1
|
Hiraiwa T, Yoshii S, Kawada J, Sugawara T, Kawasaki T, Shibata S, Shindo T, Fujimori K, Umezawa A, Akutsu H. A human iPSC-Derived myelination model for investigating fetal brain injuries. Regen Ther 2025; 29:100-107. [PMID: 40162018 PMCID: PMC11953958 DOI: 10.1016/j.reth.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Cerebral white matter injuries, such as periventricular leukomalacia, are major contributors to neurodevelopmental impairments in preterm infants. Despite the clinical significance of these conditions, human-relevant models for studying fetal brain development and injury mechanisms remain limited. This study introduces a human iPSC-derived myelination model developed using a microfluidic device. The platform combines spinal cord-patterned neuronal and oligodendrocyte spheroids to recapitulate axon-glia interactions and myelination processes in vitro. The model successfully achieved axonal fascicle formation and compact myelin deposition, as validated by immunostaining and transmission electron microscopy. Functional calcium imaging confirmed neuronal activity within the system, underscoring its physiological relevance. While myelination efficiency was partial, with some axons remaining unmyelinated under the current conditions, this model represents a significant advancement in human myelin biology, offering a foundation for investigating fetal and perinatal brain injuries and related pathologies. Future refinements, such as improved myelination coverage and incorporating additional CNS cell types, will enhance its utility for studying disease mechanisms and enabling high-throughput drug screening.
Collapse
Affiliation(s)
- Tsuyoshi Hiraiwa
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima, Japan
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Shoko Yoshii
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jiro Kawada
- Jiksak Bioengineering, Inc., Kanagawa, Japan
| | - Tohru Sugawara
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Tomoyuki Kawasaki
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Keiya Fujimori
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima, Japan
| | - Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hidenori Akutsu
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
2
|
Mihailov A, Pron A, Lefèvre J, Deruelle C, Desnous B, Bretelle F, Manchon A, Milh M, Rousseau F, Girard N, Auzias G. Burst of gyrification in the human brain after birth. Commun Biol 2025; 8:805. [PMID: 40419689 DOI: 10.1038/s42003-025-08155-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 05/01/2025] [Indexed: 05/28/2025] Open
Abstract
Gyrification, the intricate folding of the brain's cortex, begins mid-gestation and surges dramatically throughout the perinatal period. Yet, a critical factor has been largely overlooked in neurodevelopmental research: the profound impact of birth on brain structure. Leveraging the largest known perinatal MRI dataset-819 sessions spanning 21 to 45 postconceptional weeks-we reveal a burst in gyrification immediately following birth (~37 weeks post-conception), amounting to half the entire gyrification expansion occurring during the fetal period. Using state-of-the-art, homogenized imaging processing tools across varied acquisition protocols, and applying a regression discontinuity design approach that is novel to neuroimaging, we provide the first evidence of a sudden, birth-triggered shift in cortical development. Investigation of additional cortical features confirms that this effect is uniquely confined to gyrification. This finding sheds light onto the understanding of early brain development, suggesting that the neurobiological consequences of birth may hold significant behavioral and physiological relevance.
Collapse
Affiliation(s)
- Angeline Mihailov
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France.
| | - Alexandre Pron
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France
| | - Julien Lefèvre
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France
| | - Christine Deruelle
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France
| | - Béatrice Desnous
- APHM, Service de Neurologie Pédiatrique, Hôpital de la Timone, Aix-Marseille University, Marseille, France
| | - Florence Bretelle
- APHM, Service de Gynécologie Obstétrique, Hôpital Nord, Aix-Marseille University, Marseille, France
| | - Aurélie Manchon
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France
- APHM, Service de Neuroradiologie Diagnostique et Interventionnelle, Hôpital de la Timone 2, Aix-Marseille University, Marseille, France
| | - Mathieu Milh
- APHM, Service de Neurologie Pédiatrique, Hôpital de la Timone, Aix-Marseille University, Marseille, France
| | | | - Nadine Girard
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France
- APHM, Service de Neuroradiologie Diagnostique et Interventionnelle, Hôpital de la Timone 2, Aix-Marseille University, Marseille, France
| | - Guillaume Auzias
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France
| |
Collapse
|
3
|
Ikebara JM, Jorge RS, Marinho LSR, Higa GSV, Adhikari A, Reis FMCV, Borges FS, Ulrich H, Takada SH, De Pasquale R, Kihara AH. Hippocampal Interneurons Shape Spatial Coding Alterations in Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05020-2. [PMID: 40392508 DOI: 10.1007/s12035-025-05020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Hippocampal interneurons (INs) play a fundamental role in regulating neural oscillations, modulating excitatory circuits, and shaping spatial representation. While historically overshadowed by excitatory pyramidal cells in spatial coding research, recent advances have demonstrated that inhibitory INs not only coordinate network dynamics but also contribute directly to spatial information processing. This review aims to provide a novel integrative perspective on how distinct IN subtypes participate in spatial coding and how their dysfunction contributes to cognitive deficits in neurological disorders such as epilepsy, Alzheimer's disease (AD), traumatic brain injury (TBI), and cerebral hypoxia-ischemia. We synthesize recent findings demonstrating that different IN classes-including parvalbumin (PV)-, somatostatin (SST)-, cholecystokinin (CCK)-, and calretinin (CR)-expressing neurons-exhibit spatially selective activity, challenging traditional views of spatial representation, and influence memory consolidation through network-level interactions. By leveraging cutting-edge techniques such as in vivo calcium imaging and optogenetics, new evidence suggests that INs encode spatial information with a level of specificity previously attributed only to pyramidal cells. Furthermore, we investigate the impact of inhibitory circuit dysfunction in neurological disorders, examining how disruptions in interneuronal activity lead to impaired theta-gamma coupling, altered sharp wave ripples, and destabilized place cell representations, ultimately resulting in spatial memory deficits. This review advances the field by shifting the focus from pyramidal-centered models to a more nuanced understanding of the hippocampal network, emphasizing the active role of INs in spatial coding. By highlighting the translational potential of targeting inhibitory circuits for therapeutic interventions, we propose novel strategies for restoring hippocampal network function in neurological conditions. Readers will gain a comprehensive understanding of the emerging role of INs in spatial representation and the critical implications of their dysfunction, paving the way for future research on interneuron-targeted treatments for cognitive disorders.
Collapse
Affiliation(s)
- Juliane Midori Ikebara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Renata Silva Jorge
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Luciana Simões Rafagnin Marinho
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Guilherme Shigueto Vilar Higa
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Avishek Adhikari
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Fernando M C V Reis
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Fernando S Borges
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Roberto De Pasquale
- Neurophysiology Laboratory, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil.
| |
Collapse
|
4
|
Abi Habib P, Bucak M, Crispi F, Gomez O, Turan O, Turan S. Fetal heart, brain and placenta: introducing a three-way (patho)physiological pairing. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2025. [PMID: 40387119 DOI: 10.1002/uog.29250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/29/2025] [Accepted: 04/08/2025] [Indexed: 05/20/2025]
Affiliation(s)
- P Abi Habib
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M Bucak
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - F Crispi
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - O Gomez
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clinic and Hospital Sant Joan de Déu, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - O Turan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - S Turan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Yousefpour N, Tansley SN, Locke S, Sharif B, Parisien M, Bourojeni FB, Deamond H, Mathur V, Arana NRK, Austin JS, Bourassa V, Wang C, Cabana VC, Wong C, Lister KC, Rodrigues R, St-Louis M, Paquet ME, Carroll MC, Andrews-Zwilling Y, Seguela P, Kania A, Yednock T, Mogil JS, De Koninck Y, Diatchenko L, Khoutorsky A, Ribeiro-da-Silva A. Targeting C1q prevents microglia-mediated synaptic removal in neuropathic pain. Nat Commun 2025; 16:4590. [PMID: 40382320 PMCID: PMC12085617 DOI: 10.1038/s41467-025-59849-1] [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: 11/10/2021] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Activation of spinal microglia following peripheral nerve injury is a central component of neuropathic pain pathology. While the contributions of microglia-mediated immune and neurotrophic signalling have been well-characterized, the phagocytic and synaptic pruning roles of microglia in neuropathic pain remain less understood. Here, we show that peripheral nerve injury induces microglial engulfment of dorsal horn synapses, leading to a preferential loss of inhibitory synapses and a shift in the balance between inhibitory and excitatory synapse density. This synapse removal is dependent on the microglial complement-mediated synapse pruning pathway, as mice deficient in complement C3 and C4 do not exhibit synapse elimination. Furthermore, pharmacological inhibition of the complement protein C1q prevents dorsal horn inhibitory synapse loss and attenuates neuropathic pain. Therefore, these results demonstrate that the complement pathway promotes persistent pain hypersensitivity via microglia-mediated engulfment of dorsal horn synapses in the spinal cord, revealing C1q as a therapeutic target in neuropathic pain.
Collapse
Affiliation(s)
- Noosha Yousefpour
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Annexon Biosciences, Brisbane, CA, USA
| | - Shannon N Tansley
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Dept. of Psychology, McGill University, Montréal, QC, Canada
- Dept. of Anesthesia, McGill University, Montréal, QC, Canada
| | - Samantha Locke
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| | - Behrang Sharif
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, Dept. of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| | - Marc Parisien
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Farin B Bourojeni
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Haley Deamond
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| | | | | | | | - Valerie Bourassa
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| | - Chengyang Wang
- Dept. of Psychology, McGill University, Montréal, QC, Canada
| | - Valérie C Cabana
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Calvin Wong
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Dept. of Anesthesia, McGill University, Montréal, QC, Canada
| | - Kevin C Lister
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Dept. of Psychology, McGill University, Montréal, QC, Canada
- Dept. of Anesthesia, McGill University, Montréal, QC, Canada
| | - Rose Rodrigues
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Dept. of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| | - Manon St-Louis
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Marie-Eve Paquet
- Dép. de biochimie, microbiologie et bioinformatique, Université Laval, Québec, QC, Canada
- CERVO Brain Research Centre, Québec, QC, Canada
| | - Michael C Carroll
- Harvard Medical School and Boston Children's Hospital, Boston, MA, USA
| | | | - Philippe Seguela
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, Dept. of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Artur Kania
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
- Dept. of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Science, McGill University, Montréal, QC, Canada
| | | | - Jeffrey S Mogil
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Dept. of Psychology, McGill University, Montréal, QC, Canada
| | - Yves De Koninck
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- CERVO Brain Research Centre, Québec, QC, Canada
- Dép. de psychiatrie et neurosciences, Université Laval, Québec, QC, Canada
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Dept. of Anesthesia, McGill University, Montréal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Arkady Khoutorsky
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
- Dept. of Anesthesia, McGill University, Montréal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Alfredo Ribeiro-da-Silva
- Dept. of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada.
- Dept. of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada.
| |
Collapse
|
6
|
Si Y, Zhang H, Du L, Deng Z. Abnormalities of brain dynamics based on large-scale cortical network modeling in autism spectrum disorder. Neural Netw 2025; 189:107561. [PMID: 40388872 DOI: 10.1016/j.neunet.2025.107561] [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: 10/21/2024] [Revised: 03/12/2025] [Accepted: 04/27/2025] [Indexed: 05/21/2025]
Abstract
Synaptic increase is a common phenomenon in the brain of autism spectrum disorder (ASD). However, the impact of increased synapses on the neurophysiological activity of ASD remains unclear. To address this, we propose a large-scale cortical network model based on empirical structural connectivity data using the Wendling model, which successfully simulates both pathological and physiological electroencephalography (EEG) signals. Building on this, the EEG functional network is constructed using the phase lag index, effectively characterizing the functional connectivity. Our modeling results indicate that EEG activity and functional network properties undergo significant changes by globally increasing synaptic coupling strength. Specifically, it leads to abnormal neural oscillations clinically reported in ASD, including the decreased dominant frequency, the decreased relative power in the α band and the increased relative power in the δ+θ band, particularly in the frontal lobe. At the same time, the clustering coefficient and global efficiency of the functional network decrease, while the characteristic path length increases, suggesting that the functional network of ASD is inefficient and poorly integrated. Additionally, we find insufficient functional connectivity across multiple brain regions in ASD, along with decreased wavelet coherence in the α band within the frontal lobe and between the frontal and temporal lobes. Considering that most of the synaptic increases in ASD are limited, brain regions are further randomly selected to increase the local synaptic coupling strength. The results show that disturbances in local brain regions can also facilitate the development of ASD. This study reveals the intrinsic link between synapse increase and abnormal brain activity in ASD, and inspires treatments related to synapse pruning.
Collapse
Affiliation(s)
- Youyou Si
- School of Mathematics and Statistics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China
| | - Honghui Zhang
- School of Mathematics and Statistics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China.
| | - Lin Du
- School of Mathematics and Statistics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China
| | - Zichen Deng
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China
| |
Collapse
|
7
|
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
|
8
|
Lean RE, Kenley JK, Latham AR, Smyser TA, Neil J, Nielsen AN, Sylvester CM, Miller JP, Shimony JJ, Luby J, Barch DM, Warner BB, Smyser CD, Rogers CE. Prenatal Adversity and Neonatal Brain Connectivity Relate to Emerging Executive Function at Age 2 Years. J Am Acad Child Adolesc Psychiatry 2025:S0890-8567(25)00224-2. [PMID: 40334775 DOI: 10.1016/j.jaac.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 02/18/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVE Early life adversity alters the structure and function of higher-order brain networks that subserve executive function (EF). The extent that prenatal exposure to adversity and neonatal white matter (WM) microstructure and resting-state functional connectivity (rs-fc) underlie problems in emerging EF remains unclear. METHOD This prospective study includes 164 infants (45% female, 85% term-born) who were recruited prenatally and underwent neonatal diffusion and rs-fc magnetic resonance imaging scans. Social disadvantage and maternal psychosocial stress were assessed in the prenatal period. At age 2 years, children completed the Minnesota Executive Function Scale. Multivariable regression, moderation, and mediation analyses examined associations between prenatal adversity, neonatal WM microstructure and rs-fc, and emerging EF outcome. RESULTS Prenatal social disadvantage (PSD), but not maternal psychosocial stress, was associated with poorer emerging EF. After multiple comparison correction, higher mean diffusivity (MD) and lower fractional anisotropy (FA) in the corpus callosum, as well as higher MD in the inferior fronto-occipital fasciculus and corticospinal tract and lower FA in the uncinate, related to poorer emerging EF. In moderation analysis, associations between neonatal WM microstructure and emerging EF did not vary as a function of PSD. In mediation analyses, neonatal WM microstructure did not attenuate the association between PSD and emerging EF. The rs-fc findings did not pass multiple comparison correction. CONCLUSION PSD was related to poorer emerging EF outcomes. Neonatal WM microstructure was also related to emerging EF, with similar associations for children with lower or higher PSD. Prenatal social welfare programs may support neonatal brain development and early neurodevelopmental outcomes. DIVERSITY & INCLUSION We worked to ensure sex and gender balance in the recruitment of human participants. We worked to ensure race, ethnic, and/or other types of diversity in the recruitment of human participants. We worked to ensure that the study questionnaires were prepared in an inclusive way.
Collapse
Affiliation(s)
- Rachel E Lean
- Washington University School of Medicine, St. Louis, Missouri.
| | | | - Aidan R Latham
- Washington University School of Medicine, St. Louis, Missouri
| | - Tara A Smyser
- Washington University School of Medicine, St. Louis, Missouri
| | - Jeff Neil
- Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Chad M Sylvester
- Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - J Philip Miller
- Washington University School of Medicine, St. Louis, Missouri
| | - Joshua J Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Joan Luby
- Washington University School of Medicine, St. Louis, Missouri
| | - Deanna M Barch
- Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | | | | |
Collapse
|
9
|
Cao S, Wan Y, Xiong Z, Li R, Wang Y, Qian X, Chen R, Wang J, Zhang L, Cui Y, Cheng R, Li Y, Xu S, Xia W. Trimester-specific associations of exposure to epoxide alkanes, alkenals, and 1,3-butadiene with preschool children's intellectual development: A birth cohort study in Wuhan, China. ENVIRONMENT INTERNATIONAL 2025; 199:109456. [PMID: 40252551 DOI: 10.1016/j.envint.2025.109456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
The impact of prenatal exposure to contaminants with neurotoxicity like epoxide alkanes (ethylene oxide, propylene oxide), alkenals (acrolein, crotonaldehyde), and 1,3-butadiene on children's intellectual development remains underreported, and related sensitive window is of interest. In this cohort study, metabolites of these contaminants were measured in 3,081 urine samples from 1,027 pregnant women across three trimesters. Children's intelligence quotient was evaluated at 4-6 years old. Generalized estimating equation models showed that higher urinary concentrations of 2-hydroxypropyl mercapturic acid (a metabolite of propylene oxide), 3-hydroxypropyl mercapturic acid (HPMMA, a metabolite of crotonaldehyde), and the sum of acrolein metabolites in the first trimester were associated with lower visual spatial index (VSI), working memory index (WMI), or processing speed index scores. Quantile g-computation models revealed that co-exposure to these contaminants in the first trimester were associated with lower VSI (β = -0.98, 95 % CI: -1.94, -0.03) and WMI (β = -0.86, 95 % CI: -1.66, -0.06) scores, with HPMMA as the major contributor. These results suggested that early pregnancy could be a sensitive window during which exposure to propylene oxide, crotonaldehyde, and acrolein may impair offspring's intellectual development.
Collapse
Affiliation(s)
- Shuting Cao
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Yanjian Wan
- Center for Public Health Laboratory Service, Institute of Environmental Health, Wuhan Centers for Disease Control & Prevention, Wuhan, Hubei 430024, PR China.
| | - Zhaoying Xiong
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Ruiying Li
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Yin Wang
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Xi Qian
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Ruixin Chen
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Jingyu Wang
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Liping Zhang
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Yuan Cui
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Rongrong Cheng
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Yuanyuan Li
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| | - Shunqing Xu
- School of Environmental Science and Engineering, Hainan University, Haikou, Hainan 570228, PR China.
| | - Wei Xia
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
10
|
Tu JC, Myers MJ, Li W, Li J, Wang X, Dierker D, Day TKM, Snyder A, Latham A, Kenley JK, Sobolewski CM, Wang Y, Labonte AK, Feczko E, Kardan O, Moore LA, Sylvester CM, Fair DA, Elison JT, Warner BB, Barch DM, Rogers CE, Luby JL, Smyser CD, Gordon EM, Laumann TO, Eggebrecht AT, Wheelock MD. The generalizability of cortical area parcellations across early childhood. Cereb Cortex 2025; 35:bhaf116. [PMID: 40422981 DOI: 10.1093/cercor/bhaf116] [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: 10/01/2024] [Revised: 03/03/2025] [Accepted: 04/04/2025] [Indexed: 05/28/2025] Open
Abstract
The cerebral cortex consists of distinct areas that develop through intrinsic embryonic patterning and postnatal experiences. Accurate parcellation of these areas in neuroimaging studies improves statistical power and cross-study comparability. Given significant brain changes in volume, microstructure, and connectivity during early life, we hypothesized that cortical areas in 1- to 3-year-olds would differ markedly from neonates and increasingly resemble adult patterns as development progresses. Here, we parcellated the cerebral cortex into putative areas using local functional connectivity (FC) gradients in 92 toddlers at 2 years old. We demonstrate high reproducibility of these cortical areas across 1- to 3-year-olds in two independent datasets. The area boundaries in 1- to 3-year-olds were more similar to those in adults than those in neonates. While the age-specific group area parcellation better fits the underlying FC in individuals during the first 3 years, adult area parcellations still have utility in developmental studies, especially in children older than 6 years. Additionally, we provide connectivity-based community assignments of the area parcels, showing fragmented anterior and posterior components based on the strongest connectivity, yet alignment with adult systems when weaker connectivity was included.
Collapse
Affiliation(s)
- Jiaxin Cindy Tu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| | - Michael J Myers
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| | - Wei Li
- Department of Mathematics and Statistics, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, United States
| | - Jiaqi Li
- Department of Mathematics and Statistics, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, United States
- Department of Statistics, University of Chicago, 5747 S Ellis Ave, Chicago, IL 60637, United States
| | - Xintian Wang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| | - Donna Dierker
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| | - Trevor K M Day
- Masonic Institute for the Developing Brain, University of Minnesota, 2025 E River Pkwy, Minneapolis, MN 55414, United States
- Institute of Child Development, University of Minnesota, Campbell Hall, 51 E River Rd, Minneapolis, MN 55455, United States
- Center for Brain Plasticity and Recovery, Georgetown University, Department of Neurology Building D, Suite 145, 4000 Reservoir Road, N.W. Washington, DC 20007, United States
| | - Abraham Snyder
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| | - Aidan Latham
- Department of Neurology, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, United States
| | - Jeanette K Kenley
- Department of Neurology, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, United States
| | - Chloe M Sobolewski
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
- Department of Psychology, Virginia Commonwealth University, White House 806 W. Franklin St. Box 842018. Richmond, Virginia 23284-2018, United States
| | - Yu Wang
- Department of Mathematics and Statistics, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, United States
| | - Alyssa K Labonte
- Department of Psychiatry, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO 63110-1010, United States
| | - Eric Feczko
- Masonic Institute for the Developing Brain, University of Minnesota, 2025 E River Pkwy, Minneapolis, MN 55414, United States
| | - Omid Kardan
- Department of Psychiatry, University of Michigan, 250 Plymouth Road, Ann Arbor 48109, United States
| | - Lucille A Moore
- Masonic Institute for the Developing Brain, University of Minnesota, 2025 E River Pkwy, Minneapolis, MN 55414, United States
| | - Chad M Sylvester
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
- Department of Psychiatry, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO 63110-1010, United States
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, 4444 Forest Park Ave #2600, St. Louis, MO 63108, United States
| | - Damien A Fair
- Masonic Institute for the Developing Brain, University of Minnesota, 2025 E River Pkwy, Minneapolis, MN 55414, United States
- Institute of Child Development, University of Minnesota, Campbell Hall, 51 E River Rd, Minneapolis, MN 55455, United States
| | - Jed T Elison
- Masonic Institute for the Developing Brain, University of Minnesota, 2025 E River Pkwy, Minneapolis, MN 55414, United States
- Institute of Child Development, University of Minnesota, Campbell Hall, 51 E River Rd, Minneapolis, MN 55455, United States
| | - Barbara B Warner
- Department of Pediatrics, Washington University in St. Louis, 660 S Euclid Ave, St. Louis, MO 63110, United States
| | - Deanna M Barch
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
- Department of Psychiatry, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO 63110-1010, United States
- Department of Psychological and Brain Sciences, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, United States
| | - Cynthia E Rogers
- Department of Psychiatry, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO 63110-1010, United States
| | - Joan L Luby
- Department of Psychiatry, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO 63110-1010, United States
| | - Christopher D Smyser
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
- Department of Neurology, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, United States
- Department of Psychiatry, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO 63110-1010, United States
- Department of Pediatrics, Washington University in St. Louis, 660 S Euclid Ave, St. Louis, MO 63110, United States
| | - Evan M Gordon
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| | - Timothy O Laumann
- Department of Psychiatry, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO 63110-1010, United States
| | - Adam T Eggebrecht
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| | - Muriah D Wheelock
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave, St. Louis, MO 63110, United States
| |
Collapse
|
11
|
Peterson BS, Delavari S, Sadik J, Ersland L, Elgen IB, Sawardekar S, Bansal R, Aukland SM. Brain tissue microstructure in a prospective, longitudinal, population-based cohort of preterm and term-born young adults. J Child Psychol Psychiatry 2025; 66:635-649. [PMID: 39561978 PMCID: PMC12018296 DOI: 10.1111/jcpp.14069] [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] [Accepted: 07/12/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Fifteen million infants annually are born prematurely, placing them at high risk for life-long adverse neurodevelopmental outcomes. Whether brain tissue abnormalities that accompany preterm birth persist into young adulthood and are associated with long-term cognitive or psychiatric outcomes is not known. METHODS From infancy into young adulthood, we followed a population-based sample of consecutively identified preterm infants and their matched term controls. The preterm group was born at an average gestational age of 31.5 ± 2.6 weeks. We obtained Diffusion Tensor Imaging scans and assessed cognitive and psychiatric outcomes in young adulthood, at a mean age of 19 (range 17.6-20.8) years. Usable data were acquired from 180 participants (89 preterm, 91 term). RESULTS Preterm birth was associated with lower fractional anisotropy (FA) and higher average diffusion coefficient (ADC) values in deep white matter tracts of the internal capsule, cerebral peduncles, inferior frontal-occipital fasciculus, sagittal stratum and splenium of the corpus callosum, as well as in grey matter of the caudate, putamen and thalamus. A younger gestational age at birth accentuated these tissue abnormalities. Perinatal characteristics, including lower 5-min APGAR score, history of bronchopulmonary dysplasia, more days of oxygen supplementation and multiple births all increased ADC values in deep white matter tracts and grey matter throughout the brain. Preterm individuals had significantly lower full-scale IQ and more frequent lifetime psychiatric disorders. Those with psychiatric illnesses had significantly higher ADC and lower FA values throughout the deep posterior white matter. CONCLUSIONS Abnormalities in brain tissue microstructure associated with preterm birth persist into young adulthood and likely represent disordered myelination and accompanying axonal pathology. These disturbances are associated with a higher likelihood of developing a psychiatric disorder by young adulthood. Brain tissue disturbances were accentuated in those born at younger gestational ages and in those with a history of perinatal complications associated with infection and inflammation.
Collapse
Affiliation(s)
- Bradley S. Peterson
- Institute for the Developing MindChildren's Hospital Los AngelesLos AngelesCAUSA
- Department of PsychiatryKeck School of Medicine at the University of Southern CaliforniaLos AngelesCAUSA
| | - Sahar Delavari
- Institute for the Developing MindChildren's Hospital Los AngelesLos AngelesCAUSA
- Department of PsychiatryKeck School of Medicine at the University of Southern CaliforniaLos AngelesCAUSA
| | - Jonathan Sadik
- Institute for the Developing MindChildren's Hospital Los AngelesLos AngelesCAUSA
- Department of PsychiatryKeck School of Medicine at the University of Southern CaliforniaLos AngelesCAUSA
| | - Lars Ersland
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
- Department of Clinical EngineeringHaukeland University HospitalBergenNorway
| | - Irene B. Elgen
- Division of Psychiatry, Department of Child and Adolescent PsychiatryHaukeland University HospitalBergenNorway
| | - Siddhant Sawardekar
- Institute for the Developing MindChildren's Hospital Los AngelesLos AngelesCAUSA
| | - Ravi Bansal
- Institute for the Developing MindChildren's Hospital Los AngelesLos AngelesCAUSA
- Department of PsychiatryKeck School of Medicine at the University of Southern CaliforniaLos AngelesCAUSA
| | - Stein Magnus Aukland
- Department of RadiologyHaukeland University HospitalBergenNorway
- Department of Clinical MedicineUniversity of BergenBergenNorway
| |
Collapse
|
12
|
Geertsema J, Juncker HG, Wilmes L, Burchell GL, de Rooij SR, van Goudoever JB, O'Riordan KJ, Clarke G, Cryan JF, Korosi A. Nutritional interventions to counteract the detrimental consequences of early-life stress. Mol Psychiatry 2025:10.1038/s41380-025-03020-1. [PMID: 40289212 DOI: 10.1038/s41380-025-03020-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 03/19/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
Exposure to stress during sensitive developmental periods comes with long term consequences for neurobehavioral outcomes and increases vulnerability to psychopathology later in life. While we have advanced our understanding of the mechanisms underlying the programming effects of early-life stress (ES), these are not yet fully understood and often hard to target, making the development of effective interventions challenging. In recent years, we and others have suggested that nutrition might be instrumental in modulating and possibly combatting the ES-induced increased risk to psychopathologies and neurobehavioral impairments. Nutritional strategies are very promising as they might be relatively safe, cheap and easy to implement. Here, we set out to comprehensively review the existing literature on nutritional interventions aimed at counteracting the effects of ES on neurobehavioral outcomes in preclinical and clinical settings. We identified eighty six rodent and ten human studies investigating a nutritional intervention to ameliorate ES-induced impairments. The human evidence to date, is too few and heterogeneous in terms of interventions, thus not allowing hard conclusions, however the preclinical studies, despite their heterogeneity in terms of designs, interventions used, and outcomes measured, showed nutritional interventions to be promising in combatting ES-induced impairments. Furthermore, we discuss the possible mechanisms involved in the beneficial effects of nutrition on the brain after ES, including neuroinflammation, oxidative stress, hypothalamus-pituitary-adrenal axis regulation and the microbiome-gut-brain axis. Lastly, we highlight the critical gaps in our current knowledge and make recommendations for future research to move the field forward.
Collapse
Affiliation(s)
- Jorine Geertsema
- Brain Plasticity group, Centre for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Hannah G Juncker
- Brain Plasticity group, Centre for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Lars Wilmes
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - George L Burchell
- Medical Library, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susanne R de Rooij
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Epidemiology and Data Science, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Public Health research institute, Aging and Later Life, Health Behaviors and Chronic Diseases, Amsterdam, The Netherlands
| | - J B van Goudoever
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | | | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Aniko Korosi
- Brain Plasticity group, Centre for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Godeanu S, Cătălin B. The Complementary Role of Morphology in Understanding Microglial Functional Heterogeneity. Int J Mol Sci 2025; 26:3811. [PMID: 40332469 PMCID: PMC12027755 DOI: 10.3390/ijms26083811] [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: 03/13/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
A search of the PubMed database for publications on microglia reveals an intriguing shift in scientific interest over time. Dividing microglia into categories such as "resting" and "activated" or M1 versus M2 is nowadays obsolete, with the current research focusing on unraveling microglial heterogeneity. The onset of transcriptomics, especially single-cell RNA sequencing (scRNA-seq), has profoundly reshaped our understanding of microglia heterogeneity. Conversely, microglia morphology analysis can offer important insights regarding their activation state or involvement in tissue responses. This review explores microglial heterogeneity under homeostatic conditions, developmental stages, and disease states, with a focus on integrating transcriptomic data with morphological analysis. Beyond the core gene expression profile, regional differences are observed with cerebellar microglia exhibiting a uniquely immune-vigilant profile. During development, microglia express homeostatic genes before birth, yet the bushy appearance is a characteristic that appears later on. In neurodegeneration, microglia alternate between proinflammatory and neuroprotective roles, influenced by regional factors and disease onset. Understanding these structural adaptations may help identify specific microglial subpopulations for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sânziana Godeanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Bogdan Cătălin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Building 48, University of Saarland, 66421 Homburg, Germany
| |
Collapse
|
14
|
Shimamura T, Kitashiba M, Nishizawa K, Hattori Y. Physiological roles of embryonic microglia and their perturbation by maternal inflammation. Front Cell Neurosci 2025; 19:1552241. [PMID: 40260079 PMCID: PMC12009865 DOI: 10.3389/fncel.2025.1552241] [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: 12/27/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
The interplay between the nervous and immune systems is well documented in the context of adult physiology and disease. Recent advances in understanding immune cell development have highlighted a significant interaction between neural lineage cells and microglia, the resident brain macrophages, during developmental stages. Throughout development, particularly from the embryonic to postnatal stages, diverse neural lineage cells are sequentially generated, undergo fate determination, migrate dynamically to their appropriate locations while maturing, and establish connections with their surroundings to form neural circuits. Previous studies have demonstrated that microglia contribute to this highly orchestrated process, ensuring the proper organization of brain structure. These findings underscore the need to further investigate how microglia behave and function within a broader framework of neurodevelopment. Importantly, recent epidemiological studies have suggested that maternal immune activation (MIA), triggered by various factors, such as viral or bacterial infections, environmental stressors, or other external influences, can affect neurogenesis and neural circuit formation, increasing the risk of neurodevelopmental disorders (NDDs) in offspring. Notably, many studies have revealed that fetal microglia undergo significant changes in response to MIA. Given their essential roles in neurogenesis and vascular development, inappropriate activation or disruption of microglial function may impair these critical processes, potentially leading to abnormal neurodevelopment. This review highlights recent advances in rodent models and human studies that have shed light on the behaviors and multifaceted roles of microglia during brain development, with a particular focus on the embryonic stage. Furthermore, drawing on insights from rodent MIA models, this review explores how MIA disrupts microglial function and how such disturbances may impair brain development, ultimately contributing to the onset of NDDs.
Collapse
Affiliation(s)
| | | | | | - Yuki Hattori
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
15
|
Palomero-Sierra B, Sánchez-Gómez V, Magán-Maganto M, Bejarano-Martín Á, Ruiz-Ayúcar I, de Vena-Díez VB, Mannarino GV, Díez-Villoria E, Canal-Bedia R. Early social communication and language development in moderate-to-late preterm infants: a longitudinal study. Front Psychol 2025; 16:1556416. [PMID: 40248834 PMCID: PMC12003367 DOI: 10.3389/fpsyg.2025.1556416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
This study investigates early development and language acquisition in moderate-to-late preterm (MLPT) infants, focusing on social communication as a key factor. Using a longitudinal design, social communicative, cognitive and language outcomes were assessed at 12, 18, and 24 months in 106 infants, including 49 MLPT and 57 full-term (FT) infants. Standardized tools, including the Bayley Scales of Infant and Toddler Development (Bayley-III), the Vineland Adaptive Behavior Scales (Vineland-3), and the Social Attention and Communication Surveillance-Revised (SACS-R), were used to assess early developmental performance. Group differences and the interaction between group and assessment time points were analyzed to examine developmental patterns over time. Additionally, predictive models identified early indicators of receptive and expressive language performance at 24 months. The results revealed significant developmental delays in the MLPT group compared to their FT peers, with receptive language showing the most pronounced deficits. Early social communication behaviors, such as pointing, following a point, and attending to sounds at 12 months, emerged as strong predictors of both receptive and expressive language performance. Cognitive abilities also played a significant role, particularly in receptive language development. These findings underscore the utility of tools like the SACS-R in identifying early communication challenges and guiding tailored support strategies. Sustained developmental monitoring and targeted interventions that foster communication skills may promote positive language outcomes in MLPT infants, supporting their long-term developmental potential within this population with increased developmental needs.
Collapse
Affiliation(s)
- Blanca Palomero-Sierra
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Department of Basic Psychology, Psychobiology, and Behavioral Science Methodology, University of Salamanca, Salamanca, Spain
| | - Victoria Sánchez-Gómez
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Department of Basic Psychology, Psychobiology, and Behavioral Science Methodology, University of Salamanca, Salamanca, Spain
| | - María Magán-Maganto
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Department of Basic Psychology, Psychobiology, and Behavioral Science Methodology, University of Salamanca, Salamanca, Spain
| | - Álvaro Bejarano-Martín
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Department of Personality, Assessment, and Psychological Treatments, University of Salamanca, Salamanca, Spain
| | | | - Victoria B. de Vena-Díez
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Giselle V. Mannarino
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Emiliano Díez-Villoria
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Department of Basic Psychology, Psychobiology, and Behavioral Science Methodology, University of Salamanca, Salamanca, Spain
| | - Ricardo Canal-Bedia
- Institute for Community Inclusion (INICO), University of Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Department of Personality, Assessment, and Psychological Treatments, University of Salamanca, Salamanca, Spain
| |
Collapse
|
16
|
Yoon SB, Park JB, Kang P, Jang YE, Kim EH, Lee JH, Lee HC, Kim JT, Kim HS, Ji SH. Electroencephalography and Anesthetic Depth in Children Under 2 Years of Age: A Prospective Observational Study. Paediatr Anaesth 2025; 35:294-301. [PMID: 39723638 DOI: 10.1111/pan.15058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/25/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Processed electroencephalogram (EEG) indices are widely used to monitor anesthetic depth. However, their reliability in children under 2 years of age remains questionable. During anesthesia maintenance in this age group, processed EEG indices frequently exhibit unexpectedly elevated values that exceed the intended target range. AIM This study aimed to identify EEG spectral parameters associated with false positive elevations in processed EEG indices and investigate their differences from true positive elevations during emergence. METHODS This prospective observational study included 50 children aged 4-24 months undergoing general anesthesia. Bispectral index (BIS), patient state index (PSi), and raw EEG were continuously recorded throughout anesthesia. False positive was defined as elevated processed EEG indices when end-tidal sevoflurane concentration was maintained at 0.7-1.3 minimum alveolar concentration, with heart rate and mean blood pressure between 80% and 120% of baseline values. We analyzed EEG power spectra and band power values during periods of false positives and compared them with those of true positives during emergence. Bonferroni-corrected p < 0.05 was considered significant. RESULTS False positives in processed EEG indices were observed in 35 (70%) of the children during anesthesia maintenance, occupying 28% of the maintenance phase. These false positives were associated with decreased power in delta (269-174 dB) and theta (115-97 dB) bands, but widespread increases in alpha and beta bands, resulting in elevated spectral edge frequency (19-22 Hz). Notably, EEG band power during false positives significantly differed from those observed during emergence (delta: 52 dB, theta: 38 dB) (all p < 0.001). CONCLUSIONS Processed EEG indices may exhibit unexpectedly elevated values during anesthesia maintenance in children under 2 years of age. Quantitative assessments derived from raw EEG data may improve the evaluation of anesthetic depth in this population.
Collapse
Affiliation(s)
- Soo-Bin Yoon
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung-Bin Park
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Pyoyoon Kang
- Department of Anesthesiology and Pain Medicine, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Young-Eun Jang
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun-Hee Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji-Hyun Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyung-Chul Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Tae Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hee-Soo Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang-Hwan Ji
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Nivins S, Padilla N, Kvanta H, Ådén U. Gestational Age and Cognitive Development in Childhood. JAMA Netw Open 2025; 8:e254580. [PMID: 40227687 PMCID: PMC11997729 DOI: 10.1001/jamanetworkopen.2025.4580] [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: 10/15/2024] [Accepted: 02/10/2025] [Indexed: 04/15/2025] Open
Abstract
Importance Preterm and early-term births are known risk factors for cognitive impairment, but studies that comprehensively include genetics, prenatal risk, and child-specific factors in high-risk populations are lacking. Objective To investigate the long-term cognitive outcomes of children born at various gestational ages, including very preterm (28-31 weeks), moderately preterm (32-33 weeks), late preterm (34-36 weeks), and early term (37-38 weeks), compared with full-term (≥39 weeks), accounting for genetics and other risk factors. Design, Setting, and Participants In this prospective, multicenter, longitudinal cross-sectional study, children aged 9 to 10 years were recruited from the Adolescent Brain and Cognitive Development Study between January 1, 2016, and December 31, 2018. Children underwent cognitive assessments using the National Institutes of Health Toolbox, Little Man Task, and Rey Auditory Verbal Learning Test. Polygenic scores for cognitive performance (cogPGS) were generated using results of a genome-wide association study from the genetic variants related to cognitive performance, educational attainment, and mathematical ability. Data analysis was performed from March to June 2024. Exposure Preterm (very preterm, moderately preterm, late preterm) and early-term birth status, with full-term birth status as the reference group. Main Outcomes and Measures The primary outcome of interest was the composite cognitive score, while secondary outcomes included individual cognitive domain scores. Hierarchical regression models were used to examine associations between gestational age and cognitive outcomes, adjusting for socioeconomic status (SES), cogPGS, prenatal risks, and child-specific factors. Results Among 5946 children included in the study (mean [SD] age, 9.9 [0.6] years; 3083 [51.8%] male), 55 (0.9%) were born very preterm, 110 (1.8%) were born moderately preterm, 454 (7.6%) were born late preterm, 261 (4.4%) were born early term, and 5066 (85.2%) were born full term. The cogPGS was positively associated with the composite cognitive score (β = 0.14; 95% CI, 0.12-0.17; P < .001) in the overall cohort. Compared with full-term children, those born moderately preterm had lower composite cognitive scores (β = -0.39; 95% CI, -0.55 to -0.22; P < .001) and lower scores in vocabulary (β = -0.36; 95% CI, -0.53 to -0.19; P < .001), working memory (β = -0.27; 95% CI, -0.45 to -0.09; P = .003), episodic memory (β = -0.32; 95% CI, -0.50 to -0.14; P < .001), and both short-delay recall (β = -0.36; 95% CI, -0.54 to -0.18; P < .001) and long-delay recall (β = -0.29; 95% CI, -0.48 to -0.11; P = .002). These associations were independent of SES, cogPGS, and other risk factors. Importantly, the lowest cognitive scores appeared among children born at 32 weeks or less. In contrast, late-preterm and early-term children performed similarly to full-term peers. Conclusions and Relevance In this cross-sectional study of children aged 9 to 10 years, moderately preterm birth was associated with long-term cognitive problems independent of SES, genetics, and other risk factors. These findings underscore the need for continued follow-up of all preterm children, with particular focus on those born before 34 weeks' gestational age, because they may face greater developmental challenges over time.
Collapse
Affiliation(s)
- Samson Nivins
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Nelly Padilla
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Hedvig Kvanta
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Ådén
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Vasquez LS, Stack S, Taylor WW, Dias BG. Intergenerational Effects of Stress - A Focus on Learning and Memory. Curr Top Behav Neurosci 2025. [PMID: 40119217 DOI: 10.1007/7854_2025_578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Stress is a ubiquitous facet of life. Ranging in form (e.g., psychosocial, physical, nutritional, economic) and longevity (e.g., acute, chronic), stressors affect the biology of those directly in their line of attack. As is becoming increasingly appreciated, the pernicious effects of stress echo across generations (Dias et al. 2015; Yehuda and Lehrner 2018; Jawaid et al. 2021; Dion et al. 2022; Zhou and Ryan 2023; Dias 2024). With a focus on learning and memory, this chapter addresses how stressors derail learning and memory in the generation directly exposed to them andin future generations. To do so, with a specific emphasis on associative fear conditioning in humans and rodents, we touch upon the relevance of extinction training in the aftermath of such conditioning and the recall of such extinction training as windows into normative and disrupted learning. Next, we briefly discuss underlying neuroanatomical substrates mediating these processes. We then draw attention to influences of postnatal, in utero, and pre-conceptional stress on learning and memory across generations. Finally, we briefly outline biological factors that underlie how learning and memory is derailed by these stressors.
Collapse
Affiliation(s)
- L S Vasquez
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - S Stack
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - W W Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - B G Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA.
- Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Wood EK, Nomura O, Ablow JC, Elliot A, Horgan A, Nigg JT, Gustafsson HC, Sullivan EL. Higher prenatal dietary glycemic index in the third trimester of pregnancy is associated with infant negative affect at 6 months. Sci Rep 2025; 15:8357. [PMID: 40069302 PMCID: PMC11897411 DOI: 10.1038/s41598-025-91886-0] [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: 02/26/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
The dietary glycemic index (GI) reflects post-prandial plasma glucose generation rate, with higher-GI foods rapidly increasing blood sugar. Prenatal consumption of high-GI foods is associated with offspring risk for obesity and metabolic disorders. The impact of prenatal dietary GI exposure on infant neurodevelopment remains unclear. Maternal dietary intake, percent adiposity, and insulin resistance were prospectively assessed during the second and third trimesters in a sample of women with healthy, singleton pregnancies (N = 302). Infant negative affect was prospectively assessed at six months using observer ratings (Still Face Paradigm) and caregiver-reports (Infant-Behavior Questionnaire-Revised). Structural equation models assessed the independent effects of second and third trimester maternal dietary GI, adiposity, insulin resistance on infant negative affect, adjusted for relevant covariates. Higher third, but not second, trimester dietary GI was associated with increased observer-rated infant negative affect (β = 0.14, p = .04) and with higher caregiver-reported infant sadness (β = 0.17, p = .01), suggesting a programming effect of prenatal dietary GI on infant neurodevelopment. Targeted interventions that decrease dietary GI in later pregnancy may prove more effective for optimizing infant behavioral health compared to longer-term changes needed to alter metabolic state. Identifying modifiable early contributors to infant negative affect supports proactive strategies for mitigating future psychopathology risk.
Collapse
Affiliation(s)
- Elizabeth K Wood
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Olivia Nomura
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jennifer C Ablow
- Department of Psychology, University of Oregon, Eugene, OR, 97403, USA
| | - Annaliese Elliot
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Angela Horgan
- Clinical & Translational Research Center, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Joel T Nigg
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA.
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, 97239, USA.
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA.
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA.
- Departments of Psychiatry and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
20
|
Mireku MO, Boivin MJ, Zoumenou R, Garrison A, Cot M, Alao J, Fievet N, Massougbodji A, Bodeau-Livinec F. Prenatal Hemoglobin Concentration and Long-Term Child Neurocognitive Development. Am J Trop Med Hyg 2025; 112:692-698. [PMID: 39689363 PMCID: PMC11884297 DOI: 10.4269/ajtmh.24-0643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/25/2024] [Indexed: 12/19/2024] Open
Abstract
Anemia in pregnancy, defined by a hemoglobin level (Hb) of less than 110 g/L, contributes to infant mortality and morbidity in sub-Saharan Africa. Maternal Hb changes physiologically and pathologically during pregnancy. However, the impact of these changes on long-term child neurocognitive function is unknown. This study therefore investigates the association between Hb at specific antenatal care visits and prenatal Hb trajectories during pregnancy and long-term child neurocognitive function. We analyzed data from a prospective cohort study that included 6-year-old singleton children born to women enrolled before 29 weeks of gestation into an antimalarial drug clinical trial. Hemoglobin level was analyzed from venous blood collected at least twice during pregnancy and at delivery. We used group-based trajectory modeling to identify distinct prenatal Hb trajectories. In total, 478 children (75.1% of eligible children) had assessment of cognitive and motor functions at 6 years of age. Three distinct Hb trajectories were identified: persistently anemic (Hb <110 g/L throughout the second and third trimesters), anemic to nonanemic (Hb <110 g/L at second trimester with increasing Hb toward the third trimester to Hb ≥110 g/L), and persistently nonanemic (Hb ≥110 g/L throughout the second and third trimesters). Children of women in the persistently anemic and anemic-to-nonanemic groups had significantly lower neurocognitive scores than children of women in the persistently nonanemic group (β = -6.8, 95% CI: -11.7 to -1.8; and β = -6.3, 95% CI: -10.4 to -2.2, respectively). The study shows that maintaining an elevation of Hb at or above 110 g/L from the second to third trimester of pregnancy may be associated with optimal long-term child neurocognitive function.
Collapse
Affiliation(s)
- Michael O. Mireku
- College of Health and Science, University of Lincoln, Lincoln, United Kingdom
| | - Michael J. Boivin
- Department of Psychiatry, Michigan State University, East Lansing, Michigan
| | - Roméo Zoumenou
- UMR 261-MERIT, Institut de Recherche pour le Développement (IRD), Université Paris Cité, Paris, France
- Institute of Psychology, Laboratoire de Psychopathologie et Processus de Santé, Boulogne, France
| | - Amanda Garrison
- Université Rennes, EHESP, Inserm, IRSET (Institut de Recherche en Santé, Environnement et Travail)–UMR_S 1085, Rennes, France
| | - Michel Cot
- UMR 261-MERIT, Institut de Recherche pour le Développement (IRD), Université Paris Cité, Paris, France
| | - Jules Alao
- Service de Pédiatrie, CHU de la Mère et de l’Enfant-Lagune de Cotonou, Cotonou, Benin
| | - Nadine Fievet
- UMR 261-MERIT, Institut de Recherche pour le Développement (IRD), Université Paris Cité, Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Abomey-Calavi, Benin
| | | | - Florence Bodeau-Livinec
- UMR 261-MERIT, Institut de Recherche pour le Développement (IRD), Université Paris Cité, Paris, France
- Université Rennes, EHESP, Inserm, IRSET (Institut de Recherche en Santé, Environnement et Travail)–UMR_S 1085, Rennes, France
| |
Collapse
|
21
|
Li N, Du J, Yang Y, Zhao T, Wu D, Peng F, Wang D, Kong L, Zhou W, Hao A. Microglial PCGF1 alleviates neuroinflammation associated depressive behavior in adolescent mice. Mol Psychiatry 2025; 30:914-926. [PMID: 39215186 PMCID: PMC11835731 DOI: 10.1038/s41380-024-02714-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Epigenetics plays a crucial role in regulating gene expression during adolescent brain maturation. In adolescents with depression, microglia-mediated chronic neuroinflammation may contribute to the activation of cellular signaling cascades and cause central synapse loss. However, the exact mechanisms underlying the epigenetic regulation of neuroinflammation leading to adolescent depression remain unclear. In this study, we found that the expression of polycomb group 1 (PCGF1), an important epigenetic regulator, was decreased both in the plasma of adolescent major depressive disorder (MDD) patients and in the microglia of adolescent mice in a mouse model of depression. We demonstrated that PCGF1 alleviates neuroinflammation mediated by microglia in vivo and in vitro, reducing neuronal damage and improving depression-like behavior in adolescent mice. Mechanistically, PCGF1 inhibits the transcription of MMP10 by upregulating RING1B/H2AK119ub and EZH2/H3K27me3 in the MMP10 promoter region, specifically inhibiting microglia-mediated neuroinflammation. These results provide valuable insights into the pathogenesis of adolescent depression, highlighting potential links between histone modifications, neuroinflammation and nerve damage. Potential mechanisms of microglial PCGF1 regulates depression-like behavior in adolescent mice. Microglial PCGF1 inhibits NF-κB/MAPK pathway activation through regulation of RING1B/H2AK119ub and EZH2/H3K27me3 in the MMP10 promoter region, which attenuates neuroinflammation and ameliorates depression-like behaviors in adolescent mice.
Collapse
Affiliation(s)
- Naigang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingyi Du
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Yang
- Childhood Psychiatry Unit, Shandong Mental Health Center, Jinan, China
| | - Tiantian Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dong Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fan Peng
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dongshuang Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Linghua Kong
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Wenjuan Zhou
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Aijun Hao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
22
|
Erboz A, Kesekler E, Gentili PL, Uversky VN, Coskuner-Weber O. Electromagnetic radiation and biophoton emission in neuronal communication and neurodegenerative diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:87-99. [PMID: 39732343 DOI: 10.1016/j.pbiomolbio.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
The intersection of electromagnetic radiation and neuronal communication, focusing on the potential role of biophoton emission in brain function and neurodegenerative diseases is an emerging research area. Traditionally, it is believed that neurons encode and communicate information via electrochemical impulses, generating electromagnetic fields detectable by EEG and MEG. Recent discoveries indicate that neurons may also emit biophotons, suggesting an additional communication channel alongside the regular synaptic interactions. This dual signaling system is analyzed for its potential in synchronizing neuronal activity and improving information transfer, with implications for brain-like computing systems. The clinical relevance is explored through the lens of neurodegenerative diseases and intrinsically disordered proteins, where oxidative stress may alter biophoton emission, offering clues for pathological conditions, such as Alzheimer's and Parkinson's diseases. The potential therapeutic use of Low-Level Laser Therapy (LLLT) is also examined for its ability to modulate biophoton activity and mitigate oxidative stress, presenting new opportunities for treatment. Here, we invite further exploration into the intricate roles the electromagnetic phenomena play in brain function, potentially leading to breakthroughs in computational neuroscience and medical therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Aysin Erboz
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Elif Kesekler
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Pier Luigi Gentili
- Department of Chemistry, Biology, and Biotechnology, Università degli Studi di Perugia, 06123, Perugia, Italy.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC07, Tampa, FL 33612, USA.
| | - Orkid Coskuner-Weber
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey.
| |
Collapse
|
23
|
Hassan M, Flanagan TW, Eshaq AM, Altamimi OK, Altalag H, Alsharif M, Alshammari N, Alkhalidi T, Boulifa A, El Jamal SM, Haikel Y, Megahed M. Reduction of Prostate Cancer Risk: Role of Frequent Ejaculation-Associated Mechanisms. Cancers (Basel) 2025; 17:843. [PMID: 40075690 PMCID: PMC11898507 DOI: 10.3390/cancers17050843] [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: 01/17/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Prostate cancer (PCa) accounts for roughly 15% of diagnosed cancers among men, with disease incidence increasing worldwide. Age, family history and ethnicity, diet, physical activity, and chemoprevention all play a role in reducing PCa risk. The prostate is an exocrine gland that is characterized by its multi-functionality, being involved in reproductive aspects such as male ejaculation and orgasmic ecstasy, as well as playing key roles in the regulation of local and systemic concentrations of 5α-dihydrotestosterone. The increase in androgen receptors at the ventral prostate is the first elevated response induced by copulation. The regulation of prostate growth and function is mediated by an androgen-dependent mechanism. Binding 5-DHT to androgen receptors (AR) results in the formation of a 5α-DHT:AR complex. The interaction of the 5α-DHT:AR complex with the specific DNA enhancer element of androgen-regulated genes leads to the regulation of androgen-specific target genes to maintain prostate homeostasis. Consequently, ejaculation may play a significant role in the reduction of PCa risk. Thus, frequent ejaculation in the absence of risky sexual behavior is a possible approach for the prevention of PCa. In this review, we provide an insight into possible mechanisms regulating the impact of frequent ejaculation on reducing PCa risk.
Collapse
Affiliation(s)
- Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Abdulaziz M. Eshaq
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA
| | - Osama K. Altamimi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.K.A.); (H.A.); (M.A.); (N.A.); (T.A.)
| | - Hassan Altalag
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.K.A.); (H.A.); (M.A.); (N.A.); (T.A.)
| | - Mohamed Alsharif
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.K.A.); (H.A.); (M.A.); (N.A.); (T.A.)
| | - Nouf Alshammari
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.K.A.); (H.A.); (M.A.); (N.A.); (T.A.)
| | - Tamadhir Alkhalidi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.K.A.); (H.A.); (M.A.); (N.A.); (T.A.)
| | - Abdelhadi Boulifa
- Berlin Institute of Health, Charité University Hospital, 10117 Berlin, Germany;
- Competence Center of Immuno-Oncology and Translational Cell Therapy (KITZ), Charité-University Hospital, 10117 Berlin, Germany
| | - Siraj M. El Jamal
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA;
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| |
Collapse
|
24
|
Bhatoa RS, Nijjar S, Bathelt J, de Haan M. The impact of gestational age on executive function in infancy and early-to-middle childhood following preterm birth: a systematic review. Child Neuropsychol 2025:1-41. [PMID: 40012110 DOI: 10.1080/09297049.2025.2467950] [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: 05/10/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Lower gestational age (GA) is a risk factor for cognitive and developmental concerns following preterm birth. However, its impact on executive function (EF) is unclear based on conflicting conclusions across the literature. Moreover, as children below 4 years have largely been neglected from previous reviews, the impact of GA on EF within this early developmental period remains unclear. Hence, this systematic review investigated the impact of GA on EF following preterm birth in infancy and early-to-middle childhood. PubMed, Web of Science, and PsycInfo were searched for articles investigating the impact of GA on EF (inhibition, working memory, shifting) in preterm-born (<37 week gestation) and term-born participants aged 0-10 years. Eighteen studies were included. Most of the studies (n = 10) found no significant association between EF and GA. However, several limitations hindered conclusions to be drawn about the strength of this interpretation. Examples include inconsistencies in the theoretical underpinnings and operationalisations of EF, discrepancies in the reporting and measurement of GA, recruitment biases, and a paucity of infant or longitudinal studies available. Consequently, these issues may have contributed to inconsistent or null findings, and they must be addressed in future research to better clarify the impact of GA on EF in preterm-born infants and children.
Collapse
Affiliation(s)
- Raj Seraya Bhatoa
- Department of Psychology, Royal Holloway University London, Egham, UK
| | - Simrit Nijjar
- EGA Institute for Women's Health, University College London, London, UK
| | - Joe Bathelt
- Department of Psychology, Royal Holloway University London, Egham, UK
- Department of Psychology, University of Amsterdam, Amsterdam, Netherlands
| | - Michelle de Haan
- UCL Great Ormond Street Institute of Child Health, University College London, Egham, UK
| |
Collapse
|
25
|
Tu JC, Myers M, Li W, Li J, Wang X, Dierker D, Day TKM, Snyder AZ, Latham A, Kenley JK, Sobolewski CM, Wang Y, Labonte AK, Feczko E, Kardan O, Moore LA, Sylvester CM, Fair DA, Elison JT, Warner BB, Barch DM, Rogers CE, Luby JL, Smyser CD, Gordon EM, Laumann TO, Eggebrecht AT, Wheelock MD. The Generalizability of Cortical Area Parcellations Across Early Childhood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.09.612056. [PMID: 39314355 PMCID: PMC11419084 DOI: 10.1101/2024.09.09.612056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The cerebral cortex consists of distinct areas that develop through intrinsic embryonic patterning and postnatal experiences. Accurate parcellation of these areas in neuroimaging studies improves statistical power and cross-study comparability. Given significant brain changes in volume, microstructure, and connectivity during early life, we hypothesized that cortical areas in 1- to 3-year-olds would differ markedly from neonates and increasingly resemble adult patterns as development progresses. Here, we parcellated the cerebral cortex into putative areas using local functional connectivity gradients in 92 toddlers at 2 years old. We demonstrate high reproducibility of these cortical regions across 1- to 3-year-olds in two independent datasets. The area boundaries in 1- to 3-year-olds were more similar to those in adults than those in neonates. While the age-specific group area parcellation better fit the underlying functional connectivity in individuals during the first 3 years, adult area parcellations might still have some utility in developmental studies, especially in children older than 6 years. Additionally, we provide connectivity-based community assignments of the parcels, showing fragmented anterior and posterior components based on the strongest connectivity, yet alignment with adult systems when weaker connectivity was included.
Collapse
Affiliation(s)
| | - Michael Myers
- Department of Psychiatry, Washington University in St. Louis
| | - Wei Li
- Department of Mathematics and Statistics, Washington University in St. Louis
| | - Jiaqi Li
- Department of Mathematics and Statistics, Washington University in St. Louis
- Department of Statistics, University of Chicago
| | - Xintian Wang
- Department of Radiology, Washington University in St. Louis
| | - Donna Dierker
- Department of Radiology, Washington University in St. Louis
| | - Trevor K M Day
- Masonic Institute for the Developing Brain, University of Minnesota
- Institute of Child Development, University of Minnesota
- Center for Brain Plasticity and Recovery, Georgetown University
| | | | - Aidan Latham
- Department of Neurology, Washington University in St. Louis
| | | | - Chloe M Sobolewski
- Department of Radiology, Washington University in St. Louis
- Department of Psychology, Virginia Commonwealth University
| | - Yu Wang
- Department of Mathematics and Statistics, Washington University in St. Louis
| | | | - Eric Feczko
- Masonic Institute for the Developing Brain, University of Minnesota
| | - Omid Kardan
- Department of Psychiatry, University of Michigan
| | - Lucille A Moore
- Masonic Institute for the Developing Brain, University of Minnesota
| | | | - Damien A Fair
- Masonic Institute for the Developing Brain, University of Minnesota
- Institute of Child Development, University of Minnesota
| | - Jed T Elison
- Masonic Institute for the Developing Brain, University of Minnesota
- Institute of Child Development, University of Minnesota
| | | | - Deanna M Barch
- Department of Psychological and Brain Sciences, Washington University in St Louis
| | | | - Joan L Luby
- Department of Psychiatry, Washington University in St. Louis
| | - Christopher D Smyser
- Department of Radiology, Washington University in St. Louis
- Department of Psychiatry, Washington University in St. Louis
- Department of Neurology, Washington University in St. Louis
- Department of Pediatrics, Washington University in St. Louis
| | - Evan M Gordon
- Department of Radiology, Washington University in St. Louis
| | | | | | | |
Collapse
|
26
|
Garcia-Martínez T, Gornatti DG, Ortiz M, Cañellas G, Heine-Suñer D, Vives-Bauzà C. The Triad of Blood-Brain Barrier Integrity: Endothelial Cells, Astrocytes, and Pericytes in Perinatal Stroke Pathophysiology. Int J Mol Sci 2025; 26:1886. [PMID: 40076511 PMCID: PMC11900453 DOI: 10.3390/ijms26051886] [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: 01/29/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Pediatric stroke, a significant cause of long-term neurological deficits in children, often arises from disruptions within neurovascular unit (NVU) components. The NVU, a dynamic ensemble of astrocytes, endothelial cells, pericytes, and microglia, is vital for maintaining cerebral homeostasis and regulating vascular brain development. Its structural integrity, particularly at the blood-brain barrier (BBB), depends on intercellular junctions and the basement membrane, which together restrict paracellular transport and shield the brain from systemic insults. Dysfunction in this intricate system is increasingly linked to pediatric stroke and related cerebrovascular conditions. Mutations disrupting endothelial cell adhesion or pericyte-endothelial interactions can compromise BBB stability, leading to pathological outcomes such as intraventricular hemorrhage in the germinal matrix, a hallmark of vascular brain immaturity. Additionally, inflammation, ferroptosis, necroptosis, and autophagy are key cellular processes influencing brain damage and repair. Excessive activation of these mechanisms can exacerbate NVU injury, whereas targeted therapeutic modulation offers potential pathways to mitigate damage and support recovery. This review explores the cellular and molecular mechanisms underlying NVU dysfunction, BBB disruption, and subsequent brain injury in pediatric stroke. Understanding the interplay between genetic mutations, environmental stressors, and NVU dynamics provides new insights into stroke pathogenesis. The susceptibility of the germinal matrix to vascular rupture further emphasizes the critical role of NVU integrity in early brain development. Targeting inflammatory pathways and cell death mechanisms presents promising strategies to preserve NVU function and improve outcomes for affected neonates.
Collapse
Affiliation(s)
- Tania Garcia-Martínez
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Denise G. Gornatti
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Marina Ortiz
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Guillem Cañellas
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), 07122 Palma, Spain
| | - Damià Heine-Suñer
- Genomics of Health Research, Unit of Molecular Diagnostics and Clinical Genetics, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07013 Palma, Spain;
| | - Cristòfol Vives-Bauzà
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), 07122 Palma, Spain
| |
Collapse
|
27
|
Dos Santos AAA, Torrezan R, Rodrigues WDNDS, Ribeiro MVG, Ferreira ARO, Peres MNC, Saavedra LPJ, Raposo SR, Almeida DL, Malta A, Mathias PCDF. High-fat diet during lactation, as opposed to during adolescence or gestation, programs cardiometabolic and autonomic dysfunctions in adult offspring. Brain Res 2025; 1849:149354. [PMID: 39603318 DOI: 10.1016/j.brainres.2024.149354] [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: 05/15/2024] [Revised: 09/06/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
The Developmental Origins of Health and Disease (DOHaD) concept has been established for three decades. Many studies have shown that, besides pregnancy, other plastic phases (mainly preconception, lactation, and infancy-adolescence) are also sensitive to environmental changes, including nutritional conditions, that can program health or disease later in life. This study compared the susceptibility of the gestation, lactation and adolescence to a high-fat diet (HFD) intervention to program rats into autonomic nervous system imbalance and cardiometabolic dysfunction in adulthood. Four groups of rats were studied: offspring from mothers exposed to a HFD (35% fat) or a standard chow diet (4.5% fat) during gestation (GEST and CONT groups, respectively), offspring from mothers exposed to the HFD during lactation (LAC), and adolescent rats exposed to the HFD (ADOL). Mothers treated during pregnancy exhibited a higher body mass, but nursing mothers presented the highest food energy intake and higher adiposity. Compared to the other groups, the LAC rats showed increased body mass gain, food energy intake, body fat, glucose intolerance and blood pressure. LAC group also showed increased parasympathetic activity. In contrast, LAC sympathetic nerve activity decreased compared with the other groups. The ADOL group exhibited mostly similar responses but of a smaller magnitude. This suggests that the lactation phase is the most sensitive to HFD programming for cardiometabolic dysfunction in adulthood and that early overnutrition may affect neural connections by altering the autonomic nervous system balance.
Collapse
Affiliation(s)
- Annie Araújo Alves Dos Santos
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | - Rosana Torrezan
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | | | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | - Maria Natália Chimirri Peres
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | - Scarlett Rodrigues Raposo
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | - Douglas Lopes Almeida
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| | - Ananda Malta
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil.
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Experimental DOHaD, Department of Biotechnology, Genetics, and Cell Biology, State University of Maringá, Maringá 87090-020, Brazil
| |
Collapse
|
28
|
Eisen A, Kiernan MC. The Neonatal Microbiome: Implications for Amyotrophic Lateral Sclerosis and Other Neurodegenerations. Brain Sci 2025; 15:195. [PMID: 40002527 PMCID: PMC11852589 DOI: 10.3390/brainsci15020195] [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: 01/13/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Most brain development occurs in the "first 1000 days", a critical period from conception to a child's second birthday. Critical brain processes that occur during this time include synaptogenesis, myelination, neural pruning, and the formation of functioning neuronal circuits. Perturbations during the first 1000 days likely contribute to later-life neurodegenerative disease, including sporadic amyotrophic lateral sclerosis (ALS). Neurodevelopment is determined by many events, including the maturation and colonization of the infant microbiome and its metabolites, specifically neurotransmitters, immune modulators, vitamins, and short-chain fatty acids. Successful microbiome maturation and gut-brain axis function depend on maternal factors (stress and exposure to toxins during pregnancy), mode of delivery, quality of the postnatal environment, diet after weaning from breast milk, and nutritional deficiencies. While the neonatal microbiome is highly plastic, it remains prone to dysbiosis which, once established, may persist into adulthood, thereby inducing the development of chronic inflammation and abnormal excitatory/inhibitory balance, resulting in neural excitation. Both are recognized as key pathophysiological processes in the development of ALS.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Matthew C. Kiernan
- Neuroscience Research Australia, University of New South Wales, Randwick, Sydney, NSW 2031, Australia;
| |
Collapse
|
29
|
Risk B, Li L, Jones W, Shultz S. Dynamics of infant white matter maturation from birth to 6 months. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638114. [PMID: 39990497 PMCID: PMC11844443 DOI: 10.1101/2025.02.13.638114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The first months after a baby's birth encompass the most rapid period of postnatal change in the human lifespan, but longitudinal trajectories of white matter maturation in this period remain uncharted. Using densely sampled diffusion tensor images collected longitudinally at a mean rate of 1 scan per 1.55 days, we measured non-linear growth and growth rate trajectories of major white matter tracts from birth to 6 months. Growth rates at birth were 6 to 11 times faster than at 6 months, with tracts less mature at birth developing fastest. When matched on chronological age, shorter gestation infants had less mature white matter at birth but faster growth rates than their longer gestation peers; however, growth trajectories were highly similar when corrected for gestational age. This is the first study to estimate white matter trajectories using dense sampling in the first 6 post-natal months, which can inform the study of neurodevelopmental disorders beginning in infancy.
Collapse
Affiliation(s)
- Benjamin Risk
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Longchuan Li
- Marcus Autism Center, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Warren Jones
- Marcus Autism Center, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sarah Shultz
- Marcus Autism Center, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
30
|
Bu S, Li X, Pang H, Zhao M, Wang J, Liu Y, Yu H, Jiang Y, Fan G. Motor Functional Hierarchical Organization of Cerebrum and Its Underlying Genetic Architecture in Parkinson's Disease. J Neurosci 2025; 45:e1492242024. [PMID: 39824632 PMCID: PMC11823334 DOI: 10.1523/jneurosci.1492-24.2024] [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: 08/06/2024] [Revised: 11/02/2024] [Accepted: 12/05/2024] [Indexed: 01/20/2025] Open
Abstract
Hierarchy has been identified as a principle underlying the organization of human brain networks. However, it remains unclear how the network hierarchy is disrupted in Parkinson's disease (PD) motor symptoms and how it is modulated by the underlying genetic architecture. The aim of this study was to explore alterations in the motor functional hierarchical organization of the cerebrum and their underlying genetic mechanism. In this study, the brain network hierarchy of each group was described through a connectome gradient analysis among 68 healthy controls (HC), 70 postural instability and gait difficulty (PIGD) subtype, and 69 tremor-dominant (TD) subtype, including both male and female participants, according to its motor symptoms. Furthermore, transcription-neuroimaging association analyses using gene expression data from Allen Human Brain Atlas and case-control gradient differences were performed to identify genes associated with gradient alterations. Different PD motor subtypes exhibited contracted principal and secondary functional gradients relative to HC. The identified genes in different PD motor subtypes enriched for shared biological processes like metal ion transport and inorganic ion transmembrane transport. In addition, these genes were overexpressed in Ntsr+ neurons cell, enriched in extensive cortical regions and wide developmental time windows. Aberrant cerebral functional gradients in PD-related motor symptoms have been detected, and the motor-disturbed genes have shared biological functions. The present findings may contribute to a more comprehensive understanding of the molecular mechanisms underlying hierarchical alterations in PD.
Collapse
Affiliation(s)
- Shuting Bu
- Departments of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaolu Li
- Departments of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Huize Pang
- Departments of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Mengwan Zhao
- Departments of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Juzhou Wang
- Departments of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yu Liu
- Departments of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Hongmei Yu
- Neurology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yueluan Jiang
- MR Research Collaboration, Siemens Healthineers, Beijing 100102, China
| | - Guoguang Fan
- Departments of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
31
|
Cendra-Duarte E, Canals J, Becerra-Tomás N, Mateu-Fabregat J, Bulló M, Arija V. Dietary glycemic index and load during pregnancy and offspring behavioral outcomes: exploring sex differences. Eur J Pediatr 2025; 184:178. [PMID: 39909936 PMCID: PMC11799018 DOI: 10.1007/s00431-025-06005-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
Given the importance of carbohydrates during pregnancy and the limited evidence on the impact of its excessive intake on offspring neurodevelopment, this study aimed to assess the associations between maternal glycemic index (GI) and glycemic load (GL) during early and late pregnancy and behavior problems in 4-year-old children, considering potential sex-related differences in susceptibility to maternal diet. This observational study included 188 mother-child pairs from the ECLIPSES study. GI and GL were estimated from a validated food frequency questionnaire. Offspring behavior was assessed using the Child Behavior Checklist 1.5-5. Multivariable linear and logistic regression analyses were employed to assess the association between GI, GL, and child behavior. Children of mothers in the highest tertile of GL during the first trimester of pregnancy showed elevated scores of both internalizing (β = 5.77; 95% CI, 2.28-9.26) and externalizing (β = 3.95; 95% CI, 0.70-7.19) problems, including anxiety and depression problems, withdrawn, attention problems, aggressive behavior, and attention-deficit/hyperactivity problems, as well as total (β = 5.24; 95% CI, 1.71-8.77) and autism spectrum problems (β = 3.30; 95% CI, 1.11-5.50). Similarly, higher odd ratios were observed for internalizing (OR = 2.37; 95% CI, 1.09-5.18), externalizing (OR = 3.46; 95% CI, 1.49-8.00), and total problems (OR = 3.83; 95% CI, 1.68-8.71). These associations were more pronounced in girls. No associations were observed during the third trimester. Regarding GI, no associations were found for the evaluated outcomes in any of the trimesters. CONCLUSION These findings indicated that elevated maternal GL during the early pregnancy, but not later stages, was associated with adverse behavioral outcomes in offspring. TRIAL REGISTRATION EUCTR-2012-005480-28, NCT03196882. WHAT IS KNOWN • Carbohydrate intake is important during pregnancy as glucose is the main energy source for an optimal fetal brain development. • Elevated prenatal glycemic index and glycemic load have been associated with adverse offspring outcomes but their impact on behavioral development remains insufficiently explored. WHAT IS NEW • A high maternal glycemic load during pregnancy may increase the risk of behavioral impairments in preschool-aged offspring. • Female offspring may be more vulnerable to behavioral disturbances to elevated maternal glycemic load during gestation.
Collapse
Affiliation(s)
- Esther Cendra-Duarte
- Universitat Rovira i Virgili, Unitat de Salut Pública i Epidemiologia Nutricional, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain
- Collaborative Group On Lifestyles, Nutrition, and Tobacco (CENIT), Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Institut Català de La Salut (ICS), Reus, Spain
| | - Josefa Canals
- Universitat Rovira i Virgili, Unitat de Salut Pública i Epidemiologia Nutricional, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Universitat Rovira i Virgili, Centre de Recerca en Avaluació i Mesura de La Conducta (CRAMC), Department of Psychology, Tarragona, Spain
| | - Nerea Becerra-Tomás
- Universitat Rovira i Virgili, Unitat de Salut Pública i Epidemiologia Nutricional, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Javier Mateu-Fabregat
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Universitat Rovira i Virgili, Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Reus, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Rovira i Virgili University, Reus, Spain
| | - Mònica Bulló
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Universitat Rovira i Virgili, Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Reus, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Rovira i Virgili University, Reus, Spain
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid, Spain
| | - Victoria Arija
- Universitat Rovira i Virgili, Unitat de Salut Pública i Epidemiologia Nutricional, Nutrition and Mental Health (NUTRISAM) Research Group, Reus, Spain.
- Collaborative Group On Lifestyles, Nutrition, and Tobacco (CENIT), Institut d'Investigació en Atenció Primària IDIAP Jordi Gol, Institut Català de La Salut (ICS), Reus, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.
| |
Collapse
|
32
|
Byeon K, Park H, Park S, Cluce J, Mehta K, Cieslak M, Cui Z, Hong SJ, Chang C, Smallwood J, Satterthwaite TD, Milham MP, Xu T. Developmental Variations in Recurrent Spatiotemporal Brain Propagations from Childhood to Adulthood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.04.635765. [PMID: 39975397 PMCID: PMC11838599 DOI: 10.1101/2025.02.04.635765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The brain undergoes profound structural and functional transformations from childhood to adolescence. Convergent evidence suggests that neurodevelopment proceeds in a hierarchical manner, characterized by heterogeneous maturation patterns across brain regions and networks. However, the maturation of the intrinsic spatiotemporal propagations of brain activity remains largely unexplored. This study aims to bridge this gap by delineating spatiotemporal propagations from childhood to early adulthood. By leveraging a recently developed approach that captures time-lag dynamic propagations, we characterized intrinsic dynamic propagations along three axes: sensory-association (S-A), 'task-positive' to default networks (TP-D), and somatomotor-visual (SM-V) networks, which progress towards adult-like brain dynamics from childhood to early adulthood. Importantly, we demonstrated that as participants mature, there is a prolonged occurrence of the S-A and TP-D propagation states, indicating that they spend more time in these states. Conversely, the prevalence of SM-V propagation states declines during development. Notably, top-down propagations along the S-A axis exhibited an age-dependent increase in occurrence, serving as a superior predictor of cognitive scores compared to bottom-up S-A propagation. These findings were replicated across two independent cohorts (N = 677 in total), emphasizing the robustness and generalizability of these findings. Our results provide new insights into the emergence of adult-like functional dynamics during youth and their role in supporting cognition.
Collapse
Affiliation(s)
| | - Hyunjin Park
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, South Korea
- IBS Center for Neuroscience Imaging Research, Sungkyunkwan University, Suwon, South Korea
| | - Shinwon Park
- Child Mind Institute, New York, NY, United States
| | - Jon Cluce
- Child Mind Institute, New York, NY, United States
| | - Kahini Mehta
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn-CHOP Lifespan Brain Institute, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Cieslak
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn-CHOP Lifespan Brain Institute, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zaixu Cui
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Seok-Jun Hong
- Child Mind Institute, New York, NY, United States
- IBS Center for Neuroscience Imaging Research, Sungkyunkwan University, Suwon, South Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, South Korea
| | - Catie Chang
- Departments of Electrical and Computer Engineering, Computer Science, and Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | | | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn-CHOP Lifespan Brain Institute, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Milham
- Child Mind Institute, New York, NY, United States
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Ting Xu
- Child Mind Institute, New York, NY, United States
| |
Collapse
|
33
|
Mofatteh M, Mohamed A, Mashayekhi MS, Skandalakis GP, Neudorfer C, Arfaie S, MohanaSundaram A, Sabahi M, Anand A, Aboulhosn R, Liao X, Horn A, Ashkan K. Deep brain stimulation of the hypothalamic region: a systematic review. Acta Neurochir (Wien) 2025; 167:33. [PMID: 39904782 PMCID: PMC11794333 DOI: 10.1007/s00701-025-06430-w] [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: 12/06/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Deep brain stimulation (DBS) has been successfully used for the treatment of circuitopathies including movement, anxiety, and behavioral disorders. The hypothalamus is a crucial integration center for many peripheral and central pathways relating to cardiovascular, metabolic, and behavioral functions and constitutes a potential target for neuromodulation in treatment-refractory conditions. To conduct a systematic review, investigating hypothalamic targets in DBS, their indications, and the primary clinical findings. METHODS PubMed, Scopus, and Web of Science databases were searched in accordance with the PRISMA guideline to identify papers published in English studying DBS of the hypothalamus in humans. RESULTS After screening 3,148 papers, 34 studies consisting of 412 patients published over two decades were included in the final review. Hypothalamic DBS was indicated in refractory headaches (n = 238, 57.8%), aggressive behavior (n = 100, 24.3%), mild Alzheimer's disease (n = 58, 14.1%), trigeminal neuralgia in multiple sclerosis (n = 5, 1.2%), Prader-Willi syndrome (n = 4, 0.97%), and atypical facial pain (n = 3, 0.73%). The posterior hypothalamus was the most common DBS target site across 30 studies (88.2%). 262 (63.6%) participants were males, and 110 (26.7%) were females. 303 (73.5%) patients were adults whereas 33 (8.0%) were pediatrics. The lowest mean age of participants was 15.25 ± 4.6 years for chronic refractory aggressiveness, and the highest was 68.5 ± 7.9 years in Alzheimer's disease patients. The mean duration of the disease ranged from 2.2 ± 1.7 (mild Alzheimer's disease) to 19.8 ± 10.1 years (refractory headaches). 213 (51.7%) patients across 29 studies (85.3%) reported symptom improvements which ranged from 23.1% to 100%. 25 (73.5%) studies reported complications, most of which were associated with higher voltage stimulations. CONCLUSIONS DBS of the hypothalamus is feasible in selected patients with various refractory conditions ranging from headaches to aggression in both pediatric and adult populations. Future large-scale studies with long-term follow-up are required to validate the safety and efficacy data and extend these findings.
Collapse
Affiliation(s)
- Mohammad Mofatteh
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
- Neuro International Collaboration (NIC), London, UK.
| | - Abdulkadir Mohamed
- Medical Sciences Division, University of Oxford, Oxford, UK
- Neuro International Collaboration (NIC), Oxford, UK
| | - Mohammad Sadegh Mashayekhi
- Faculty of Medicine, Division of Neurosurgery, University of Ottawa, Ottawa, ON, Canada
- Neuro International Collaboration (NIC), Vancouver, Ottawa, ON, Canada
| | - Georgios P Skandalakis
- Department of Neurosurgery, Evangelismos General Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Clemens Neudorfer
- Brain Modulation Lab, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité -Universitätsmedizin Berlin, corporate member of, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Saman Arfaie
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Division of Neurosurgery, Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Neuro International Collaboration (NIC), Montreal, QC, Canada
| | | | - Mohammadmahdi Sabahi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Ayush Anand
- Koirala Institute of Health Sciences, B. P, Dharan, Nepal
| | | | - Xuxing Liao
- Department of Neurosurgery, First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Andreas Horn
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Center for Brain Circuit Therapeutics, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Departments of Neurology and Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Keyoumars Ashkan
- Neuro International Collaboration (NIC), London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- King's Health Partners Academic Health Sciences Centre, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
34
|
Babiszewska-Aksamit M, Szołtysik M, Apanasewicz A, Piosek M, Winczowska P, Cierniak A, Danel DP, Ziomkiewicz A. The Concentration of Docosahexaenoic Acid (DHA) and Arachidonic Acid (AA) in Human Milk Is Associated With the Size of Maternal Social Network. Am J Hum Biol 2025; 37:e70006. [PMID: 39915968 DOI: 10.1002/ajhb.70006] [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: 05/07/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION Social factors, such as kin and non-kin support in helping the mother of a newborn baby, impact the duration of lactation and may affect human milk composition. Recent studies suggest that maternal stress negatively affects the level of polyunsaturated fatty acids in human milk, which are crucial for infant vision and brain development. We suggest that social support may have the potential to attenuate a negative effect of stress on the composition of human milk fatty acids. METHODS We studied 129 exclusively breastfeeding mothers and their healthy, term infants to explore the relationship between support from significant others (structural and functional) and the concentration of docosahexaenoic acid (DHA, N = 49) and arachidonic acid (AA, N = 129) in human milk. We also examined whether maternal stress reactivity (log Cort. AUC) may be related to these fatty acids. Gas chromatography was used to analyze the concentration of DHA and AA in human milk samples. RESULTS Analyses revealed a positive association between the number of helpers (structural support) and the concentration of DHA and AA. Maternal stress reactivity was not a statistically significant predictor of DHA and AA contents in milk and was unrelated to the number of helpers. CONCLUSION Our results show for the first time that human milk composition, particularly DHA and AA concentrations, may be associated with the size of mothers' immediate social network of kin and non-kin helpers. This result is consistent with evolutionary studies that emphasize the role of cooperative breeding in human reproduction.
Collapse
Affiliation(s)
- Magdalena Babiszewska-Aksamit
- Department of Anthropology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wroclaw, Poland
- Department of Medical Biology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Szołtysik
- Department of Functional Food Products Development, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Anna Apanasewicz
- Department of Anthropology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wroclaw, Poland
| | | | - Patrycja Winczowska
- Department of Anthropology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wroclaw, Poland
| | - Agnieszka Cierniak
- Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, Krakow, Poland
| | - Dariusz P Danel
- Department of Anthropology, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, Wroclaw, Poland
| | - Anna Ziomkiewicz
- Institute of Zoology and Biomedical Research, Laboratory of Anthropology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
35
|
Borzage MT, Peterson BS. A Scoping Review of the Mechanisms Underlying Developmental Anesthetic Neurotoxicity. Anesth Analg 2025; 140:409-426. [PMID: 38536739 PMCID: PMC11427602 DOI: 10.1213/ane.0000000000006897] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 09/28/2024]
Abstract
Although anesthesia makes painful or uncomfortable diagnostic and interventional health care procedures tolerable, it may also disrupt key cellular processes in neurons and glia, harm the developing brain, and thereby impair cognition and behavior in children. Many years of studies using in vitro, animal behavioral, retrospective database studies in humans, and several prospective clinical trials in humans have been invaluable in discerning the potential toxicity of anesthetics. The objective of this scoping review was to synthetize the evidence from preclinical studies for various mechanisms of toxicity across diverse experimental designs and relate their findings to those of recent clinical trials in real-world settings.
Collapse
Affiliation(s)
- Matthew Thomas Borzage
- From the Fetal and Neonatal Institute, Division of Neonatology, Children’s Hospital Los Angeles, Los Angeles, California
| | - Bradley S. Peterson
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Institute for the Developing Mind, Children’s Hospital Los Angeles, Los Angeles, California
- Department of Psychiatry, Keck School of Medicine at the University of Southern California, Los Angeles, California
| |
Collapse
|
36
|
Lin Q, Gao Y, Liu Y, Huang S, Su Y, Luo W, Shi C, Yang Y, Lin H, Su X, Zhang Z. Heat wave exposure during pregnancy and neurodevelopmental delay in young children: A birth cohort study. ENVIRONMENTAL RESEARCH 2025; 266:120541. [PMID: 39643258 DOI: 10.1016/j.envres.2024.120541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/16/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Gestation is a critical period for fetal brain development, and extreme heat exposure during this stage may have adverse impact on neurodevelopment in children. However, current evidence is scarce. METHODS We examined the associations between maternal exposure to heat wave during pregnancy and neurodevelopmental delay in young children in a birth cohort study of 67,453 child-mother pairs from Foshan, China. Specifically, temperature data (spatial resolution: 0.0625° × 0.0625°) were assigned to study participants based on residential addresses. Then, heat wave events were defined by combining the intensity (temperature thresholds: ≥90th, 92.5th or 95th percentile) and duration (number of consecutive days: 2, 3 or 4 days). Neurodevelopmental status was assessed using a five-domain scale by trained medical professionals. Logistic regression was used to investigate the associations between gestational heat wave exposure and neurodevelopmental delay in children. RESULTS We found that exposure to heat wave during early and late pregnancy was associated with increased risks of neurodevelopmental delay in children. By contrast, the results for mid-pregnancy heat wave exposure were mixed. The observed associations remained stable in a group of sensitivity analyses. CONCLUSIONS Our study adds some suggestive evidence that prenatal exposure to heat wave may have detrimental impact on children's neurodevelopment. More investigations are needed to verify our findings.
Collapse
Affiliation(s)
- Qingmei Lin
- Foshan Women and Children Hospital Affiliated to Guangdong Medical University, Foshan, 528315, China
| | - Yonggui Gao
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuewei Liu
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Saijun Huang
- Foshan Women and Children Hospital Affiliated to Guangdong Medical University, Foshan, 528315, China
| | - Yang Su
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Weidong Luo
- Foshan Women and Children Hospital Affiliated to Guangdong Medical University, Foshan, 528315, China
| | - Chunxiang Shi
- Meteorological Data Laboratory, National Meteorological Information Center, Beijing, 100081, China
| | - Yin Yang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xi Su
- Foshan Women and Children Hospital Affiliated to Guangdong Medical University, Foshan, 528315, China.
| | - Zilong Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
37
|
Jantz PB, Bigler ED. A case of severe TBI: Recovery? APPLIED NEUROPSYCHOLOGY. CHILD 2025:1-24. [PMID: 39874021 DOI: 10.1080/21622965.2025.2455115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Chronic stage neuropsychological assessments of children with severe TBI typically center around a referral question and focus on assessing cognitive, behavioral, and emotional functioning, making differential diagnoses, and planning treatment. When severe TBI-related neurological deficits are subtle and fall outside commonly assessed behavioral indicators, as can happen with theory of mind and social information processing, they can go unobserved and subsequently fail to be assessed. Additionally, should chronic stage cognitive, behavioral, and emotional assessment findings fall within the average to above average range, a child experiencing ongoing significant unassessed severe TBI-related subtle deficits could be mistakenly judged to have "recovered" from their injury; and to be experiencing no significant ongoing residual neurological deficits. To illustrate how this could happen, and how subacute neuroimaging and brain network theory might be early indicators of emergent chronic stage neuropsychological deficits, we present a child with a severe TBI and average to above average cognitive, behavioral, and emotional assessment findings who has comorbid significant deficits in theory of mind and social functioning.
Collapse
Affiliation(s)
- Paul B Jantz
- Department of Counseling, Leadership, Adult Education, and School Psychology, Texas State University, San Marcos, USA
| | - E D Bigler
- Department of Psychology and Neuroscience Center, Brigham Young University, Provo, USA
- Departments of Neurology, Psychiatry, and Radiology University of Utah Salt Lake City, UT, USA
| |
Collapse
|
38
|
Li H, Cui Z, Cieslak M, Salo T, Moore TM, Gur RE, Gur RC, Shinohara RT, Oathes DJ, Davatzikos C, Satterthwaite TD, Fan Y. Spatial Heterogeneity and Subtypes of Functional Connectivity Development in Youth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634828. [PMID: 39896611 PMCID: PMC11785253 DOI: 10.1101/2025.01.24.634828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The brain functional connectome development is fundamental to neurocognitive growth in youth. While brain age prediction has been widely used to assess connectome development at the individual level, traditional approaches providing a global index overlook the spatial variability and inter-individual heterogeneity of functional connectivity (FC) development across the cortex. In this study, we introduced a regional brain development index to assess spatially fine-grained FC development. We examined the spatial variability of FC development and stratified individuals into subtypes with distinct patterns of spatial heterogeneity in region-wise FC development across the cortex through clustering. An evaluation conducted on a sample of youths aged 8-23 years using fMRI data from the Philadelphia Neurodevelopmental Cohort (PNC) revealed three distinct FC development subtypes. Individuals with advanced FC development aligned with the hierarchical brain organization along the sensorimotor-association (S-A) axis demonstrated superior cognitive performance compared to those with other patterns. These patterns were replicated in the Human Connectome Project Development (HCP-D) cohort, confirming their robustness. Further analysis revealed associations between FC development and gene expression, with enriched genes linked to neural differentiation, synaptogenesis, and myelination. These findings suggest that spatial heterogeneity in FC development reflects underlying cortical microstructure and hierarchical cortical organization, underscoring its critical role in understanding neurocognitive maturation and individual variability during youth.
Collapse
Affiliation(s)
- Hongming Li
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zaixu Cui
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Matthew Cieslak
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Taylor Salo
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler M. Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell T. Shinohara
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Desmond J. Oathes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Brain Imaging and Stimulation, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Brain Science, Translation, Innovation, and Modulation Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Neuromodulation in Depression and Stress, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Neuroscience, Neurosurgery and Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theodore D. Satterthwaite
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yong Fan
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Nguyen QD, Kaiser RH, Snyder HR. Stress generation and subsequent repetitive negative thinking link poor executive functioning and depression. ANXIETY, STRESS, AND COPING 2025:1-15. [PMID: 39841611 DOI: 10.1080/10615806.2025.2450308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND AND OBJECTIVE Poor executive functioning (EF) has been consistently linked to depression, but questions remain regarding mechanisms driving this association. The current study tested whether poor EF is linked to depression symptoms six weeks later via dependent stressors (model 1) and stressors perceived to be uncontrollable (model 2) at week two (W2) and repetitive negative thinking (RNT) at W4 during early COVID-19 in college students. DESIGN This was a longitudinal study with four timepoints spanning six weeks (April-June 2020). METHODS Participants (N = 154) completed online questionnaires measuring EF, dependent stress frequency, stress controllability appraisals, brooding rumination, worry, and depression. RESULTS Supporting model 1, poorer baseline EF predicted higher dependent stress frequency at W2; W2 dependent stress frequency, in turn, predicted increases in W4 RNT, which predicted increases in W6 depression. Model 2 was not supported: Baseline EF did not predict W2 perceived stress uncontrollability, which did not predict W4 RNT; however, W4 RNT predicted increases in W6 depression. LIMITATIONS The sample was relatively small and EF was measured using only self-reports. CONCLUSIONS Findings supported a model in which poor EF conferred risk for depression via dependent stress and subsequent RNT, highlighting these processes as risk mechanisms for depression.
Collapse
Affiliation(s)
- Quynh D Nguyen
- Department of Psychology, Brandeis University, Waltham, MA, USA
| | - Roselinde H Kaiser
- Department of Psychology & Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Hannah R Snyder
- Department of Psychology, Brandeis University, Waltham, MA, USA
| |
Collapse
|
40
|
Berger PK, Bansal R, Sawardekar S, Monk C, Peterson BS. Associations of Maternal Prenatal Zinc Consumption with Infant Brain Tissue Organization and Neurodevelopmental Outcomes. Nutrients 2025; 17:303. [PMID: 39861433 PMCID: PMC11767866 DOI: 10.3390/nu17020303] [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: 12/02/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES While studies in rat pups suggest that early zinc exposure is critical for optimal brain structure and function, associations of prenatal zinc intake with measures of brain development in infants are unknown. This study aimed to assess the associations of maternal zinc intake during pregnancy with MRI measures of brain tissue microstructure and neurodevelopmental outcomes, as well as to determine whether MRI measures of the brain mediated the relationship between maternal zinc intake and neurodevelopmental indices. METHODS Forty-one adolescent mothers were recruited for a longitudinal study during pregnancy. Maternal zinc intake was assessed during the third trimester of pregnancy using a 24 h dietary recall. Infant MRI scans were acquired at 3 weeks postpartum using a 3.0 Tesla scanner to measure fractional anisotropy (FA) and mean diffusivity (MD). Cognitive, language, and motor skills were assessed at 4, 14, and 24 months postpartum using the Bayley Scales of Infant Development. RESULTS Greater prenatal zinc intake was associated with reduced FA in cortical gray matter, particularly in the frontal lobe [medial superior frontal gyrus; β (95% CI) = -1.0 (-1.5, -0.5)], in developing white matter, and in subcortical gray matter nuclei. Greater prenatal zinc intake was associated with reduced MD in cortical gray matter and developing white matter [superior longitudinal fasciculus; -4.4 (-7.1, -1.7)]. Greater maternal zinc intake also was associated with higher cognitive development scores at 14 [0.1 (0.0, 0.1)] and 24 [0.1 (0.0, 0.2)] months of age; MRI indices of FA and MD did not mediate this relationship. CONCLUSIONS Maternal prenatal zinc intake was associated with more favorable measures of brain tissue microstructural maturation and cognitive development during infancy.
Collapse
Affiliation(s)
- Paige K. Berger
- Department of Pediatrics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ravi Bansal
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Child & Adolescent Psychiatry, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - Siddhant Sawardekar
- Division of Child & Adolescent Psychiatry, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - Catherine Monk
- Departments of Obstetrics and Gynecology and Psychiatry, Columbia University Medical Center, New York, NY 10032, USA;
| | - Bradley S. Peterson
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Child & Adolescent Psychiatry, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| |
Collapse
|
41
|
Yuan Z, Su T, Yang L, Xi L, Wang HJ, Ji Y. Maternal Glycemia and Its Pattern Associated with Offspring Neurobehavioral Development: A Chinese Birth Cohort Study. Nutrients 2025; 17:257. [PMID: 39861387 PMCID: PMC11767945 DOI: 10.3390/nu17020257] [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: 11/30/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study investigates the impact of maternal glycemic levels during early and late pregnancy on offspring neurodevelopment in China. METHODS Fasting plasma glucose (FPG) and triglyceride (TG) levels were measured in maternal blood during pregnancy, and the TyG index was calculated to assess insulin resistance. Hyperglycemia was defined as FPG > 5.1 mmol/L. Neurodevelopmental outcomes in offspring aged 6-36 months were evaluated using the China Developmental Scale for Children, focusing on developmental delay (DD) and developmental quotient (DQ). Mothers were categorized into four glycemic groups: healthy glycemia group (HGG), early pregnancy hyperglycemia group (EHG), late pregnancy hyperglycemia group (LHG), and full-term hyperglycemia group (FHG). Linear and logistic regression models were applied. RESULTS Among 1888 mother-child pairs, hyperglycemia and FPG were associated with an increased risk of overall DD (aOR = 1.68; 95% CI 1.07-2.64) and lower DQ (aBeta = -1.53; 95% CI -2.70 to -0.36). Elevated FPG was linked to DD in fine motor and social behaviors. Compared to HGG, LHG and FHG significantly increased the risk of overall DD (aOR = 2.18; 95% CI 1.26-3.77; aOR = 2.64; 95% CI 1.38-5.05), whereas EHG did not. Male offspring were particularly vulnerable to early pregnancy hyperglycemia (aBeta = -2.80; 95% CI -4.36 to -1.34; aOR = 2.05; 95% CI 1.10-3.80). CONCLUSIONS Maternal glycemic levels during pregnancy influence offspring neurodevelopment, with persistent hyperglycemia significantly increasing DD risk. Early pregnancy hyperglycemia particularly affects male offspring, underscoring the need for glycemic management during pregnancy.
Collapse
Affiliation(s)
- Zhichao Yuan
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Tao Su
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Li Yang
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Lei Xi
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Hai-Jun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Yuelong Ji
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| |
Collapse
|
42
|
Shahabi B, Hernández-Martínez C, Jardí C, Aparicio E, Arija V. Maternal Omega-6/Omega-3 Concentration Ratio During Pregnancy and Infant Neurodevelopment: The ECLIPSES Study. Nutrients 2025; 17:170. [PMID: 39796604 PMCID: PMC11723212 DOI: 10.3390/nu17010170] [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: 12/05/2024] [Revised: 12/28/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND The balance of omega-6/omega-3 (n-6/n-3) is crucial for proper brain function as they have opposite physiological roles. OBJECTIVES To analyze the association between maternal serum ratios of n-6/n-3 in the first and third trimesters of pregnancy and the neurodevelopment of their children in the early days after birth in the population of Northern Spain's Mediterranean region. METHODS Longitudinal study in which 336 mother-child pairs participated. Mother serum concentrations of long-chain polyunsaturated fatty acids (LCPUFAs), docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (ARA) were determined. Sociodemographic, clinical, lifestyle habits, and obstetrical variables were collected. The Bayley Scales of Infant and Toddler Development (BSID-III) was used to assess infant neurodevelopment. Multiple linear regression models adjusting for confounding factors were performed. RESULTS In the third trimester, a higher maternal n-6/n-3 ratio was negatively associated with infant motor development (β = -0.124, p = 0.023). Similarly, higher ARA/DHA ratios were negatively associated with total motor (β = -2.005, p = 0.002) and fine motor development (β = -0.389, p = 0.001). No significant associations were observed in the first trimester nor for the ARA/EPA ratio in the third trimester. CONCLUSIONS Our findings indicate that an elevated n-6/n-3 ratio and ARA/DHA ratio in the third trimester of pregnancy are associated with poorer motor development outcomes in infants. These results highlight the importance of optimizing maternal fatty acid balance during pregnancy to support fetal neurodevelopment, suggesting a need for further research to verify these associations and elucidate underlying mechanisms.
Collapse
Affiliation(s)
- Behnaz Shahabi
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43201 Reus, Spain; (B.S.); (C.H.-M.); (C.J.); (E.A.)
| | - Carmen Hernández-Martínez
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43201 Reus, Spain; (B.S.); (C.H.-M.); (C.J.); (E.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43003 Tarragona, Spain
- Research Center for Behavioural Assessment (CRAMC), Department of Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Cristina Jardí
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43201 Reus, Spain; (B.S.); (C.H.-M.); (C.J.); (E.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43003 Tarragona, Spain
| | - Estefanía Aparicio
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43201 Reus, Spain; (B.S.); (C.H.-M.); (C.J.); (E.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43003 Tarragona, Spain
| | - Victoria Arija
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43201 Reus, Spain; (B.S.); (C.H.-M.); (C.J.); (E.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43003 Tarragona, Spain
| |
Collapse
|
43
|
Tarumi T, Tomoto T, Sugawara J, Zhang R. Aerobic Exercise Training for the Aging Brain: Effective Dosing and Vascular Mechanism. Exerc Sport Sci Rev 2025; 53:31-40. [PMID: 39254652 DOI: 10.1249/jes.0000000000000349] [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: 09/11/2024]
Abstract
This article presents evidence supporting the hypothesis that starting aerobic exercise in early adulthood and continuing it throughout life leads to significant neurocognitive benefits compared with starting exercise later in life. Regular aerobic exercise at moderate-to-vigorous intensity during midlife is associated with significant improvement in cardiorespiratory fitness, which may create a favorable brain microenvironment promoting neuroplasticity through enhanced vascular function.
Collapse
|
44
|
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
|
45
|
An WW, Bhowmik AC, Nelson CA, Wilkinson CL. EEG-based brain age prediction in infants-toddlers: Implications for early detection of neurodevelopmental disorders. Dev Cogn Neurosci 2025; 71:101493. [PMID: 39721149 PMCID: PMC11732522 DOI: 10.1016/j.dcn.2024.101493] [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: 07/29/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
The infant brain undergoes rapid developmental changes in the first three years of life. Understanding these changes through the prediction of chronological age using neuroimaging can provide insights into typical and atypical brain development. We utilized 938 resting-state EEG recordings from 457 typically developing infants, 2 to 38 months old, to develop age prediction models. The multilayer perceptron model demonstrated the highest accuracy with an R2 of 0.83 and a mean absolute error of 91.7 days. Feature importance analysis that combined hierarchical clustering and Shapley values identified two feature clusters describing periodic alpha and low beta activity as key predictors of age. Application of the model to EEG data from infants later diagnosed with autism or Down syndrome revealed significant underestimations of chronological age, supporting its potential as a clinical tool for early identification of alterations in brain development.
Collapse
Affiliation(s)
- Winko W An
- Developmental Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, 02115, MA, USA; Harvard Medical School, 25 Shattuck St, Boston, 02115, MA, USA
| | - Aprotim C Bhowmik
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, 11549, NY, USA; Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, 21205, MD, USA
| | - Charles A Nelson
- Developmental Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, 02115, MA, USA; Harvard Medical School, 25 Shattuck St, Boston, 02115, MA, USA; Harvard Graduate School of Education, 13 Appian Way, Cambridge, 02138, MA, USA
| | - Carol L Wilkinson
- Developmental Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, 02115, MA, USA; Harvard Medical School, 25 Shattuck St, Boston, 02115, MA, USA.
| |
Collapse
|
46
|
Sadegh-Zadeh SA, Bahrami M, Soleimani O, Ahmadi S. Neural reshaping: the plasticity of human brain and artificial intelligence in the learning process. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2024; 13:34-48. [PMID: 39850545 PMCID: PMC11751442 DOI: 10.62347/nhkd7661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/25/2024] [Indexed: 01/25/2025]
Abstract
This study explores the concept of neural reshaping and the mechanisms through which both human and artificial intelligence adapt and learn. OBJECTIVES To investigate the parallels and distinctions between human brain plasticity and artificial neural network plasticity, with a focus on their learning processes. METHODS A comparative analysis was conducted using literature reviews and machine learning experiments, specifically employing a multi-layer perceptron neural network to examine regression and classification problems. RESULTS Experimental findings demonstrate that machine learning models, similar to human neuroplasticity, enhance performance through iterative learning and optimization, drawing parallels in strengthening and adjusting connections. CONCLUSIONS Understanding the shared principles and limitations of neural and artificial plasticity can drive advancements in AI design and cognitive neuroscience, paving the way for future interdisciplinary innovations.
Collapse
Affiliation(s)
- Seyed-Ali Sadegh-Zadeh
- Department of Computing, School of Digital, Technologies and Arts, Staffordshire UniversityStoke-on-Trent ST4 2DE, UK
| | - Mahboobe Bahrami
- Behavioral Sciences Research Centre, School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| | | | - Sahar Ahmadi
- School of Electrical Engineering, Iran University of Science and TechnologyTehran, Iran
| |
Collapse
|
47
|
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
|
48
|
Vahidi H, Kowalczyk A, Stubbs K, Musabi M, Roychaudhuri S, Kent M, Bhattacharya S, de Ribaupierre S, Lawrence KS, Mohsenzadeh Y, Duerden EG. Investigating Task-Free Functional Connectivity Patterns in Newborns Using Functional Near-Infrared Spectroscopy. Brain Behav 2024; 14:e70180. [PMID: 39690863 DOI: 10.1002/brb3.70180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Resting-state networks (RSNs), particularly the sensorimotor network, begin to strengthe in the third trimester of pregnancy and mature extensively by term age. The integrity and structure of these networks have been repeatedly linked to neurological health outcomes in neonates, highlighting the importance of understanding the normative variations in RSNs in healthy development. Specifically, robust bilateral functional connectivity in the sensorimotor RSN has been linked to optimal neurodevelopmental outcomes in neonates. AIM In the current study, we aimed to map the developmental trajectory of the sensorimotor RSN in awake neonates using functional near-infrared spectroscopy (fNIRS). MATERIALS & METHODS We acquired fNIRS resting-state data from 41 healthy newborns (17 females, gestational age ranging from 36 + 0 to 42 + 1 weeks) within the first week after birth. We performed both single channel and hemispheric analyses to investigate the relationship between functional connectivity and both gestational and postnatal age. RESULTS We observed robust positive connectivity in numerous channel-pairs across the sensorimotor network, especially in the left hemisphere. Next, we examined the relationship between functional connectivity, gestational age, and postnatal age, while controlling for sex and subject effects. We found both gestational and postnatal age to be significantly associated with changes in functional connectivity in the sensorimotor RSN. In our hemispheric analysis (Ninterhemispheric = 10, Nleft intrahemispheric = 15, and Nright intrahemispheric = 9), we observed a significant positive relationship between interhemispheric connectivity and postnatal age. DISCUSSION AND CONCLUSION In summary, our findings demonstrate the utility of fNIRS for monitoring early developmental changes in functional networks in awake newborns.
Collapse
Affiliation(s)
- Homa Vahidi
- Department of Neuroscience, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Alexandra Kowalczyk
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Kevin Stubbs
- Brain and Mind Centre, Western University, London, Ontario, Canada
| | - Melab Musabi
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Sriya Roychaudhuri
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Michaela Kent
- Department of Neuroscience, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Soume Bhattacharya
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Sandrine de Ribaupierre
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Keith St Lawrence
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Yalda Mohsenzadeh
- Brain and Mind Centre, Western University, London, Ontario, Canada
- Department of Computer Science, Western University, London, Ontario, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario, Canada
| | - Emma G Duerden
- Brain and Mind Centre, Western University, London, Ontario, Canada
- Applied Psychology, Faculty of Education, Western University, London, Ontario, Canada
| |
Collapse
|
49
|
Riggins T, Ratliff EL, Horger MN, Spencer RMC. The importance of sleep for the developing brain. CURRENT SLEEP MEDICINE REPORTS 2024; 10:437-446. [PMID: 40123674 PMCID: PMC11928160 DOI: 10.1007/s40675-024-00307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2024] [Indexed: 03/25/2025]
Abstract
Purpose of review This paper summarizes recent research regarding the possible contribution of sleep to brain development. Major milestones in brain development and the methods used to track these changes are reviewed. Changes in sleep, at both behavioral and neural levels, that take place during the same developmental periods are discussed. Finally, a few empirical examples that have contributed new knowledge regarding how sleep contributes to brain development are highlighted. Recent findings Empirical examples demonstrating associations between development of sleep and the brain include: predictive associations between SWA topography and myelin development, associations between SWS and hippocampal development, and links between sleep duration and both white matter volume and whole-brain functional connectivity in developing populations. Summary There is evidence that sleep is important for the developing brain. However, studies utilizing longitudinal, objective measures of sleep, high-resolution brain imaging, and behavioral measures across developmental are critical for understanding sleep function.
Collapse
|
50
|
Ji X, Lakuleswaran M, Cowell W, Kahn LG, Sirota M, Abrahamsson D. Insights into the Chemical Exposome during Pregnancy: A Non-Targeted Analysis of Preterm and Term Births. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:20883-20893. [PMID: 39526929 PMCID: PMC11603774 DOI: 10.1021/acs.est.4c08534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Human-made chemicals are ubiquitous, leading to chronic exposure to complex mixtures of potentially harmful substances. We investigated chemical exposures in pregnant women in New York City by applying a non-targeted analysis (NTA) workflow to 95 paired prenatal urine and serum samples (35 pairs of preterm birth) collected as part of the New York University Children's Health and Environment Study. We analyzed all samples using liquid chromatography coupled with Orbitrap high-resolution mass spectrometry in both positive and negative electrospray ionization modes, employing full scan and data-dependent MS/MS fragmentation scans. We detected a total of 1524 chemical features for annotation, with 12 chemicals confirmed by authentic standards. Two confirmed chemicals dodecyltrimethylammonium and N,N-dimethyldecylamine N-oxide appear to not have been previously reported in human blood samples. We observed a statistically significant differential enrichment between urine and serum samples, as well as between preterm and term birth (p < 0.0001) in serum samples. When comparing between preterm and term births, an exogenous contaminant, 1,4-cyclohexanedicarboxylic acid (tentative), showed a statistical significance difference (p = 0.003) with more abundance in preterm birth in serum. An example of chemical associations (12 associations in total) observed was between surfactants (tertiary amines) and endogenous metabolites (fatty acid amides).
Collapse
Affiliation(s)
- Xiaowen Ji
- Division
of Environmental Pediatrics, Department of Pediatrics, Grossman School of Medicine, New York University, New York, New York 10016, United States
| | - Mathusa Lakuleswaran
- Division
of Environmental Pediatrics, Department of Pediatrics, Grossman School of Medicine, New York University, New York, New York 10016, United States
| | - Whitney Cowell
- Division
of Environmental Pediatrics, Department of Pediatrics, Grossman School of Medicine, New York University, New York, New York 10016, United States
| | - Linda G. Kahn
- Division
of Environmental Pediatrics, Department of Pediatrics, Grossman School of Medicine, New York University, New York, New York 10016, United States
| | - Marina Sirota
- Bakar
Computational Health Sciences Institute, UCSF, San Francisco, California 94158, United States
- Department
of Pediatrics, University of California,
San Francisco, San Francisco, California 94158, United States
| | - Dimitri Abrahamsson
- Division
of Environmental Pediatrics, Department of Pediatrics, Grossman School of Medicine, New York University, New York, New York 10016, United States
- Department
of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|