1
|
Hui SC, Andescavage N, Limperopoulos C. The Role of Proton Magnetic Resonance Spectroscopy in Neonatal and Fetal Brain Research. J Magn Reson Imaging 2025; 61:2404-2424. [PMID: 39835523 PMCID: PMC12063769 DOI: 10.1002/jmri.29709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/22/2025] Open
Abstract
The biochemical composition and structure of the brain are in a rapid change during the exuberant stage of fetal and neonatal development. 1H-MRS is a noninvasive tool that can evaluate brain metabolites in healthy fetuses and infants as well as those with neurological diseases. This review aims to provide readers with an understanding of 1) the basic principles and technical considerations relevant to 1H-MRS in the fetal-neonatal brain and 2) the role of 1H-MRS in early fetal-neonatal development brain research. We performed a PubMed search to identify original studies using 1H-MRS in neonates and fetuses to establish the clinical applications of 1H-MRS. The eligible studies for this review included original research with 1H-MRS applications to the fetal-neonatal brain in healthy and high-risk conditions. We ran our search between 2000 and 2023, then added in several high-impact landmark publications from the 1990s. A total of 366 results appeared. After, we excluded original studies that did not include fetuses or neonates, non-proton MRS and non-neurological studies. Eventually, 110 studies were included in this literature review. Overall, the function of 1H-MRS in healthy fetal-neonatal brain studies focuses on measuring the change of metabolite concentrations during neurodevelopment and the physical properties of the metabolites such as T1/T2 relaxation times. For high-risk neonates, studies in very low birth weight preterm infants and full-term neonates with hypoxic-ischemic encephalopathy, along with examining the associations between brain biochemistry and cognitive neurodevelopment are most common. Additional high-risk conditions included infants with congenital heart disease or metabolic diseases, as well as fetuses of pregnant women with hypertensive disorders were of specific interest to researchers using 1H-MRS. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Steve C.N. Hui
- Developing Brain Institute, Children's National HospitalWashingtonD.C.USA
- Department of RadiologyThe George Washington University School of Medicine and Health SciencesWashingtonD.C.USA
- Department of PediatricsThe George Washington University School of Medicine and Health SciencesWashingtonD.C.USA
| | - Nickie Andescavage
- Developing Brain Institute, Children's National HospitalWashingtonD.C.USA
- Department of PediatricsThe George Washington University School of Medicine and Health SciencesWashingtonD.C.USA
- Division of NeonatologyChildren's National HospitalWashingtonD.C.USA
| | - Catherine Limperopoulos
- Developing Brain Institute, Children's National HospitalWashingtonD.C.USA
- Department of RadiologyThe George Washington University School of Medicine and Health SciencesWashingtonD.C.USA
- Department of PediatricsThe George Washington University School of Medicine and Health SciencesWashingtonD.C.USA
- Prenatal Pediatric Institute, Children's National HospitalWashingtonD.C.USA
| |
Collapse
|
2
|
Yu Y, Koyama Y, Shimada S. Development of the thermoregulatory mechanism - Raising the possibility that it is acquired at birth. Neuroscience 2025; 577:123-131. [PMID: 40345478 DOI: 10.1016/j.neuroscience.2025.04.044] [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: 11/25/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Whether the human thermoregulation mechanism in response to environmental temperature stimuli originates from learning or evolution remains an intriguing research question. Body temperature regulation depends not only on innate temperature sensation but also on acquired conditioning. Maintaining body temperature is essential for homeostasis, and the brain coordinates this process through a network of interconnected regulatory systems. In this review, we discuss how humans perceive temperature and establish thermoregulatory mechanisms at birth. We also propose an acquired connectivity structure perspective for the development of neonatal thermoregulatory mechanisms, particularly for brown adipose tissue thermogenesis. This perspective will enhance our understanding of the various acquired mechanisms of thermoregulation and adaptation to environmental temperature. Ultimately, this knowledge may contribute to the development of effective interventions for thermal balance disruptions, such as neonatal hypothermia.
Collapse
Affiliation(s)
- Yong Yu
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China; Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita 565-0871, Japan.
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
3
|
Champaud JLY, Asite S, Fabrizi L. Development of brain metastable dynamics during the equivalent of the third gestational trimester. Dev Cogn Neurosci 2025; 73:101556. [PMID: 40252359 PMCID: PMC12023897 DOI: 10.1016/j.dcn.2025.101556] [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: 11/28/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/21/2025] Open
Abstract
Metastability, a concept from dynamical systems theory, provides a framework for understanding how the brain shifts between various functional states and underpins essential cognitive, behavioural, and social function. While studied in adults, metastability in early brain development has only received recent attention. As the brain undergoes dramatic functional and structural changes over the third gestational trimester, here we review how these are reflected in changes in brain metastable dynamics in preterm, preterm at term-equivalent and full-term neonates. We synthesize findings from EEG, fMRI, fUS, and computational models, focusing on the spatial distribution and temporal dynamics of metastable states, which include functional integration and segregation, signal predictability and complexity. Despite fragmented evidence, studies suggest that neonatal metastability develops over the equivalent of the third gestational trimester, with increasing ability for integration-segregation, broader range of metastable states, faster metastable state transitions and greater signal complexity. Preterms at term-equivalent age exhibit immature metastability features compared to full-terms. We explain and interpret these changes in terms of maturation of the brain in a free energy landscape and establishment of cognitive functions.
Collapse
Affiliation(s)
- Juliette L Y Champaud
- Department of Neuroscience, Psychology and Pharmacology, University College London, UK; Centre for the Developing Brain, King's College London, UK
| | - Samanta Asite
- Department of Neuroscience, Psychology and Pharmacology, University College London, UK
| | - Lorenzo Fabrizi
- Department of Neuroscience, Psychology and Pharmacology, University College London, UK.
| |
Collapse
|
4
|
Nelson PM, Scheiber F, Demir-Lira ÖE, Harmon HM. Early medical risks to language development in extremely preterm infants. J Perinatol 2025; 45:378-385. [PMID: 39672899 DOI: 10.1038/s41372-024-02191-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/11/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE To study the mechanistic role of neonatal morbidities on language performance in extremely preterm (EP) infants. STUDY DESIGN We conducted secondary analyses on EP infants born at a single tertiary care center, investigating whether neonatal morbidities mediated associations between gestational age and language performance at 18-26 months corrected age. RESULTS Infants born at 25-26 weeks (n = 298) outperformed those born at 22-24 weeks (n = 177) in expressive communication, receptive communication, composite language, and cognition. Retinopathy of prematurity (ROP), grade 2/3 bronchopulmonary dysplasia (BPD), and cognition partially explained gestational age effects on expressive and receptive communication. In the final sequential path models, infants born 22-24 weeks gestation were more likely to be diagnosed with grade 2/3 BPD, which was linked to diminished cognitive skills, and reduced language skills. CONCLUSION Families of EP infants born under 25 weeks or with ROP and/or grade 2/3 BPD should be counseled about higher language impairment risk and receive proactive intervention.
Collapse
Affiliation(s)
- Paige M Nelson
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA.
| | - Francesca Scheiber
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Ö Ece Demir-Lira
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Heidi M Harmon
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
5
|
Wang D, Jiang Y, Jiang J, Pan Y, Yang Y, Fang X, Liang L, Li H, Dong Z, Fan S, Ma D, Zhang XS, Li H, He Y, Li N. Gut microbial GABA imbalance emerges as a metabolic signature in mild autism spectrum disorder linked to overrepresented Escherichia. Cell Rep Med 2025; 6:101919. [PMID: 39809266 PMCID: PMC11866488 DOI: 10.1016/j.xcrm.2024.101919] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 08/31/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
Gut microbiota (GM) alterations have been implicated in autism spectrum disorder (ASD), yet the specific functional architecture remains elusive. Here, employing multi-omics approaches, we investigate stool samples from two distinct cohorts comprising 203 children with mild ASD or typical development. In our screening cohort, regression-based analysis for metabolomic profiling identifies an elevated γ-aminobutyric acid (GABA) to glutamate (Glu) ratio as a metabolic signature of ASD, independent of age and gender. In the validating cohort, we affirm the GABA/Glu ratio as an ASD diagnostic indicator after adjusting for geography, age, gender, and specific food-consuming frequency. Integrated analysis of metabolomics, 16S rRNA sequencing, and metagenomics reveals a correlation between overrepresented Escherichia and disrupted GABA metabolism. Furthermore, we observe social behavioral impairments in weaning mice transplanted with E. coli, suggesting a potential link to ASD symptomatology. Collectively, these findings provide insights into potential diagnostic and therapeutic strategies aimed at evaluating and restoring gut microbial neurotransmitter homeostasis.
Collapse
Affiliation(s)
- Dilong Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Department of Pediatrics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Youheng Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Center for Digestive Disease, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jian Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yihang Pan
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Center for Digestive Disease, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yanming Yang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xiaoyi Fang
- Department of Neonatology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Liyang Liang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Hai Li
- Neurorehabilitation Laboratory, Department of Rehabilitation Medicine, Shenzhen Hospital, Shenzhen, Guangdong 518107, China
| | - Zepeng Dong
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Shilu Fan
- ARK Autism & Rehabilitation Institute, Taiyuan, Shanxi 030000, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, UK; Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA.
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, Division of Medicine, Faculty of Medical Sciences, University College London, London WC1E 6AE, UK.
| | - Yulong He
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Center for Digestive Disease, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China; China-UK Institute for Frontier Science, Shenzhen 518107, China.
| |
Collapse
|
6
|
Luotonen S, Railo H, Acosta H, Huotilainen M, Lavonius M, Karlsson L, Karlsson H, Tuulari JJ. Gestational Duration and Postnatal Age-Related Changes in Aperiodic and Periodic Parameters in Neonatal and Toddler Electroencephalogram (EEG). Hum Brain Mapp 2025; 46:e70130. [PMID: 39764646 PMCID: PMC11705402 DOI: 10.1002/hbm.70130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
The brain develops most rapidly during pregnancy and early neonatal months. While prior electrophysiological studies have shown that aperiodic brain activity undergoes changes across infancy to adulthood, the role of gestational duration in aperiodic and periodic activity remains unknown. In this study, we aimed to bridge this gap by examining the associations between gestational duration and aperiodic and periodic activity in the EEG power spectrum in both neonates and toddlers. This cross-sectional study involved EEG data from 73 neonates (postnatal age 1-5 days, 40 females) and 56 toddlers (postnatal age of 2.9-3.2 years, 28 females) from the FinnBrain Birth Cohort Study. EEG power spectra were parameterized to aperiodic and periodic components using the SpecParam tool. We tested the associations between gestational duration as well as postnatal age and SpecParam parameters in neonates and toddlers while including birth weight and child sex as covariates. For neonates, multilevel models were employed, considering different data acquisitions (sleep and auditory paradigm + sleep), while in toddlers, regression models were used as only data from the auditory paradigm was available. We found that longer gestational duration was associated with a steeper power spectrum across EEG frequencies both in neonates and toddlers. Effect was especially strong in toddlers (β = 0.45, p = 0.004), while in neonates, it remained nearly statistically significant (p = 0.061). In neonates, a quadratic association between gestational duration and beta center frequency (12.5-30 Hz) was found. In toddlers, beta center frequencies were overall higher in females compared to males. Offset (calculated as the power of the aperiodic curve at 2.5 Hz) and theta center frequency had negative associations with postnatal age in neonates, but not in toddlers. Our results suggest that gestational duration may have significant and relatively long-lasting effects on brain physiology. The possible behavioral and cognitive consequences of these changes are enticing topics for future research.
Collapse
Affiliation(s)
- Silja Luotonen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of Pediatric NeurologyTurku University HospitalTurkuFinland
| | - Henry Railo
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Department of Clinical NeurophysiologyUniversity of TurkuTurkuFinland
| | - Henriette Acosta
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Department of Psychiatry and PsychotherapyPhilipps University of MarburgMarburgGermany
| | - Minna Huotilainen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre of Excellence in Music, Mind, Body, and Brain, Faculty of Educational SciencesUniversity of HelsinkiHelsinkiFinland
- Cognitive Brain Research Unit, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Maria Lavonius
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of Clinical Medicine, Unit of Public HealthUniversity of TurkuTurkuFinland
- Department of Child PsychiatryTurku University HospitalTurkuFinland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of PsychiatryTurku University Hospital and University of TurkuTurkuFinland
| | - Jetro J. Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical MedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchTurku University Hospital and University of TurkuTurkuFinland
- Department of PsychiatryTurku University Hospital and University of TurkuTurkuFinland
- Turku Collegium for Science, Medicine and TechnologyUniversity of TurkuTurkuFinland
| |
Collapse
|
7
|
Jiao Y, Wang X, Yu A, Wu L, Li H. Multi-omics insights into beagle dog fed with a sucking-rewarded automatic feeding device. Front Pediatr 2024; 12:1467581. [PMID: 39670188 PMCID: PMC11634582 DOI: 10.3389/fped.2024.1467581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Background Facilitating the development of the sucking function in early stages of preterm infants holds substantial potential for influencing their long-term outcomes. To this end, our team has devised a sucking-rewarded automatic feeding device specifically tailored for preterm infants. The present study is designed to investigate the impacts of this innovative device, utilizing a multi-omics profiling approach, on beagle dogs as a surrogate model. Methods This study involved seven-day-old male newborn beagle puppies, carefully selected and matched in terms of body weights. The participants were stratified into two groups: the experimental group (AFG, sucking-rewarded feeding group) and the control group (PFG). After a 14-day intervention period, fecal and blood samples were systematically collected from each dog. The collected samples were then subjected to distinct profiling analyses, encompassing the assessment of gut microbial composition, plasma metabolic profiles, and proteomic expression profiles. Results The gut microbial data showed a significant difference between the AFG and PFG groups based on Bray-Curtis dissimilarity (P = 0.048), and the relative abundance of Lactobacillus was significantly more abundant in the AFG group compared to the PFG group. The significantly different metabolites between the two groups were enriched in functional metabolic pathways related to amino acids, fatty acid metabolism, and the nervous system. Notably, neurotransmitter L-glutamic acid was significantly up-regulated in the AFG group. Moreover, the significantly different proteins between the two groups were enriched in GO terms related to oxygen transport, oxygen binding, iron ion binding, hemoglobin complex, and heme binding. Among them, proteins A0A8C0MTD2, P60524, P60529 were significantly up-regulated in the AFG group. Notably, Lactobacillus, L-glutamic acid, A0A8C0MTD2, P60524, and P60529 were correlated with each other through correlation analysis, these molecules play important roles in the neural function and neurodevelopment. Conclusion Our investigation elucidated discernible modifications in gut microbial composition, plasma metabolic profiles, and proteomic expression patterns in beagle dogs subjected to the sucking-rewarded automatic feeding device.
Collapse
Affiliation(s)
- Yang Jiao
- Kashgar People’s Hospital, Kashgar Prefecture, Xinjiang Uygur Autonomous Region, Kashgar, China
| | - Xin Wang
- Neonatology Department, Affiliated Shenzhen Children's Hospital of Shantou University Medical College, Shenzhen, Guangdong, China
| | - Aizhen Yu
- Neonatology Department, Affiliated Shenzhen Children's Hospital of Shantou University Medical College, Shenzhen, Guangdong, China
| | - Li Wu
- Neonatology Department, Affiliated Shenzhen Children's Hospital of Shantou University Medical College, Shenzhen, Guangdong, China
| | - Hongping Li
- Neonatology Department, Affiliated Shenzhen Children's Hospital of Shantou University Medical College, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
de Groot ER, Dudink J, Austin T. Sleep as a driver of pre- and postnatal brain development. Pediatr Res 2024; 96:1503-1509. [PMID: 38956219 PMCID: PMC11624135 DOI: 10.1038/s41390-024-03371-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
In 1966, Howard Roffwarg proposed the ontogenic sleep hypothesis, relating neural plasticity and development to rapid eye movement (REM) sleep, a hypothesis that current fetal and neonatal sleep research is still exploring. Recently, technological advances have enabled researchers to automatically quantify neonatal sleep architecture, which has caused a resurgence of research in this field as attempts are made to further elucidate the important role of sleep in pre- and postnatal brain development. This article will review our current understanding of the role of sleep as a driver of brain development and identify possible areas for future research. IMPACT: The evidence to date suggests that Roffwarg's ontogenesis hypothesis of sleep and brain development is correct. A better understanding of the relationship between sleep and the development of functional connectivity is needed. Reliable, non-invasive tools to assess sleep in the NICU and at home need to be tested in a real-world environment and the best way to promote healthy sleep needs to be understood before clinical trials promoting and optimizing sleep quality in neonates could be undertaken.
Collapse
Affiliation(s)
- Eline R de Groot
- 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
- Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Topun Austin
- NeoLab, Evelyn Perinatal Imaging Centre, The Rosie Hospital, Cambridge University Hospitals, Cambridge, UK.
| |
Collapse
|
9
|
Zhang M, Chen Y, Xu T, Jiang J, Zhang D, Huang H, Kurth CD, Yuan I, Wang R, Liu J, Zhu T, Zhou C. γ-Aminobutyric Acid-Ergic Development Contributes to the Enhancement of Electroencephalogram Slow-Delta Oscillations Under Volatile Anesthesia in Neonatal Rats. Anesth Analg 2024; 138:198-209. [PMID: 36753442 DOI: 10.1213/ane.0000000000006396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
BACKGROUND General anesthetics (eg, propofol and volatile anesthetics) enhance the slow-delta oscillations of the cortical electroencephalogram (EEG), which partly results from the enhancement of (γ-aminobutyric acid [GABA]) γ-aminobutyric acid-ergic (GABAergic) transmission. There is a GABAergic excitatory-inhibitory shift during postnatal development. Whether general anesthetics can enhance slow-delta oscillations in the immature brain has not yet been unequivocally determined. METHODS Perforated patch-clamp recording was used to confirm the reversal potential of GABAergic currents throughout GABAergic development in acute brain slices of neonatal rats. The power density of the electrocorticogram and the minimum alveolar concentrations (MAC) of isoflurane and/or sevoflurane were measured in P4-P21 rats. Then, the effects of bumetanide, an inhibitor of the Na + -K + -2Cl - cotransporter (NKCC1) and K + -Cl - cotransporter (KCC2) knockdown on the potency of volatile anesthetics and the power density of the EEG were determined in vivo. RESULTS Reversal potential of GABAergic currents were gradually hyperpolarized from P4 to P21 in cortical pyramidal neurons. Bumetanide enhanced the hypnotic effects of volatile anesthetics at P5 (for MAC LORR , isoflurane: 0.63% ± 0.07% vs 0.81% ± 0.05%, 95% confidence interval [CI], -0.257 to -0.103, P < .001; sevoflurane: 1.46% ± 0.12% vs 1.66% ± 0.09%, 95% CI, -0.319 to -0.081, P < .001); while knockdown of KCC2 weakened their hypnotic effects at P21 in rats (for MAC LORR , isoflurane: 0.58% ± 0.05% to 0.77% ± 0.20%, 95% CI, 0.013-0.357, P = .003; sevoflurane: 1.17% ± 0.04% to 1.33% ± 0.04%, 95% CI, 0.078-0.244, P < .001). For cortical EEG, slow-delta oscillations were the predominant components of the EEG spectrum in neonatal rats. Isoflurane and/or sevoflurane suppressed the power density of slow-delta oscillations rather than enhancement of it until GABAergic maturity. Enhancement of slow-delta oscillations under volatile anesthetics was simulated by preinjection of bumetanide at P5 (isoflurane: slow-delta changed ratio from -0.31 ± 0.22 to 1.57 ± 1.15, 95% CI, 0.67-3.08, P = .007; sevoflurane: slow-delta changed ratio from -0.46 ± 0.25 to 0.95 ± 0.97, 95% CI, 0.38-2.45, P = .014); and suppressed by KCC2-siRNA at P21 (isoflurane: slow-delta changed ratio from 16.13 ± 5.69 to 3.98 ± 2.35, 95% CI, -18.50 to -5.80, P = .002; sevoflurane: slow-delta changed ratio from 0.13 ± 2.82 to 3.23 ± 2.49, 95% CI, 3.02-10.79, P = .003). CONCLUSIONS Enhancement of cortical EEG slow-delta oscillations by volatile anesthetics may require mature GABAergic inhibitory transmission during neonatal development.
Collapse
Affiliation(s)
- Mingyue Zhang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yali Chen
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Ting Xu
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingyao Jiang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Donghang Zhang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, West China Second Hospital of Sichuan University, Chengdu, China
| | - Charles D Kurth
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ian Yuan
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Rurong Wang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jin Liu
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Zhu
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Dinh J, Johnson TN, Grimstein M, Lewis T. Physiologically Based Pharmacokinetics Modeling in the Neonatal Population-Current Advances, Challenges, and Opportunities. Pharmaceutics 2023; 15:2579. [PMID: 38004559 PMCID: PMC10675397 DOI: 10.3390/pharmaceutics15112579] [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: 09/26/2023] [Revised: 10/24/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is an approach to predicting drug pharmacokinetics, using knowledge of the human physiology involved and drug physiochemical properties. This approach is useful when predicting drug pharmacokinetics in under-studied populations, such as pediatrics. PBPK modeling is a particularly important tool for dose optimization for the neonatal population, given that clinical trials rarely include this patient population. However, important knowledge gaps exist for neonates, resulting in uncertainty with the model predictions. This review aims to outline the sources of variability that should be considered with developing a neonatal PBPK model, the data that are currently available for the neonatal ontogeny, and lastly to highlight the data gaps where further research would be needed.
Collapse
Affiliation(s)
- Jean Dinh
- Certara UK Limited, Sheffield S1 2BJ, UK; (J.D.); (T.N.J.)
| | | | - Manuela Grimstein
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Tamorah Lewis
- Pediatric Clinical Pharmacology & Toxicology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
11
|
Basu SK, Pradhan S, Sharker YM, Kapse KJ, Murnick J, Chang T, Lopez CA, Andescavage N, duPlessis AJ, Limperopoulos C. Severity of prematurity and age impact early postnatal development of GABA and glutamate systems. Cereb Cortex 2023; 33:7386-7394. [PMID: 36843135 PMCID: PMC10267637 DOI: 10.1093/cercor/bhad046] [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: 07/02/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/28/2023] Open
Abstract
Gamma-aminobutyric acid (GABA) and glutamatergic system perturbations following premature birth may explain neurodevelopmental deficits in the absence of structural brain injury. Using GABA-edited spectroscopy (MEscher-GArwood Point Resolved Spectroscopy [MEGA-PRESS] on 3 T MRI), we have described in-vivo brain GABA+ (+macromolecules) and Glx (glutamate + glutamine) concentrations in term-born infants. We report previously unavailable comparative data on in-vivo GABA+ and Glx concentrations in the cerebellum, the right basal ganglia, and the right frontal lobe of preterm-born infants without structural brain injury. Seventy-five preterm-born (gestational age 27.8 ± 2.9 weeks) and 48 term-born (39.6 ± 0.9 weeks) infants yielded reliable MEGA-PRESS spectra acquired at post-menstrual age (PMA) of 40.2 ± 2.3 and 43.0 ± 2 weeks, respectively. GABA+ (median 2.44 institutional units [i.u.]) concentrations were highest in the cerebellum and Glx higher in the cerebellum (5.73 i.u.) and basal ganglia (5.16 i.u.), with lowest concentrations in the frontal lobe. Metabolite concentrations correlated positively with advancing PMA and postnatal age at MRI (Spearman's rho 0.2-0.6). Basal ganglia Glx and NAA, and frontal GABA+ and NAA concentrations were lower in preterm compared with term infants. Moderate preterm infants had lower metabolite concentrations than term and extreme preterm infants. Our findings emphasize the impact of premature extra-uterine stimuli on GABA-glutamate system development and may serve as early biomarkers of neurodevelopmental deficits.
Collapse
Affiliation(s)
- Sudeepta K Basu
- Neonatology, Children’s National Hospital, Washington, D.C., United States
- Developing Brain Institute, Children’s National Hospital, Washington, D.C. 20010, United States
- The George Washington University School of Medicine, Washington, D.C. 20037, United States
| | - Subechhya Pradhan
- Developing Brain Institute, Children’s National Hospital, Washington, D.C. 20010, United States
- The George Washington University School of Medicine, Washington, D.C. 20037, United States
| | - Yushuf M Sharker
- Developing Brain Institute, Children’s National Hospital, Washington, D.C. 20010, United States
| | - Kushal J Kapse
- Developing Brain Institute, Children’s National Hospital, Washington, D.C. 20010, United States
| | - Jonathan Murnick
- The George Washington University School of Medicine, Washington, D.C. 20037, United States
- Division of Diagnostic Imaging and Radiology, Children’s National Hospital, Washington, D.C. 20010, United States
| | - Taeun Chang
- The George Washington University School of Medicine, Washington, D.C. 20037, United States
- Division of Neurology, Children’s National Hospital, Washington, D.C. 20010, United States
| | - Catherine A Lopez
- Developing Brain Institute, Children’s National Hospital, Washington, D.C. 20010, United States
| | - Nickie Andescavage
- Neonatology, Children’s National Hospital, Washington, D.C., United States
- Developing Brain Institute, Children’s National Hospital, Washington, D.C. 20010, United States
- The George Washington University School of Medicine, Washington, D.C. 20037, United States
- Perinatal Pediatrics institute, Children’s National Hospital, Washington, D.C. 20010, United States
| | - Adre J duPlessis
- The George Washington University School of Medicine, Washington, D.C. 20037, United States
- Division of Neurology, Children’s National Hospital, Washington, D.C. 20010, United States
- Perinatal Pediatrics institute, Children’s National Hospital, Washington, D.C. 20010, United States
| | - Catherine Limperopoulos
- Developing Brain Institute, Children’s National Hospital, Washington, D.C. 20010, United States
- The George Washington University School of Medicine, Washington, D.C. 20037, United States
- Division of Diagnostic Imaging and Radiology, Children’s National Hospital, Washington, D.C. 20010, United States
| |
Collapse
|
12
|
Xiao T, Dong X, Lu Y, Zhou W. High-Resolution and Multidimensional Phenotypes Can Complement Genomics Data to Diagnose Diseases in the Neonatal Population. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:204-215. [PMID: 37197647 PMCID: PMC10110825 DOI: 10.1007/s43657-022-00071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 05/19/2023]
Abstract
Advances in genomic medicine have greatly improved our understanding of human diseases. However, phenome is not well understood. High-resolution and multidimensional phenotypes have shed light on the mechanisms underlying neonatal diseases in greater details and have the potential to optimize clinical strategies. In this review, we first highlight the value of analyzing traditional phenotypes using a data science approach in the neonatal population. We then discuss recent research on high-resolution, multidimensional, and structured phenotypes in neonatal critical diseases. Finally, we briefly introduce current technologies available for the analysis of multidimensional data and the value that can be provided by integrating these data into clinical practice. In summary, a time series of multidimensional phenome can improve our understanding of disease mechanisms and diagnostic decision-making, stratify patients, and provide clinicians with optimized strategies for therapeutic intervention; however, the available technologies for collecting multidimensional data and the best platform for connecting multiple modalities should be considered.
Collapse
Affiliation(s)
- Tiantian Xiao
- Division of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai, 201102 China
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000 China
| | - Xinran Dong
- Center for Molecular Medicine, Pediatric Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, 201102 China
| | - Yulan Lu
- Center for Molecular Medicine, Pediatric Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, 201102 China
| | - Wenhao Zhou
- Division of Neonatology, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai, 201102 China
- Center for Molecular Medicine, Pediatric Research Institute, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, 201102 China
| |
Collapse
|
13
|
Ren Q, Wan B, Luo X, Liu Q, Gong H, Li H, Luo M, Xu D, Liu P, Wang J, Yin Z, Li X. Glutamate alterations in the premature infant brain during different gestational ages with glutamate chemical exchange saturation transfer imaging: a pilot study. Eur Radiol 2023; 33:4214-4222. [PMID: 36600123 DOI: 10.1007/s00330-022-09374-2] [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: 08/05/2022] [Revised: 11/02/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVES To elucidate the change in glutamate levels in preterm infants at different gestational ages by glutamate chemical exchange saturated transfer (GluCEST) magnetic resonance imaging and to compare the difference in glutamate levels among different brain regions between very early preterm infants and middle and late preterm infants. METHODS Fifty-three preterm infants (59% males; median gestational age = 33.6 weeks) underwent MRI, including conventional MRI and GluCEST. The original data were postprocessed in MATLAB. Correlation analysis was used to determine the relationship between the MTRasym and gestational age. The differences in MTRasym signals among different ROIs were statistically analysed by one-way analysis of variance (ANOVA). The MTRasym difference of the bilateral hemispherical ROI was compared by a paired T test. RESULTS In all ROIs, glutamate concentration was positively correlated with gestational age. The glutamate concentration in the thalamus was higher than that in the frontal lobe in very early, middle and late preterm infants. A difference in glutamate concentration was not found in the bilateral ROIs. CONCLUSIONS The concentration of glutamate in the brains of preterm infants of different gestational ages increased with gestational age, which may be one of the factors contributing to the higher incidence of neurodevelopmental dysfunction in very early preterm infants compared to that in middle and late preterm infants. Meanwhile, the glutamate concentrations among different brain regions were also diverse. KEY POINTS • The glutamate concentration was positively correlated with gestational age in preterm infants of the brain. • Glutamate concentrations were dissimilar in different brain regions of preterm infants. • Glutamate concentration during the process of brain development in premature infants was not found to be asymmetric.
Collapse
Affiliation(s)
- Qingfa Ren
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Laishan District, Yantai, 264003, China
| | - Bin Wan
- Neonatal Intensive Care Unit, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Bincheng District, Binzhou, 256600, China
| | - Xunrong Luo
- Department of Radiology, Affiliated Cancer Hospital of Chongqing University, No. 181 Hanyu Road, Shapingba District, Chongqing, 400016, China
| | - Quanyuan Liu
- Department of Radiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Bincheng District, Binzhou, 256600, China
| | - He Gong
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Laishan District, Yantai, 264003, China
| | - Hao Li
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Laishan District, Yantai, 264003, China
| | - Mingfang Luo
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Qingyang District, Chengdu, 610072, China
| | - Donghao Xu
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Laishan District, Yantai, 264003, China
| | - Pan Liu
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Laishan District, Yantai, 264003, China
| | - Jing Wang
- Department of Radiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Bincheng District, Binzhou, 256600, China.
| | - Zhijie Yin
- Department of Radiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Bincheng District, Binzhou, 256600, China.
| | - Xianglin Li
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Laishan District, Yantai, 264003, China.
| |
Collapse
|
14
|
Dimitroglou M, Iliodromiti Z, Christou E, Volaki P, Petropoulou C, Sokou R, Boutsikou T, Iacovidou N. Human Breast Milk: The Key Role in the Maturation of Immune, Gastrointestinal and Central Nervous Systems: A Narrative Review. Diagnostics (Basel) 2022; 12:diagnostics12092208. [PMID: 36140609 PMCID: PMC9498242 DOI: 10.3390/diagnostics12092208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/22/2022] Open
Abstract
Premature birth is a major cause of mortality and morbidity in the pediatric population. Because their immune, gastrointestinal and nervous systems are not fully developed, preterm infants (<37 weeks of gestation) and especially very preterm infants (VPIs, <32 weeks of gestation) are more prone to infectious diseases, tissue damage and future neurodevelopmental impairment. The aim of this narrative review is to report the immaturity of VPI systems and examine the role of Human Breast Milk (HBM) in their development and protection against infectious diseases, inflammation and tissue damage. For this purpose, we searched and synthesized the data from the existing literature published in the English language. Studies revealed the significance of HBM and indicate HBM as the best dietary choice for VPIs.
Collapse
|
15
|
Purushotham SS, Reddy NMN, D'Souza MN, Choudhury NR, Ganguly A, Gopalakrishna N, Muddashetty R, Clement JP. A perspective on molecular signalling dysfunction, its clinical relevance and therapeutics in autism spectrum disorder. Exp Brain Res 2022; 240:2525-2567. [PMID: 36063192 DOI: 10.1007/s00221-022-06448-x] [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: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022]
Abstract
Intellectual disability (ID) and autism spectrum disorder (ASD) are neurodevelopmental disorders that have become a primary clinical and social concern, with a prevalence of 2-3% in the population. Neuronal function and behaviour undergo significant malleability during the critical period of development that is found to be impaired in ID/ASD. Human genome sequencing studies have revealed many genetic variations associated with ASD/ID that are further verified by many approaches, including many mouse and other models. These models have facilitated the identification of fundamental mechanisms underlying the pathogenesis of ASD/ID, and several studies have proposed converging molecular pathways in ASD/ID. However, linking the mechanisms of the pathogenic genes and their molecular characteristics that lead to ID/ASD has progressed slowly, hampering the development of potential therapeutic strategies. This review discusses the possibility of recognising the common molecular causes for most ASD/ID based on studies from the available models that may enable a better therapeutic strategy to treat ID/ASD. We also reviewed the potential biomarkers to detect ASD/ID at early stages that may aid in diagnosis and initiating medical treatment, the concerns with drug failure in clinical trials, and developing therapeutic strategies that can be applied beyond a particular mutation associated with ASD/ID.
Collapse
Affiliation(s)
- Sushmitha S Purushotham
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Neeharika M N Reddy
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Michelle Ninochka D'Souza
- Centre for Brain Research, Indian Institute of Science Campus, CV Raman Avenue, Bangalore, 560 012, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - Nilpawan Roy Choudhury
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Anusa Ganguly
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Niharika Gopalakrishna
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Ravi Muddashetty
- Centre for Brain Research, Indian Institute of Science Campus, CV Raman Avenue, Bangalore, 560 012, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India.
| |
Collapse
|
16
|
Polese D, Riccio ML, Fagioli M, Mazzetta A, Fagioli F, Parisi P, Fagioli M. The Newborn's Reaction to Light as the Determinant of the Brain's Activation at Human Birth. Front Integr Neurosci 2022; 16:933426. [PMID: 36118115 PMCID: PMC9478760 DOI: 10.3389/fnint.2022.933426] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Developmental neuroscience research has not yet fully unveiled the dynamics involved in human birth. The trigger of the first breath, often assumed to be the marker of human life, has not been characterized nor has the process entailing brain modification and activation at birth been clarified yet. To date, few researchers only have investigated the impact of the extrauterine environment, with its strong stimuli, on birth. This ‘hypothesis and theory' article assumes the role of a specific stimulus activating the central nervous system (CNS) at human birth. This stimulus must have specific features though, such as novelty, efficacy, ubiquity, and immediacy. We propose light as a robust candidate for the CNS activation via the retina. Available data on fetal and neonatal neurodevelopment, in particular with reference to retinal light-responsive pathways, will be examined together with the GABA functional switch, and the subplate disappearance, which, at an experimental level, differentiate the neonatal brain from the fetal brain. In this study, we assume how a very rapid activation of retinal photoreceptors at birth initiates a sudden brain shift from the prenatal pattern of functions to the neonatal setup. Our assumption implies the presence of a photoreceptor capable of capturing and transducing light/photon stimulus, transforming it into an effective signal for the activation of new brain functions at birth. Opsin photoreception or, more specifically, melanopsin-dependent photoreception, which is provided by intrinsically photosensitive retinal ganglion cells (ipRGCs), is considered as a valid candidate. Although what is assumed herein cannot be verified in humans based on knowledge available so far, proposing an important and novel function can trigger a broad range of diversified research in different domains, from neurophysiology to neurology and psychiatry.
Collapse
Affiliation(s)
- Daniela Polese
- PhD Program on Sensorineural Plasticity, Department of Neuroscience, Mental Health and Sensory Organs NESMOS, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
- *Correspondence: Daniela Polese
| | | | - Marcella Fagioli
- Department of Mental Health, National Health System ASL Rome 1, Rome, Italy
| | - Alessandro Mazzetta
- PhD Program on Neuroscience, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Francesca Fagioli
- Department of Mental Health, National Health System ASL Rome 1, Rome, Italy
| | - Pasquale Parisi
- Chair of Pediatrics, Department of Neuroscience, Mental Health and Sensory Organs NESMOS, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
17
|
Shyu C, Chavez S, Boileau I, Foll BL. Quantifying GABA in Addiction: A Review of Proton Magnetic Resonance Spectroscopy Studies. Brain Sci 2022; 12:918. [PMID: 35884725 PMCID: PMC9316447 DOI: 10.3390/brainsci12070918] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 02/04/2023] Open
Abstract
Gamma-aminobutyric acid (GABA) signaling plays a crucial role in drug reward and the development of addiction. Historically, GABA neurochemistry in humans has been difficult to study due to methodological limitations. In recent years, proton magnetic resonance spectroscopy (1H-MRS, MRS) has emerged as a non-invasive imaging technique that can detect and quantify human brain metabolites in vivo. Novel sequencing and spectral editing methods have since been developed to allow for quantification of GABA. This review outlines the clinical research utilization of 1H-MRS in understanding GABA neurochemistry in addiction and summarizes current literature that reports GABA measurements by MRS in addiction. Research on alcohol, nicotine, cocaine, and cannabis addiction all suggest medications that modulate GABA signaling may be effective in reducing withdrawal, craving, and other addictive behaviors. Thus, we discuss how improvements in current MRS techniques and design can optimize GABA quantification in future studies and explore how monitoring changes to brain GABA could help identify risk factors, improve treatment efficacy, further characterize the nature of addiction, and provide crucial insights for future pharmacological development.
Collapse
Affiliation(s)
- Claire Shyu
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON M5S 2S1, Canada;
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sofia Chavez
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON M5S 2S1, Canada;
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, ON M5T 1R8, Canada
- Centre for Addiction and Mental Health, Concurrent Outpatient Medical & Psychosocial Addiction Support Services, Toronto, ON M6J 1H4, Canada
- Centre for Addiction and Mental Health, Acute Care Program, Toronto, ON M6J 1H3, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Waypoint Centre for Mental Health Care, Waypoint Research Institute, 500 Church Street, Penetanguishene, ON L9M 1G3, Canada
| |
Collapse
|
18
|
Cavaliere S, Lori S, Bastianelli M, Cossu C, Gabbanini S, Dani C, Bertini G. Unilateral Transient Enhanced SEP during Integrated Multiparameter Neurophysiological Monitoring in a Newborn with Symptomatic Seizure. Pediatr Rep 2022; 14:254-261. [PMID: 35736655 PMCID: PMC9230835 DOI: 10.3390/pediatric14020033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/05/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
During Integrated Multiparametric Neurophysiological Monitoring (IMNA), a newborn with suspected hypoxia at birth and microhaemorrhagic and ischaemic lesions presented some clonic-tonic episodes with specific EEG patterns characterized by rolandic and temporal spikes and the appearance of a unilateral enhanced Somatosensory Evoked Potential (SEP) (10.45 µv). Since the literature does not seem to describe cases of giant SEP in newborns, in this case report, we will discuss the hypotheses underlying this potential. It could be assumed that the ischaemic and haemorrhagic lesions presented by the newborn may have developed as a result of neurotransmitter balance failure. This may be the origin of the EEG picture, which, consequently, could have triggered a potential with high amplitude.
Collapse
Affiliation(s)
- Sara Cavaliere
- Department of Neurosciences, Psychology, Drug Research and Children’s Health, University of Florence, 50134 Florence, Italy; (S.C.); (C.D.)
| | - Silvia Lori
- Neurophysiology Unit, Neuro-Musculo-Skeletal Department, Careggi University Hospital, 50134 Florence, Italy; (S.L.); (M.B.); (C.C.); (S.G.)
| | - Maria Bastianelli
- Neurophysiology Unit, Neuro-Musculo-Skeletal Department, Careggi University Hospital, 50134 Florence, Italy; (S.L.); (M.B.); (C.C.); (S.G.)
| | - Cesarina Cossu
- Neurophysiology Unit, Neuro-Musculo-Skeletal Department, Careggi University Hospital, 50134 Florence, Italy; (S.L.); (M.B.); (C.C.); (S.G.)
| | - Simonetta Gabbanini
- Neurophysiology Unit, Neuro-Musculo-Skeletal Department, Careggi University Hospital, 50134 Florence, Italy; (S.L.); (M.B.); (C.C.); (S.G.)
| | - Carlo Dani
- Department of Neurosciences, Psychology, Drug Research and Children’s Health, University of Florence, 50134 Florence, Italy; (S.C.); (C.D.)
| | - Giovanna Bertini
- Department of Neurosciences, Psychology, Drug Research and Children’s Health, University of Florence, 50134 Florence, Italy; (S.C.); (C.D.)
- Correspondence:
| |
Collapse
|
19
|
Neonatal Oxidative Stress Impairs Cortical Synapse Formation and GABA Homeostasis in Parvalbumin-Expressing Interneurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8469756. [PMID: 35663195 PMCID: PMC9159830 DOI: 10.1155/2022/8469756] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/08/2022] [Indexed: 11/28/2022]
Abstract
Neonatal brain injury is often caused by preterm birth. Brain development is vulnerable to increased environmental stress, including oxidative stress challenges. Due to a premature change of the fetal living environment from low oxygen in utero into postnatal high-oxygen room air conditions ex utero, the immature preterm brain is exposed to a relative hyperoxia, which can induce oxidative stress and impair neuronal cell development. To simulate the drastic increase of oxygen exposure in the immature brain, 5-day-old C57BL/6 mice were exposed to hyperoxia (80% oxygen) for 48 hours or kept in room air (normoxia, 21% oxygen) and mice were analyzed for maturational alterations of cortical GABAergic interneurons. As a result, oxidative stress was indicated by elevated tyrosine nitration of proteins. We found perturbation of perineuronal net formation in line with decreased density of parvalbumin-expressing (PVALB) cortical interneurons in hyperoxic mice. Moreover, maturational deficits of cortical PVALB+ interneurons were obtained by decreased glutamate decarboxylase 67 (GAD67) protein expression in Western blot analysis and lower gamma-aminobutyric acid (GABA) fluorescence intensity in immunostaining. Hyperoxia-induced oxidative stress affected cortical synaptogenesis by decreasing synapsin 1, synapsin 2, and synaptophysin expression. Developmental delay of synaptic marker expression was demonstrated together with decreased PI3K-signaling as a pathway being involved in synaptogenesis. These results elucidate that neonatal oxidative stress caused by increased oxygen exposure can lead to GABAergic interneuron damage which may serve as an explanation for the high incidence of psychiatric and behavioral alterations found in preterm infants.
Collapse
|
20
|
Mohammadian F, Golitabari N, Abedi A, Saadati H, Milan HS, Salari AA, Amani M. Early life GABA A blockade alters the synaptic plasticity and cognitive functions in male and female rats. Eur J Pharmacol 2022; 925:174992. [PMID: 35513017 DOI: 10.1016/j.ejphar.2022.174992] [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: 01/17/2022] [Revised: 04/14/2022] [Accepted: 04/28/2022] [Indexed: 11/03/2022]
Abstract
Gamma-aminobutyric acid (GABA), the major inhibitory neurotransmitter in adults, has a critical contribution to balanced excitatory-inhibitory networks in the brain. Alteration in depolarizing action of GABA during early life is connected to a wide variety of neurodevelopmental disorders. Additionally, the effects of postnatal GABA blockade on neuronal synaptic plasticity are not known and therefore, we set out to determine whether postnatal exposure to bicuculline, a competitive antagonist of GABAA receptors, affects electrophysiologic changes in hippocampal CA1 neurons later on. To this end, male and female Wistar rats received vehicle or bicuculline (300 μg/kg) on postnatal days (PNDs) 7, 9 and 11, and then underwent different behavioral and electrophysiological examinations in adulthood. Postnatal exposure to bicuculline did not affect basic synaptic transmission but led to a pronounced decrease in paired-pulse facilitation (PPF) in CA1 pyramidal neurons. Bicuculline treatment also attenuated the long-term potentiation (LTP) and long-term depression (LTD) of CA1 neurons accompanied by decreased theta-burst responses in male and female adult rats. These electrophysiology findings together with the reduced brain-derived neurotrophic factor (BDNF) levels in the hippocampus and prefrontal cortex reliably explain the disturbance in spatial reference and working memories of bicuculline-treated animals. This study suggests that postnatal GABAA blockade deteriorates short- and long-term synaptic plasticity of hippocampal CA1 neurons and related encoding of spatial memory in adulthood.
Collapse
Affiliation(s)
- Forouzan Mohammadian
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nastaran Golitabari
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Abedi
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hakimeh Saadati
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Ali-Akbar Salari
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran; Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mohammad Amani
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
21
|
Montanari M, Martella G, Bonsi P, Meringolo M. Autism Spectrum Disorder: Focus on Glutamatergic Neurotransmission. Int J Mol Sci 2022; 23:ijms23073861. [PMID: 35409220 PMCID: PMC8998955 DOI: 10.3390/ijms23073861] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Disturbances in the glutamatergic system have been increasingly documented in several neuropsychiatric disorders, including autism spectrum disorder (ASD). Glutamate-centered theories of ASD are based on evidence from patient samples and postmortem studies, as well as from studies documenting abnormalities in glutamatergic gene expression and metabolic pathways, including changes in the gut microbiota glutamate metabolism in patients with ASD. In addition, preclinical studies on animal models have demonstrated glutamatergic neurotransmission deficits and altered expression of glutamate synaptic proteins. At present, there are no approved glutamatergic drugs for ASD, but several ongoing clinical trials are currently focusing on evaluating in autistic patients glutamatergic pharmaceuticals already approved for other conditions. In this review, we provide an overview of the literature concerning the role of glutamatergic neurotransmission in the pathophysiology of ASD and as a potential target for novel treatments.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Department of Systems Neuroscience, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| |
Collapse
|
22
|
McGrath T, Baskerville R, Rogero M, Castell L. Emerging Evidence for the Widespread Role of Glutamatergic Dysfunction in Neuropsychiatric Diseases. Nutrients 2022; 14:nu14050917. [PMID: 35267893 PMCID: PMC8912368 DOI: 10.3390/nu14050917] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
The monoamine model of depression has long formed the basis of drug development but fails to explain treatment resistance or associations with stress or inflammation. Recent animal research, clinical trials of ketamine (a glutamate receptor antagonist), neuroimaging research, and microbiome studies provide increasing evidence of glutamatergic dysfunction in depression and other disorders. Glutamatergic involvement across diverse neuropathologies including psychoses, neurodevelopmental, neurodegenerative conditions, and brain injury forms the rationale for this review. Glutamate is the brain's principal excitatory neurotransmitter (NT), a metabolic and synthesis substrate, and an immune mediator. These overlapping roles and multiple glutamate NT receptor types complicate research into glutamate neurotransmission. The glutamate microcircuit comprises excitatory glutamatergic neurons, astrocytes controlling synaptic space levels, through glutamate reuptake, and inhibitory GABA interneurons. Astroglia generate and respond to inflammatory mediators. Glutamatergic microcircuits also act at the brain/body interface via the microbiome, kynurenine pathway, and hypothalamus-pituitary-adrenal axis. Disruption of excitatory/inhibitory homeostasis causing neuro-excitotoxicity, with neuronal impairment, causes depression and cognition symptoms via limbic and prefrontal regions, respectively. Persistent dysfunction reduces neuronal plasticity and growth causing neuronal death and tissue atrophy in neurodegenerative diseases. A conceptual overview of brain glutamatergic activity and peripheral interfacing is presented, including the common mechanisms that diverse diseases share when glutamate homeostasis is disrupted.
Collapse
Affiliation(s)
- Thomas McGrath
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| | - Richard Baskerville
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
- Correspondence:
| | - Marcelo Rogero
- School of Public Health, University of Sao Paulo, Sao Paulo 01246-904, Brazil;
| | - Linda Castell
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| |
Collapse
|
23
|
Basu S, Pradhan S, Barnett S, Mikkelsen M, Kapse K, Murnick J, Quistorff J, Lopez C, du Plessis A, Limperopoulos C. Regional Differences in Gamma-Aminobutyric Acid and Glutamate Concentrations in the Healthy Newborn Brain. AJNR Am J Neuroradiol 2022; 43:125-131. [PMID: 34764083 PMCID: PMC8757541 DOI: 10.3174/ajnr.a7336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND PURPOSE Gamma-aminobutyric acid and glutamate system disruptions may underlie neonatal brain injury. However, in vivo investigations are challenged by the need for special 1H-MR spectroscopy sequences for the reliable measurement of the neurotransmitters in this population. We used J-edited 1H-MR spectroscopy (Mescher-Garwood point-resolved spectroscopy) to quantify regional in vivo gamma-aminobutyric acid and glutamate concentrations during the early postnatal period in healthy neonates. MATERIALS AND METHODS We prospectively enrolled healthy neonates and acquired Mescher-Garwood point-resolved spectroscopy spectra on a 3T MR imaging scanner from voxels located in the cerebellum, the right basal ganglia, and the right frontal lobe. CSF-corrected metabolite concentrations were compared for regional variations and cross-sectional temporal trends with advancing age. RESULTS Fifty-eight neonates with acceptable spectra acquired at postmenstrual age of 39.1 (SD, 1.3) weeks were included for analysis. Gamma-aminobutyric acid (+ macromolecule) (2.56 [SD, 0.1]) i.u., glutamate (3.80 [SD, 0.2]), Cho, and mIns concentrations were highest in the cerebellum, whereas NAA (6.72 [SD, 0.2]), NAA/Cho, Cr/Cho, and Glx/Cho were highest in the basal ganglia. Frontal gamma-aminobutyric acid (1.63 [SD, 0.1]), Glx (4.33 [SD, 0.3]), Cr (3.64 [SD, 0.2]), and Cho concentrations were the lowest among the ROIs. Glx, NAA, and Cr demonstrated a significant adjusted increase with postmenstrual age (β = 0.2-0.35), whereas gamma-aminobutyric acid and Cho did not. CONCLUSIONS We report normative regional variations and temporal trends of in vivo gamma-aminobutyric acid and glutamate concentrations reflecting the functional and maturational status of 3 distinct brain regions of the neonate. These measures will serve as important normative values to allow early detection of subtle neurometabolic alterations in high-risk neonates.
Collapse
Affiliation(s)
- S.K. Basu
- From the Department of Neonatology (S.K.B.),Developing Brain Institute (S.K.B., S.P., S.D.B., K.J.K., J.L.Q., C.A.L., C.L.),George Washington University School of Medicine (S.K.B. S.P., S.D.B., J.M., A.J.d.P., C.L.), Washington, DC
| | - S. Pradhan
- Developing Brain Institute (S.K.B., S.P., S.D.B., K.J.K., J.L.Q., C.A.L., C.L.),George Washington University School of Medicine (S.K.B. S.P., S.D.B., J.M., A.J.d.P., C.L.), Washington, DC
| | - S.D. Barnett
- Developing Brain Institute (S.K.B., S.P., S.D.B., K.J.K., J.L.Q., C.A.L., C.L.),George Washington University School of Medicine (S.K.B. S.P., S.D.B., J.M., A.J.d.P., C.L.), Washington, DC
| | - M. Mikkelsen
- Department of Radiology (M.M., J.M.), Weill Cornell Medicine, New York, New York
| | - K.J. Kapse
- Developing Brain Institute (S.K.B., S.P., S.D.B., K.J.K., J.L.Q., C.A.L., C.L.)
| | - J. Murnick
- George Washington University School of Medicine (S.K.B. S.P., S.D.B., J.M., A.J.d.P., C.L.), Washington, DC,Department of Radiology (M.M., J.M.), Weill Cornell Medicine, New York, New York
| | - J.L. Quistorff
- Developing Brain Institute (S.K.B., S.P., S.D.B., K.J.K., J.L.Q., C.A.L., C.L.)
| | - C.A. Lopez
- Developing Brain Institute (S.K.B., S.P., S.D.B., K.J.K., J.L.Q., C.A.L., C.L.)
| | - A.J. du Plessis
- Fetal Medicine Institute (A.J.d.P.), Children’s National Hospital, Washington, DC,George Washington University School of Medicine (S.K.B. S.P., S.D.B., J.M., A.J.d.P., C.L.), Washington, DC
| | - C. Limperopoulos
- Developing Brain Institute (S.K.B., S.P., S.D.B., K.J.K., J.L.Q., C.A.L., C.L.),Division of Diagnostic Imaging and Radiology (C.L.),George Washington University School of Medicine (S.K.B. S.P., S.D.B., J.M., A.J.d.P., C.L.), Washington, DC
| |
Collapse
|
24
|
Gustorff C, Scheuer T, Schmitz T, Bührer C, Endesfelder S. GABA B Receptor-Mediated Impairment of Intermediate Progenitor Maturation During Postnatal Hippocampal Neurogenesis of Newborn Rats. Front Cell Neurosci 2021; 15:651072. [PMID: 34421540 PMCID: PMC8377254 DOI: 10.3389/fncel.2021.651072] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
The neurotransmitter GABA and its receptors assume essential functions during fetal and postnatal brain development. The last trimester of a human pregnancy and early postnatal life involves a vulnerable period of brain development. In the second half of gestation, there is a developmental shift from depolarizing to hyperpolarizing in the GABAergic system, which might be disturbed by preterm birth. Alterations of the postnatal GABA shift are associated with several neurodevelopmental disorders. In this in vivo study, we investigated neurogenesis in the dentate gyrus (DG) in response to daily administration of pharmacological GABAA (DMCM) and GABAB (CGP 35348) receptor inhibitors to newborn rats. Six-day-old Wistar rats (P6) were daily injected (i.p.) to postnatal day 11 (P11) with DMCM, CGP 35348, or vehicle to determine the effects of both antagonists on postnatal neurogenesis. Due to GABAB receptor blockade by CGP 35348, immunohistochemistry revealed a decrease in the number of NeuroD1 positive intermediate progenitor cells and a reduction of proliferative Nestin-positive neuronal stem cells at the DG. The impairment of hippocampal neurogenesis at this stage of differentiation is in line with a significantly decreased RNA expression of the transcription factors Pax6, Ascl1, and NeuroD1. Interestingly, the number of NeuN-positive postmitotic neurons was not affected by GABAB receptor blockade, although strictly associated transcription factors for postmitotic neurons, Tbr1, Prox1, and NeuroD2, displayed reduced expression levels, suggesting impairment by GABAB receptor antagonization at this stage of neurogenesis. Antagonization of GABAB receptors decreased the expression of neurotrophins (BDNF, NT-3, and NGF). In contrast to the GABAB receptor blockade, the GABAA receptor antagonization revealed no significant changes in cell counts, but an increased transcriptional expression of Tbr1 and Tbr2. We conclude that GABAergic signaling via the metabotropic GABAB receptor is crucial for hippocampal neurogenesis at the time of rapid brain growth and of the postnatal GABA shift. Differentiation and proliferation of intermediate progenitor cells are dependent on GABA. These insights become more pertinent in preterm infants whose developing brains are prematurely exposed to spostnatal stress and predisposed to poor neurodevelopmental disorders, possibly as sequelae of early disruption in GABAergic signaling.
Collapse
Affiliation(s)
- Charlotte Gustorff
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Till Scheuer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|