1
|
Tousley AR, Deykin I, Koc B, Yeh PWL, Yeh HH. Prenatal ethanol exposure results in cell-type, age, and sex-dependent differences in the neonatal striatum that coincide with early motor deficits. eNeuro 2025; 12:ENEURO.0448-24.2025. [PMID: 40086875 PMCID: PMC11949650 DOI: 10.1523/eneuro.0448-24.2025] [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: 10/21/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
Delayed motor development is an early clinical sign of Fetal Alcohol Spectrum Disorders (FASD). However, changes at the neural circuit level that underlie early motor differences are underexplored. The striatum, the principal input nucleus of the basal ganglia, plays an important role in motor learning in adult animals, and the maturation of the striatal circuit has been associated with the development of early motor behaviors. Here, we briefly exposed pregnant C57BL/6 dams to ethanol (5% w/w) in a liquid diet on embryonic days (E)13.5-16.5, and assessed the mouse progeny using a series of 9 brief motor behavior tasks on postnatal days (P)2-14. Live brain slices were then obtained from behaviorally-tested mice for whole cell-voltage and current clamp electrophysiology to assess GABAergic/glutamatergic synaptic activity, and passive/active properties in two populations of striatal neurons: GABAergic interneurons and spiny striatal projection neurons. Electrophysiologically-recorded spiny striatal projection neurons were also filled intracellularly with biocytin for post-hoc analysis of dendritic morphology. We found that prenatal ethanol exposure resulted in developmental motor delays that were more severe in male mice and coincided with sex-dependent differences in the maturation of striatal neurons. Our findings indicate that prenatal ethanol exposure results in dynamic morphological and functional changes to the developmental trajectories of striatal neurons commensurate with the development of motor behaviors that differ between male and female mice.Significance Statement Developmental differences in motor behaviors are an early clinical sign of Fetal Alcohol Spectrum Disorders (FASD) but the neural circuit level changes that contribute to these differences have not yet been determined. Here we demonstrate that a brief binge exposure to ethanol alters the motor development of neonatal mice in a sex-dependent manner, and identify concurrent differences in the functional, synaptic and morphological development of striatal GABAergic interneurons and medium spiny striatal projection neurons. These data suggest that altered development of striatal neurons may contribute to differences in early motor development observed in individuals with FASD.
Collapse
Affiliation(s)
- Adelaide R. Tousley
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755
| | - Ilana Deykin
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755
| | - Betul Koc
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755
| | - Pamela W. L. Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755
| | - Hermes H. Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755
| |
Collapse
|
2
|
Hwang HM, Yamashita S, Matsumoto Y, Ito M, Edwards A, Sasaki J, Dutta DJ, Mohammad S, Yamashita C, Wetherill L, Schwantes-An TH, Abreu M, Mahnke AH, Mattson SN, Foroud T, Miranda RC, Chambers C, Torii M, Hashimoto-Torii K. Reduction of APOE accounts for neurobehavioral deficits in fetal alcohol spectrum disorders. Mol Psychiatry 2024; 29:3364-3380. [PMID: 38734844 PMCID: PMC11541007 DOI: 10.1038/s41380-024-02586-6] [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: 10/10/2023] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024]
Abstract
A hallmark of fetal alcohol spectrum disorders (FASD) is neurobehavioral deficits that still do not have effective treatment. Here, we present that reduction of Apolipoprotein E (APOE) is critically involved in neurobehavioral deficits in FASD. We show that prenatal alcohol exposure (PAE) changes chromatin accessibility of Apoe locus, and causes reduction of APOE levels in both the brain and peripheral blood in postnatal mice. Of note, postnatal administration of an APOE receptor agonist (APOE-RA) mitigates motor learning deficits and anxiety in those mice. Several molecular and electrophysiological properties essential for learning, which are altered by PAE, are restored by APOE-RA. Our human genome-wide association study further reveals that the interaction of PAE and a single nucleotide polymorphism in the APOE enhancer which chromatin is closed by PAE in mice is associated with lower scores in the delayed matching-to-sample task in children. APOE in the plasma is also reduced in PAE children, and the reduced level is associated with their lower cognitive performance. These findings suggest that controlling the APOE level can serve as an effective treatment for neurobehavioral deficits in FASD.
Collapse
Grants
- F31AA027693 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AA021886 NIAAA NIH HHS
- R01 AA026272 NIAAA NIH HHS
- U01 AA014834 NIAAA NIH HHS
- U01AA014834 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U24AA030169 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AA014835 NIAAA NIH HHS
- P50HD105328 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AA014835 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 AA025215 NIAAA NIH HHS
- R01AA026272 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AA026103 NIAAA NIH HHS
- R01AA025215 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AA014809 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AA025103 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P50 HD105328 NICHD NIH HHS
- U24 AA030169 NIAAA NIH HHS
- U01 AA014809 NIAAA NIH HHS
- F31 AA027693 NIAAA NIH HHS
- District of Columbia Intellectual and Developmental Disabilities Research Center (DC-IDDRC) Award program
- Lilly Endowment
- Texas A&M University’s Accountability, Climate, Equity, and Scholarship (ACES) Faculty Fellows Program
Collapse
Affiliation(s)
- Hye M Hwang
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Yu Matsumoto
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Mariko Ito
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Alex Edwards
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Junko Sasaki
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Dipankar J Dutta
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Shahid Mohammad
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Chiho Yamashita
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Leah Wetherill
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marco Abreu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Amanda H Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Sarah N Mattson
- Center for Behavioral Teratology, San Diego State University, San Diego, CA, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Christina Chambers
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Masaaki Torii
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA.
- Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA.
- Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
3
|
Zhang ZH, Wang B, Peng Y, Xu YW, Li CH, Ning YL, Zhao Y, Shan FB, Zhang B, Yang N, Zhang J, Chen X, Xiong RP, Zhou YG, Li P. Identification of a Hippocampus-to-Zona Incerta Projection involved in Motor Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307185. [PMID: 38958448 PMCID: PMC11434110 DOI: 10.1002/advs.202307185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 05/19/2024] [Indexed: 07/04/2024]
Abstract
Motor learning (ML), which plays a fundamental role in growth and physical rehabilitation, involves different stages of learning and memory processes through different brain regions. However, the neural mechanisms that underlie ML are not sufficiently understood. Here, a previously unreported neuronal projection from the dorsal hippocampus (dHPC) to the zona incerta (ZI) involved in the regulation of ML behaviors is identified. Using recombinant adeno-associated virus, the projections to the ZI are surprisingly identified as originating from the dorsal dentate gyrus (DG) and CA1 subregions of the dHPC. Furthermore, projection-specific chemogenetic and optogenetic manipulation reveals that the projections from the dorsal CA1 to the ZI play key roles in the acquisition and consolidation of ML behaviors, whereas the projections from the dorsal DG to the ZI mediate the retrieval/retention of ML behaviors. The results reveal new projections from the dorsal DG and dorsal CA1 to the ZI involved in the regulation of ML and provide insight into the stages over which this regulation occurs.
Collapse
Affiliation(s)
- Zhuo-Hang Zhang
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong, Chongqing, 400010, China
| | - Bo Wang
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Yan Peng
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Ya-Wei Xu
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Chang-Hong Li
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Ya-Lei Ning
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Yan Zhao
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Fa-Bo Shan
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Bo Zhang
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Nan Yang
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Jing Zhang
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Xing Chen
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Ren-Ping Xiong
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Yuan-Guo Zhou
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| | - Ping Li
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing, 400042, China
| |
Collapse
|
4
|
Vargas-Foitzick R, García-Ordenes B, Iratchet D, Acuña A, Alcayaga S, Fernández C, Toledo K, Rodríguez M, Naranjo C, Bustamante R, Haeger PA. Exercise reduces physical alterations in a rat model of fetal alcohol spectrum disorders. Biol Res 2024; 57:41. [PMID: 38907274 PMCID: PMC11193177 DOI: 10.1186/s40659-024-00520-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/04/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) has serious physical consequences for children such as behavioral disabilities, growth disorders, neuromuscular problems, impaired motor coordination, and decreased muscle tone. However, it is not known whether loss of muscle strength occurs, and which interventions will effectively mitigate physical PAE impairments. We aimed to investigate whether physical alteration persists during adolescence and whether exercise is an effective intervention. RESULTS Using paradigms to evaluate different physical qualities, we described that early adolescent PAE animals have significant alterations in agility and strength, without alterations in balance and coordination compared to CTRL animals. We evaluated the effectiveness of 3 different exercise protocols for 4 weeks: Enrichment environment (EE), Endurance exercise (EEX), and Resistance exercise (REX). The enriched environment significantly improved the strength in the PAE group but not in the CTRL group whose strength parameters were maintained even during exercise. Resistance exercise showed the greatest benefits in gaining strength, and endurance exercise did not. CONCLUSION PAE induced a significant decrease in strength compared to CTRL in PND21. Resistance exercise is the most effective to reverse the effects of PAE on muscular strength. Our data suggests that individualized, scheduled, and supervised training of resistance is more beneficial than endurance or enriched environment exercise for adolescents FASD.
Collapse
Affiliation(s)
- Ronald Vargas-Foitzick
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Bayron García-Ordenes
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo, Chile
| | - Donovan Iratchet
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Angie Acuña
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Spencer Alcayaga
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Cristian Fernández
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Karla Toledo
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Marianela Rodríguez
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Carolina Naranjo
- Departamento de Ciencias Clínicas, Universidad Católica del Norte, Coquimbo, Chile
| | - René Bustamante
- Carrera de Kinesiología, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo, Chile
| | - Paola A Haeger
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo, Chile.
| |
Collapse
|
5
|
Imamura F. Effects of prenatal alcohol exposure on the olfactory system development. Front Neural Circuits 2024; 18:1408187. [PMID: 38818309 PMCID: PMC11138157 DOI: 10.3389/fncir.2024.1408187] [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: 03/27/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Fetal Alcohol Spectrum Disorders (FASD), resulting from maternal alcohol consumption during pregnancy, are a prominent non-genetic cause of physical disabilities and brain damage in children. Alongside common symptoms like distinct facial features and neurocognitive deficits, sensory anomalies, including olfactory dysfunction, are frequently noted in FASD-afflicted children. However, the precise mechanisms underpinning the olfactory abnormalities induced by prenatal alcohol exposure (PAE) remain elusive. Utilizing rodents as a model organism with varying timing, duration, dosage, and administration routes of alcohol exposure, prior studies have documented impairments in olfactory system development caused by PAE. Many reported a reduction in the olfactory bulb (OB) volume accompanied by reduced OB neuron counts, suggesting the OB is a brain region vulnerable to PAE. In contrast, no significant olfactory system defects were observed in some studies, though subtle alterations might exist. These findings suggest that the timing, duration, and extent of fetal alcohol exposure can yield diverse effects on olfactory system development. To enhance comprehension of PAE-induced olfactory dysfunctions, this review summarizes key findings from previous research on the olfactory systems of offspring prenatally exposed to alcohol.
Collapse
Affiliation(s)
- Fumiaki Imamura
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
6
|
Dutta DJ, Sasaki J, Bansal A, Sugai K, Yamashita S, Li G, Lazarski C, Wang L, Sasaki T, Yamashita C, Carryl H, Suzuki R, Odawara M, Imamura Kawasawa Y, Rakic P, Torii M, Hashimoto-Torii K. Alternative splicing events as peripheral biomarkers for motor learning deficit caused by adverse prenatal environments. Proc Natl Acad Sci U S A 2023; 120:e2304074120. [PMID: 38051767 PMCID: PMC10723155 DOI: 10.1073/pnas.2304074120] [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: 03/13/2023] [Accepted: 10/25/2023] [Indexed: 12/07/2023] Open
Abstract
Severity of neurobehavioral deficits in children born from adverse pregnancies, such as maternal alcohol consumption and diabetes, does not always correlate with the adversity's duration and intensity. Therefore, biological signatures for accurate prediction of the severity of neurobehavioral deficits, and robust tools for reliable identification of such biomarkers, have an urgent clinical need. Here, we demonstrate that significant changes in the alternative splicing (AS) pattern of offspring lymphocyte RNA can function as accurate peripheral biomarkers for motor learning deficits in mouse models of prenatal alcohol exposure (PAE) and offspring of mother with diabetes (OMD). An aptly trained deep-learning model identified 29 AS events common to PAE and OMD as superior predictors of motor learning deficits than AS events specific to PAE or OMD. Shapley-value analysis, a game-theory algorithm, deciphered the trained deep-learning model's learnt associations between its input, AS events, and output, motor learning performance. Shapley values of the deep-learning model's input identified the relative contribution of the 29 common AS events to the motor learning deficit. Gene ontology and predictive structure-function analyses, using Alphafold2 algorithm, supported existing evidence on the critical roles of these molecules in early brain development and function. The direction of most AS events was opposite in PAE and OMD, potentially from differential expression of RNA binding proteins in PAE and OMD. Altogether, this study posits that AS of lymphocyte RNA is a rich resource, and deep-learning is an effective tool, for discovery of peripheral biomarkers of neurobehavioral deficits in children of diverse adverse pregnancies.
Collapse
Affiliation(s)
- Dipankar J. Dutta
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
| | - Junko Sasaki
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo160-8402, Japan
| | - Ankush Bansal
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
| | - Keiji Sugai
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo160-8402, Japan
| | - Satoshi Yamashita
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
| | - Guojiao Li
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo160-8402, Japan
| | - Christopher Lazarski
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC20010
| | - Li Wang
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
| | - Toru Sasaki
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo160-8402, Japan
| | - Chiho Yamashita
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
| | - Heather Carryl
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
| | - Ryo Suzuki
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo160-8402, Japan
| | - Masato Odawara
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo160-8402, Japan
| | - Yuka Imamura Kawasawa
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA17033
| | - Pasko Rakic
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT06520
| | - Masaaki Torii
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
- Department of Pediatrics, Pharmacology and Physiology, George Washington University, Washington, DC20010
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s National Hospital,Washington, DC20010
- Department of Pediatrics, Pharmacology and Physiology, George Washington University, Washington, DC20010
| |
Collapse
|
7
|
Bariselli S, Mateo Y, Reuveni N, Lovinger DM. Gestational ethanol exposure impairs motor skills in female mice through dysregulated striatal dopamine and acetylcholine function. Neuropsychopharmacology 2023; 48:1808-1820. [PMID: 37188849 PMCID: PMC10579353 DOI: 10.1038/s41386-023-01594-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/29/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023]
Abstract
Fetal alcohol exposure has deleterious consequences on the motor skills of patients affected by Fetal Alcohol Spectrum Disorder (FASD) and in pre-clinical models of gestational ethanol exposure (GEE). Deficits in striatal cholinergic interneurons (CINs) and dopamine function impair action learning and execution, yet the effects of GEE on acetylcholine (ACh) and striatal dopamine release remain unexplored. Here, we report that alcohol exposure during the first ten postnatal days (GEEP0-P10), which mimics ethanol consumption during the last gestational trimester in humans, induces sex-specific anatomical and motor skill deficits in female mice during adulthood. Consistent with these behavioral impairments, we observed increased stimulus evoked-dopamine levels in the dorsolateral striatum (DLS) of GEEP0-P10 female, but not male, mice. Further experiments revealed sex-specific deficits in β2-containing nicotinic ACh receptor (nAChR)-modulation of electrically evoked dopamine release. Moreover, we found a reduced decay of ACh transients and a decreased excitability of striatal CINs in DLS of GEEP0-P10 females, indicating striatal CIN dysfunctions. Finally, the administration of varenicline, a β2-containing nAChR partial agonist, and chemogenetic-mediated increase in CIN activity improved motor performance in adult GEEP0-P10 females. Altogether, these data shed new light on GEE-induced striatal deficits and establish potential pharmacological and circuit-specific interventions to ameliorate motor symptoms of FASD.
Collapse
Affiliation(s)
- Sebastiano Bariselli
- Laboratory for Integrative Neuroscience (LIN), NIH-NIAAA, 5625 Fishers Lane, Bethesda, MD, 20892, USA.
| | - Yolanda Mateo
- Laboratory for Integrative Neuroscience (LIN), NIH-NIAAA, 5625 Fishers Lane, Bethesda, MD, 20892, USA
| | - Noa Reuveni
- Laboratory for Integrative Neuroscience (LIN), NIH-NIAAA, 5625 Fishers Lane, Bethesda, MD, 20892, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience (LIN), NIH-NIAAA, 5625 Fishers Lane, Bethesda, MD, 20892, USA
| |
Collapse
|
8
|
Altounian M, Bellon A, Mann F. Neuronal miR-17-5p contributes to interhemispheric cortical connectivity defects induced by prenatal alcohol exposure. Cell Rep 2023; 42:113020. [PMID: 37610874 DOI: 10.1016/j.celrep.2023.113020] [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: 01/13/2023] [Revised: 06/30/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
Structural and functional deficits in brain connectivity are reported in patients with fetal alcohol spectrum disorders (FASDs), but whether and how prenatal alcohol exposure (PAE) affects axonal development of neurons and disrupts wiring between brain regions is unknown. Here, we develop a mouse model of moderate alcohol exposure during prenatal brain wiring to study the effects of PAE on corpus callosum (CC) development. PAE induces aberrant navigation of interhemispheric CC axons that persists even after exposure ends, leading to ectopic termination in the contralateral cortex. The neuronal miR-17-5p and its target ephrin type A receptor 4 (EphA4) mediate the effect of alcohol on the contralateral targeting of CC axons. Thus, altered microRNA-mediated regulation of axonal guidance may have implications for interhemispheric cortical connectivity and associated behaviors in FASD.
Collapse
Affiliation(s)
| | - Anaïs Bellon
- Aix Marseille University, INSERM, INMED, Marseille, France
| | - Fanny Mann
- Aix Marseille University, CNRS, IBDM, Marseille, France.
| |
Collapse
|
9
|
Mukherjee S, Tarale P, Sarkar DK. Neuroimmune Interactions in Fetal Alcohol Spectrum Disorders: Potential Therapeutic Targets and Intervention Strategies. Cells 2023; 12:2323. [PMID: 37759545 PMCID: PMC10528917 DOI: 10.3390/cells12182323] [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: 07/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a set of abnormalities caused by prenatal exposure to ethanol and are characterized by developmental defects in the brain that lead to various overt and non-overt physiological abnormalities. Growing evidence suggests that in utero alcohol exposure induces functional and structural abnormalities in gliogenesis and neuron-glia interactions, suggesting a possible role of glial cell pathologies in the development of FASD. However, the molecular mechanisms of neuron-glia interactions that lead to the development of FASD are not clearly understood. In this review, we discuss glial cell pathologies with a particular emphasis on microglia, primary resident immune cells in the brain. Additionally, we examine the involvement of several neuroimmune molecules released by glial cells, their signaling pathways, and epigenetic mechanisms responsible for FASD-related alteration in brain functions. Growing evidence suggests that extracellular vesicles (EVs) play a crucial role in the communication between cells via transporting bioactive cargo from one cell to the other. This review emphasizes the role of EVs in the context of neuron-glia interactions during prenatal alcohol exposure. Finally, some potential applications involving nutritional, pharmacological, cell-based, and exosome-based therapies in the treatment of FASD are discussed.
Collapse
Affiliation(s)
- Sayani Mukherjee
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
- Hormone Laboratory Research Group, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Jonas Lies vei 91B, 5021 Bergen, Norway
| | - Prashant Tarale
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
| | - Dipak K. Sarkar
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901-1573, USA; (S.M.); (P.T.)
| |
Collapse
|
10
|
Hwang HM, Kawasawa YI, Basha A, Mohammad S, Ito M, Hashimoto-Torii K. Fatty acid metabolism changes in association with neurobehavioral deficits in animal models of fetal alcohol spectrum disorders. Commun Biol 2023; 6:736. [PMID: 37460609 DOI: 10.1038/s42003-023-05127-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: 11/27/2022] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) show behavioral problems due to prenatal alcohol exposure (PAE). A previous study reports changes in gene expressions linked to fatty acid (FA) metabolism in the cerebral cortex of the PAE mouse model. We find an increase of palmitic acid and arachidonic acid in phospholipid in the cerebral cortex of PAE at postnatal day 30. The increase of palmitic acid is consistent with increase of the producing enzyme, Fasn (fatty acid synthase). Decrease of 26:6 FA is also consistent with the increase of the enzyme which uses 26:6 as a substrate for making very long chain FAs, Elovl4 (elongation of very long chain fatty acids protein 4). However, there is no increase in the elongated products. Rather, lipid droplets (LDs) accumulated in the brain. Although FA-associated metabolic measurements are not affected by PAE, the abundance of FA-related gut microbiota is altered. This suggests that the gut microbiome could serve as a tool to facilitate uncovering the brain pathophysiology of FASD and a potential target to mitigate neurobehavioral problems.
Collapse
Affiliation(s)
- Hye Mee Hwang
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Aiesha Basha
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Shahid Mohammad
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Mariko Ito
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA.
- Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
11
|
Smimih K, El-Mansoury B, Saad FEZ, Khanouchi M, El Amine S, Aimrane A, Zouhairi N, Ferssiwi A, Bitar A, Merzouki M, El Hiba O. Sensory Motor Function Disturbances in Mice Prenatally Exposed to Low Dose of Ethanol: A Neurobehavioral Study in Postnatal and Adult Stages. Neurol Int 2023; 15:580-594. [PMID: 37092508 PMCID: PMC10123635 DOI: 10.3390/neurolint15020036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/01/2023] [Accepted: 04/09/2023] [Indexed: 04/25/2023] Open
Abstract
Prenatal alcohol exposure (PAE) refers to fetal exposure to alcohol during pregnancy through placental barrier transfer from maternal blood. The postnatal outcomes of PAE differ among exposed individuals and range from overt (serious) alcohol-related behavioral and neurophysiological impairments to covert (silenced) symptoms. The aims of the present investigation were to assess the postnatal neurobehavioral disturbances, particularly, motor coordination and sensory-motor function in mice with PAE. Female mice with positive vaginal plugs were divided into three groups: group 1: Et + Pyr: received two i.p injections of ethanol (1 g/kg) followed by pyrazole (100 mg/kg). Group 2: Pyr: received an i.p injection of pyrazole (100 mg/kg). Group 3: C: of saline controls received, in equal volume, saline solution (NaCl 0.9%). After birth, mice pups were weighed and subjected to behavioral tests for motor function screening using the motor ambulation test, cliff aversion, surface righting, and negative geotaxis, while at the adult stage, mice were subjected to the open field, rotarod, parallel bars, and static rods tests. Our data show an obvious decrement of body weight from the first post-natal day (P1) and continues over the adult stage. This was accompanied by an obvious impaired sensory-motor function which was maintained even at the adult stage with alteration of the locomotor and coordination abilities. The current data demonstrate the powerful neurotoxic effect of prenatal ethanol exposure on the sensory-motor and coordination functions, leading to suppose possible structural and/or functional neuronal disturbances, particularly the locomotor network.
Collapse
Affiliation(s)
- Kamal Smimih
- Biological Engineering Laboratory, Faculty of Sciences and Techniques (FST), Sultan Moulay Slimane University, Beni Mellal 23000, Morocco
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Bilal El-Mansoury
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Fatima Ez-Zahraa Saad
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Manal Khanouchi
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Souad El Amine
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Abdelmohcine Aimrane
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Nadia Zouhairi
- Biological Engineering Laboratory, Faculty of Sciences and Techniques (FST), Sultan Moulay Slimane University, Beni Mellal 23000, Morocco
| | - Abdessalam Ferssiwi
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Abdelali Bitar
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| | - Mohamed Merzouki
- Biological Engineering Laboratory, Faculty of Sciences and Techniques (FST), Sultan Moulay Slimane University, Beni Mellal 23000, Morocco
| | - Omar El Hiba
- Laboratory of Anthropogenic, Biotechnology and Health, Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, El Jadida 24000, Morocco
| |
Collapse
|
12
|
Popova S, Charness ME, Burd L, Crawford A, Hoyme HE, Mukherjee RAS, Riley EP, Elliott EJ. Fetal alcohol spectrum disorders. Nat Rev Dis Primers 2023; 9:11. [PMID: 36823161 DOI: 10.1038/s41572-023-00420-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/25/2023]
Abstract
Alcohol readily crosses the placenta and may disrupt fetal development. Harm from prenatal alcohol exposure (PAE) is determined by the dose, pattern, timing and duration of exposure, fetal and maternal genetics, maternal nutrition, concurrent substance use, and epigenetic responses. A safe dose of alcohol use during pregnancy has not been established. PAE can cause fetal alcohol spectrum disorders (FASD), which are characterized by neurodevelopmental impairment with or without facial dysmorphology, congenital anomalies and poor growth. FASD are a leading preventable cause of birth defects and developmental disability. The prevalence of FASD in 76 countries is >1% and is high in individuals living in out-of-home care or engaged in justice and mental health systems. The social and economic effects of FASD are profound, but the diagnosis is often missed or delayed and receives little public recognition. Future research should be informed by people living with FASD and be guided by cultural context, seek consensus on diagnostic criteria and evidence-based treatments, and describe the pathophysiology and lifelong effects of FASD. Imperatives include reducing stigma, equitable access to services, improved quality of life for people with FASD and FASD prevention in future generations.
Collapse
Affiliation(s)
- Svetlana Popova
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada.
| | - Michael E Charness
- VA Boston Healthcare System, West Roxbury, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.,Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Larry Burd
- North Dakota Fetal Alcohol Syndrome Center, Department of Pediatrics, University of North Dakota School of Medicine and Health Sciences, Pediatric Therapy Services, Altru Health System, Grand Forks, ND, USA
| | - Andi Crawford
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - H Eugene Hoyme
- Sanford Children's Genomic Medicine Consortium, Sanford Health, and University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| | - Raja A S Mukherjee
- National UK FASD Clinic, Surrey and Borders Partnership NHS Foundation Trust, Redhill, Surrey, UK
| | - Edward P Riley
- Center for Behavioral Teratology, San Diego State University, San Diego, CA, USA
| | - Elizabeth J Elliott
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,New South Wales FASD Assessment Service, CICADA Centre for Care and Intervention for Children and Adolescents affected by Drugs and Alcohol, Sydney Children's Hospitals Network, Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Wu X, Yan Q, Liu L, Xue X, Yao W, Li X, Li W, Ding S, Xia Y, Zhang D, Zhu F. Domesticated HERV-W env contributes to the activation of the small conductance Ca 2+-activated K + type 2 channels via decreased 5-HT4 receptor in recent-onset schizophrenia. Virol Sin 2023; 38:9-22. [PMID: 36007838 PMCID: PMC10006216 DOI: 10.1016/j.virs.2022.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022] Open
Abstract
The human endogenous retroviruses type W family envelope (HERV-W env) gene is located on chromosome 7q21-22. Our previous studies show that HERV-W env is elevated in schizophrenia and HERV-W env can increase calcium influx. Additionally, the 5-HTergic system and particularly 5-hydroxytryptamine (5-HT) receptors play a prominent role in the pathogenesis and treatment of schizophrenia. 5-hydroxytryptamine receptor 4 (5-HT4R) agonist can block calcium channels. However, the underlying relationship between HERV-W env and 5-HT4R in the etiology of schizophrenia has not been revealed. Here, we used enzyme-linked immunosorbent assay to detect the concentration of HERV-W env and 5-HT4R in the plasma of patients with schizophrenia and we found that there were decreased levels of 5-HT4R and a negative correlation between 5-HT4R and HERV-W env in schizophrenia. Overexpression of HERV-W env decreased the transcription and protein levels of 5-HT4R but increased small conductance Ca2+-activated K+ type 2 channels (SK2) expression levels. Further studies revealed that HERV-W env could interact with 5-HT4R. Additionally, luciferase assay showed that an essential region (-364 to -176 from the transcription start site) in the SK2 promoter was required for HERV-W env-induced SK2 expression. Importantly, 5-HT4R participated in the regulation of SK2 expression and promoter activity. Electrophysiological recordings suggested that HERV-W env could increase SK2 channel currents and the increase of SK2 currents was inhibited by 5-HT4R. In conclusion, HERV-W env could activate SK2 channels via decreased 5-HT4R, which might exhibit a novel mechanism for HERV-W env to influence neuronal activity in schizophrenia.
Collapse
Affiliation(s)
- Xiulin Wu
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Qiujin Yan
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | | | - Xing Xue
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wei Yao
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xuhang Li
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wenshi Li
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuang Ding
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yaru Xia
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Dongyan Zhang
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Fan Zhu
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China; Hubei Province Key Laboratory of Allergy & Immunology, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
14
|
Kaji AA, Torii M, Ishii S. Caspase-3 Inhibition toward Perinatal Protection of the Developing Brain from Environmental Stress. Dev Neurosci 2023; 45:66-75. [PMID: 36642064 PMCID: PMC10521911 DOI: 10.1159/000529125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Throughout our lives, we are exposed to a variety of hazards, such as environmental pollutants and chemical substances that affect our health, and viruses and bacteria that cause infectious diseases. These external factors that are undesirable to an organism are called environmental stress. During the perinatal period, when neural networks are drastically reorganized and refined, the tolerance of the developing brain to various environmental stresses is lower than in adulthood. Thus, exposure to environmental stress during this vulnerable period is strongly associated with cognitive and behavioral deficits in later life. Recent studies have uncovered various mechanisms underlying the adverse impacts of environmental stress during the perinatal period on brain development. In this mini-review, we will present the findings from these studies, focusing on caspase-mediated apoptotic and nonapoptotic effects of environmental stress, and discuss several compounds that mitigate these caspase-mediated effects as examples of potential therapeutic approaches.
Collapse
Affiliation(s)
- Anna Arjun Kaji
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, D.C., United States
| | - Masaaki Torii
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, D.C., United States
- Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, D.C., United States
| | - Seiji Ishii
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
How Alcohol Damages Brain Development in Children. PRILOZI (MAKEDONSKA AKADEMIJA NA NAUKITE I UMETNOSTITE. ODDELENIE ZA MEDICINSKI NAUKI) 2022; 43:29-42. [PMID: 36473036 DOI: 10.2478/prilozi-2022-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The world over, people drink in order to socialize, celebrate, and relax, despite the negative health effects of alcohol. Three periods of dynamic brain changes are evidenced to be particularly sensitive to the harmful effects of alcohol: gestation (from conception to birth), later adolescence (15-19 years), and older adulthood (over 65 years). This article is concentrated only on the negative effects of alcohol in children who have been exposed to alcohol before birth, known as foetal alcohol syndrome (FAS). This is a review based on published data in PubMed over the last two decades and is an analysis of more than 150 published papers. Alcohol use during pregnancy can cause miscarriage, stillbirth, and a range of lifelong physical, behavioural, and intellectual disabilities. The effects of ethanol are expressed on a set of molecules involved in neuroinflammation, myelination, neurotransmission, and neuron function. Modern neuroimaging techniques are able to specify some fine structural changes in the affected areas of the brain: volume reductions in the frontal lobe, including the middle frontal gyri in the prefrontal cortex, hippocampal structure, interhemispheric connectivity, abnormalities in glial cells, white matter deficits etc. Corpus callosum myelination is affected, resulting in a lack of the inter-hemispheric connectivity. This is known to facilitate autism, stroke, schizophrenia, as well as dementia, disrupts cognitive performance, and may lead to neurobehavioral deficits. It was pointed out that many symptoms and neuroimaging characteristics are similar in ADHD and FAS, thus the anamnesis for prenatal alcohol and nicotine exposure must be taken very seriously in order to better understand and interpret clinical symptoms.
Collapse
|
16
|
Identification of the Novel Gene Markers Based on the Gene Profile among Different Severity of Obstructive Sleep Apnea. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6517965. [PMID: 36245838 PMCID: PMC9554663 DOI: 10.1155/2022/6517965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/24/2022] [Accepted: 09/04/2022] [Indexed: 11/25/2022]
Abstract
Obstructive sleep apnea (OSA) is caused by repeated blockage of the upper respiratory airways during sleep. The traditional evaluation methods for OSA severity are yet limited. This study aimed to screen gene signatures to effectively evaluate OSA severity. Expression profiles of peripheral blood mononuclear cells in the different severities of OSA patients were accessed from Gene Expression Omnibus (GEO) database. A total of 446 differentially expressed genes (DEGs) were screened among the varying severities of OSA samples by analysis of variance (ANOVA) test. A total of 1,152 DEGs were screened between the pre- and post-treatment OSA samples by using t test. Overlap of the two groups of DEGs was selected (88 DEGs) for Metascape enrichment analysis. Afterwards, Mfuzz package was used to perform soft clustering analysis on these 88 genes, by which 6 clusters were obtained. It was observed that the gene expression condition of the cluster 3 was positively associated with OSA severity degree; also, the gene expression condition in cluster 4 was negatively correlated with OSA severity. A total of 10 gene markers related to OSA progression were selected from cluster 3 and cluster 4. Their expression levels and correlation were analyzed. The marker genes in cluster 3 and cluster 4 were examined, finding that most genes were significantly correlated with apnea hypopnea index (AHI). An accurate and objective assessment of the severity of OSA is of great significance for formulating follow-up treatment strategies for patients with OSA. In this paper, a set of marker genes that can detect the severity of OSA were screened by bioinformatics methods, which could be jointly used with the traditional OSA diagnostic index to achieve a more reliable OSA severity evaluation.
Collapse
|
17
|
Torii S, Rakic P. Tracking the Activation of Heat Shock Signaling in Cellular Protection and Damage. Cells 2022; 11:1561. [PMID: 35563865 PMCID: PMC9104565 DOI: 10.3390/cells11091561] [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: 03/15/2022] [Revised: 05/02/2022] [Accepted: 05/02/2022] [Indexed: 01/27/2023] Open
Abstract
Heat Shock (HS) signaling is activated in response to various types of cellular stress. This activation serves to protect cells from immediate threats in the surrounding environment. However, activation of HS signaling occurs in a heterogeneous manner within each cell population and can alter the epigenetic state of the cell, ultimately leading to long-term abnormalities in body function. Here, we summarize recent research findings obtained using molecular and genetic tools to track cells where HS signaling is activated. We then discuss the potential further applications of these tools, their limitations, and the necessary caveats in interpreting data obtained with these tools.
Collapse
Affiliation(s)
| | - Pasko Rakic
- Department of Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA;
| |
Collapse
|
18
|
Bottom RT, Kozanian OO, Rohac DJ, Erickson MA, Huffman KJ. Transgenerational Effects of Prenatal Ethanol Exposure in Prepubescent Mice. Front Cell Dev Biol 2022; 10:812429. [PMID: 35386207 PMCID: PMC8978834 DOI: 10.3389/fcell.2022.812429] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Fetal alcohol spectrum disorders (FASD) represent a leading cause of non-genetic neuropathologies. Recent preclinical evidence from suggests that prenatal ethanol exposure (PrEE), like other environmental exposures, may have a significant, transgenerational impact on the offspring of directly exposed animals, including altered neocortical development at birth and behavior in peri-pubescent mice. How these adverse behavioral outcomes are manifested within the brain at the time of behavioral disruption remains unknown. Methods: A transgenerational mouse model of FASD was used to generate up to a third filial generation of offspring to study. Using a multi-modal battery of behavioral assays, we assessed motor coordination/function, sensorimotor processing, risk-taking behavior, and depressive-like behavior in postnatal day (P) 20 pre-pubescent mice. Additionally, sensory neocortical area connectivity using dye tracing, neocortical gene expression using in situ RNA hybridization, and spine density of spiny stellate cells in the somatosensory cortex using Golgi-Cox staining were examined in mice at P20. Results: We found that PrEE induces behavioral abnormalities including abnormal sensorimotor processing, increased risk-taking behavior, and increased depressive-like behaviors that extend to the F3 generation in 20-day old mice. Assessment of both somatosensory and visual cortical connectivity, as well as cortical RZRβ expression in pre-pubescent mice yielded no significant differences among any experimental generations. In contrast, only directly-exposed F1 mice displayed altered cortical expression of Id2 and decreased spine density among layer IV spiny stellate cells in somatosensory cortex at this pre-pubescent, post weaning age. Conclusion: Our results suggest that robust, clinically-relevant behavioral abnormalities are passed transgenerationally to the offspring of mice directly exposed to prenatal ethanol. Additionally, in contrast to our previous findings in the newborn PrEE mouse, a lack of transgenerational findings within the brain at this later age illuminates the critical need for future studies to attempt to discover the link between neurological function and the described behavioral changes. Overall, our study suggests that multi-generational effects of PrEE may have a substantial impact on human behavior as well as health and well-being and that these effects likely extend beyond early childhood.
Collapse
Affiliation(s)
- Riley T Bottom
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, CA, United States
| | - Olga O Kozanian
- Department of Psychology, University of California, Riverside, Riverside, CA, United States
| | - David J Rohac
- Department of Psychology, University of California, Riverside, Riverside, CA, United States
| | - Michael A Erickson
- Department of Psychology, University of California, Riverside, Riverside, CA, United States
| | - Kelly J Huffman
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, CA, United States.,Department of Psychology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
19
|
Hwang HM, Hashimoto-Torii K. Activation of the anterior cingulate cortex ameliorates anxiety in a preclinical model of fetal alcohol spectrum disorders. Transl Psychiatry 2022; 12:24. [PMID: 35058425 PMCID: PMC8776849 DOI: 10.1038/s41398-022-01789-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/21/2021] [Accepted: 01/07/2022] [Indexed: 11/30/2022] Open
Abstract
People with fetal alcohol spectrum disorders (FASD) are suffered from a wide range of interlinked cognitive and psychological problems. However, few therapeutic options are available for those patients due to limited dissection of its underlying etiology. Here we found that prenatal alcohol exposure (PAE) increases anxiety in mice due to a dysregulated functional connectivity between the anterior cingulate cortex (ACC) and basolateral amygdala (BLA). We also show that chemogenetic activation of excitatory neurons in the ACC reduced this anxiety behavior in the PAE mice. Interestingly, although the level of plasma corticosterone correlated with the increase in anxiety in the PAE, this level was not altered by chemogenetic activation of the ACC, suggesting that the functional connectivity between the ACC and the BLA does not alter the activity of the hypothalamic-pituitary-adrenal axis. Altogether, this study demonstrated that reduced excitation in the ACC is a cause of anxiety in the PAE mice, providing critical insights into the ACC-BLA neural circuit as a potential target for treating anxiety in FASD patients.
Collapse
Affiliation(s)
- Hye M. Hwang
- grid.239560.b0000 0004 0482 1586Center for Neuroscience Research, The Children’s Research Institute, Children’s National Hospital, Washington, DC USA ,grid.253615.60000 0004 1936 9510The Institute for Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
20
|
Machado L, Relaix F, Mourikis P. Stress relief: emerging methods to mitigate dissociation-induced artefacts. Trends Cell Biol 2021; 31:888-897. [PMID: 34074577 DOI: 10.1016/j.tcb.2021.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/29/2022]
Abstract
The rapid progress of single-cell RNA-sequencing (scRNA-seq) at large scales has led to what seemed impossible until recently: the generation of comprehensive transcriptional maps of nearly all cells in multicellular tissues. We pinpoint three key elements as being critical to the production of these maps: scalability, spatial information, and accuracy of the transcriptome of the individual cells. Here, we discuss the ramifications of traditional cell-isolation protocols when capturing the transcriptional signature of cells as they exist in their native tissue context, the methods that have been developed to avoid these distortions, and the biological processes that have unraveled on account of these upgraded methodological approaches.
Collapse
Affiliation(s)
- Léo Machado
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), F-94010 Créteil, France
| | - Frederic Relaix
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), F-94010 Créteil, France; EnvA, IMRB, F-94700 Maisons-Alfort, France; Etablissement Français du Sang (EFS), IMRB, F-94010 Creteil, France; Assistance Publique-Hôpitaux de Paris, Hopital Mondor, Service d'Histologie, F-94010 Creteil, France.
| | - Philippos Mourikis
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), F-94010 Créteil, France.
| |
Collapse
|
21
|
Primary cilia safeguard cortical neurons in neonatal mouse forebrain from environmental stress-induced dendritic degeneration. Proc Natl Acad Sci U S A 2020; 118:2012482118. [PMID: 33443207 DOI: 10.1073/pnas.2012482118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The developing brain is under the risk of exposure to a multitude of environmental stressors. While perinatal exposure to excessive levels of environmental stress is responsible for a wide spectrum of neurological and psychiatric conditions, the developing brain is equipped with intrinsic cell protection, the mechanisms of which remain unknown. Here we show, using neonatal mouse as a model system, that primary cilia, hair-like protrusions from the neuronal cell body, play an essential role in protecting immature neurons from the negative impacts of exposure to environmental stress. More specifically, we found that primary cilia prevent the degeneration of dendritic arbors upon exposure to alcohol and ketamine, two major cell stressors, by activating cilia-localized insulin-like growth factor 1 receptor and downstream Akt signaling. We also found that activation of this pathway inhibits Caspase-3 activation and caspase-mediated cleavage/fragmentation of cytoskeletal proteins in stress-exposed neurons. These results indicate that primary cilia play an integral role in mitigating adverse impacts of environmental stressors such as drugs on perinatal brain development.
Collapse
|
22
|
Mohammad S, Page SJ, Sasaki T, Ayvazian N, Rakic P, Kawasawa YI, Hashimoto-Torii K, Torii M. Long-term spatial tracking of cells affected by environmental insults. J Neurodev Disord 2020; 12:38. [PMID: 33327938 PMCID: PMC7745478 DOI: 10.1186/s11689-020-09339-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/13/2020] [Indexed: 11/15/2022] Open
Abstract
Background Harsh environments surrounding fetuses and children can induce cellular damage in the developing brain, increasing the risk of intellectual disability and other neurodevelopmental disorders such as schizophrenia. However, the mechanisms by which early damage leads to disease manifestation in later life remain largely unknown. Previously, we demonstrated that the activation of heat shock (HS) signaling can be utilized as a unique reporter to label the cells that undergo specific molecular/cellular changes upon exposure to environmental insults throughout the body. Since the activation of HS signaling is an acute and transient event, this approach was not intended for long-term tracing of affected cells after the activation has diminished. In the present study, we generated new reporter transgenic mouse lines as a novel tool to achieve systemic and long-term tracking of affected cells and their progeny. Methods The reporter transgenic mouse system was designed so that the activation of HS signaling through HS response element (HSE) drives flippase (FLPo)-flippase recognition target (FRT) recombination-mediated permanent expression of the red fluorescent protein (RFP), tdTomato. With a priority on consistent and efficient assessment of the reporter system, we focused on intraperitoneal (i.p.) injection models of high-dose, short prenatal exposure to alcohol (ethanol) and sodium arsenite (ethanol at 4.0 g/kg/day and sodium arsenite at 5.0 mg/kg/day, at embryonic day (E) 12 and 13). Long-term reporter expression was examined in the brain of reporter mice that were prenatally exposed to these insults. Electrophysiological properties were compared between RFP+ and RFP− cortical neurons in animals prenatally exposed to arsenite. Results We detected RFP+ neurons and glia in the brains of postnatal mice that had been prenatally exposed to alcohol or sodium arsenite. In animals prenatally exposed to sodium arsenite, we also detected reduced excitability in RFP+ cortical neurons. Conclusion The reporter transgenic mice allowed us to trace the cells that once responded to prenatal environmental stress and the progeny derived from these cells long after the exposure in postnatal animals. Tracing of these cells indicates that the impact of prenatal exposure on neural progenitor cells can lead to functional abnormalities in their progeny cells in the postnatal brain. Further studies using more clinically relevant exposure models are warranted to explore this mechanism. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-020-09339-w.
Collapse
Affiliation(s)
- Shahid Mohammad
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Stephen J Page
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Toru Sasaki
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.,Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan
| | - Nicholas Ayvazian
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.,Institute of Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, CT, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| | - Masaaki Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
23
|
Mochel F, Rastetter A, Ceulemans B, Platzer K, Yang S, Shinde DN, Helbig KL, Lopergolo D, Mari F, Renieri A, Benetti E, Canitano R, Waisfisz Q, Plomp AS, Huisman SA, Wilson GN, Cathey SS, Louie RJ, Gaudio DD, Waggoner D, Kacker S, Nugent KM, Roeder ER, Bruel AL, Thevenon J, Ehmke N, Horn D, Holtgrewe M, Kaiser FJ, Kamphausen SB, Abou Jamra R, Weckhuysen S, Dalle C, Depienne C. Variants in the SK2 channel gene (KCNN2) lead to dominant neurodevelopmental movement disorders. Brain 2020; 143:3564-3573. [PMID: 33242881 DOI: 10.1093/brain/awaa346] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/17/2020] [Accepted: 09/08/2020] [Indexed: 11/14/2022] Open
Abstract
KCNN2 encodes the small conductance calcium-activated potassium channel 2 (SK2). Rodent models with spontaneous Kcnn2 mutations show abnormal gait and locomotor activity, tremor and memory deficits, but human disorders related to KCNN2 variants are largely unknown. Using exome sequencing, we identified a de novo KCNN2 frameshift deletion in a patient with learning disabilities, cerebellar ataxia and white matter abnormalities on brain MRI. This discovery prompted us to collect data from nine additional patients with de novo KCNN2 variants (one nonsense, one splice site, six missense variants and one in-frame deletion) and one family with a missense variant inherited from the affected mother. We investigated the functional impact of six selected variants on SK2 channel function using the patch-clamp technique. All variants tested but one, which was reclassified to uncertain significance, led to a loss-of-function of SK2 channels. Patients with KCNN2 variants had motor and language developmental delay, intellectual disability often associated with early-onset movement disorders comprising cerebellar ataxia and/or extrapyramidal symptoms. Altogether, our findings provide evidence that heterozygous variants, likely causing a haploinsufficiency of the KCNN2 gene, lead to novel autosomal dominant neurodevelopmental movement disorders mirroring phenotypes previously described in rodents.
Collapse
Affiliation(s)
- Fanny Mochel
- Institut du Cerveau et de la Moelle épinière (ICM), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, F-75013 Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Département de Génétique and Centre de Référence Neurométabolique Adulte, F-75013, Paris, France
| | - Agnès Rastetter
- Institut du Cerveau et de la Moelle épinière (ICM), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, F-75013 Paris, France
| | - Berten Ceulemans
- Division of Paediatric Neurology, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | | | - Deepali N Shinde
- Department of Clinical Diagnostics, Ambry Genetics, Aliso Viejo, CA, USA
| | - Katherine L Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,The Epilepsy Neurogenetics Initiative, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Diego Lopergolo
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Francesca Mari
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Elisa Benetti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Roberto Canitano
- Division of Child and Adolescent Neuropsychiatry, University Hospital of Siena, Siena, Italy
| | - Quinten Waisfisz
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Astrid S Plomp
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sylvia A Huisman
- Department of Pediatrics, Amsterdam UMC, Amsterdam, The Netherlands.,Prinsenstichting, Purmerend, The Netherlands
| | - Golder N Wilson
- Department of Pediatrics, Texas Tech University Health Science Center, Lubbock, Texas, USA
| | - Sara S Cathey
- Greenwood Genetic Center, Greenwood, South Carolina, 29646, USA
| | - Raymond J Louie
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Daniela Del Gaudio
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Darrel Waggoner
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Shawn Kacker
- Department of Pediatrics, Section of Child Neurology, University of Chicago, Chicago, IL, 60637, USA
| | - Kimberly M Nugent
- Department of Pediatrics, Baylor College of Medicine, San Antonio, TX, 78207, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Elizabeth R Roeder
- Department of Pediatrics, Baylor College of Medicine, San Antonio, TX, 78207, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ange-Line Bruel
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Centre de référence maladies rares 'déficiences intellectuelles de causes rares', Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Julien Thevenon
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France.,INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, 38706 Grenoble, France
| | - Nadja Ehmke
- Institute for Human Genetics and Medical Genetics, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Denise Horn
- Institute for Human Genetics and Medical Genetics, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Manuel Holtgrewe
- Core Unit Bioinformatics - CUBI, Berlin Institute of Health (BIH), Berlin, Germany
| | - Frank J Kaiser
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| | | | - Rami Abou Jamra
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Sarah Weckhuysen
- Applied and Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.,Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium.,Department of Neurology, University Hospital Antwerp, Antwerp, Belgium
| | - Carine Dalle
- Institut du Cerveau et de la Moelle épinière (ICM), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, F-75013 Paris, France
| | - Christel Depienne
- Institut du Cerveau et de la Moelle épinière (ICM), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, F-75013 Paris, France.,Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| |
Collapse
|
24
|
Otto G. Channel problems. Nat Rev Neurosci 2020; 21:300. [PMID: 32286549 DOI: 10.1038/s41583-020-0305-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|