria Luisa Tataranno
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands.
| |
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 PMCID: PMC11640791 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; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | | | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - 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; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| |
Collapse
|
3
|
Wallois F, Moghimi S. Revisiting the functional monitoring of brain development in premature neonates. A new direction in clinical care and research. Semin Fetal Neonatal Med 2024; 29:101556. [PMID: 39528364 DOI: 10.1016/j.siny.2024.101556] [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: 11/16/2024]
Abstract
The first 1000 days of life are of paramount importance for neonatal development. Premature newborns are exposed early to the external environment, modifying the fetal exposome and leading to overexposure in some sensory domains and deprivation in others. The resulting neurodevelopmental effects may persist throughout the individual's lifetime. Several neonatal neuromonitoring techniques can be used to investigate neural mechanisms in early postnatal development. EEG is the most widely used, as it is easy to perform, even at the patient's bedside. It is not expensive and provides information with a high temporal resolution and relatively good spatial resolution when performed in high-density mode. Functional near-infrared spectroscopy (fNIRS), a technique for monitoring vascular network dynamics, can also be used at the patient's bedside. It is not expensive and has a good spatial resolution at the cortical surface. These two techniques can be combined for simultaneous monitoring of the neuronal and vascular networks in premature newborns, providing insight into neurodevelopment before term. However, the extent to which more general conclusions about fetal development can be drawn from findings for premature neonates remains unclear due to considerable differences in environmental and medical situations. Fetal MEG (fMEG, as an alternative to EEG for preterm infants) and fMRI (as an alternative to fNIRS for preterm infants) can also be used to investigate fetal neurodevelopment on a trimester-specific basis. These techniques should be used for validation purposes as they are the only tools available for evaluating neuronal dysfunction in the fetus at the time of the gene-environment interactions influencing transient neuronal progenitor populations in brain structures. But what do these techniques tell us about early neurodevelopment? We address this question here, from two points of view. We first discuss spontaneous neural activity and its electromagnetic and hemodynamic correlates. We then explore the effects of stimulating the immature developing brain with information from exogenous sources, reviewing the available evidence concerning the characteristics of electromagnetic and hemodynamic responses. Once the characteristics of the correlates of neural dynamics have been determined, it will be essential to evaluate their possible modulation in the context of disease and in at-risk populations. Evidence can be collected with various neuroimaging techniques targeting both spontaneous and exogenously driven neural activity. A multimodal approach combining the neuromonitoring of different functional compartments (neuronal and vascular) is required to improve our understanding of the normal functioning and dysfunction of the brain and to identify neurobiomarkers for predicting the neurodevelopmental outcome of premature neonate and fetus. Such an approach would provide a framework for exploring early neurodevelopment, paving the way for the development of tools for earlier diagnosis in these vulnerable populations, thereby facilitating preventive, rescue and reparative neurotherapeutic interventions.
Collapse
Affiliation(s)
- Fabrice Wallois
- Inserm U 1105, Department of Pediatric Clinical Neurophysiology, University Hospital, Amiens, France; Inserm U 1105, Multimodal Analysis of Brain Function Research Group (GRAMFC), Université de Picardie, Amiens, France.
| | - Sahar Moghimi
- Inserm U 1105, Multimodal Analysis of Brain Function Research Group (GRAMFC), Université de Picardie, Amiens, France
| |
Collapse
|
4
|
White P, Ranasinghe S, Chen J, Van de Looij Y, Sizonenko S, Prasad J, Berry M, Bennet L, Gunn A, Dean J. Comparative utility of MRI and EEG for early detection of cortical dysmaturation after postnatal systemic inflammation in the neonatal rat. Brain Behav Immun 2024; 121:104-118. [PMID: 39043347 DOI: 10.1016/j.bbi.2024.07.028] [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: 05/02/2024] [Revised: 07/10/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Exposure to postnatal systemic inflammation is associated with increased risk of brain injury in preterm infants, leading to impaired maturation of the cerebral cortex and adverse neurodevelopmental outcomes. However, the optimal method for identifying cortical dysmaturation is unclear. Herein, we compared the utility of electroencephalography (EEG), diffusion tensor imaging (DTI), and neurite orientation dispersion and density imaging (NODDI) at different recovery times after systemic inflammation in newborn rats. METHODS Sprague Dawley rat pups of both sexes received single-daily lipopolysaccharide (LPS; 0.3 mg/kg i.p.; n = 51) or saline (n = 55) injections on postnatal days (P)1, 2, and 3. A subset of these animals were implanted with EEG electrodes. Cortical EEG was recorded for 30 min from unanesthetized, unrestrained pups at P7, P14, and P21, and in separate groups, brain tissues were collected at these ages for ex-vivo MRI analysis (9.4 T) and Golgi-Cox staining (to assess neuronal morphology) in the motor cortex. RESULTS Postnatal inflammation was associated with reduced cortical pyramidal neuron arborization from P7, P14, and P21. These changes were associated with dysmature EEG features (e.g., persistence of delta waveforms, higher EEG amplitude, reduced spectral edge frequency) at P7 and P14, and higher EEG power in the theta and alpha ranges at P21. By contrast, there were no changes in cortical DTI or NODDI in LPS rats at P7 or P14, while there was an increase in cortical fractional anisotropy (FA) and decrease in orientation dispersion index (ODI) at P21. CONCLUSIONS EEG may be useful for identifying the early evolution of impaired cortical development after early life postnatal systemic inflammation, while DTI and NODDI seem to be more suited to assessing established cortical changes.
Collapse
Affiliation(s)
- Petra White
- University of Auckland, Auckland, New Zealand
| | | | - Joseph Chen
- University of Auckland, Auckland, New Zealand
| | - Yohan Van de Looij
- University of Geneva, Geneva, Switzerland; Lausanne Federal Polytechnic School, Lausanne, Switzerland
| | | | - Jaya Prasad
- University of Auckland, Auckland, New Zealand
| | - Mary Berry
- University of Otago, Wellington, New Zealand
| | | | | | - Justin Dean
- University of Auckland, Auckland, New Zealand.
| |
Collapse
|
5
|
Usman F, Marchant S, Baxter L, Salihu HM, Aliyu MH, Adams E, Hartley C. The effect of acute respiratory events and respiratory stimulants on EEG-recorded brain activity in neonates: A systematic review. Clin Neurophysiol Pract 2023; 8:203-225. [PMID: 38125677 PMCID: PMC10730387 DOI: 10.1016/j.cnp.2023.11.002] [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: 03/15/2023] [Revised: 10/16/2023] [Accepted: 11/01/2023] [Indexed: 12/23/2023] Open
Abstract
Objective We conducted a systematic review to investigate electroencephalography (EEG) changes during periods of acute respiratory events such as apnoea and the effect of respiratory stimulants on EEG features in infants. Methods Studies examining respiration and EEG-recorded brain activity in human neonates between 28 and 42 weeks postmenstrual age were included. Two reviewers independently screened all records and included studies were assessed using the Joanna Briggs Institute Critical Appraisal Tool. The protocol was registered in PROSPERO (CRD42022339873). Results We identified 14 studies with a total of 534 infants. Nine articles assessed EEG changes in relation to apnoea, one assessed hiccups, and four investigated the effect of respiratory stimulants. The relationship between neonatal apnoea and EEG changes was inconsistent; EEG suppression and decreased amplitude and frequency were observed during some, but not all, apnoeas. Respiratory stimulants increased EEG continuity compared with before use. Conclusions Current studies in this area are constrained by small sample sizes. Diverse exposure definitions and outcome measures impact inference. Significance This review highlights the need for further work; understanding the relationship between respiration and the developing brain is key to mitigating the long-term effects of apnoea.
Collapse
Affiliation(s)
- Fatima Usman
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Simon Marchant
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Luke Baxter
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Muktar H. Aliyu
- Department of Health Policy and Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eleri Adams
- Newborn Care Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | |
Collapse
|
6
|
Koskela T, Meek J, Huertas-Ceballos A, Kendall GS, Whitehead K. Clinical value of cortical bursting in preterm infants with intraventricular haemorrhage. Early Hum Dev 2023; 184:105840. [PMID: 37556995 DOI: 10.1016/j.earlhumdev.2023.105840] [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: 05/29/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND In healthy preterm infants, cortical burst rate and temporal dynamics predict important measures such as brain growth. We hypothesised that in preterm infants with germinal matrix-intraventricular haemorrhage (GM-IVH), cortical bursting could provide prognostic information. AIMS We determined how cortical bursting was influenced by the injury, and whether this was related to developmental outcome. STUDY DESIGN Single-centre retrospective cohort study at University College London Hospitals, UK. SUBJECTS 33 infants with GM-IVH ≥ grade II (median gestational age: 25 weeks). OUTCOME MEASURES We identified 47 EEGs acquired between 24 and 40 weeks corrected gestational age as part of routine clinical care. In a subset of 33 EEGs from 25 infants with asymmetric injury, we used the least-affected hemisphere as an internal comparison. We tested whether cortical burst rate predicted survival without severe impairment (median 2 years follow-up). RESULTS In asymmetric injury, cortical burst rate was lower over the worst- than least-affected hemisphere, and bursts over the worst-affected hemisphere were less likely to immediately follow bursts over the least-affected hemisphere than vice versa. Overall, burst rate was lower in cases of GM-IVH with parenchymal involvement, relative to milder structural injury grades. Higher burst rate modestly predicted survival without severe language (AUC 0.673) or motor impairment (AUC 0.667), which was partly mediated by structural injury grade. CONCLUSIONS Cortical bursting can index the functional injury after GM-IVH: perturbed burst initiation (rate) and propagation (inter-hemispheric dynamics) likely reflect associated grey matter and white matter damage. Higher cortical burst rate is reassuring for a positive outcome.
Collapse
Affiliation(s)
- Tuomas Koskela
- Research IT Services, University College London, London WC1E 7HB, UK.
| | - Judith Meek
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK; Academic Neonatology, Institute for Women's Health, University College London, London WC1E 6HU, UK.
| | - Angela Huertas-Ceballos
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK.
| | - Giles S Kendall
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK; Academic Neonatology, Institute for Women's Health, University College London, London WC1E 6HU, UK.
| | - Kimberley Whitehead
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK; Department of Neuroscience, Physiology & Pharmacology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
7
|
Kumar BS, O'Herron PJ, Kara P, Chakravarthy VS. The development of bi-directionally coupled self-organizing neurovascular networks captures orientation-selective neural and hemodynamic cortical responses. Eur J Neurosci 2023; 57:1929-1946. [PMID: 37070156 DOI: 10.1111/ejn.15993] [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: 09/23/2022] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/19/2023]
Abstract
Networks of neurons are the primary substrate of information processing. Conversely, blood vessels in the brain are generally viewed to have physiological functions unrelated to information processing, such as the timely supply of oxygen, and other nutrients to the neural tissue. However, recent studies have shown that cerebral microvessels, like neurons, exhibit tuned responses to sensory stimuli. Tuned neural responses to sensory stimuli may be enhanced with experience-dependent Hebbian plasticity and other forms of learning. Hence, it is possible that the microvascular network might also be subject to some form of competitive learning rules during early postnatal development such that its fine-scale structure becomes optimized for metabolic delivery to a given neural micro-architecture. To explore the possibility of adaptive lateral interactions and tuned responses in cerebral microvessels, we modelled the cortical neurovascular network by interconnecting two laterally connected self-organizing networks. The afferent and lateral connections of the neural and vascular networks were defined by trainable weights. By varying the topology of lateral connectivity in the vascular network layer, we observed that the partial correspondence of feature selectivity between neural and hemodynamic responses could be explained by lateral coupling across local blood vessels such that the central domain receives an excitatory drive of more blood flow and a distal surrounding region where blood flow is reduced. Critically, our simulations suggest a new role for feedback from the vascular to the neural network because the radius of vascular perfusion determines whether the cortical neural map develops into a clustered vs. salt-and-pepper organization.
Collapse
Affiliation(s)
- Bhadra S Kumar
- Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, India
| | | | - Prakash Kara
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - V Srinivasa Chakravarthy
- Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, India
- Center for Complex Systems and Dynamics, Indian Institute of Technology Madras (IITM), Chennai, India
| |
Collapse
|
8
|
Lear BA, Lear CA, Dhillon SK, Davidson JO, Gunn AJ, Bennet L. Evolution of grey matter injury over 21 days after hypoxia-ischaemia in preterm fetal sheep. Exp Neurol 2023; 363:114376. [PMID: 36889575 DOI: 10.1016/j.expneurol.2023.114376] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/05/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
Reduced grey matter volume in preterm infants is associated with later disability, but its time course and relationship with white matter injury are not well understood. We recently showed that moderate-severe hypoxia-ischaemia (HI) in preterm fetal sheep led to severe cystic injury 2-3 weeks later. In the same cohort we now show profound hippocampal neuronal loss from 3 days after HI. By contrast, reduction in cortical area and perimeter developed much more slowly, with maximum reduction at day 21. There was transient upregulation of cleaved caspase-3-positive apoptosis in the cortex at day 3 but no change in neuronal density or macroscopic injury of the cortex. Both microglia and astrocytes were transiently upregulated in the grey matter. EEG power was initially profoundly suppressed but partially recovered by 21 days of recovery, and final power was correlated with white matter area (p < 0.001, r2 = 0.75, F = 24.19), cortical area (p = 0.004, r2 = 0.44, F = 11.90) and hippocampi area (p = 0.049, r2 = 0.23, F = 4.58). In conclusion, the present study suggests that in preterm fetal sheep, hippocampal injury is established within a few days of acute HI, but impaired cortical growth develops slowly, in a similar time course to severe white matter injury.
Collapse
Affiliation(s)
- Benjamin A Lear
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Simerdeep K Dhillon
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Joanne O Davidson
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- The Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
9
|
Mukherjee D, Kanold PO. Changing subplate circuits: Early activity dependent circuit plasticity. Front Cell Neurosci 2023; 16:1067365. [PMID: 36713777 PMCID: PMC9874351 DOI: 10.3389/fncel.2022.1067365] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Early neural activity in the developing sensory system comprises spontaneous bursts of patterned activity, which is fundamental for sculpting and refinement of immature cortical connections. The crude early connections that are initially refined by spontaneous activity, are further elaborated by sensory-driven activity from the periphery such that orderly and mature connections are established for the proper functioning of the cortices. Subplate neurons (SPNs) are one of the first-born mature neurons that are transiently present during early development, the period of heightened activity-dependent plasticity. SPNs are well integrated within the developing sensory cortices. Their structural and functional properties such as relative mature intrinsic membrane properties, heightened connectivity via chemical and electrical synapses, robust activation by neuromodulatory inputs-place them in an ideal position to serve as crucial elements in monitoring and regulating spontaneous endogenous network activity. Moreover, SPNs are the earliest substrates to receive early sensory-driven activity from the periphery and are involved in its modulation, amplification, and transmission before the maturation of the direct adult-like thalamocortical connectivity. Consequently, SPNs are vulnerable to sensory manipulations in the periphery. A broad range of early sensory deprivations alters SPN circuit organization and functions that might be associated with long term neurodevelopmental and psychiatric disorders. Here we provide a comprehensive overview of SPN function in activity-dependent development during early life and integrate recent findings on the impact of early sensory deprivation on SPNs that could eventually lead to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Didhiti Mukherjee
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Patrick O. Kanold
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States,Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, United States,*Correspondence: Patrick O. Kanold ✉
| |
Collapse
|
10
|
Zayachkivsky A, Lehmkuhle MJ, Ekstrand JJ, Dudek FE. Background suppression of electrical activity is a potential biomarker of subsequent brain injury in a rat model of neonatal hypoxia-ischemia. J Neurophysiol 2022; 128:118-130. [PMID: 35675445 DOI: 10.1152/jn.00024.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Electrographic seizures and abnormal background activity in the neonatal electroencephalogram (EEG) may differentiate between harmful versus benign brain insults. Using two animal models of neonatal seizures, electrical activity was recorded in freely behaving rats and examined quantitatively during successive time periods with field-potential recordings obtained shortly after the brain insult (i.e., 0-4 days). Single-channel, differential recordings with miniature wireless telemetry were used to analyze spontaneous electrographic seizures and background suppression of electrical activity after 1) hypoxia-ischemia (HI), which is a model of neonatal encephalopathy that causes acute seizures and a large brain lesion with possible development of epilepsy, 2) hypoxia alone (Ha), which causes severe acute seizures without an obvious lesion or subsequent epilepsy, and 3) sham control rats. Background EEG exhibited increases in power as a function of age in control animals. Although background electrical activity was depressed in all frequency bands immediately after HI, suppression in the β and γ bands was greatest and lasted longest. Spontaneous electrographic seizures were recorded, but only in a few HI-treated animals. Ha-treated rat pups were similar to sham controls, they had no subsequent spontaneous electrographic seizures after the treatment and background suppression was only briefly observed in one frequency band. Thus, the normal age-dependent maturation of electrical activity patterns in control animals was significantly disrupted after HI. Suppression of the background EEG observed here after HI-induced acute seizures and subsequent brain injury may be a noninvasive biomarker for detecting severe brain injuries and may help predict subsequent epilepsy.NEW & NOTEWORTHY Biomarkers of neonatal brain injury are needed. Hypoxia-ischemia (HI) in immature rat pups caused severe brain injury, which was associated with strongly suppressed background EEG. The suppression was most robust in the β and γ bands; it started immediately after the HI injury and persisted for days. Thus, background suppression may be a noninvasive biomarker for detecting severe brain injuries and may help predict subsequent epilepsy.
Collapse
Affiliation(s)
- A Zayachkivsky
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - M J Lehmkuhle
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - J J Ekstrand
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - F E Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
11
|
Luhmann HJ, Kanold PO, Molnár Z, Vanhatalo S. Early brain activity: Translations between bedside and laboratory. Prog Neurobiol 2022; 213:102268. [PMID: 35364141 PMCID: PMC9923767 DOI: 10.1016/j.pneurobio.2022.102268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/01/2022] [Accepted: 03/25/2022] [Indexed: 01/29/2023]
Abstract
Neural activity is both a driver of brain development and a readout of developmental processes. Changes in neuronal activity are therefore both the cause and consequence of neurodevelopmental compromises. Here, we review the assessment of neuronal activities in both preclinical models and clinical situations. We focus on issues that require urgent translational research, the challenges and bottlenecks preventing translation of biomedical research into new clinical diagnostics or treatments, and possibilities to overcome these barriers. The key questions are (i) what can be measured in clinical settings versus animal experiments, (ii) how do measurements relate to particular stages of development, and (iii) how can we balance practical and ethical realities with methodological compromises in measurements and treatments.
Collapse
Affiliation(s)
- Heiko J. Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz, Germany.,Correspondence:, , ,
| | - Patrick O. Kanold
- Department of Biomedical Engineering and Kavli Neuroscience Discovery Institute, Johns Hopkins University, School of Medicine, 720 Rutland Avenue / Miller 379, Baltimore, MD 21205, USA.,Correspondence:, , ,
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford OX1 3PT, UK.
| | - Sampsa Vanhatalo
- BABA Center, Departments of Physiology and Clinical Neurophysiology, Children's Hospital, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
12
|
Wang X, Liu H, Kota S, Das Y, Liu Y, Zhang R, Chalak L. EEG phase-amplitude coupling to stratify encephalopathy severity in the developing brain. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 214:106593. [PMID: 34959157 PMCID: PMC11614140 DOI: 10.1016/j.cmpb.2021.106593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 11/19/2021] [Accepted: 12/15/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Neonatal hypoxic ischemic encephalopathy (HIE) is difficult to classify within the narrow therapeutic window of hypothermia. Neurophysiological biomarkers are needed for timely differentiation of encephalopathy severity within the short therapeutic window for initiation of hypothermia therapy. METHODS A novel analysis of mean Phase Amplitude Coupling index, PACm, of amplitudes high frequencies (12-30 Hz) coupled with phases of low (1,2 Hz) frequencies was calculated from the 6 h EEG recorded during the first day of life. PACm values were compared to identify differences between mild versus higher-grade HIE, respectively, for each of the EEG electrodes. A receiver operating characteristic curve was generated to examine the performance of PACm. RESULTS 38 newborns with different HIE grades were enrolled in the first 6 h of life. Threshold PACm 0.001 at Fz, O1, O2, P3, and P4 had AUC >0.9 to differentiate HIE severity and predict the persistence of moderate to severe encephalopathy that requires treatment with hypothermia. CONCLUSION PAC is a promising biomarker to identify mild from higher severity of HIE after birth.
Collapse
Affiliation(s)
- Xinlong Wang
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Hanli Liu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Srinivas Kota
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yudhajit Das
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Yulun Liu
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Rong Zhang
- Departments of Internal Medicine and Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Lina Chalak
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
13
|
Warm D, Schroer J, Sinning A. Gabaergic Interneurons in Early Brain Development: Conducting and Orchestrated by Cortical Network Activity. Front Mol Neurosci 2022; 14:807969. [PMID: 35046773 PMCID: PMC8763242 DOI: 10.3389/fnmol.2021.807969] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/06/2021] [Indexed: 01/22/2023] Open
Abstract
Throughout early phases of brain development, the two main neural signaling mechanisms—excitation and inhibition—are dynamically sculpted in the neocortex to establish primary functions. Despite its relatively late formation and persistent developmental changes, the GABAergic system promotes the ordered shaping of neuronal circuits at the structural and functional levels. Within this frame, interneurons participate first in spontaneous and later in sensory-evoked activity patterns that precede cortical functions of the mature brain. Upon their subcortical generation, interneurons in the embryonic brain must first orderly migrate to and settle in respective target layers before they can actively engage in cortical network activity. During this process, changes at the molecular and synaptic level of interneurons allow not only their coordinated formation but also the pruning of connections as well as excitatory and inhibitory synapses. At the postsynaptic site, the shift of GABAergic signaling from an excitatory towards an inhibitory response is required to enable synchronization within cortical networks. Concomitantly, the progressive specification of different interneuron subtypes endows the neocortex with distinct local cortical circuits and region-specific modulation of neuronal firing. Finally, the apoptotic process further refines neuronal populations by constantly maintaining a controlled ratio of inhibitory and excitatory neurons. Interestingly, many of these fundamental and complex processes are influenced—if not directly controlled—by electrical activity. Interneurons on the subcellular, cellular, and network level are affected by high frequency patterns, such as spindle burst and gamma oscillations in rodents and delta brushes in humans. Conversely, the maturation of interneuron structure and function on each of these scales feeds back and contributes to the generation of cortical activity patterns that are essential for the proper peri- and postnatal development. Overall, a more precise description of the conducting role of interneurons in terms of how they contribute to specific activity patterns—as well as how specific activity patterns impinge on their maturation as orchestra members—will lead to a better understanding of the physiological and pathophysiological development and function of the nervous system.
Collapse
|
14
|
Sheikh A, Meng X, Kao JPY, Kanold PO. Neonatal Hypoxia-Ischemia Causes Persistent Intracortical Circuit Changes in Layer 4 of Rat Auditory Cortex. Cereb Cortex 2021; 32:2575-2589. [PMID: 34729599 DOI: 10.1093/cercor/bhab365] [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: 05/12/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/12/2022] Open
Abstract
The connection between early brain injury and subsequent development of disorders is unknown. Neonatal hypoxia-ischemia (HI) alters circuits associated with subplate neurons (SPNs). SPNs are among the first maturing cortical neurons, project to thalamorecipient layer 4 (L4), and are required for the development of thalamocortical connections. Thus, early HI might influence L4 and such influence might persist. We investigated functional circuits to L4 neurons in neonatal rat HI models of different severities (mild and moderate) shortly after injury and at adolescence. We used laser-scanning photostimulation in slices of auditory cortex during P5-10 and P18-23. Mild injuries did not initially (P6/P7) alter the convergence of excitatory inputs from L2/3, but hyperconnectivity emerged by P8-10. Inputs from L4 showed initial hypoconnectivity which resolved by P8-10. Moderate injuries resulted in initial hypoconnectivity from both layers which resolved by P8-10 and led to persistent strengthening of connections. Inhibitory inputs to L4 cells showed similar changes. Functional changes were mirrored by reduced dendritic complexity. We also observed a persistent increase in similarity of L4 circuits, suggesting that HI interferes with developmental circuit refinement and diversification. Altogether, our results show that neonatal HI injuries lead to persistent changes in intracortical connections.
Collapse
Affiliation(s)
- Aminah Sheikh
- Department of Biology, University of Maryland, College Park, MD 20742, USA.,Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD 20742, USA
| | - Xiangying Meng
- Department of Biology, University of Maryland, College Park, MD 20742, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joseph P Y Kao
- Center for Biomedical Engineering and Technology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Patrick O Kanold
- Department of Biology, University of Maryland, College Park, MD 20742, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
15
|
Learning Disabilities in Reading and Writing and Type of Delivery in Twin Births. CHILDREN-BASEL 2021; 8:children8100834. [PMID: 34682099 PMCID: PMC8534999 DOI: 10.3390/children8100834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023]
Abstract
The aim of this study was to analyse the relationship between the type of delivery (vaginal or caesarean), as a risk factor, and the likelihood of having learning disabilities in reading (reading accuracy) and writing (phonetic and visual orthography), controlling for the interaction and/or confounding effect of gestational, obstetric, and neonatal variables (maternal age at delivery, gestational age, foetal presentation, Apgar 1, and newborn weight) among six-year-old children born in twin births. In this retrospective cohort study, the exposed and non-exposed cohorts consisted of children born by caesarean section and vaginal delivery, respectively. A total of 124 children born in twin births were evaluated in year one of primary education. Intelligence was measured using the K-BIT test; reading and writing variables were evaluated using the Evalúa-1 battery of tests, and clinical records were used to measure gestational, obstetric, and neonatal variables. Binary logistic regressions applied to each dependent variable indicated that caesarean delivery is a possible independent risk factor for difficulties in reading accuracy and phonetic and visual orthography. Future research using larger samples of younger children is required to analyse the relationship between obstetric and neonatal variables and the different basic indicators of reading and writing.
Collapse
|
16
|
Koskela T, Kendall GS, Memon S, Sokolska M, Mabuza T, Huertas-Ceballos A, Mitra S, Robertson NJ, Meek J, Whitehead K. Prognostic value of neonatal EEG following therapeutic hypothermia in survivors of hypoxic-ischemic encephalopathy. Clin Neurophysiol 2021; 132:2091-2100. [PMID: 34284244 PMCID: PMC8407358 DOI: 10.1016/j.clinph.2021.05.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Early prediction of neurological deficits following neonatal hypoxic-ischemic encephalopathy (HIE) may help to target support. Neonatal animal models suggest that recovery following hypoxia-ischemia depends upon cortical bursting. To test whether this holds in human neonates, we correlated the magnitude of cortical bursting during recovery (≥postnatal day 3) with neurodevelopmental outcomes. METHODS We identified 41 surviving infants who received therapeutic hypothermia for HIE (classification at hospital discharge: 19 mild, 18 moderate, 4 severe) and had 9-channel electroencephalography (EEG) recordings as part of their routine care. We correlated burst power with Bayley-III cognitive, motor and language scores at median 24 months. To examine whether EEG offered additional prognostic information, we controlled for structural MRI findings. RESULTS Higher power of central and occipital cortical bursts predicted worse cognitive and language outcomes, and higher power of central cortical bursts predicted worse motor outcome, all independently of structural MRI findings. CONCLUSIONS Clinical EEG after postnatal day 3 may provide additional prognostic information by indexing persistent active mechanisms that either support recovery or exacerbate brain damage, especially in infants with less severe encephalopathy. SIGNIFICANCE These findings could allow for the effect of clinical interventions in the neonatal period to be studied instantaneously in the future.
Collapse
Affiliation(s)
- Tuomas Koskela
- Research IT Services, University College London, London WC1E 7HB, UK.
| | - Giles S Kendall
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK; Academic Neonatology, Institute for Women's Health, University College London, London WC1E 6HU, UK.
| | - Sara Memon
- Department of Neuroscience, Physiology & Pharmacology, University College London, London WC1E 6BT, UK.
| | - Magdalena Sokolska
- Department of Medical Physics and Biomedical Engineering, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK.
| | - Thalitha Mabuza
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK.
| | - Angela Huertas-Ceballos
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK.
| | - Subhabrata Mitra
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK; Academic Neonatology, Institute for Women's Health, University College London, London WC1E 6HU, UK.
| | - Nicola J Robertson
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK; Academic Neonatology, Institute for Women's Health, University College London, London WC1E 6HU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellors Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK.
| | - Judith Meek
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK.
| | - Kimberley Whitehead
- Neonatal Intensive Care Unit, Elizabeth Garrett Anderson Wing, University College London Hospitals, London WC1E 6DB, UK; Department of Neuroscience, Physiology & Pharmacology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
17
|
Kumar BS, Khot A, Chakravarthy VS, Pushpavanam S. A Network Architecture for Bidirectional Neurovascular Coupling in Rat Whisker Barrel Cortex. Front Comput Neurosci 2021; 15:638700. [PMID: 34211384 PMCID: PMC8241226 DOI: 10.3389/fncom.2021.638700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Neurovascular coupling is typically considered as a master-slave relationship between the neurons and the cerebral vessels: the neurons demand energy which the vessels supply in the form of glucose and oxygen. In the recent past, both theoretical and experimental studies have suggested that the neurovascular coupling is a bidirectional system, a loop that includes a feedback signal from the vessels influencing neural firing and plasticity. An integrated model of bidirectionally connected neural network and the vascular network is hence required to understand the relationship between the informational and metabolic aspects of neural dynamics. In this study, we present a computational model of the bidirectional neurovascular system in the whisker barrel cortex and study the effect of such coupling on neural activity and plasticity as manifest in the whisker barrel map formation. In this model, a biologically plausible self-organizing network model of rate coded, dynamic neurons is nourished by a network of vessels modeled using the biophysical properties of blood vessels. The neural layer which is designed to simulate the whisker barrel cortex of rat transmits vasodilatory signals to the vessels. The feedback from the vessels is in the form of available oxygen for oxidative metabolism whose end result is the adenosine triphosphate (ATP) necessary to fuel neural firing. The model captures the effect of the feedback from the vascular network on the neuronal map formation in the whisker barrel model under normal and pathological (Hypoxia and Hypoxia-Ischemia) conditions.
Collapse
Affiliation(s)
- Bhadra S. Kumar
- Computational Neuroscience Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Aditi Khot
- Department of Chemical Engineering, Purdue University, West Lafayette, IN, United States
| | - V. Srinivasa Chakravarthy
- Computational Neuroscience Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - S. Pushpavanam
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
18
|
Kidokoro H. Delta brushes are not just a hallmark of EEG in human preterm infants. Pediatr Int 2021; 63:130-136. [PMID: 32749014 DOI: 10.1111/ped.14420] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 02/02/2023]
Abstract
The delta brush, a well-known characteristic waveform of the human preterm electroencephalogram, represents spontaneous electrical activity. Recent experimental animal model evidence suggests that delta brushes are not only spontaneous intrinsic activity but are also evoked by external sensory stimulation or spontaneous movement. They are also likely to reflect the activity of subplate neurons, which play an important role in early brain development and network organization. Here, evidence about delta brushes in human preterm electroencephalogram is provided along with future perspectives.
Collapse
Affiliation(s)
- Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
19
|
Wallois F, Routier L, Heberlé C, Mahmoudzadeh M, Bourel-Ponchel E, Moghimi S. Back to basics: the neuronal substrates and mechanisms that underlie the electroencephalogram in premature neonates. Neurophysiol Clin 2020; 51:5-33. [PMID: 33162287 DOI: 10.1016/j.neucli.2020.10.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Electroencephalography is the only clinically available technique that can address the premature neonate normal and pathological functional development week after week. The changes in the electroencephalogram (EEG) result from gradual structural and functional modifications that arise during the last trimester of pregnancy. Here, we review the structural changes over time that underlie the establishment of functional immature neural networks, the impact of certain anatomical specificities (fontanelles, connectivity, etc.) on the EEG, limitations in EEG interpretation, and the utility of high-resolution EEG (HR-EEG) in premature newborns (a promising technique with a high degree of spatiotemporal resolution). In particular, we classify EEG features according to whether they are manifestations of endogenous generators (i.e. theta activities that coalesce with a slow wave or delta brushes) or come from a broader network. Furthermore, we review publications on EEG in premature animals because the data provide a better understanding of what is happening in premature newborns. We then discuss the results and limitations of functional connectivity analyses in premature newborns. Lastly, we report on the magnetoelectroencephalographic studies of brain activity in the fetus. A better understanding of complex interactions at various structural and functional levels during normal neurodevelopment (as assessed using electroencephalography as a benchmark method) might lead to better clinical care and monitoring for premature neonates.
Collapse
Affiliation(s)
- Fabrice Wallois
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France.
| | - Laura Routier
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Claire Heberlé
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Mahdi Mahmoudzadeh
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Emilie Bourel-Ponchel
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| | - Sahar Moghimi
- INSERM U1105, Research Group on Multimodal Analysis of Brain Function, Jules Verne University of Picardie, Amiens, France; Service d'Explorations Fonctionnelles du Système Nerveux Pédiatrique, Amiens-Picardie Medical Center, Amiens, France
| |
Collapse
|
20
|
Hamdy N, Eide S, Sun HS, Feng ZP. Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol 2020; 334:113457. [PMID: 32889009 DOI: 10.1016/j.expneurol.2020.113457] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.
Collapse
Affiliation(s)
- Nancy Hamdy
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
21
|
Sun H, Gonzalez F, McQuillen PS. Caffeine Restores Background EEG Activity Independent of Infarct Reduction after Neonatal Hypoxic Ischemic Brain Injury. Dev Neurosci 2020; 42:72-82. [PMID: 32810862 DOI: 10.1159/000509365] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/10/2020] [Indexed: 12/19/2022] Open
Abstract
In human preterm newborns, caffeine increases brain activity and improves neurodevelopmental outcomes. In animal models of hypoxic ischemic brain injury, caffeine pretreatment reduces infarct volume. We studied the relationship between tissue neuroprotection and brain activity after injury to further understand caffeine neuroprotection. Rat dams received caffeine prior to birth or on postnatal day 3 (P3) through P16. Caffeine-treated and -untreated pups underwent the Vannucci procedure (unilateral carotid ligation, global hypoxia) on P2. A subset had EEG recordings. Brain hemispheric infarct volume was measured on P16. P2 hypoxic ischemia (HI) results in histologic brain injury (mean ± standard deviation infarct volume 10.3 ± 4.6%) and transient suppression of EEG activity. Caffeine pretreatment reduces brain injury (mean ± standard deviation infarct volume 1.6 ± 4.5%, p < 0.001) and improves amplitude-integrated EEG (aEEG) and EEG burst duration and amplitude. Caffeine treatment after HI does not reduce infarct volume (mean ± standard deviation 8.3 ± 4.1%, p = 1.0). However, caffeine posttreatment was equally effective at restoring aEEG amplitude and EEG burst duration and amplitude. Thus, caffeine supports brain background electrical activity independent of tissue neuroprotection.
Collapse
Affiliation(s)
- Haiyan Sun
- Institute of Pediatrics, Xuzhou Medical University, Xuzhou, China
| | - Fernando Gonzalez
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Patrick S McQuillen
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA,
| |
Collapse
|
22
|
Fleiss B, Gressens P, Stolp HB. Cortical Gray Matter Injury in Encephalopathy of Prematurity: Link to Neurodevelopmental Disorders. Front Neurol 2020; 11:575. [PMID: 32765390 PMCID: PMC7381224 DOI: 10.3389/fneur.2020.00575] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Preterm-born infants frequently suffer from an array of neurological damage, collectively termed encephalopathy of prematurity (EoP). They also have an increased risk of presenting with a neurodevelopmental disorder (e.g., autism spectrum disorder; attention deficit hyperactivity disorder) later in life. It is hypothesized that it is the gray matter injury to the cortex, in addition to white matter injury, in EoP that is responsible for the altered behavior and cognition in these individuals. However, although it is established that gray matter injury occurs in infants following preterm birth, the exact nature of these changes is not fully elucidated. Here we will review the current state of knowledge in this field, amalgamating data from both clinical and preclinical studies. This will be placed in the context of normal processes of developmental biology and the known pathophysiology of neurodevelopmental disorders. Novel diagnostic and therapeutic tactics required integration of this information so that in the future we can combine mechanism-based approaches with patient stratification to ensure the most efficacious and cost-effective clinical practice.
Collapse
Affiliation(s)
- Bobbi Fleiss
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Université de Paris, NeuroDiderot, Inserm, Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Pierre Gressens
- Université de Paris, NeuroDiderot, Inserm, Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Helen B. Stolp
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
23
|
Pineda R, Liszka L, Inder T. Early neurobehavior at 30 weeks postmenstrual age is related to outcome at term equivalent age. Early Hum Dev 2020; 146:105057. [PMID: 32470768 PMCID: PMC7377927 DOI: 10.1016/j.earlhumdev.2020.105057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/03/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
AIMS To determine 1) the relationship between infant medical factors and early neurobehavior, and 2) the relationship between early neurobehavior at 30 weeks postmenstrual age (PMA) and neurobehavior at term equivalent age. STUDY DESIGN In this prospective longitudinal study, 88 very preterm infants born ≤30 weeks estimated gestational age (EGA) had neurobehavioral assessments at 30 weeks PMA using the Premie-Neuro and at term equivalent age using the NICU Network Neurobehavioral Scale (NNNS) and Hammersmith Neonatal Neurological Evaluation (HNNE). RESULTS Lower Premie-Neuro scores at 30 weeks PMA were related to being more immature at birth (p = 0.01; β = 3.87); the presence of patent ductus arteriosus (PDA; p < 0.01; β = -16.50) and cerebral injury (p < 0.01; β = -20.46); and prolonged exposure to oxygen therapy (p < 0.01; β = -0.01), endotracheal intubation (p < 0.01; β = -0.23), and total parenteral nutrition (p < 0.01; β = -0.35). After controlling for EGA, PDA, and number of days of endotracheal intubation, lower Premie-Neuro scores at 30 weeks PMA were independently related to lower total HNNE scores at term (p < 0.01; β = 0.12) and worse outcome on the NNNS with poorer quality of movement (p < 0.01; β = 0.02) and more stress (p < 0.01; ß = -0.004), asymmetry (p = 0.01; β = -0.04), excitability (p < 0.01; β = -0.05) and suboptimal reflexes (p < 0.01; ß = -0.06). CONCLUSION Medical factors were associated with early neurobehavioral performance at 30 weeks PMA. Early neurobehavior at 30 weeks PMA was a good marker of adverse neurobehavior at NICU discharge.
Collapse
Affiliation(s)
- Roberta Pineda
- University of Southern California, Chan Division of Occupational Science and Occupational Therapy, Los Angeles, CA, United States of America; Keck School of Medicine, Department of Pediatrics, Los Angeles, CA, United States of America; Washington University School of Medicine, Program in Occupational Therapy, St. Louis, MO, United States of America.
| | - Lara Liszka
- Washington University School of Medicine, Program in Occupational Therapy, St. Louis, MO, United States of America; Seattle Children's Hospital, Seattle, WA, United States of America
| | - Terrie Inder
- Brigham and Women's Hospital, Department of Pediatric Newborn Medicine, Boston, MA, United States of America; Harvard University, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
24
|
Tang W, Xin X, O'Connor M, Zhang N, Lai B, Man HY, Xie Y, Wei Y. Transient sublethal hypoxia in neonatal rats causes reduced dendritic spines, aberrant synaptic plasticity, and impairments in memory. J Neurosci Res 2020; 98:1588-1604. [PMID: 32495348 DOI: 10.1002/jnr.24652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 01/06/2023]
Abstract
Hypoxic/ischemic insult, a leading cause of functional brain defects, has been extensively studied in both clinical and experimental animal research, including its etiology, neuropathogenesis, and pharmacological interventions. Transient sublethal hypoxia (TSH) is a common clinical occurrence in the perinatal period. However, its effect on early developing brains remains poorly understood. The present study was designed to investigate the effect of TSH on the dendrite and dendritic spine formation, neuronal and synaptic activity, and cognitive behavior of early postnatal Day 1 rat pups. While TSH showed no obvious effect on gross brain morphology, neuron cell density, or glial activation in the hippocampus, we found transient hypoxia did cause significant changes in neuronal structure and function. In brains exposed to TSH, hippocampal neurons developed shorter and thinner dendrites, with decreased dendritic spine density, and reduced strength in excitatory synaptic transmission. Moreover, TSH-treated rats showed impaired cognitive performance in spatial learning and memory. Our findings demonstrate that TSH in newborn rats can cause significant impairments in synaptic formation and function, and long-lasting brain functional deficits. Therefore, this study provides a useful animal model for the study of TSH on early developing brains and to explore potential pharmaceutical interventions for patients subjected to TSH insult.
Collapse
Affiliation(s)
- Wenjie Tang
- Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoming Xin
- Shanghai University of Medicine and Health Sciences, Shanghai, China
| | | | - Nana Zhang
- Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Lai
- Institute of Brain science, Fudan University, Shanghai, China
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA
| | - Yuanyun Xie
- National Clinic and Medicine Research Institute for Geriatric Diseases, Gannan Health Promotion and Translational Laboratory, The First Affiliated Hospital, Gannan University of Medical sciences, Ganzhou, China
| | - Youzhen Wei
- Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Andreoli E, Petrenko V, Constanthin PE, Contestabile A, Bocchi R, Egervari K, Quairiaux C, Salmon P, Kiss JZ. Transplanted Embryonic Neurons Improve Functional Recovery by Increasing Activity in Injured Cortical Circuits. Cereb Cortex 2020; 30:4708-4725. [PMID: 32266929 DOI: 10.1093/cercor/bhaa075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 02/07/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Transplantation of appropriate neuronal precursors after injury is a promising strategy to reconstruct cortical circuits, but the efficiency of these approaches remains limited. Here, we applied targeted apoptosis to selectively ablate layer II/III pyramidal neurons in the rat juvenile cerebral cortex and attempted to replace lost neurons with their appropriate embryonic precursors by transplantation. We demonstrate that grafted precursors do not migrate to replace lost neurons but form vascularized clusters establishing reciprocal synaptic contacts with host networks and show functional integration. These heterotopic neuronal clusters significantly enhance the activity of the host circuits without causing epileptic seizures and attenuate the apoptotic injury-induced functional deficits in electrophysiological and behavioral tests. Chemogenetic activation of grafted neurons further improved functional recovery, and the persistence of the graft was necessary for maintaining restored functions in adult animals. Thus, implanting neuronal precursors capable to form synaptically integrated neuronal clusters combined with activation-based approaches represents a useful strategy for helping long-term functional recovery following brain injury.
Collapse
Affiliation(s)
- Evgenia Andreoli
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Volodymyr Petrenko
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Paul Eugène Constanthin
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Alessandro Contestabile
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Riccardo Bocchi
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Kristof Egervari
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Charles Quairiaux
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Patrick Salmon
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Jozsef Zoltan Kiss
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| |
Collapse
|
26
|
Sheikh A, Meng X, Liu J, Mikhailova A, Kao JPY, McQuillen PS, Kanold PO. Neonatal Hypoxia-Ischemia Causes Functional Circuit Changes in Subplate Neurons. Cereb Cortex 2020; 29:765-776. [PMID: 29365081 DOI: 10.1093/cercor/bhx358] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/21/2017] [Indexed: 01/16/2023] Open
Abstract
Neonatal hypoxia-ischemia (HI) in the preterm human results in damage to subcortical developing white matter and cognitive impairments. Subplate neurons (SPNs) are among the first-born cortical neurons and are necessary for normal cerebral development. While moderate or severe HI at P1 in rats leads to SPN loss, it is unclear if HI, esp. forms not associated with overt cell loss lead to altered SPN circuits. Thus, we used two HI models with different severities in P1 rats. Cauterization of the common carotid artery (CCA) causes a largely transient and thus milder ischemia (HI-Caut) while CCA ligation causes more severe ischemia (HI-Lig). While HI-Lig caused subplate damage, HI-Caut did not cause overt histological damage on the light microscopic level. We used laser-scanning photostimulation (LSPS) in acute thalamocortical slices of auditory cortex during P5-10 to study the functional connectivity of SPNs. Both HI categories resulted in hyperconnectivity of excitatory and inhibitory circuits to SPNs. Thus, alterations on the circuit level are present in the absence of cell loss. Our results show that SPN circuits are uniquely susceptible to HI. Given the key developmental role of SPNs, our results suggest that altered SPN circuits might underlie the abnormal development of cortical function after HI.
Collapse
Affiliation(s)
- Aminah Sheikh
- Department of Biology, University of Maryland, College Park, MD, USA.,Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD, USA
| | - Xiangying Meng
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Ji Liu
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Alexandra Mikhailova
- Departments of Pediatrics and Neurology, University of California, San Francisco, CA, USA
| | - Joseph P Y Kao
- Center for Biomedical Engineering and Technology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Patrick S McQuillen
- Departments of Pediatrics and Neurology, University of California, San Francisco, CA, USA
| | - Patrick O Kanold
- Department of Biology, University of Maryland, College Park, MD, USA.,Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD, USA
| |
Collapse
|
27
|
Wallois F, Routier L, Bourel-Ponchel E. Impact of prematurity on neurodevelopment. HANDBOOK OF CLINICAL NEUROLOGY 2020; 173:341-375. [PMID: 32958184 DOI: 10.1016/b978-0-444-64150-2.00026-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The consequences of prematurity on brain functional development are numerous and diverse, and impact all brain functions at different levels. Prematurity occurs between 22 and 36 weeks of gestation. This period is marked by extreme dynamics in the physiologic maturation, structural, and functional processes. These different processes appear sequentially or simultaneously. They are dependent on genetic and/or environmental factors. Disturbance of these processes or of the fine-tuning between them, when caring for premature children, is likely to induce disturbances in the structural and functional development of the immature neural networks. These will appear as impairments in learning skills progress and are likely to have a lasting impact on the development of children born prematurely. The level of severity depends on the initial alteration, whether structural or functional. In this chapter, after having briefly reviewed the neurodevelopmental, structural, and functional processes, we describe, in a nonexhaustive manner, the impact of prematurity on the different brain, motor, sensory, and cognitive functions.
Collapse
Affiliation(s)
- Fabrice Wallois
- Research Group on Multimodal Analysis of Brain Function, Jules Verne Picardie University, Amiens, France; Department of Pediatric Functional Exploration of the Nervous System, University Hospital, Picardie, Amiens, France.
| | - Laura Routier
- Research Group on Multimodal Analysis of Brain Function, Jules Verne Picardie University, Amiens, France; Department of Pediatric Functional Exploration of the Nervous System, University Hospital, Picardie, Amiens, France
| | - Emilie Bourel-Ponchel
- Research Group on Multimodal Analysis of Brain Function, Jules Verne Picardie University, Amiens, France; Department of Pediatric Functional Exploration of the Nervous System, University Hospital, Picardie, Amiens, France
| |
Collapse
|
28
|
Abbasi H, Unsworth CP. Electroencephalogram studies of hypoxic ischemia in fetal and neonatal animal models. Neural Regen Res 2020; 15:828-837. [PMID: 31719243 PMCID: PMC6990791 DOI: 10.4103/1673-5374.268892] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alongside clinical achievements, experiments conducted on animal models (including primate or non-primate) have been effective in the understanding of various pathophysiological aspects of perinatal hypoxic/ischemic encephalopathy (HIE). Due to the reasonably fair degree of flexibility with experiments, most of the research around HIE in the literature has been largely concerned with the neurodevelopmental outcome or how the frequency and duration of HI seizures could relate to the severity of perinatal brain injury, following HI insult. This survey concentrates on how EEG experimental studies using asphyxiated animal models (in rodents, piglets, sheep and non-human primate monkeys) provide a unique opportunity to examine from the exact time of HI event to help gain insights into HIE where human studies become difficult.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| | - Charles P Unsworth
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| |
Collapse
|
29
|
González-Valenzuela MJ, López-Montiel D, Cazorla-Granados O, González-Mesa E. Type of delivery and reading, writing, and arithmetic learning in twin births. Dev Psychobiol 2019; 62:484-495. [PMID: 31674012 DOI: 10.1002/dev.21932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 12/27/2022]
Abstract
This study analyses, in children born in twin births, the relationship between reading, writing and arithmetic learning, on the one hand, and type of delivery, on the other, controlling for the effect of interaction and/or confusion of third variables (maternal age at delivery, gestational age, fetal position, birthweight, 1-min Apgar score). In the planned retrospective cohort design, the exposed cohort consisted of children born by caesarean section, and the non-exposed cohort was comprised of children born vaginally. One hundred and twenty-four children born of twin births were evaluated during their first year of primary school: K-BIT tests were used to measure intelligence; the Evalúa-1 battery was used to assess reading, writing, and arithmetic ability; and the children's clinical histories were analysed for obstetric and neonatal variables. After applying binary logistic regressions for each dependent variable, it was found that caesarean delivery in twin births appeared as a possible independent risk factor for specific learning disabilities (LDs) in reading, writing, and arithmetic. Based on these results, further research using larger samples and at more advanced ages is required in order to analyse the influence of obstetric and neonatal variables on the processes underlying specific LDs.
Collapse
Affiliation(s)
| | - Dolores López-Montiel
- Dpto. Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Olga Cazorla-Granados
- Colegio Santa Rosa de Lima, Fundación de Enseñanza Santa Mª de la Victoria, Málaga, Spain
| | - Ernesto González-Mesa
- Área de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
30
|
Cavarsan CF, Gorassini MA, Quinlan KA. Animal models of developmental motor disorders: parallels to human motor dysfunction in cerebral palsy. J Neurophysiol 2019; 122:1238-1253. [PMID: 31411933 PMCID: PMC6766736 DOI: 10.1152/jn.00233.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cerebral palsy (CP) is the most common motor disability in children. Much of the previous research on CP has focused on reducing the severity of brain injuries, whereas very few researchers have investigated the cause and amelioration of motor symptoms. This research focus has had an impact on the choice of animal models. Many of the commonly used animal models do not display a prominent CP-like motor phenotype. In general, rodent models show anatomically severe injuries in the central nervous system (CNS) in response to insults associated with CP, including hypoxia, ischemia, and neuroinflammation. Unfortunately, most rodent models do not display a prominent motor phenotype that includes the hallmarks of spasticity (muscle stiffness and hyperreflexia) and weakness. To study motor dysfunction related to developmental injuries, a larger animal model is needed, such as rabbit, pig, or nonhuman primate. In this work, we describe and compare various animal models of CP and their potential for translation to the human condition.
Collapse
Affiliation(s)
- Clarissa F Cavarsan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Monica A Gorassini
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Katharina A Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| |
Collapse
|
31
|
Martino MSD, Alves LLF, Ortiz Sobrinho C, Barbosa ADM. Comparison on cognitive performance among schoolchildren born prematurely according to the presence of intracranial hemorrhage in the neonatal period. REVISTA BRASILEIRA DE SAÚDE MATERNO INFANTIL 2019. [DOI: 10.1590/1806-93042019000300002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract Objectives: to compare the cognitive performance of schoolchildren born prematurely according to the presence of intracranial hemorrhage (ICH) during the neonatal period. Methods: a cross-sectional cohort study of schoolchildren between the ages of 6-8 years old, born prematurely with or without a history of neonatal ICH. Between January and December 2015, some children were followed up at the outpatient clinic of a tertiary hospital and underwent a cognitive evaluation by using the Wechsler Intelligence Scale for Children, Third Edition (WISC-III) and they were divided into two groups: those with no history of ICH (control group) and those with ICH (case group), confirmed by a transfontanelar ultrasound in the prenatal period. Results: 39 schoolchildren were included, 21 cases and 18 controls. There was no difference in gestational age or chronological age at evaluation between the groups. Also there was no significant difference in subtest scores between the groups. Conclusions: WISC-III evaluated the cognitive performance in children, born preterm, aged 6-8 years old, and had neonatal ICH did not differ from those of their peers without a history of ICH. These findings suggest that, in preterm infants, a neonatal diagnosis of ICH may not be associated with cognitive performance at school age and this should be investigated through a longitudinal study.
Collapse
|
32
|
Doran SJ, Jandzinski M, Patrizz A, Trammel C, Sharmeen R, Mamun AA, Capozzi LA, Venna VR, Liu F, McCullough LD. Deficits in ultrasonic vocalization development and production following neonatal hypoxic ischemic insult. Behav Brain Res 2019; 369:111931. [PMID: 31047922 DOI: 10.1016/j.bbr.2019.111931] [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: 03/07/2019] [Revised: 04/22/2019] [Accepted: 04/28/2019] [Indexed: 10/26/2022]
Abstract
Neonatal hypoxic ischemia encephalopathy (HIE) leads to major deficits in language development. While clinically there is a known correlation in the degree of HIE injury and subsequent language disability, there are no treatments beyond speech and language therapy; therefore, experimental studies with a HIE animal model to test new interventions and therapeutics are warranted. Neonatal rodents normally ultrasonically vocalize at postnatal day 7 (PND 7) to PND 14 in response to removal from their mothers. At 6-8 weeks of age juvenile male rodents ultrasonically vocalize in response to exposure to a mature female mouse. Changes in ultrasonic vocalization (USV) production after neonatal brain injury, such ashypoxic ischemia (HI), have not been studied. This study examines the acute and long-term ultrasonic vocalization ability of mice after HI at PND 10. Pups were subjected to HI, sham, or naïve conditions; where in HI and sham surgeries the right common carotid artery was exposed, in the HI this artery was double ligated. The HI and sham pups were then exposed to60minof hypoxia. Naïve pups did not undergo surgery and were subjected to60minof room air. At 3 days following surgery, HI and sham pups vocalize less than nonsurgical naïve controls; yet "juvenile" mice of 6-8 weeks old that underwent HI at PND 10 vocalize less than sham and naïve mice. We conclude that HI injury has significant impact on later adult vocalization.
Collapse
Affiliation(s)
- Sarah J Doran
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States
| | - Mike Jandzinski
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States
| | - Anthony Patrizz
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States; Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, United States
| | - Cassandra Trammel
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States
| | - Romana Sharmeen
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, United States
| | - Abdullah A Mamun
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, United States
| | - Lori A Capozzi
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States
| | - Venugopal Reddy Venna
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States; Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, United States
| | - Fudong Liu
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States; Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, United States
| | - Louise D McCullough
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States; Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX 77030, United States.
| |
Collapse
|
33
|
Kanold PO, Deng R, Meng X. The Integrative Function of Silent Synapses on Subplate Neurons in Cortical Development and Dysfunction. Front Neuroanat 2019; 13:41. [PMID: 31040772 PMCID: PMC6476909 DOI: 10.3389/fnana.2019.00041] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
The thalamocortical circuit is of central importance in relaying information to the cortex. In development, subplate neurons (SPNs) form an integral part of the thalamocortical pathway. These early born cortical neurons are the first neurons to receive thalamic inputs and excite neurons in the cortical plate. This feed-forward circuit topology of SPNs supports the role of SPNs in shaping the formation and plasticity of thalamocortical connections. Recently it has been shown that SPNs also receive inputs from the developing cortical plate and project to the thalamus. The cortical inputs to SPNs in early ages are mediated by N-methyl-D-aspartate (NMDA)-receptor only containing synapses while at later ages α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-receptors are present. Thus, SPNs perform a changing integrative function over development. NMDA-receptor only synapses are crucially influenced by the resting potential and thus insults to the developing brain that causes depolarizations, e.g., hypoxia, can influence the integrative function of SPNs. Since such insults in humans cause symptoms of neurodevelopmental disorders, NMDA-receptor only synapses on SPNs might provide a crucial link between early injuries and later circuit dysfunction. We thus here review subplate associated circuits, their changing functions, and discuss possible roles in development and disease.
Collapse
Affiliation(s)
- Patrick O. Kanold
- Department of Biology, University of Maryland, College Park, MD, United States
| | | | | |
Collapse
|
34
|
Zhang Y, Xu N, Ding Y, Doycheva DM, Zhang Y, Li Q, Flores J, Haghighiabyaneh M, Tang J, Zhang JH. Chemerin reverses neurological impairments and ameliorates neuronal apoptosis through ChemR23/CAMKK2/AMPK pathway in neonatal hypoxic-ischemic encephalopathy. Cell Death Dis 2019; 10:97. [PMID: 30718467 PMCID: PMC6362229 DOI: 10.1038/s41419-019-1374-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 12/12/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a devastating neurological event that contributes to the prolonged neurodevelopmental consequences in infants. Therapeutic strategies focused on attenuating neuronal apoptosis in the penumbra appears to be promising. Given the increasingly recognized neuroprotective roles of adipokines in HIE, we investigated the potential anti-apoptotic roles of a novel member of adipokines, Chemerin, in an experimental model of HIE. In the present study, 10-day-old rat pups underwent right common carotid artery ligation followed by 2.5 h hypoxia. At 1 h post hypoxia, pups were intranasally administered with human recombinant chemerin (rh-chemerin). Here, we showed that rh-chemerin prevented the neuronal apoptosis and degeneration as evidenced by the decreased expression of the pro-apoptotic markers, cleaved caspase 3 and Bax, as well as the numbers of Fluoro-Jade C and TUNEL-positive neurons. Furthermore, rh-Chemerin reversed neurological and morphological impairments induced by hypoxia-ischemia in neonatal rats at 24 h and 4 weeks after HIE. In addition, chemerin-mediated neuronal survival correlated with the elevation of chemerin receptor 23 (chemR23), phosphorylated calmodulin-dependent protein kinase kinase 2 (CAMKK2), as well as phosphorylated adenosine monophosphate-activated protein kinase (AMPK). Specific inhibition of chemR23, CAMKK2, and AMPK abolished the anti-apoptotic effects of rh-chemerin at 24 h after HIE, demonstrating that rh-chemerin ameliorated neuronal apoptosis partially via activating chemR23/CAMKK2/AMPK signaling pathway. Neuronal apoptosis is a well-established contributing factor of pathological changes and the neurological impairment after HIE. These results revealed mechanisms of neuroprotection by rh-chemerin, and indicated that activation of chemR23 might be harnessed to protect from neuronal apoptosis in HIE.
Collapse
Affiliation(s)
- Yixin Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Ningbo Xu
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Yan Ding
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Yiting Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Qian Li
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Jerry Flores
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Mina Haghighiabyaneh
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA.
- Departments of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
35
|
Boylan GB, Kharoshankaya L, Mathieson SR. Diagnosis of seizures and encephalopathy using conventional EEG and amplitude integrated EEG. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:363-400. [PMID: 31324321 DOI: 10.1016/b978-0-444-64029-1.00018-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seizures are more common in the neonatal period than at any other time of life, partly due to the relative hyperexcitability of the neonatal brain. Brain monitoring of sick neonates in the NICU using either conventional electroencephalography or amplitude integrated EEG is essential to accurately detect seizures. Treatment of seizures is important, as evidence increasingly indicates that seizures damage the brain in addition to that caused by the underlying etiology. Prompt treatment has been shown to reduce seizure burden with the potential to ameliorate seizure-mediated damage. Neonatal encephalopathy most commonly caused by a hypoxia-ischemia results in an alteration of mental status and problems such as seizures, hypotonia, apnea, and feeding difficulties. Confirmation of encephalopathy with EEG monitoring can act as an important adjunct to other investigations and the clinical examination, particularly when considering treatment strategies such as therapeutic hypothermia. Brain monitoring also provides useful early prognostic indicators to clinicians. Recent use of machine learning in algorithms to continuously monitor the neonatal EEG, detect seizures, and grade encephalopathy offers the exciting prospect of real-time decision support in the NICU in the very near future.
Collapse
Affiliation(s)
- Geraldine B Boylan
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland.
| | - Liudmila Kharoshankaya
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
| | - Sean R Mathieson
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Dempsey EM, Kooi EMW, Boylan G. It's All About the Brain-Neuromonitoring During Newborn Transition. Semin Pediatr Neurol 2018; 28:48-59. [PMID: 30522728 DOI: 10.1016/j.spen.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- E M Dempsey
- Department of Paediatrics and Child Health, Neonatal Intensive Care Unit, University College Cork, Cork, Ireland; INFANT, Irish Centre for Fetal and Neonatal Translational Research, University College Cork, Cork, Ireland
| | - E M W Kooi
- Department of Paediatrics and Child Health, Neonatal Intensive Care Unit, University College Cork, Cork, Ireland; Division of Neonatology, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, Groningen, The Netherlands
| | - Geraldine Boylan
- Department of Paediatrics and Child Health, Neonatal Intensive Care Unit, University College Cork, Cork, Ireland; INFANT, Irish Centre for Fetal and Neonatal Translational Research, University College Cork, Cork, Ireland.
| |
Collapse
|
37
|
Luhmann HJ, Kirischuk S, Kilb W. The Superior Function of the Subplate in Early Neocortical Development. Front Neuroanat 2018; 12:97. [PMID: 30487739 PMCID: PMC6246655 DOI: 10.3389/fnana.2018.00097] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022] Open
Abstract
During early development the structure and function of the cerebral cortex is critically organized by subplate neurons (SPNs), a mostly transient population of glutamatergic and GABAergic neurons located below the cortical plate. At the molecular and morphological level SPNs represent a rather diverse population of cells expressing a variety of genetic markers and revealing different axonal-dendritic morphologies. Electrophysiologically SPNs are characterized by their rather mature intrinsic membrane properties and firing patterns. They are connected via electrical and chemical synapses to local and remote neurons, e.g., thalamic relay neurons forming the first thalamocortical input to the cerebral cortex. Therefore SPNs are robustly activated at pre- and perinatal stages by the sensory periphery. Although SPNs play pivotal roles in early neocortical activity, development and plasticity, they mostly disappear by programmed cell death during further maturation. On the one hand, SPNs may be selectively vulnerable to hypoxia-ischemia contributing to brain damage, on the other hand there is some evidence that enhanced survival rates or alterations in SPN distribution may contribute to the etiology of neurological or psychiatric disorders. This review aims to give a comprehensive and up-to-date overview on the many functions of SPNs during early physiological and pathophysiological development of the cerebral cortex.
Collapse
Affiliation(s)
- Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
38
|
Rensing N, Moy B, Friedman JL, Galindo R, Wong M. Longitudinal analysis of developmental changes in electroencephalography patterns and sleep-wake states of the neonatal mouse. PLoS One 2018; 13:e0207031. [PMID: 30399187 PMCID: PMC6219806 DOI: 10.1371/journal.pone.0207031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/23/2018] [Indexed: 11/19/2022] Open
Abstract
The neonatal brain undergoes rapid maturational changes that facilitate the normal development of the nervous system and also affect the pathological response to brain injury. Electroencephalography (EEG) and analysis of sleep-wake vigilance states provide important insights into the function of the normal and diseased immature brain. While developmental changes in EEG and vigilance states are well-described in people, less is known about the normal maturational properties of rodent EEG, including the emergence and evolution of sleep-awake vigilance states. In particular, a number of developmental EEG studies have been performed in rats, but there is limited comparable research in neonatal mice, especially as it pertains to longitudinal EEG studies performed within the same mouse. In this study, we have attempted to provide a relatively comprehensive assessment of developmental changes in EEG background activity and vigilance states in wild-type mice from postnatal days 9-21. A novel EEG and EMG method allowed serial recording from the same mouse pups. EEG continuity and power and vigilance states were analyzed by quantitative assessment and fast Fourier transforms. During this developmental period, we demonstrate the timing of maturational changes in EEG background continuity, frequencies, and power and the emergence of identifiable wake, NREM, and REM sleep states. These results should serve as important control data for physiological studies of mouse models of normal brain development and neurological disease.
Collapse
Affiliation(s)
- Nicholas Rensing
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Brianna Moy
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joseph L. Friedman
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Rafael Galindo
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael Wong
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
39
|
Thalamus Controls Development and Expression of Arousal States in Visual Cortex. J Neurosci 2018; 38:8772-8786. [PMID: 30150360 DOI: 10.1523/jneurosci.1519-18.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/13/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Two major checkpoints of development in cerebral cortex are the acquisition of continuous spontaneous activity and the modulation of this activity by behavioral state. Despite the critical importance of these functions, the circuit mechanisms of their development remain unknown. Here we use the rodent visual system as a model to test the hypothesis that the locus of circuit change responsible for the developmental acquisition of continuity and state dependence measured in sensory cortex is relay thalamus, rather than the local cortical circuitry or the interconnectivity of the two structures. We conducted simultaneous recordings in the dorsal lateral geniculate nucleus (dLGN) and primary visual cortex (VC) of awake, head-fixed male and female rats using linear multielectrode arrays throughout early development. We find that activity in dLGN becomes continuous and positively correlated with movement (a measure of state dependence) on P13, the same day as VC, and that these properties are not dependent on VC activity. By contrast, silencing dLGN after P13 causes activity in VC to become discontinuous and movement to suppress, rather than augment, cortical firing, effectively reversing development. Thalamic bursting, a core characteristic of non-aroused states, emerged later, on P16, suggesting these processes are developmentally independent. Together our results indicate that cellular or circuit changes in relay thalamus are critical drivers for the maturation of background activity, which occurs around term in humans.SIGNIFICANCE STATEMENT The developing brain acquires two crucial features, continuous spontaneous activity and its modulation by arousal state, around term in humans and before the onset of sensory experience in rodents. This developmental transition in cortical activity, as measured by electroencephalogram (EEG), is an important milestone for normal brain development and indicates a good prognosis for babies born preterm and/or suffering brain damage such as hypoxic-ischemic encephalopathy. By using the awake rodent visual system as a model, we identify changes occurring at the level of relay thalamus, the major input to cortex, as the critical driver of EEG maturation. These results could help understand the circuit basis of human EEG development to improve diagnosis and treatment of infants in vulnerable situations.
Collapse
|
40
|
Colonnese MT, Phillips MA. Thalamocortical function in developing sensory circuits. Curr Opin Neurobiol 2018; 52:72-79. [PMID: 29715588 DOI: 10.1016/j.conb.2018.04.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022]
Abstract
Thalamocortical activity patterns, both spontaneous and evoked, undergo a dramatic shift in preparation for the onset of rich sensory experience (e.g. birth in humans; eye-opening in rodents). This change is the result of a switch from thalamocortical circuits tuned for transmission of spontaneous bursting in sense organs, to circuits capable of high resolution, active sensory processing. Early 'pre-sensory' tuning uses amplification generated by corticothalamic excitatory feedback and early-born subplate neurons to ensure transmission of bursts, at the expense of stimulus discrimination. The switch to sensory circuits is due, at least in part, to the coordinated remodeling of inhibitory circuits in thalamus and cortex. Appreciation of the distinct rules that govern early circuit function can, and should, inform translational studies of genetic and acquired developmental dysfunction.
Collapse
Affiliation(s)
- Matthew T Colonnese
- Department of Pharmacology and Physiology, Institute for Neuroscience, The George Washington University, United States.
| | - Marnie A Phillips
- Department of Pharmacology and Physiology, Institute for Neuroscience, The George Washington University, United States
| |
Collapse
|
41
|
Changes in brain morphology and microstructure in relation to early brain activity in extremely preterm infants. Pediatr Res 2018; 83:834-842. [PMID: 29244803 DOI: 10.1038/pr.2017.314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 11/27/2017] [Indexed: 01/18/2023]
Abstract
Background and ObjectiveTo investigate the relation of early brain activity with structural (growth of the cortex and cerebellum) and white matter microstructural brain development.MethodsA total of 33 preterm neonates (gestational age 26±1 weeks) without major brain abnormalities were continuously monitored with electroencephalography during the first 48 h of life. Rate of spontaneous activity transients per minute (SAT rate) and inter-SAT interval (ISI) in seconds per minute were calculated. Infants underwent brain magnetic resonance imaging ∼30 (mean 30.5; min: 29.3-max: 32.0) and 40 (41.1; 40.0-41.8) weeks of postmenstrual age. Increase in cerebellar volume, cortical gray matter volume, gyrification index, fractional anisotropy (FA) of posterior limb of the internal capsule, and corpus callosum (CC) were measured.ResultsSAT rate was positively associated with cerebellar growth (P=0.01), volumetric growth of the cortex (P=0.027), increase in gyrification (P=0.043), and increase in FA of the CC (P=0.037). ISI was negatively associated with cerebellar growth (P=0.002).ConclusionsIncreased early brain activity is associated with cerebellar and cortical growth structures with rapid development during preterm life. Higher brain activity is related to FA microstructural changes in the CC, a region responsible for interhemispheric connections. This study underlines the importance of brain activity for microstructural brain development.
Collapse
|
42
|
Khazipov R, Milh M. Early patterns of activity in the developing cortex: Focus on the sensorimotor system. Semin Cell Dev Biol 2018; 76:120-129. [DOI: 10.1016/j.semcdb.2017.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 02/08/2023]
|
43
|
Bennet L, Dhillon S, Lear CA, van den Heuij L, King V, Dean JM, Wassink G, Davidson JO, Gunn AJ. Chronic inflammation and impaired development of the preterm brain. J Reprod Immunol 2018; 125:45-55. [DOI: 10.1016/j.jri.2017.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 11/13/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
|
44
|
Kirischuk S, Sinning A, Blanquie O, Yang JW, Luhmann HJ, Kilb W. Modulation of Neocortical Development by Early Neuronal Activity: Physiology and Pathophysiology. Front Cell Neurosci 2017; 11:379. [PMID: 29238291 PMCID: PMC5712676 DOI: 10.3389/fncel.2017.00379] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/13/2017] [Indexed: 12/23/2022] Open
Abstract
Animal and human studies revealed that patterned neuronal activity is an inherent feature of developing nervous systems. This review summarizes our current knowledge about the mechanisms generating early electrical activity patterns and their impact on structural and functional development of the cerebral cortex. All neocortical areas display distinct spontaneous and sensory-driven neuronal activity patterns already at early phases of development. At embryonic stages, intermittent spontaneous activity is synchronized within small neuronal networks, becoming more complex with further development. This transition is accompanied by a gradual shift from electrical to chemical synaptic transmission, with a particular role of non-synaptic tonic currents before the onset of phasic synaptic activity. In this review article we first describe functional impacts of classical neurotransmitters (GABA, glutamate) and modulatory systems (e.g., acetylcholine, ACh) on early neuronal activities in the neocortex with special emphasis on electrical synapses, nonsynaptic and synaptic currents. Early neuronal activity influences probably all developmental processes and is crucial for the proper formation of neuronal circuits. In the second part of our review, we illustrate how specific activity patterns might interfere with distinct neurodevelopmental processes like proliferation, migration, axonal and dendritic sprouting, synapse formation and neurotransmitter specification. Finally, we present evidence that transient alterations in neuronal activity during restricted perinatal periods can lead to persistent changes in functional connectivity and therefore might underlie the manifestation of neurological and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anne Sinning
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Oriane Blanquie
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
45
|
Luhmann HJ, Khazipov R. Neuronal activity patterns in the developing barrel cortex. Neuroscience 2017; 368:256-267. [PMID: 28528963 DOI: 10.1016/j.neuroscience.2017.05.025] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 11/26/2022]
Abstract
The developing barrel cortex reveals a rich repertoire of neuronal activity patterns, which have been also found in other sensory neocortical areas and in other species including the somatosensory cortex of preterm human infants. The earliest stage is characterized by asynchronous, sparse single-cell firing at low frequencies. During the second stage neurons show correlated firing, which is initially mediated by electrical synapses and subsequently transforms into network bursts depending on chemical synapses. Activity patterns during this second stage are synchronous plateau assemblies, delta waves, spindle bursts and early gamma oscillations (EGOs). In newborn rodents spindle bursts and EGOs occur spontaneously or can be elicited by sensory stimulation and synchronize the activity in a barrel-related columnar network with topographic organization at the day of birth. Interfering with this early activity causes a disturbance in the development of the cortical architecture, indicating that spindle bursts and EGOs influence the formation of cortical columns. Early neuronal activity also controls the rate of programed cell death in the developing barrel cortex, suggesting that spindle bursts and EGOs are physiological activity patterns particularly suited to suppress apoptosis. It remains to be studied in more detail how these different neocortical activity patterns control early developmental processes such as formation of synapses, microcircuits, topographic maps and large-scale networks.
Collapse
Affiliation(s)
- Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Rustem Khazipov
- INMED - INSERM, Aix-Marseille University, Marseille 13273, France; Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
| |
Collapse
|
46
|
Lee JY, Xu K, Nguyen H, Guedes VA, Borlongan CV, Acosta SA. Stem Cell-Induced Biobridges as Possible Tools to Aid Neuroreconstruction after CNS Injury. Front Cell Dev Biol 2017; 5:51. [PMID: 28540289 PMCID: PMC5424542 DOI: 10.3389/fcell.2017.00051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Notch-induced mesenchymal stromal cells (MSCs) mediate a distinct mechanism of repair after brain injury by forming a biobridge that facilitates biodistribution of host cells from a neurogenic niche to the area of injury. We have observed the biobridge in an area between the subventricular zone and the injured cortex using immunohistochemistry and laser capture. Cells in the biobridge express high levels of extracellular matrix metalloproteinases (MMPs), specifically MMP-9, which co-localized with a trail of MSCs graft. The transplanted stem cells then become almost undetectable, being replaced by newly recruited host cells. This stem cell-paved biobridge provides support for distal migration of host cells from the subventricular zone to the site of injury. Biobridge formation by transplanted stem cells seems to have a fundamental role in initiating endogenous repair processes. Two major stem cell-mediated repair mechanisms have been proposed thus far: direct cell replacement by transplanted grafts and bystander effects through the secretion of trophic factors including fibroblast growth factor 2 (FGF-2), epidermal growth factor (EGF), stem cell factor (SCF), erythropoietin, and brain-derived neurotrophic factor (BDNF) among others. This groundbreaking observation of biobridge formation by transplanted stem cells represents a novel mechanism for stem cell mediated brain repair. Future studies on graft-host interaction will likely establish biobridge formation as a fundamental mechanism underlying therapeutic effects of stem cells and contribute to the scientific pursuit of developing safe and efficient therapies not only for traumatic brain injury but also for other neurological disorders. The aim of this review is to hypothetically extend concepts related to the formation of biobridges in other central nervous system disorders.
Collapse
Affiliation(s)
- Jea Y Lee
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Kaya Xu
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Hung Nguyen
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Vivian A Guedes
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Sandra A Acosta
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| |
Collapse
|
47
|
Mikhailova A, Sunkara N, McQuillen PS. Unbiased Quantification of Subplate Neuron Loss following Neonatal Hypoxia-Ischemia in a Rat Model. Dev Neurosci 2017; 39:171-181. [PMID: 28434006 DOI: 10.1159/000460815] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/08/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cellular targets of neonatal hypoxia-ischemia (HI) include both oligodendrocyte and neuronal lineages with differences in the patterns of vulnerable cells depending upon the developmental stage at which the injury occurs. Injury to the developing white matter is a characteristic feature of human preterm brain injury. Data are accumulating, however, for neuronal injury in the developing cerebral cortex. In the most widely used rodent model of preterm HI brain injury, conflicting data have been reported regarding the sensitivity of subplate neurons to early neonatal HI, with some reports of selective vulnerability and others that find no increased loss of subplate neurons in comparison with other cortical layers. Methods used to identify subplate neurons and quantify their numbers vary across studies. OBJECTIVE To use recently developed cortical layer-specific markers quantified with definitive stereologic methods to determine the magnitude and specificity of subplate neuron cell loss following neonatal HI in a rodent model. METHODS Postnatal day 2 (P2) rats underwent right common carotid artery coagulation followed by 2-3 h of hypoxia (5.6% oxygen). Categorically moderately injured brains were stained with subplate and cortical layer III-V markers (Complexin3 and Foxp1, respectively) at P8 and P21 (Foxp1 only). An Optical Fractionator was used to quantify subplate and middle/lower cortical neuronal numbers and these were compared across groups (naive control, hypoxia hemisphere, and HI hemisphere). RESULTS Following HI at P2 in rats, the total Complexin3-expressing subplate neuron number decreases significantly in the HI hemisphere compared with naive controls or hypoxia alone (HI vs. control 26,747 ± 7,952 vs. 35,468 ± 8,029, p = 0.04; HI vs. hypoxia, 26,747 ± 7,952 vs. 40,439 ± 7,363, p = 0.003). In contrast, the total Foxp1-expressing layer III-V cell number did not differ across the 3 conditions at P8 (HI vs. control 1,195,085 ± 436,609 vs. 1,234,640 ± 178,540, p = 0.19; HI vs. hypoxia, 1,195,085 ± 436,609 vs. 1,289,195 ± 468,941, p = 0.35) and at P21 (HI vs. control 1,265,190 ± 48,089 vs. 1,195,632 ± 26,912, p = 0.19; HI vs. hypoxia, 1,265,190 ± 48,089 vs. 1,309,563 ± 41,669, p = 0.49). CONCLUSIONS There is significant biological variability inherent in both the subplate neuron cell number and the pattern and severity of cortical injury following HI at P2 in rats. Despite this variability, the subplate neuron cell number is lower following P2 HI in animals with mild or moderate cortical injury, whereas the middle-to-lower-layer cortical neuronal number is unchanged. In more severe cases, neurons are lost from the lower cortical layers, suggesting a relative vulnerability of subplate neurons.
Collapse
Affiliation(s)
- Alexandra Mikhailova
- Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
48
|
Shi X, Xu L, Doycheva DM, Tang J, Yan M, Zhang JH. Sestrin2, as a negative feedback regulator of mTOR, provides neuroprotection by activation AMPK phosphorylation in neonatal hypoxic-ischemic encephalopathy in rat pups. J Cereb Blood Flow Metab 2017; 37:1447-1460. [PMID: 27381825 PMCID: PMC5453464 DOI: 10.1177/0271678x16656201] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hypoxic-ischemic encephalopathy is a condition caused by reduced oxygen and cerebral blood flow to the brain resulting in neurological impairments. Effective therapeutic treatments to ameliorate these disabilities are still lacking. We sought to investigate the role of sestrin2, a highly conserved stress-inducible protein, in a neonatal rat hypoxic-ischemic encephalopathy model. Ten-day-old rat pups underwent right common carotid artery ligation followed by 2.5 h hypoxia. At 1 h post hypoxic-ischemic encephalopathy, rats were intranasally administered with recombinant human sestrin2 and sacrificed for brain infarct area measurement, Fluoro-Jade C, immunofluorescence staining, Western blot, and neurological function testing. rh-sestrin2 reduced brain infarct area, brain atrophy, apoptosis, ventricular area enlargement, and improved neurological function. Western blot showed that sestrin2 expression levels were increased after treatment with rh-sestrin2, and sestrin2 exerts neuroprotective effects via activation of the adenosine monophosphate-activated protein kinase pathway which in turn inhibits mammalian target of rapamycin signaling resulting in the attenuation of apoptosis. In conclusions: Sestrin2 plays an important neuroprotective role after hypoxic-ischemic encephalopathy via adenosine monophosphate-activated protein kinase signaling pathway and serves as a negative feedback regulator of mammalian target of rapamycin. Administration of rh-sestrin2 not only reduced infarct area and brain atrophy, but also significantly improved neurological function.
Collapse
Affiliation(s)
- Xudan Shi
- 1 Department of Anesthesiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China.,2 Departments of Anesthesiology and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Liang Xu
- 2 Departments of Anesthesiology and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Desislava Met Doycheva
- 2 Departments of Anesthesiology and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- 2 Departments of Anesthesiology and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Min Yan
- 1 Department of Anesthesiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - John H Zhang
- 2 Departments of Anesthesiology and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
49
|
Whitehead K, Pressler R, Fabrizi L. Characteristics and clinical significance of delta brushes in the EEG of premature infants. Clin Neurophysiol Pract 2016; 2:12-18. [PMID: 30214965 PMCID: PMC6123866 DOI: 10.1016/j.cnp.2016.11.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/10/2016] [Accepted: 11/18/2016] [Indexed: 10/29/2022] Open
Abstract
Delta brushes are the hallmark of the EEG of premature infants. They are readily recognisable because of their characteristic appearance and are a key marker of neural maturation. However they are sometimes inconsistently described in the literature making identification of abnormalities challenging. The goal of this review is to provide an overview of research findings on this topic in the last five decades. Firstly, the characteristic features of delta brushes are described, including the developmental trajectory of their incidence and how they are modulated by vigilance state in normal neonates. Secondly, their clinical significance is discussed including how abnormalities in their incidence or appearance indicate particular pathophysiology. We propose that (i) the effect of age and vigilance state on the frequency, amplitude and topography of delta brushes, and (ii) heterogeneity within the cohorts of 'normal' premature infants studied, may explain the very variable descriptions of delta brush characteristics in the literature. By explicitly taking these factors into consideration to explain delta brush variability, the presented summary facilitates the clinical electrodiagnostic and prognostic use of delta brush abnormalities as a biomarker.
Collapse
Affiliation(s)
- Kimberley Whitehead
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Ronit Pressler
- Department of Clinical Neurophysiology, Great Ormond Street Hospital, London, United Kingdom.,Department of Clinical Neuroscience, UCL Great Ormond Street Institute for Child Health, London, United Kingdom
| | - Lorenzo Fabrizi
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
50
|
Sukhanova IA, Sebentsova EA, Levitskaya NG. The acute and delayed effects of perinatal hypoxic brain damage in children and in model experiments with rodents. NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416040127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|