1
|
Tadas M, Wankhede N, Chandurkar P, Kotagale N, Umekar M, Katariya R, Waghade A, Kokare D, Taksande B. Postnatal propionic acid exposure disrupts hippocampal agmatine homeostasis leading to social deficits and cognitive impairment in autism spectrum disorder-like phenotype in rats. Pharmacol Biochem Behav 2025; 252:174030. [PMID: 40318701 DOI: 10.1016/j.pbb.2025.174030] [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: 03/08/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental condition characterized by a range of symptoms including impaired social interaction and cognitive deficits. Although the exact pathogenesis of ASD is not well established, recent clinical findings suggest a decline in levels of biogenic amine agmatine in autistic patients. The present study was designed to investigate the impact of postnatal propionic acid (PPA) exposure on hippocampal agmatine homeostasis in male rat pups and to explore a new therapeutic intervention for ASD using agmatine as a biological target. PPA is commonly used in experimental models of ASD due to its ability to induce social deficits, cognitive impairments, and stereotyped behaviors, which closely resemble key characteristics of ASD. Male rat pups were administered with PPA via the intrahippocampal route bilaterally (25 μg/0.25 μl per side) on PND-21 to simulate the ASD phenotype, and its subsequent effect on the endogenous agmatinergic system. The influence of agmatine treatment and its endogenous modulation on ASD-like phenotypes was also investigated. Behavioral assessments revealed that PPA exposure reduced sociability and social preference, caused learning and memory impairment in the Morris water maze, increased anxiety-like behavior in the elevated plus maze, and reduced exploratory behavior in the hole board test. Neurochemical analyses showed a decrease in agmatine concentration and an increase in its degrading enzyme agmatinase in the hippocampus. PPA treatment altered the content of GABA, glutamate, TNF-α, IL-6, BDNF, and also resulted in increased astrogliosis and neurotoxicity within the hippocampus. Chronic agmatine treatment and its endogenous modulation ameliorated the behavioral and biochemical disruptions induced by PPA exposure. This study highlights the critical role of hippocampal agmatinergic pathway in the etiopathogenesis of ASD, positioning agmatine as a promising therapeutic target for its treatment.
Collapse
Affiliation(s)
- Manasi Tadas
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S. 441 002, India
| | - Nitu Wankhede
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S. 441 002, India
| | - Pranali Chandurkar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S. 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy, Kathora Naka, VMV Road, Amravati, M.S. 444 604, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S. 441 002, India
| | - Raj Katariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S. 441 002, India
| | - Akash Waghade
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, M.S. 440 033, India
| | - Dadasaheb Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, M.S. 440 033, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S. 441 002, India.
| |
Collapse
|
2
|
Wang ZH, Xu C, Ma YY, Xue WX, Wang HY, Fan LY, Zhang CY, Li L, Zhang XY, Zhu JN, Zhang QP. Oxytocin Improves Autistic Behaviors by Positively Shifting GABA Reversal Potential via NKCC1 in Early-Postnatal-Stage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415432. [PMID: 40305761 DOI: 10.1002/advs.202415432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/30/2025] [Indexed: 05/02/2025]
Abstract
Accumulating evidence has identified disrupted oxytocin signaling in both autistic patients and animal models of autism. Nevertheless, the specific timing of the impact of oxytocin on social behavior has remained unclear. Using mouse strains from oxytocin-Cre mice crossed with Cre-dependent chemogenetic mice, oxytocinergic neuronal activity is selectivity manipulated during the early or late postnatal stages and revealed, for the first time, that the suppression of oxytocinergic neurons in the early rather than late postnatal stage led to the emergence of autistic-like behaviors. Notably, significantly reduced oxytocin levels are identified specifically during the early postnatal stage in both valproic acid (VPA)-exposed and Fmr1-KO mouse brains, along with an impairment of the GABA reversal potential and downregulation of the Na+-K+-2Cl- cotransporter (NKCC1) post-birth. Furthermore, chemogenetic activation of oxytocinergic neurons during the early rather than late postnatal stage effectively restored the aberrant NKCC1 expression and GABAA receptor reversal potential and consequently alleviated autistic-like behaviors in VPA-exposed mice. Overall, the results demonstrate that the early postnatal stage may be the unique critical period for oxytocin signaling to regulate GABA reversal potential and promote brain development for prosocial behaviors. These findings suggest an earlier intervention window and strategy for the clinical oxytocin treatment of autism.
Collapse
Affiliation(s)
- Zi-Hui Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Chang Xu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Yao-Yao Ma
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Wei-Xuan Xue
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Hao-Yuan Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Lin-Yao Fan
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
- NJU Institute of AI Biomedicine and Biotechnology, Nanjing University, Nanjing, Jiangsu, 210023, China
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, 210023, China
| | - Liang Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Xiao-Yang Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
| | - Jing-Ning Zhu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Qi-Peng Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, 210023, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210008, China
| |
Collapse
|
3
|
Alabdali A, Ben Bacha A, Alonazi M, Al-Ayadhi LY, Alanazi ASJ, El‐Ansary A. Comparative evaluation of certain biomarkers emphasizing abnormal GABA inhibitory effect and glutamate excitotoxicity in autism spectrum disorders. Front Psychiatry 2025; 16:1562631. [PMID: 40330649 PMCID: PMC12052539 DOI: 10.3389/fpsyt.2025.1562631] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/12/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication deficits and repetitive behaviors. An imbalance between the excitatory neurotransmitter glutamate and the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) might play a crucial role in ASD. This study explores the biochemical markers associated with GABAergic and glutamatergic signaling in individuals with autism and healthy controls, aiming to identify potential diagnostic and therapeutic targets. Methods The study included 46 male individuals with autism and 26 age- and gender-matched healthy controls. The plasma levels of excitatory amino acid transporter 2 (EAAT2), potassium chloride co-transporter 2 (KCC2), Na-K-Cl co-transporter 1 (NKCC1), vitamin D3 (VD3), GABA, gamma aminobutyric acid type a receptor subunit alpha 5 (GABRA5), and glutamate were measured using ELISA. Statistical analyses, including correlation, multiple regression, and receiver operating characteristic (ROC) curve analysis, were performed to evaluate the diagnostic utility and interrelationships of these biomarkers. Results Significant biochemical differences were found between individuals with autism and healthy controls. Individuals with autism had notably lower levels of EAAT2, KCC2, NKCC1, VD3, GABA, and GABRA5, especially in the severe group. Altered KCC2/NKCC1 and GABA/glutamate ratios highlighted the imbalance in neurotransmission. The correlation and multiple regression analyses showed significant interconnections between biomarkers. The ROC analysis indicated that EAAT2, KCC2, GABA, and the ratios of KCC2/NKCC1 and GABA/glutamate have high diagnostic potential. Conclusion These findings support the hypothesis that GABA and glutamate imbalance is central to the pathophysiology of ASD. Significant disruptions in neurotransmitter signaling and chloride homeostasis, particularly in severe cases, provide insights into the neurobiological mechanisms of ASD. Restoring the GABA-glutamate balance could be an effective therapeutic strategy for ASD, warranting further research into these biochemical pathways for targeted treatments.
Collapse
Affiliation(s)
- Altaf Alabdali
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Laila Y. Al-Ayadhi
- Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Afaf El‐Ansary
- Autism Center, Lotus Holistic Alternative Medical Center, Abu Dhabi, United Arab Emirates
| |
Collapse
|
4
|
Bolla M, Colombo G, Falappa M, Pace M, Baravalle R, Martinez N, Montani F, Tucci V, Cancedda L. NKCC1 inhibition improves sleep quality and EEG information content in a Down syndrome mouse model. iScience 2025; 28:112220. [PMID: 40224007 PMCID: PMC11986984 DOI: 10.1016/j.isci.2025.112220] [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: 09/16/2024] [Revised: 12/10/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
In several brain disorders, the hyperpolarizing/inhibitory effects of GABA signaling through Cl-permeable GABAA receptors are compromised, leading to an imbalance between neuronal excitation and inhibition. For example, the Ts65Dn mouse model of Down syndrome (DS) exhibits increased expression of the Cl- importer NKCC1, leading to depolarizing gamma aminobutyric acid (GABA) signaling in the mature hippocampus and cortex. Inhibiting NKCC1 with the Food and Drug Administration (FDA)-approved diuretic bumetanide rescues inhibitory GABAergic transmission, synaptic plasticity, and cognitive functions in adult Ts65Dn mice. Given that DS individuals and Ts65Dn mice show sleep disturbances, and considering the key role of GABAergic transmission in sleep, we investigated whether NKCC1 upregulation contributes to sleep abnormalities in adult Ts65Dn mice. Chronic oral administration of bumetanide ameliorated the spectral profile of sleep, sleep architecture, and electroencephalogram (EEG) entropy/complexity, accompanied by a lower hyperactivity in trisomic mice. These results offer a potential avenue for addressing common sleep disturbances in DS.
Collapse
Affiliation(s)
- Maria Bolla
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163 Genoa, Italy
- Università Degli Studi di Genova, Via Balbi, 5, 16126 Genoa, Italy
| | - Giulia Colombo
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163 Genoa, Italy
| | - Matteo Falappa
- Università Degli Studi di Genova, Via Balbi, 5, 16126 Genoa, Italy
- Genetics and Epigenetics of Behavior Laboratory, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Marta Pace
- Genetics and Epigenetics of Behavior Laboratory, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Roman Baravalle
- Instituto de Física de La Plata (IFLP), CONICET-UNLP, La Plata, Buenos Aires, Argentina
- State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Nataniel Martinez
- IFIMAR (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar Del Plata, B7602AYL, Mar Del Plata, Argentina
| | - Fernando Montani
- Instituto de Física de La Plata (IFLP), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Valter Tucci
- Genetics and Epigenetics of Behavior Laboratory, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163 Genoa, Italy
- Dulbecco Telethon Institute, Rome, Italy
| |
Collapse
|
5
|
Ben-Ari Y, Danchin ÉÉ. Limitations of genomics to predict and treat autism: a disorder born in the womb. J Med Genet 2025; 62:303-310. [PMID: 40081874 PMCID: PMC12015019 DOI: 10.1136/jmg-2024-110224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Brain development involves the sequential expression of vulnerable biological processes including cell proliferation, programmed cell death, neuronal migration, synapse and functional unit formation. All these processes involve gene and activity-dependent events that can be distorted by many extrinsic and intrinsic environmental factors, including stress, microbiota, inflammatory signals, hormonal signals and epigenetic factors, hence leading to disorders born in the womb that are manifested later in autism spectrum disorders (ASDs) and other neurodevelopmental disorders. Predicting and treating such disorders call for a conceptual framework that includes all aspects of developmental biology. Here, taking the high incidence of ASDs as an example, we first discuss the intrinsic limitations of the genetic approach, notably the widely used twin studies and SNPs. We then review the long list of in utero events that can deviate developmental sequences, leading to persistent aberrant activity generated by immature misplaced and misconnected neuronal ensembles that are the direct cause of ASD. In a clinical perspective, we suggest analysing non-genetic maternity data to enable an early prediction of babies who will develop ASD years later, thereby facilitating early psycho-educative techniques. Subsequently, agents capable of selectively silencing malformed immature networks offer promising therapeutic perspectives. In summary, understanding developmental processes is critical to predicting, understanding and treating ASD, as well as most other disorders that arise in the womb.
Collapse
Affiliation(s)
| | - Étienne É Danchin
- Centre de biologie integrative, Centre de recherches sur la cognition animale, Toulouse University, Toulouse, France
| |
Collapse
|
6
|
Pizzamiglio L, Capitano F, Rusina E, Fossati G, Menna E, Léna I, Antonucci F, Mantegazza M. Neurodevelopmental defects in Dravet syndrome Scn1a +/- mice: Targeting GABA-switch rescues behavioral dysfunctions but not seizures and mortality. Neurobiol Dis 2025; 207:106853. [PMID: 40021096 DOI: 10.1016/j.nbd.2025.106853] [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: 12/26/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Dravet syndrome (DS) is a developmental and epileptic encephalopathy (DEE) caused by mutations of the SCN1A gene (NaV1.1 sodium channel) and characterized by seizures, motor disabilities and cognitive/behavioral deficits, including autistic traits. The relative role of seizures and neurodevelopmental defects in disease progression, as well as the role of the mutation in inducing early neurodevelopmental defects before symptoms' onset, are not clear yet. A delayed switch of GABAergic transmission from excitatory to inhibitory (GABA-switch) was reported in models of DS, but its effects on the phenotype have not been investigated. Using a multi-scale approach, here we show that targeting GABA-switch with the drugs KU55933 (KU) or bumetanide (which upregulate KCC2 or inhibits NKCC1 chloride transporters, respectively) rescues social interaction deficits and reduces hyperactivity observed in P21 Scn1a+/- DS mouse model. Bumetanide also improves spatial working memory defects. Importantly, neither KU nor bumetanide have effect on seizures or mortality rate. Also, we disclose early behavioral defects and delayed neurodevelopmental milestones well before seizure onset, at the beginning of NaV1.1 expression. We thus reveal that neurodevelopmental components in DS, in particular GABA switch, underlie some cognitive/behavioral defects, but not seizures. Our work provides further evidence that seizures and neuropsychiatric dysfunctions in DEEs can be uncoupled and can have differential pathological mechanisms. They could be treated separately with targeted pharmacological strategies.
Collapse
Affiliation(s)
- Lara Pizzamiglio
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | - Fabrizio Capitano
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | - Evgeniia Rusina
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | | | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Rozzano, Milan, Italy
| | - Isabelle Léna
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | - Flavia Antonucci
- Institute of Neuroscience - National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Rozzano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy.
| | - Massimo Mantegazza
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France.
| |
Collapse
|
7
|
Cainelli E, Stramucci G, Bisiacchi P. A light in the darkness: Early phases of development and the emergence of cognition. Dev Cogn Neurosci 2025; 72:101527. [PMID: 39933251 PMCID: PMC11869870 DOI: 10.1016/j.dcn.2025.101527] [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: 03/09/2024] [Revised: 01/18/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
During the prenatal period, the major brain development milestones are posed and calibrated through different mechanisms, among which endogenous activity, that prepares the "system" to face the external environment. However, the specific nature of the human nervous system, intended for brain plasticity that is varied by brain area and prolonged over time, requires much time for environmental experiences to shape the cerebral circuitries. Therefore, the neonate completely depends on the caregiver, and during the first months of postnatal life, it exhibits a transitory and limited repertoire of behaviors and skills that favors the mother in her new role. This transitory condition will gradually give way to more mature competencies, the milestones of which are posed within 2 years of age. This review takes a new perspective on early development and attempts to trace the remarkable changes from in-utero period to the second year of postnatal life, posing a bridge between the neurobiological substrate and behavioral development. We based our work on the "normal" development, pointing out the risks inherent in any development process.
Collapse
Affiliation(s)
- Elisa Cainelli
- Department of General Psychology, University of Padova, Padova 35131, Italy.
| | - Giulia Stramucci
- Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Unit of Neurology and Clinical Neurophysiology, Department of Medicine, Surgery and Neuroscience, University of Siena, Italy; School of Advanced Studies, Center of Neuroscience, University of Camerino, Camerino, MC, Italy; Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Unit of Neurology and Clinical Neurophysiology, Department of Medicine, Surgery and Neuroscience, Camerino, MC, Italy.
| | - Patrizia Bisiacchi
- Department of General Psychology, University of Padova, Padova 35131, Italy; Padova Neuroscience Center, PNC, Padova 35131, Italy.
| |
Collapse
|
8
|
Parkhill M, Salmaso N, D'Angiulli A, Lee V, Aguilar-Valles A. Emerging autism and Fragile X syndrome treatments. Trends Pharmacol Sci 2025; 46:357-371. [PMID: 40102109 DOI: 10.1016/j.tips.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
The limitations of current symptom-focused treatments drive the urgent need for effective therapies for autism and Fragile X syndrome (FXS). Currently, no approved pharmacological interventions target the core symptoms of these disorders. Advances in understanding the underlying biology of autism and FXS make this an important time to explore novel options. Indeed, several treatments have recently been tested in clinical trials, with promising results in treating core symptoms of autism and FXS. We focus on emerging interventions, such as gut microbiome therapies, anti-inflammatory approaches, bumetanide, phosphodiesterase 4D inhibitors, and endocannabinoid modulators. We also discuss factors, such as disorder heterogeneity, which may have contributed to poor efficacy in previously failed late-phase trials and impact recent trials, emphasizing the need for personalized treatment approaches.
Collapse
Affiliation(s)
- Michael Parkhill
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada; Department of Health Sciences, Carleton University, Ottawa, ON, Canada; Ottawa Brain and Mind Institute, Ottawa, ON, Canada
| | - Amedeo D'Angiulli
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada; Neurodevelopmental Health Unit, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Vivian Lee
- Department of Psychology, Carleton University, Ottawa, ON, Canada
| | | |
Collapse
|
9
|
Raveendran VA, Pressey JC, Woodin MA. Protocol to study chloride regulation in cultured mouse cortical neurons using electrophysiology. STAR Protoc 2025; 6:103628. [PMID: 39923240 PMCID: PMC11850218 DOI: 10.1016/j.xpro.2025.103628] [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: 11/08/2024] [Revised: 12/13/2024] [Accepted: 01/15/2025] [Indexed: 02/11/2025] Open
Abstract
Inhibitory synaptic transmission mediated by the neurotransmitter γ-aminobutyric acid (GABA) is dependent on the concentration of chloride ions (Cl-) in neurons, which can be assessed by making patch-clamp recordings of the reversal potential for GABA (EGABA). Here, we present a protocol to study the regulation of cation-chloride cotransporters and the strength of synaptic inhibition in cultured mouse cortical neurons using electrophysiology. We describe steps for culturing neurons isolated from postnatal pups and electrophysiological measurement of EGABA. For complete details on the use and execution of this protocol, please refer to Raveendran et al.1.
Collapse
Affiliation(s)
- Vineeth A Raveendran
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jessica C Pressey
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Melanie A Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
10
|
Kourdougli N, Nomura T, Wu MW, Heuvelmans A, Dobler Z, Contractor A, Portera-Cailliau C. The NKCC1 Inhibitor Bumetanide Restores Cortical Feedforward Inhibition and Lessens Sensory Hypersensitivity in Early Postnatal Fragile X Mice. Biol Psychiatry 2025; 97:507-516. [PMID: 38950809 PMCID: PMC11825064 DOI: 10.1016/j.biopsych.2024.06.023] [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: 01/25/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Exaggerated responses to sensory stimuli, a hallmark of fragile X syndrome, contribute to anxiety and learning challenges. Sensory hypersensitivity is recapitulated in the Fmr1 knockout (KO) mouse model of fragile X syndrome. Recent studies in Fmr1 KO mice have demonstrated differences in the activity of cortical interneurons and a delayed switch in the polarity of GABA (gamma-aminobutyric acid) signaling during development. Previously, we reported that blocking the chloride transporter NKCC1 with the diuretic bumetanide could rescue synaptic circuit phenotypes in the primary somatosensory cortex (S1) of Fmr1 KO mice. However, it remains unknown whether bumetanide can rescue earlier circuit phenotypes or sensory hypersensitivity in Fmr1 KO mice. METHODS We used acute and chronic systemic administration of bumetanide in Fmr1 KO mice and performed in vivo 2-photon calcium imaging to record neuronal activity, while tracking mouse behavior with high-resolution videos. RESULTS We demonstrated that layer 2/3 pyramidal neurons in the S1 of Fmr1 KO mice showed a higher frequency of synchronous events on postnatal day 6 than wild-type controls. This was reversed by acute administration of bumetanide. Furthermore, chronic bumetanide treatment (postnatal days 5-14) restored S1 circuit differences in Fmr1 KO mice, including reduced neuronal adaptation to repetitive whisker stimulation, and ameliorated tactile defensiveness. Bumetanide treatment also rectified the reduced feedforward inhibition of layer 2/3 neurons in the S1 and boosted the circuit participation of parvalbumin interneurons. CONCLUSIONS This further supports the notion that synaptic, circuit, and sensory behavioral phenotypes in Fmr1 KO can be mitigated by inhibitors of NKCC1, such as the Food and Drug Administration-approved diuretic bumetanide.
Collapse
Affiliation(s)
- Nazim Kourdougli
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | - Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Michelle W Wu
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, California; UCLA-Caltech Medical Scientist Training Program, University of California, Los Angeles, Los Angeles, California
| | - Anouk Heuvelmans
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | - Zoë Dobler
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, California
| | - Anis Contractor
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Carlos Portera-Cailliau
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Department of Neurobiology, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
11
|
Dell’Osso L, Bonelli C, Giovannoni F, Poli F, Anastasio L, Cerofolini G, Nardi B, Cremone IM, Pini S, Carpita B. Available Treatments for Autism Spectrum Disorder: From Old Strategies to New Options. Pharmaceuticals (Basel) 2025; 18:324. [PMID: 40143102 PMCID: PMC11944800 DOI: 10.3390/ph18030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 03/28/2025] Open
Abstract
Autism spectrum disorder (ASD) is a condition that is gaining increasing interest in research and clinical fields. Due to the improvement of screening programs and diagnostic procedures, an increasing number of cases are reaching clinical attention. Despite this, the available pharmacological options for treating ASD-related symptoms are still very limited, and while a wide number of studies are focused on children or adolescents, there is a need to increase research about the treatment of ASD in adult subjects. Given this framework, this work aims to review the available literature about pharmacological treatments for ASD, from older strategies to possible new therapeutic targets for this condition, which are often poorly responsive to available resources. The literature, besides confirming the efficacy of the approved drugs for ASD, shows a lack of adequate research for several psychopharmacological treatments despite possible promising results that need to be further investigated.
Collapse
Affiliation(s)
| | - Chiara Bonelli
- Department of Clinical and Experimental Medicine, University of Pisa, 67 Via Roma, 56126 Pisa, Italy; (L.D.); (F.G.); (F.P.); (L.A.); (G.C.); (B.N.); (I.M.C.); (S.P.); (B.C.)
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Matsushima T, Toji N, Wada K, Shikanai H, Izumi T. Embryonic exposure to valproic acid and neonicotinoid deteriorates the hyperpolarizing GABA shift and impairs long-term potentiation of excitatory transmission in the local circuit of intermediate medial mesopallium of chick telencephalon. Cereb Cortex 2025; 35:bhaf044. [PMID: 40037548 DOI: 10.1093/cercor/bhaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Embryonic exposure to valproic acid and imidacloprid (a neonicotinoid insecticide) impairs filial imprinting in hatchlings, and the deteriorating effects of valproic acid are mitigated by post-hatch injection of bumetanide, a blocker of the chloride intruder Na-K-2Cl cotransporter 1. Here, we report that these exposures depolarized the reversal potential of local GABAergic transmission in the neurons of the intermediate medial mesopallium, the pallial region critical for imprinting. Furthermore, exposure increased field excitatory post-synaptic potentials in pre-tetanus recordings and impaired long-term potentiation (LTP) by low-frequency tetanic stimulation. Bath-applied bumetanide rescued the impaired LTP in the valproic acid slices, whereas VU0463271, a blocker of the chloride extruder KCC2, suppressed LTP in the control slices, suggesting that hyperpolarizing GABA action is necessary for the potentiation of excitatory synaptic transmission. Whereas a steep increase in the gene expression of KCC2 appeared compared to NKCC1 during the peri-hatch development, significant differences were not found between valproic acid and control post-hatch chicks in these genes. Instead, both valproic acid and imidacloprid downregulated several transcriptional regulators (FOS, NR4A1, and NR4A2) and upregulated the RNA component of signal recognition particles (RN7SL1). Despite different chemical actions, valproic acid and imidacloprid could cause common neuronal effects that lead to impaired imprinting.
Collapse
Affiliation(s)
- Toshiya Matsushima
- Department of Biology, Faculty of Science, Hokkaido University, N10 W8 Kita-ku, 060-0810 Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, 061-0293 Tobetsu, Japan
- Center for Mind/Brain Sciences, University of Trento, Piazza Manifattura 1, 38068 Rovereto, Italy
| | - Noriyuki Toji
- Department of Biology, Faculty of Science, Hokkaido University, N10 W8 Kita-ku, 060-0810 Sapporo, Japan
| | - Kazuhiro Wada
- Department of Biology, Faculty of Science, Hokkaido University, N10 W8 Kita-ku, 060-0810 Sapporo, Japan
| | - Hiroki Shikanai
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, 061-0293 Tobetsu, Japan
| | - Takeshi Izumi
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, 061-0293 Tobetsu, Japan
| |
Collapse
|
13
|
Li H, Li Y, Wang T, Li S, Liu H, Ning S, Shen W, Zhao Z, Wu H. Spatiotemporal Mapping of the Oxytocin Receptor at Single-Cell Resolution in the Postnatally Developing Mouse Brain. Neurosci Bull 2025; 41:224-242. [PMID: 39277552 PMCID: PMC11794781 DOI: 10.1007/s12264-024-01296-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/24/2024] [Indexed: 09/17/2024] Open
Abstract
The oxytocin receptor (OXTR) has garnered increasing attention for its role in regulating both mature behaviors and brain development. It has been established that OXTR mediates a range of effects that are region-specific or period-specific. However, the current studies of OXTR expression patterns in mice only provide limited help due to limitations in resolution. Therefore, our objective was to generate a comprehensive, high-resolution spatiotemporal expression map of Oxtr mRNA across the entire developing mouse brain. We applied RNAscope in situ hybridization to investigate the spatiotemporal expression pattern of Oxtr in the brains of male mice at six distinct postnatal developmental stages (P7, P14, P21, P28, P42, P56). We provide detailed descriptions of Oxtr expression patterns in key brain regions, including the cortex, basal forebrain, hippocampus, and amygdaloid complex, with a focus on the precise localization of Oxtr+ cells and the variance of expression between different neurons. Furthermore, we identified some neuronal populations with high Oxtr expression levels that have been little studied, including glutamatergic neurons in the ventral dentate gyrus, Vgat+Oxtr+ cells in the basal forebrain, and GABAergic neurons in layers 4/5 of the cortex. Our study provides a novel perspective for understanding the distribution of Oxtr and encourages further investigations into its functions.
Collapse
Affiliation(s)
- Hao Li
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ying Li
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ting Wang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Shen Li
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Heli Liu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Shuyi Ning
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Wei Shen
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Zhe Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Haitao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
14
|
Ferrini L, Bartolini E, Mancini A, Tancredi R, Ferrari AR, Calderoni S. EEG Abnormalities and Phenotypic Correlates in Preschoolers with Autism Spectrum Disorder: A Single-Center Study. J Clin Med 2025; 14:529. [PMID: 39860535 PMCID: PMC11766335 DOI: 10.3390/jcm14020529] [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: 11/28/2024] [Revised: 12/21/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Background: The literature suggests the existence of an association between autism spectrum disorders (ASDs) and subclinical electroencephalographic abnormalities (SEAs), which show a heterogeneous prevalence rate (12.5-60.7%) within the pediatric ASD population. The aim of this study was to investigate the EEG findings in a cohort of ASD preschoolers and their correlation with the phenotypic characteristics. Methods: We retrospectively reviewed data on 141 ASD preschoolers evaluated in a tertiary care university hospital over the period 2008-2018. All participants underwent at least one standard polygraphic electroencephalogram (EEG) and a clinical multidisciplinary assessment with standardized instruments. Results: 77 patients (55%) showed SEAs, which were mainly represented by epileptiform discharges (p < 0.00001), especially focal and multifocal (p = 0.010). Abnormal EEG (p = 0.035) and epileptiform discharges (p = 0.014) were associated with seizure onset and were predominant in sleep (p < 0.00001). Patients with abnormal tracing (p = 0.031) and slow abnormalities (p < 0.001) were significantly younger. ASD severity was not found to be correlated with EEG results, which showed a potential, albeit non-significant, association with some psychometric parameters. Very similar results were found when patients were divided according to sex. Conclusions: EEG abnormalities appear to correlate more with ASD internalizing, externalizing and emotional comorbidities, rather than with ASD core symptoms; larger samples are needed to further investigate this association.
Collapse
Affiliation(s)
- Luca Ferrini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (L.F.); (A.M.); (R.T.); (A.R.F.); (S.C.)
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (L.F.); (A.M.); (R.T.); (A.R.F.); (S.C.)
- Tuscany PhD Programme in Neurosciences, NEUROFARBA Deparment, University of Florence, Viale Pieraccini, 6, 50139 Firenze, Italy
| | - Alice Mancini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (L.F.); (A.M.); (R.T.); (A.R.F.); (S.C.)
| | - Raffaella Tancredi
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (L.F.); (A.M.); (R.T.); (A.R.F.); (S.C.)
| | - Anna Rita Ferrari
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (L.F.); (A.M.); (R.T.); (A.R.F.); (S.C.)
| | - Sara Calderoni
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (L.F.); (A.M.); (R.T.); (A.R.F.); (S.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
15
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: implications for brain development and autism. Transl Psychiatry 2024; 14:482. [PMID: 39632793 PMCID: PMC11618798 DOI: 10.1038/s41398-024-03179-1] [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: 11/29/2023] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood-stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNA-seq data obtained from E12.5 fetal mouse brains 3 hours after VPA administration to the pregnant dam revealed that VPA rapidly and significantly increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of 399 autism risk genes was significantly altered by VPA as was expression of 258 genes that have been reported to modulate fetal brain development but are not otherwise linked to autism. Expression of genes associated with intracellular signaling pathways, neurogenesis, and excitation-inhibition balance as well as synaptogenesis, neuronal fate determination, axon and dendritic development, neuroinflammation, circadian rhythms, and epigenetic modulation of gene expression was dysregulated by VPA. Notably, at least 40 genes that are known to regulate embryonic neurogenesis were dysregulated by VPA. The goal of this study was to identify mouse genes that are: (a) significantly up- or downregulated by VPA in the fetal brain and (b) associated with autism and/or known to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity and, consequently behavior, in the adult. The genes meeting these criteria provide potential targets for future hypothesis-driven studies to elucidate the proximal causes of errors in brain connectivity underlying neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Susan G Dorsey
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
| | - Evelina Mocci
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
- Institute for Genome Sciences University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Malcolm V Lane
- Translational Toxicology/Department of Epidemiology and Public Health University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bruce K Krueger
- Departments of Physiology and Psychiatry University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
16
|
Nance MG, Sullivan KM, Puglia MH. The impact of the early environment on oxytocin receptor epigenetics and potential therapeutic implications. Pediatr Res 2024:10.1038/s41390-024-03563-z. [PMID: 39548294 DOI: 10.1038/s41390-024-03563-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/26/2024] [Accepted: 08/14/2024] [Indexed: 11/17/2024]
Abstract
Oxytocin research is rapidly evolving and increasingly reveals that epigenetic modifications to the oxytocin receptor gene (OXTR) are functional, plastic, and reliable components of oxytocinergic system function. This review outlines how OXTR epigenetics are shaped by the early life environment, impact social-developmental outcomes, and have strong potential to serve as therapeutic targets. We first establish the malleability of OXTR epigenetics in infancy in both animal models and humans through research demonstrating the impact of the early life environment on OXTR DNA methylation (OXTRm) and subsequent social behavior. Next, we detail how OXTRm serves as a predictive mechanism for neurodevelopmental outcomes in animal models of social behavior such as the prairie vole, and summarize the role of OXTRm in psychiatric disorders, emotional processing, and attachment behavior in humans. We discuss the potential of further OXTRm research to improve oxytocin therapeutics by highlighting how a deeper knowledge of OXTRm could improve the therapeutic potential of exogenous oxytocin, how OXTRm may impact additional cellular mechanisms with therapeutic potential including control of the perinatal GABA switch, and how early life therapies may target the tuning of endogenous OXTRm. Finally, we review limitations of previous oxytocin research and make recommendations for future research. IMPACT: Previous research into oxytocin therapeutics has been hampered by methodological difficulties that may be improved by assay of the oxytocin receptor gene (OXTR) and its methylation (OXTRm) Key sites of OXTRm modification link early life exposures to developmental and behavioral outcomes OXTRm appears to have a critical period of development in early life Epigenetic modification of the oxytocin receptor gene could serve as a powerful target for therapeutic interventions.
Collapse
Affiliation(s)
- Madelyn G Nance
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Kelsey M Sullivan
- Department of Pediatrics, Division of Neonatology, University of Virginia, Charlottesville, VA, USA.
| | - Meghan H Puglia
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
17
|
Tawata S, Sakaguchi K, Saito A. Androgyny and atypical sensory sensitivity associated with savant ability: a comparison between Klinefelter syndrome and sexual minorities assigned male at birth. FRONTIERS IN CHILD AND ADOLESCENT PSYCHIATRY 2024; 3:1356802. [PMID: 39816580 PMCID: PMC11732014 DOI: 10.3389/frcha.2024.1356802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 10/21/2024] [Indexed: 01/18/2025]
Abstract
Introduction The extreme male brain (EMB) theory, a major causal hypothesis of autism (ASD: autism spectrum disorder), attributes excess androgens during early development as one of the causes. While studies have generally followed the EMB theory in females at birth, the co-occurrence of ASD in males at birth has been observed in conditions that are assumed to be associated with reduced androgen action during early development, including Klinefelter syndrome (KS) and sexual minorities. ASD is also associated with atypical sensory sensitivity, synesthesia, and savant syndrome. Methods In the present study, we examined adult KS individuals (n = 22), sexual minorities assigned male at birth (n = 66), and control males matched for age and educational background to those with KS [Exploratory analysis (control 1st): n = 36; Reanalysis (control 2nd): n = 583]. Participants completed a self-report questionnaire assessing sensory hypersensitivity/hyposensitivity, savant tendency (developed for the present study), synesthesia, and sexual aspects, including gender identity and sexual orientation. Results The results of the exploratory analysis suggested that individuals with KS exhibited a higher tendency toward sensory hypersensitivity/hyposensitivity than the tendency exhibited by the controls. In the Reanalysis, sexual minorities were more likely to be synesthetes, and in both analyses sexual minorities exhibited a higher savant tendency and sensory hypersensitivity/hyposensitivity than the controls. Moreover, the gender dysphoric state was associated with phenotypes observed in individuals with ASD, such as synesthesia, savant tendency, and sensory hypersensitivity/hyposensitivity. Discussion These results suggest a common physiological background among gender dysphoria, synesthesia, savant tendency, and atypical sensory sensitivity. Thus, androgynous features (reduced effects of sex steroids during early development) in males at birth may be partially related to the phenotype commonly observed in individuals with ASD. Based on the present results, we propose that the reduction of sex steroids during early development may lead to atypical neurodevelopment and be involved in the atypicality of external and internal sensory perception, and thus in the atypicality of self-concept integration, through the disruption of oxytocin and the gamma-aminobutyric acid system modulating the neural excitation/inhibition balance.
Collapse
Affiliation(s)
- Shintaro Tawata
- Graduate School of Human Sciences, Sophia University, Chiyoda-ku, Tokyo, Japan
| | - Kikue Sakaguchi
- Research Department, National Institution for Academic Degrees and Quality Enhancement of Higher Education (NIAD-QE), Kodaira-shi, Tokyo, Japan
| | - Atsuko Saito
- Faculty of Human Sciences, Sophia University, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
18
|
Selfe JS, Steyn TJS, Shorer EF, Burman RJ, Düsterwald KM, Kraitzick AZ, Abdelfattah AS, Schreiter ER, Newey SE, Akerman CJ, Raimondo JV. All-optical reporting of inhibitory receptor driving force in the nervous system. Nat Commun 2024; 15:8913. [PMID: 39414774 PMCID: PMC11484818 DOI: 10.1038/s41467-024-53074-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] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
Ionic driving forces provide the net electromotive force for ion movement across receptors, channels, and transporters, and are a fundamental property of all cells. In the nervous system, fast synaptic inhibition is mediated by chloride permeable GABAA and glycine receptors, and single-cell intracellular recordings have been the only method for estimating driving forces across these receptors (DFGABAA). Here we present a tool for quantifying inhibitory receptor driving force named ORCHID: all-Optical Reporting of CHloride Ion Driving force. We demonstrate ORCHID's ability to provide accurate, high-throughput measurements of resting and dynamic DFGABAA from genetically targeted cell types over multiple timescales. ORCHID confirms theoretical predictions about the biophysical mechanisms that establish DFGABAA, reveals differences in DFGABAA between neurons and astrocytes, and affords the first in vivo measurements of intact DFGABAA. This work extends our understanding of inhibitory synaptic transmission and demonstrates the potential for all-optical methods to assess ionic driving forces.
Collapse
Affiliation(s)
- Joshua S Selfe
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Teresa J S Steyn
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Eran F Shorer
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Neurology, School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, United States of America
| | - Richard J Burman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kira M Düsterwald
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Gatsby Computational Neuroscience Unit, University College London, London, United Kingdom
| | - Ariel Z Kraitzick
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ahmed S Abdelfattah
- Department of Neuroscience, Brown University, Providence, Rhode Island, United States of America
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island, United States of America
| | - Eric R Schreiter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | - Sarah E Newey
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Colin J Akerman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Joseph V Raimondo
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa.
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
19
|
Naderi M, Nguyen TMN, Pompili C, Kwong RWM. Unraveling the socio-cognitive consequences of KCC2 disruption in zebrafish: implications for neurodevelopmental disorders and therapeutic interventions. Front Mol Neurosci 2024; 17:1483238. [PMID: 39469188 PMCID: PMC11513385 DOI: 10.3389/fnmol.2024.1483238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
During postnatal brain development, maintaining a delicate balance between excitation and inhibition (E/I) is essential for the precise formation of neuronal circuits. The K+/cl- cotransporter 2 (KCC2) is instrumental in this process, and its dysregulation is implicated in various neurological disorders. This study utilized zebrafish (Danio rerio) to investigate the socio-cognitive consequences of KCC2 disruption. Through CRISPR-Cas9 technology, biallelic kcc2a knockout zebrafish larvae were generated, revealing behavioral abnormalities, including impaired social interactions and memory deficits. Molecular analyses unveiled alterations in key genes associated with the GABAergic and glutamatergic systems, potentially contributing to E/I imbalance. Additionally, KCC2 disruption influenced the expression of oxytocin and BDNF, crucial regulators of social behaviors, synaptic plasticity, and memory formation. The study also explored the therapeutic potential of KCC2 modulation using pharmaceuticals, showing the rescuing effects of CLP-290 and LIT-001 on social abnormalities. However, the selective impact of LIT-001 on social behaviors, not memory, highlights the complexity of neurobehavioral modulation. In summary, this study sheds light on the pivotal role of KCC2 in shaping socio-cognitive functions and suggests potential therapeutic avenues for KCC2-related neurological disorders.
Collapse
|
20
|
Tsurutani M, Goto T, Hagihara M, Irie S, Miyamichi K. Selective vulnerability of parvocellular oxytocin neurons in social dysfunction. Nat Commun 2024; 15:8661. [PMID: 39370447 PMCID: PMC11456597 DOI: 10.1038/s41467-024-53092-w] [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: 11/19/2023] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
Selective vulnerability offers a conceptual framework for understanding neurodegenerative disorders such as Parkinson's disease, where specific neuronal types are selectively affected and adjacent ones are spared. However, the applicability of this framework to neurodevelopmental disorders, particularly those characterized by atypical social behaviors, such as autism spectrum disorder, remains uncertain. Here we show that an embryonic disturbance, known to induce social dysfunction in male mice, preferentially impaired the gene expression crucial for neural functions in parvocellular oxytocin (OT) neurons-a subtype linked to social rewards-while neighboring cell types experienced a lesser impact. Chemogenetic stimulation of OT neurons at the neonatal stage ameliorated social deficits in early adulthood, concurrent with cell-type-specific sustained recovery of pivotal gene expression within parvocellular OT neurons. Collectively, our data shed light on the transcriptomic selective vulnerability within the hypothalamic social behavioral center and provide a potential therapeutic target through specific neonatal neurostimulation.
Collapse
Affiliation(s)
- Masafumi Tsurutani
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto, Japan
| | - Teppei Goto
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Mitsue Hagihara
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Satsuki Irie
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan.
| |
Collapse
|
21
|
Ratsika A, Codagnone MG, Bastiaanssen TFS, Hoffmann Sarda FA, Lynch CMK, Ventura-Silva AP, Rosell-Cardona C, Caputi V, Stanton C, Fülling C, Clarke G, Cryan JF. Maternal high-fat diet-induced microbiota changes are associated with alterations in embryonic brain metabolites and adolescent behaviour. Brain Behav Immun 2024; 121:317-330. [PMID: 39032541 DOI: 10.1016/j.bbi.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
The developing central nervous system is highly sensitive to nutrient changes during the perinatal period, emphasising the potential impact of alterations of maternal diet on offspring brain development and behaviour. A growing body of research implicates the gut microbiota in neurodevelopment and behaviour. Maternal overweight and obesity during the perinatal period has been linked to changes in neurodevelopment, plasticity and affective disorders in the offspring, with implications for microbial signals from the maternal gut. Here we investigate the impact of maternal high-fat diet (mHFD)-induced changes in microbial signals on offspring brain development, and neuroimmune signals, and the enduring effects on behaviour into adolescence. We first demonstrate that maternal caecal microbiota composition at term pregnancy (embryonic day 18: E18) differs significantly in response to maternal diet. Moreover, mHFD resulted in the upregulation of microbial genes in the maternal intestinal tissue linked to alterations in quinolinic acid synthesis and elevated kynurenine levels in the maternal plasma, both neuronal plasticity mediators related to glutamate metabolism. Metabolomics of mHFD embryonic brains at E18 also detected molecules linked to glutamate-glutamine cycle, including glutamic acid, glutathione disulphide, and kynurenine. During adolescence, the mHFD offspring exhibited increased locomotor activity and anxiety-like behaviour in a sex-dependent manner, along with upregulation of glutamate-related genes compared to controls. Overall, our results demonstrate that maternal exposure to high-fat diet results in microbiota changes, behavioural imprinting, altered brain metabolism, and glutamate signalling during critical developmental windows during the perinatal period.
Collapse
Affiliation(s)
- Anna Ratsika
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Martin G Codagnone
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Fabiana A Hoffmann Sarda
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Caoimhe M K Lynch
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Cristina Rosell-Cardona
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Valentina Caputi
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | | | - Christine Fülling
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork T12YT20, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12YT20, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland.
| |
Collapse
|
22
|
Liao A, Zheng W, Wang S, Wang N, Li Y, Chen D, Wang Y. Sortilin is associated with progranulin deficiency and autism-like behaviors in valproic acid-induced autism rats. CNS Neurosci Ther 2024; 30:e70015. [PMID: 39218796 PMCID: PMC11366450 DOI: 10.1111/cns.70015] [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: 08/11/2023] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Neuroinflammation and microglial activation-related dendritic injury contribute to the pathogenesis of Autism Spectrum Disorder (ASD). Previous studies show that Progranulin (PGRN) is a growth factor associated with inflammation and synaptic development, but the role of PGRN in autism and the mechanisms underlying changes in PGRN expression remain unclear. AIMS To investigate the impact of PGRN in autism, we stereotactically injected recombinant PGRN into the hippocampus of ASD model rats. Additionally, we explored the possibility that sortilin may be the factor behind the alterations in PGRN by utilizing SORT1 knockdown. Ultimately, we aimed to identify potential targets for the treatment of autism. RESULTS PGRN could alleviate inflammatory responses, protect neuronal dendritic spines, and ameliorate autism-like behaviors. Meanwhile, elevated expression of sortilin and decreased levels of PGRN were observed in both ASD patients and rats. Enhanced sortilin levels facilitated PGRN internalization into lysosomes. Notably, suppressing SORT1 expression amplified PGRN levels, lessened microglial activation, and mitigated inflammation, thereby alleviating autism-like behaviors. CONCLUSION Collectively, our findings highlight elevated sortilin levels in ASD rat brains, exacerbating dendrite impairment by affecting PGRN expression. PGRN supplementation and SORT1 knockdown hold potential as therapeutic strategies for ASD.
Collapse
Affiliation(s)
- Ailing Liao
- NHC Key Laboratory of Birth Defects and Reproductive HealthChongqing Population and Family Planning Science and Technology Research InstituteChongqingChina
| | - Wenxia Zheng
- Institute of Neuroscience, School of Basic Medical ScienceChongqing Medical UniversityChongqingChina
| | | | - Nashi Wang
- Library/ArchiveChongqing Medical UniversityChongqingChina
| | | | - Di Chen
- Institute of Neuroscience, School of Basic Medical ScienceChongqing Medical UniversityChongqingChina
| | - Yan Wang
- Institute of Neuroscience, School of Basic Medical ScienceChongqing Medical UniversityChongqingChina
| |
Collapse
|
23
|
Godavarthi SK, Li HQ, Pratelli M, Spitzer NC. Embryonic exposure to environmental factors drives transmitter switching in the neonatal mouse cortex causing autistic-like adult behavior. Proc Natl Acad Sci U S A 2024; 121:e2406928121. [PMID: 39178233 PMCID: PMC11363343 DOI: 10.1073/pnas.2406928121] [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: 04/06/2024] [Accepted: 07/23/2024] [Indexed: 08/25/2024] Open
Abstract
Autism spectrum disorders (ASD) can be caused by environmental factors. These factors act early in the development of the nervous system and induce stereotyped repetitive behaviors and diminished social interactions, among other outcomes. Little is known about how these behaviors are produced. In pregnant women, delivery of valproic acid (VPA) (to control seizure activity or stabilize mood) or immune activation by a virus increases the incidence of ASD in offspring. We found that either VPA or Poly Inosine:Cytosine (which mimics a viral infection), administered at mouse embryonic day 12.5, induced a neurotransmitter switch from GABA to glutamate in PV- and CCK-expressing interneurons in the medial prefrontal cortex by postnatal day 10. The switch was present for only a brief period during early postnatal development, observed in male and female mice at postnatal day 21 and reversed in both males and females by postnatal day 30. At postnatal day 90, male mice exhibited stereotyped repetitive behaviors and diminished social interaction while female mice exhibited only stereotyped repetitive behavior. Transfecting GAD1 in PV- and CCK-expressing interneurons at postnatal day 10, to reintroduce GABA expression, overrode the switch and prevented expression of autistic-like behavior. These findings point to an important role of neurotransmitter switching in mediating the environmental causes of autism.
Collapse
Affiliation(s)
- Swetha K. Godavarthi
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Hui-quan Li
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Marta Pratelli
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Nicholas C. Spitzer
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
24
|
Luo Y, Wang L, Cao Y, Shen Y, Gu Y, Wang L. Reduced excitatory activity in the developing mPFC mediates a PV H-to-PV L transition and impaired social cognition in autism spectrum disorders. Transl Psychiatry 2024; 14:325. [PMID: 39107319 PMCID: PMC11303698 DOI: 10.1038/s41398-024-03043-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Understanding the neuropathogenesis of impaired social cognition in autism spectrum disorders (ASD) is challenging. Altered cortical parvalbumin-positive (PV+) interneurons have been consistently observed in ASD, but their roles and the underlying mechanisms remain poorly understood. In our study, we observed a downward-shifted spectrum of PV expression in the developing medial prefrontal cortex (mPFC) of ASD mouse models due to decreased activity of PV+ neurons. Surprisingly, chemogenetically suppressing PV+ neuron activity during postnatal development failed to induce ASD-like behaviors. In contrast, lowering excitatory activity in the developing mPFC not only dampened the activity state and PV expression of individual PV+ neurons, but also replicated ASD-like social deficits. Furthermore, enhancing excitation, but not PV+ interneuron-mediated inhibition, rescued social deficits in ASD mouse models. Collectively, our findings propose that reduced excitatory activity in the developing mPFC may serve as a shared local circuitry mechanism triggering alterations in PV+ interneurons and mediating impaired social functions in ASD.
Collapse
Affiliation(s)
- Yujian Luo
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Liangliang Wang
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yirong Cao
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou, China
| | - Ying Shen
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Gu
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou, China.
| | - Lang Wang
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China.
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
25
|
Sivayokan B, King C, Mali I, Payne M, Strating H, Warnes E, Bossmann SH, Plakke B. Aerobic exercise improves cognitive flexibility and modulates regional volume changes in a rat model of autism. Behav Brain Res 2024; 471:115136. [PMID: 38971431 PMCID: PMC12035974 DOI: 10.1016/j.bbr.2024.115136] [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: 04/25/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Gestational exposure to valproic acid (VPA) is a risk factor for autism spectrum disorder (ASD). Rodents exposed to VPA in utero display common features of ASD, including volumetric dysregulation in higher-order cognitive regions like the medial prefrontal cortex (mPFC), the anterior cingulate cortex (ACC), and the hippocampus. Exercise has been shown in elderly populations to boost cognition and to buffer against brain volume losses with age. This study employed an adolescent treadmill exercise intervention to facilitate cognitive flexibility and regional brain volume regulation in rats exposed to VPA during gestation. It was found that exercise improved performance on extra-dimensional shifts of attention on a set-shifting task, which is indicative of improved cognitive flexibility. Exercise decreased frontal cortex volume in females, whereas in males exercise increased the ventral hippocampus. These findings suggest that aerobic exercise may be an effective intervention to counteract the altered development of prefrontal and hippocampal regions often observed in ASD.
Collapse
Affiliation(s)
- Bhavana Sivayokan
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506, United States
| | - Cole King
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506, United States
| | - Ivina Mali
- Kansas State University, Department of Chemistry, 1212 Mid-Campus Dr. N, Manhattan, KS 66506, United States
| | - Macy Payne
- Kansas State University, Department of Chemistry, 1212 Mid-Campus Dr. N, Manhattan, KS 66506, United States
| | - Hunter Strating
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506, United States
| | - Ellie Warnes
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506, United States
| | - Stefan H Bossmann
- Kansas State University, Department of Chemistry, 1212 Mid-Campus Dr. N, Manhattan, KS 66506, United States
| | - Bethany Plakke
- Kansas State University, Psychological Sciences, 1114 Mid-Campus Dr. N, Manhattan, KS 66506, United States.
| |
Collapse
|
26
|
Kingsbury MA. The intertwining of oxytocin's effects on social affiliation and inflammation. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2024; 19:100239. [PMID: 38784104 PMCID: PMC11112266 DOI: 10.1016/j.cpnec.2024.100239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
•Oxytocin is an ancient adaptive hormone that promotes social affiliation to maximize fitness and longevity.•Oxytocin is a multifaceted hormone that regulates stress responses at all levels of cellular organization within individuals.•Oxytocin's dual actions on sociability and inflammation highlight its powerful capacity as a modulator of human health.
Collapse
Affiliation(s)
- Marcy A. Kingsbury
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, 02129, USA
| |
Collapse
|
27
|
Topchiy I, Mohbat J, Folorunso OO, Wang ZZ, Lazcano-Etchebarne C, Engin E. GABA system as the cause and effect in early development. Neurosci Biobehav Rev 2024; 161:105651. [PMID: 38579901 PMCID: PMC11081854 DOI: 10.1016/j.neubiorev.2024.105651] [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: 01/03/2024] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.
Collapse
Affiliation(s)
- Irina Topchiy
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Julie Mohbat
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne CH-1015, Switzerland
| | - Oluwarotimi O Folorunso
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Ziyi Zephyr Wang
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | | | - Elif Engin
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
28
|
Baud O, Knoop M. [Oxytocin as a neuroprotective strategy in neonates: concept and preclinical evidence]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2024; 52:418-424. [PMID: 38145743 DOI: 10.1016/j.gofs.2023.12.005] [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: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
OBJECTIVE Prematurity and intra-uterine growth retardation are responsible for brain damage associated with various neurocognitive and behavioral disorders in more than 9 million children each year. Most pharmacological strategies aimed at preventing perinatal brain injury have not demonstrated substantial clinical benefits so far. In contrast, enrichment of the newborn's environment appears to have positive effects on brain structure and function, influences newborn hormonal responses, and has lasting neurobehavioral consequences during infancy and adulthood. Oxytocin (OT), a neuropeptide released by the hypothalamus, may represent the hormonal basis for these long-term effects. METHOD This review of the literature summarizes the knowledge concerning the effect of OT in the newborn and the preclinical data supporting its neuroprotective effect. RESULTS OT plays a role during the perinatal period, in parent-child attachment and in social behavior. Furthermore, preclinical studies strongly suggest that endogenous and synthetic OT is capable of regulating the inflammatory response of the central nervous system in response to situations of prematurity or more generally insults to the developing brain. The long-term effect of synthetic OT administration during labor is also discussed. CONCLUSION All the conceptual and experimental data converge to indicate that OT would be a promising candidate for neonatal neuroprotection, in particular through the regulation of neuroinflammation.
Collapse
Affiliation(s)
- Olivier Baud
- Laboratoire du développement, Université de Genève, Genève, Suisse; Inserm U1141, Université Paris Cité, Paris, France; Service de Soins Intensifs Pédiatriques et Néonatologie, Hôpitaux Universitaires de Genève, Genève, Suisse.
| | - Marit Knoop
- Laboratoire du développement, Université de Genève, Genève, Suisse
| |
Collapse
|
29
|
Maal-Bared G, Yee M, Harding EK, Ghebreselassie M, Bergamini M, Choy R, Kim E, Di Vito S, Patel M, Amirzadeh M, Grieder TE, Coles BL, Nagy JI, Bonin RP, Steenland HW, van der Kooy D. Connexin-36-positive gap junctions in ventral tegmental area GABA neurons sustain opiate dependence. Eur J Neurosci 2024; 59:3422-3444. [PMID: 38679044 DOI: 10.1111/ejn.16366] [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: 05/17/2023] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Drug dependence is characterized by a switch in motivation wherein a positively reinforcing substance can become negatively reinforcing. Put differently, drug use can transform from a form of pleasure-seeking to a form of relief-seeking. Ventral tegmental area (VTA) GABA neurons form an anatomical point of divergence between two double dissociable pathways that have been shown to be functionally implicated and necessary for these respective motivations to seek drugs. The tegmental pedunculopontine nucleus (TPP) is necessary for opiate conditioned place preferences (CPP) in previously drug-naïve rats and mice, whereas dopaminergic (DA) transmission in the nucleus accumbens (NAc) is necessary for opiate CPP in opiate-dependent and withdrawn (ODW) rats and mice. Here, we show that this switch in functional anatomy is contingent upon the gap junction-forming protein, connexin-36 (Cx36), in VTA GABA neurons. Intra-VTA infusions of the Cx36 blocker, mefloquine, in ODW rats resulted in a reversion to a drug-naïve-like state wherein the TPP was necessary for opiate CPP and where opiate withdrawal aversions were lost. Consistent with these data, conditional knockout mice lacking Cx36 in GABA neurons (GAD65-Cre;Cx36 fl(CFP)/fl(CFP)) exhibited a perpetual drug-naïve-like state wherein opiate CPP was always DA independent, and opiate withdrawal aversions were absent even in mice subjected to an opiate dependence and withdrawal induction protocol. Further, viral-mediated rescue of Cx36 in VTA GABA neurons was sufficient to restore their susceptibility to an ODW state wherein opiate CPP was DA dependent. Our findings reveal a functional role for VTA gap junctions that has eluded prevailing circuit models of addiction.
Collapse
Affiliation(s)
- Geith Maal-Bared
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Mandy Yee
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Erika K Harding
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Martha Ghebreselassie
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Bergamini
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Roxanne Choy
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ethan Kim
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Di Vito
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Maryam Patel
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Mohammadreza Amirzadeh
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Taryn E Grieder
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Brenda L Coles
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - James I Nagy
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert P Bonin
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | - Derek van der Kooy
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [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: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Järvelä V, Hamze M, Komulainen-Ebrahim J, Rahikkala E, Piispala J, Kallio M, Kangas SM, Nickl T, Huttula M, Hinttala R, Uusimaa J, Medina I, Immonen EV. A novel pathogenic SLC12A5 missense variant in epilepsy of infancy with migrating focal seizures causes impaired KCC2 chloride extrusion. Front Mol Neurosci 2024; 17:1372662. [PMID: 38660387 PMCID: PMC11039960 DOI: 10.3389/fnmol.2024.1372662] [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: 01/18/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
The potassium-chloride co-transporter 2, KCC2, is a neuron-specific ion transporter that plays a multifunctional role in neuronal development. In mature neurons, KCC2 maintains a low enough intracellular chloride concentration essential for inhibitory neurotransmission. During recent years, pathogenic variants in the KCC2 encoding gene SLC12A5 affecting the functionality or expression of the transporter protein have been described in several patients with epilepsy of infancy with migrating focal seizures (EIMFS), a devastating early-onset developmental and epileptic encephalopathy. In this study, we identified a novel recessively inherited SLC12A5 c.692G>A, p. (R231H) variant in a patient diagnosed with severe and drug-resistant EIMFS and profound intellectual disability. The functionality of the variant was assessed in vitro by means of gramicidin-perforated patch-clamp experiments and ammonium flux assay, both of which indicated a significant reduction in chloride extrusion. Based on surface immunolabeling, the variant showed a reduction in membrane expression. These findings implicate pathogenicity of the SLC12A5 variant that leads to impaired inhibitory neurotransmission, increasing probability for hyperexcitability and epileptogenesis.
Collapse
Affiliation(s)
- Viivi Järvelä
- Nano and Molecular Systems Research Unit, University of Oulu, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Mira Hamze
- INMED, INSERM, Aix-Marseille University, Marseille, France
| | - Jonna Komulainen-Ebrahim
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Department of Children and Adolescents, Division of Pediatric Neurology, Oulu University Hospital, Oulu, Finland
| | - Elisa Rahikkala
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Department of Clinical Genetics, Oulu University Hospital, Oulu, Finland
| | - Johanna Piispala
- Department of Clinical Neurophysiology, Oulu University Hospital, Oulu, Finland
| | - Mika Kallio
- Department of Clinical Neurophysiology, Oulu University Hospital, Oulu, Finland
| | - Salla M. Kangas
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tereza Nickl
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Marko Huttula
- Nano and Molecular Systems Research Unit, University of Oulu, Oulu, Finland
| | - Reetta Hinttala
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Johanna Uusimaa
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Department of Children and Adolescents, Division of Pediatric Neurology, Oulu University Hospital, Oulu, Finland
| | - Igor Medina
- INMED, INSERM, Aix-Marseille University, Marseille, France
| | - Esa-Ville Immonen
- Nano and Molecular Systems Research Unit, University of Oulu, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
32
|
Mihalj D, Borbelyova V, Pirnik Z, Bacova Z, Ostatnikova D, Bakos J. Shank3 Deficiency Results in a Reduction in GABAergic Postsynaptic Puncta in the Olfactory Brain Areas. Neurochem Res 2024; 49:1008-1016. [PMID: 38183586 PMCID: PMC10902016 DOI: 10.1007/s11064-023-04097-2] [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: 10/04/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/08/2024]
Abstract
Dysfunctional sensory systems, including altered olfactory function, have recently been reported in patients with autism spectrum disorder (ASD). Disturbances in olfactory processing can potentially result from gamma-aminobutyric acid (GABA)ergic synaptic abnormalities. The specific molecular mechanism by which GABAergic transmission affects the olfactory system in ASD remains unclear. Therefore, the present study aimed to evaluate selected components of the GABAergic system in olfactory brain regions and primary olfactory neurons isolated from Shank3-deficient (-/-) mice, which are known for their autism-like behavioral phenotype. Shank3 deficiency led to a significant reduction in GEPHYRIN/GABAAR colocalization in the piriform cortex and in primary neurons isolated from the olfactory bulb, while no change of cell morphology was observed. Gene expression analysis revealed a significant reduction in the mRNA levels of GABA transporter 1 in the olfactory bulb and Collybistin in the frontal cortex of the Shank3-/- mice compared to WT mice. A similar trend of reduction was observed in the expression of Somatostatin in the frontal cortex of Shank3-/- mice. The analysis of the expression of other GABAergic neurotransmission markers did not yield statistically significant results. Overall, it appears that Shank3 deficiency leads to changes in GABAergic synapses in the brain regions that are important for olfactory information processing, which may represent basis for understanding functional impairments in autism.
Collapse
Affiliation(s)
- Denisa Mihalj
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Veronika Borbelyova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Zdeno Pirnik
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Bacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Daniela Ostatnikova
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia.
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| |
Collapse
|
33
|
van Andel DM, Sprengers JJ, Königs M, de Jonge MV, Bruining H. Effects of Bumetanide on Neurocognitive Functioning in Children with Autism Spectrum Disorder: Secondary Analysis of a Randomized Placebo-Controlled Trial. J Autism Dev Disord 2024; 54:894-904. [PMID: 36626004 PMCID: PMC10907457 DOI: 10.1007/s10803-022-05841-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2022] [Indexed: 01/11/2023]
Abstract
We present the secondary-analysis of neurocognitive tests in the 'Bumetanide in Autism Medication and Biomarker' (BAMBI;EUDRA-CT-2014-001560-35) study, a randomized double-blind placebo-controlled (1:1) trial testing 3-months bumetanide treatment (≤ 1 mg twice-daily) in unmedicated children 7-15 years with ASD. Children with IQ ≥ 70 were analyzed for baseline deficits and treatment-effects on the intention-to-treat-population with generalized-linear-models, principal component analysis and network analysis. Ninety-two children were allocated to treatment and 83 eligible for analyses. Heterogeneous neurocognitive impairments were found that were unaffected by bumetanide treatment. Network analysis showed higher modularity after treatment (mean difference:-0.165, 95%CI:-0.317 to - 0.013,p = .034) and changes in the relative importance of response inhibition in the neurocognitive network (mean difference:-0.037, 95%CI:-0.073 to - 0.001,p = .042). This study offers perspectives to include neurocognitive tests in ASD trials.
Collapse
Affiliation(s)
- Dorinde M van Andel
- Department of Psychiatry, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jan J Sprengers
- Department of Psychiatry, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Marsh Königs
- Department of Paediatrics, Emma Neuroscience Group, Amsterdam UMC Emma Children's Hospital, Amsterdam, The Netherlands
| | - Maretha V de Jonge
- Department of Psychiatry, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department Education and Child Studies, Faculty of Social and Behavioral Sciences, Leiden University, Leiden, The Netherlands
| | - Hilgo Bruining
- Department of Psychiatry, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht, The Netherlands.
- Child and Adolescent Psychiatry and Psychosocial Care, Emma Children's Hospital, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands.
- N=You Neurodevelopmental Precision Center, Amsterdam Neuroscience, Amsterdam Reproduction and Development, Amsterdam UMC, Amsterdam, Netherlands.
- Levvel, Center for Child and Adolescent Psychiatry, Amsterdam, Netherlands.
- Department of Child and Adolescent Psychiatry, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
34
|
Potenzieri A, Uccella S, Preiti D, Pisoni M, Rosati S, Lavarello C, Bartolucci M, Debellis D, Catalano F, Petretto A, Nobili L, Fellin T, Tucci V, Ramenghi LA, Savardi A, Cancedda L. Early IGF-1 receptor inhibition in mice mimics preterm human brain disorders and reveals a therapeutic target. SCIENCE ADVANCES 2024; 10:eadk8123. [PMID: 38427732 PMCID: PMC10906931 DOI: 10.1126/sciadv.adk8123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Besides recent advances in neonatal care, preterm newborns still develop sex-biased behavioral alterations. Preterms fail to receive placental insulin-like growth factor-1 (IGF-1), a major fetal growth hormone in utero, and low IGF-1 serum levels correlate with preterm poor neurodevelopmental outcomes. Here, we mimicked IGF-1 deficiency of preterm newborns in mice by perinatal administration of an IGF-1 receptor antagonist. This resulted in sex-biased brain microstructural, functional, and behavioral alterations, resembling those of ex-preterm children, which we characterized performing parallel mouse/human behavioral tests. Pharmacological enhancement of GABAergic tonic inhibition by the U.S. Food and Drug Administration-approved drug ganaxolone rescued functional/behavioral alterations in mice. Establishing an unprecedented mouse model of prematurity, our work dissects the mechanisms at the core of abnormal behaviors and identifies a readily translatable therapeutic strategy for preterm brain disorders.
Collapse
Affiliation(s)
- Alberto Potenzieri
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Università degli Studi di Genova, via Balbi, 5, 16126 Genoa, Italy
| | - Sara Uccella
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Deborah Preiti
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Matteo Pisoni
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Silvia Rosati
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Chiara Lavarello
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Lino Nobili
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Valter Tucci
- Genetics and Epigenetics of Behavior (GEB) Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Luca A. Ramenghi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| |
Collapse
|
35
|
Alavi MS, Al-Asady AM, Fanoudi S, Sadeghnia HR. Differential effects of antiseizure medications on neurogenesis: Evidence from cells to animals. Heliyon 2024; 10:e26650. [PMID: 38420427 PMCID: PMC10901100 DOI: 10.1016/j.heliyon.2024.e26650] [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: 03/05/2023] [Revised: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
Neurogenesis, the process of generating functionally integrated neurons from neural stem and progenitor cells, is involved in brain development during embryonic stages but continues throughout life. Adult neurogenesis plays essential roles in many brain functions such as cognition, brain plasticity, and repair. Abnormalities in neurogenesis have been described in many neuropsychiatric and neurological disorders, including epilepsy. While sharing a common property of suppressing seizures, accumulating evidence has shown that some antiseizure medications (ASM) exhibit neuroprotective potential in the non-epileptic models including Parkinson's disease, Alzheimer's disease, cerebral ischemia, or traumatic brain injury. ASM are a heterogeneous group of medications with different mechanisms of actions. Therefore, it remains to be revealed whether neurogenesis is a class effect or related to them all. In this comprehensive literature study, we reviewed the literature data on the influence of ASM on the neurogenesis process during brain development and also in the adult brain under physiological or pathological conditions. Meanwhile, we discussed the underlying mechanisms associated with the neurogenic effects of ASM by linking the reported in vivo and in vitro studies. PubMed, Web of Science, and Google Scholar databases were searched until the end of February 2023. A total of 83 studies were used finally. ASM can modulate neurogenesis through the increase or decrease of proliferation, survival, and differentiation of the quiescent NSC pool. The present article indicated that the neurogenic potential of ASM depends on the administered dose, treatment period, temporal administration of the drug, and normal or disease context.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdulridha Mohammed Al-Asady
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Sciences, Faculty of Nursing, University of Warith Al-Anbiyaa, Karbala, Iraq
- Department of Medical Sciences, Faculty of Dentistry, University of Kerbala, Karbala, Iraq
| | - Sahar Fanoudi
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Hamid R Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
36
|
Giri T, Maloney SE, Giri S, Goo YA, Song JH, Son M, Tycksen E, Conyers SB, Bice A, Ge X, Garbow JR, Quirk JD, Bauer AQ, Palanisamy A. Oxytocin-induced birth causes sex-specific behavioral and brain connectivity changes in developing rat offspring. iScience 2024; 27:108960. [PMID: 38327784 PMCID: PMC10847747 DOI: 10.1016/j.isci.2024.108960] [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: 08/21/2023] [Revised: 11/23/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024] Open
Abstract
Despite six decades of the use of exogenous oxytocin for management of labor, little is known about its effects on the developing brain. Motivated by controversial reports suggesting a link between oxytocin use during labor and autism spectrum disorders (ASDs), we employed our recently validated rat model for labor induction with oxytocin to address this important concern. Using a combination of molecular biological, behavioral, and neuroimaging assays, we show that induced birth with oxytocin leads to sex-specific disruption of oxytocinergic signaling in the developing brain, decreased communicative ability of pups, reduced empathy-like behaviors especially in male offspring, and widespread sex-dependent changes in functional cortical connectivity. Contrary to our hypothesis, social behavior, typically impaired in ASDs, was largely preserved. Collectively, our foundational studies provide nuanced insights into the neurodevelopmental impact of birth induction with oxytocin and set the stage for mechanistic investigations in animal models and prospective longitudinal clinical studies.
Collapse
Affiliation(s)
- Tusar Giri
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan E. Maloney
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Saswat Giri
- Graduate Student, School of Public Health and Social Justice, St. Louis University, St. Louis, MO, USA
| | - Young Ah Goo
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Jong Hee Song
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Minsoo Son
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Tycksen
- Genome Technology Access Center (GTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Sara B. Conyers
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Annie Bice
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xia Ge
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel R. Garbow
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - James D. Quirk
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Adam Q. Bauer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Arvind Palanisamy
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
37
|
Chen M, Lin Y, Yu C, Fu R, Shentu H, Yao J, Huang J, He Y, Yu M. Effect of cesarean section on the risk of autism spectrum disorders/attention deficit hyperactivity disorder in offspring: a meta-analysis. Arch Gynecol Obstet 2024; 309:439-455. [PMID: 37219611 DOI: 10.1007/s00404-023-07059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023]
Abstract
PURPOSE This study was conducted to investigate the relationship between cesarean section (CS) offspring and autism spectrum disorders (ASD)/attention deficit hyperactivity disorder (ADHD). METHODS Searching of the databases (PubMed, Web of Science, Embase, and Cochrane Library) for studies on the relationship between mode of delivery and ASD/ADHD until August 2022. The primary outcome was the incidence of ASD/ADHD in the offspring. RESULTS This meta-analysis included 35 studies (12 cohort studies and 23 case-control studies). Statistical results showed a higher risk of ASD (odds ratio (OR) = 1.25, P < 0.001) and ADHD (OR = 1.11, P < 0.001) in CS offspring compared to the VD group. Partial subgroup analysis showed no difference in ASD risk between CS and VD offspring in sibling-matched groups (OR = 0.98, P = 0.625). The risk of ASD was higher in females (OR = 1.66, P = 0.003) than in males (OR = 1.17, P = 0.004) in the CS offspring compared with the VD group. There was no difference in the risk of ASD between CS under regional anesthesia group and VD group (OR = 1.07, P = 0.173). However, the risk of ASD was higher in the CS offspring under general anesthesia than in the VD offspring (OR = 1.62, P < 0.001). CS offspring developed autism (OR = 1.38, P = 0.011) and pervasive developmental disorder-not otherwise specified (OR = 1.46, P = 0.004) had a higher risk than VD offspring, but there was no difference in Asperger syndrome (OR = 1.19, P = 0.115). Offspring born via CS had a higher incidence of ADHD in different subgroup analyses (sibling-matched, type of CS, and study design). CONCLUSIONS In this meta-analysis, CS was a risk factor for ASD/ADHD in offspring compared with VD.
Collapse
Affiliation(s)
- Meiling Chen
- The Public Health College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yeting Lin
- Anesthesiology Department, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
| | - Chiyuan Yu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Rongrong Fu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haojie Shentu
- The Medical Imaging College, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jie Yao
- The Public Health College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianing Huang
- The Public Health College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yujing He
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mengjiao Yu
- Emergency Medical Center, Ningbo Yinzhou No. 2 Hospital, 998 North Qianhe Road, Yinzhou District, Ningbo, 315100, Zhejiang, China.
| |
Collapse
|
38
|
McArdle CJ, Arnone AA, Heaney CF, Raab-Graham KF. A paradoxical switch: the implications of excitatory GABAergic signaling in neurological disorders. Front Psychiatry 2024; 14:1296527. [PMID: 38268565 PMCID: PMC10805837 DOI: 10.3389/fpsyt.2023.1296527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/04/2023] [Indexed: 01/26/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. In the mature brain, inhibitory GABAergic signaling is critical in maintaining neuronal homeostasis and vital human behaviors such as cognition, emotion, and motivation. While classically known to inhibit neuronal function under physiological conditions, previous research indicates a paradoxical switch from inhibitory to excitatory GABAergic signaling that is implicated in several neurological disorders. Various mechanisms have been proposed to contribute to the excitatory switch such as chloride ion dyshomeostasis, alterations in inhibitory receptor expression, and modifications in GABAergic synaptic plasticity. Of note, the hypothesized mechanisms underlying excitatory GABAergic signaling are highlighted in a number of neurodevelopmental, substance use, stress, and neurodegenerative disorders. Herein, we present an updated review discussing the presence of excitatory GABAergic signaling in various neurological disorders, and their potential contributions towards disease pathology.
Collapse
Affiliation(s)
- Colin J. McArdle
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Alana A. Arnone
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of General Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Chelcie F. Heaney
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kimberly F. Raab-Graham
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
39
|
Bosetti C, Ferrini L, Ferrari AR, Bartolini E, Calderoni S. Children with Autism Spectrum Disorder and Abnormalities of Clinical EEG: A Qualitative Review. J Clin Med 2024; 13:279. [PMID: 38202286 PMCID: PMC10779511 DOI: 10.3390/jcm13010279] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/22/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
Over the last decade, the comorbidity between Autism Spectrum Disorder (ASD) and epilepsy has been widely demonstrated, and many hypotheses regarding the common neurobiological bases of these disorders have been put forward. A variable, but significant, prevalence of abnormalities on electroencephalogram (EEG) has been documented in non-epileptic children with ASD; therefore, several scientific studies have recently tried to demonstrate the role of these abnormalities as a possible biomarker of altered neural connectivity in ASD individuals. This narrative review intends to summarize the main findings of the recent scientific literature regarding abnormalities detected with standard EEG in children/adolescents with idiopathic ASD. Research using three different databases (PubMed, Scopus and Google Scholar) was conducted, resulting in the selection of 10 original articles. Despite an important lack of studies on preschoolers and a deep heterogeneity in results, some authors speculated on a possible association between EEG abnormalities and ASD characteristics, in particular, the severity of symptoms. Although this correlation needs to be more strongly elucidated, these findings may encourage future studies aimed at demonstrating the role of electrical brain abnormalities as an early biomarker of neural circuit alterations in ASD, highlighting the potential diagnostic, prognostic and therapeutic value of EEG in this field.
Collapse
Affiliation(s)
- Chiara Bosetti
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Luca Ferrini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Anna Rita Ferrari
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
| | - Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Tuscany PhD Programme in Neurosciences, 50139 Florence, Italy
| | - Sara Calderoni
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
40
|
Comas M, De Pietri Tonelli D, Berdondini L, Astiz M. Ontogeny of the circadian system: a multiscale process throughout development. Trends Neurosci 2024; 47:36-46. [PMID: 38071123 DOI: 10.1016/j.tins.2023.11.004] [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: 07/23/2023] [Revised: 10/02/2023] [Accepted: 11/12/2023] [Indexed: 01/12/2024]
Abstract
The 24 h (circadian) timing system develops in mammals during the perinatal period. It carries out the essential task of anticipating daily recurring environmental changes to identify the best time of day for each molecular, cellular, and systemic process. Although significant knowledge has been acquired about the organization and function of the adult circadian system, relatively little is known about its ontogeny. During the perinatal period, the circadian system progressively gains functionality under the influence of the early environment. This review explores current evidence on the development of the circadian clock in mammals, highlighting the multilevel complexity of the process and the importance of gaining a better understanding of its underlying biology.
Collapse
Affiliation(s)
- Maria Comas
- Circadian Physiology of Neurons and Glia Laboratory, Achucarro Basque Center for Neuroscience, 48940 Leioa, Basque Country, Spain
| | | | - Luca Berdondini
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | - Mariana Astiz
- Circadian Physiology of Neurons and Glia Laboratory, Achucarro Basque Center for Neuroscience, 48940 Leioa, Basque Country, Spain; Ikerbasque - Basque Foundation for Science, Bilbao, Spain; Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany.
| |
Collapse
|
41
|
Ruiz-Sobremazas D, Ruiz Coca M, Morales-Navas M, Rodulfo-Cárdenas R, López-Granero C, Colomina MT, Perez-Fernandez C, Sanchez-Santed F. Neurodevelopmental consequences of gestational exposure to particulate matter 10: Ultrasonic vocalizations and gene expression analysis using a bayesian approach. ENVIRONMENTAL RESEARCH 2024; 240:117487. [PMID: 37918762 DOI: 10.1016/j.envres.2023.117487] [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: 10/02/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Air pollution has been associated with a wide range of health issues, particularly regarding cardio-respiratory diseases. Increasing evidence suggests a potential link between gestational exposure to environmental pollutants and neurodevelopmental disorders such as autism spectrum disorder. The respiratory pathway is the most commonly used exposure model regarding PM due to valid and logical reasons. However, PM deposition on food (vegetables, fruits, cereals, etc.) and water has been previously described. Although this justifies the need of unforced, oral models of exposure, preclinical studies using oral exposure are uncommon. Specifically, air pollution can modify normal brain development at genetic, cellular, and structural levels. The present work aimed to investigate the effects of oral gestational exposure to particulate matter (PM) on ultrasonic vocalizations (USV). To this end, pregnant rats were exposed to particulate matter during gestation. The body weight of the pups was monitored until the day of recording the USVs. The results revealed that the exposed group emitted more USV calls when compared to the control group. Furthermore, the calls from the exposed group were longer in duration and started earlier than those from the non-exposed group. Gene expression analyses showed that PM exposure down-regulates the expression of Gabrg2 and Maoa genes in the brain, but no effect was detected on glutamate or other neurotransmission systems. These findings suggest that gestational exposure to PM10 may be related to social deficits or other phenomena that can be analyzed with USV. In addition, we were able to detect abnormalities in the expression of genes related to different neurotransmitter systems, such as the GABAergic and monoaminergic systems. Further research is needed to fully understand the possible effects of air pollutant exposure on neurodevelopmental disorders as well as the way in which these effects are linked to differences in neurotransmission systems.
Collapse
Affiliation(s)
- Diego Ruiz-Sobremazas
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain; University of Zaragoza, Department of Psychology and Sociology, Teruel, Spain
| | - Mario Ruiz Coca
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - Miguel Morales-Navas
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - Rocío Rodulfo-Cárdenas
- Universitat Rovira I Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira I Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira I Virgili, Center of Environmental, Food and Toxicological Technology (TECNATOX), Reus, Spain
| | | | - Maria Teresa Colomina
- Universitat Rovira I Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira I Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira I Virgili, Center of Environmental, Food and Toxicological Technology (TECNATOX), Reus, Spain
| | - Cristian Perez-Fernandez
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - Fernando Sanchez-Santed
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain.
| |
Collapse
|
42
|
Zarate-Lopez D, Torres-Chávez AL, Gálvez-Contreras AY, Gonzalez-Perez O. Three Decades of Valproate: A Current Model for Studying Autism Spectrum Disorder. Curr Neuropharmacol 2024; 22:260-289. [PMID: 37873949 PMCID: PMC10788883 DOI: 10.2174/1570159x22666231003121513] [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: 08/04/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 10/25/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with increased prevalence and incidence in recent decades. Its etiology remains largely unclear, but it seems to involve a strong genetic component and environmental factors that, in turn, induce epigenetic changes during embryonic and postnatal brain development. In recent decades, clinical studies have shown that inutero exposure to valproic acid (VPA), a commonly prescribed antiepileptic drug, is an environmental factor associated with an increased risk of ASD. Subsequently, prenatal VPA exposure in rodents has been established as a reliable translational model to study the pathophysiology of ASD, which has helped demonstrate neurobiological changes in rodents, non-human primates, and brain organoids from human pluripotent stem cells. This evidence supports the notion that prenatal VPA exposure is a valid and current model to replicate an idiopathic ASD-like disorder in experimental animals. This review summarizes and describes the current features reported with this animal model of autism and the main neurobiological findings and correlates that help elucidate the pathophysiology of ASD. Finally, we discuss the general framework of the VPA model in comparison to other environmental and genetic ASD models.
Collapse
Affiliation(s)
- David Zarate-Lopez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Ana Laura Torres-Chávez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Alma Yadira Gálvez-Contreras
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara 44340, México
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
| |
Collapse
|
43
|
Yenkoyan K, Ounanian Z, Mirumyan M, Hayrapetyan L, Zakaryan N, Sahakyan R, Bjørklund G. Advances in the Treatment of Autism Spectrum Disorder: Current and Promising Strategies. Curr Med Chem 2024; 31:1485-1511. [PMID: 37888815 PMCID: PMC11092563 DOI: 10.2174/0109298673252910230920151332] [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: 03/20/2023] [Revised: 07/04/2023] [Accepted: 08/26/2023] [Indexed: 10/28/2023]
Abstract
Autism spectrum disorder (ASD) is an umbrella term for developmental disorders characterized by social and communication impairments, language difficulties, restricted interests, and repetitive behaviors. Current management approaches for ASD aim to resolve its clinical manifestations based on the type and severity of the disability. Although some medications like risperidone show potential in regulating ASD-associated symptoms, a comprehensive treatment strategy for ASD is yet to be discovered. To date, identifying appropriate therapeutic targets and treatment strategies remains challenging due to the complex pathogenesis associated with ASD. Therefore, a comprehensive approach must be tailored to target the numerous pathogenetic pathways of ASD. From currently viable and basic treatment strategies, this review explores the entire field of advancements in ASD management up to cutting-edge modern scientific research. A novel systematic and personalized treatment approach is suggested, combining the available medications and targeting each symptom accordingly. Herein, summarize and categorize the most appropriate ways of modern ASD management into three distinct categories: current, promising, and prospective strategies.
Collapse
Affiliation(s)
- Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
- Department of Biochemistry, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Zadik Ounanian
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Margarita Mirumyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
- Department of Biochemistry, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Liana Hayrapetyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
- Department of Radiation Oncology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Naira Zakaryan
- Department of Biochemistry, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Raisa Sahakyan
- Department of Biochemistry, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Geir Bjørklund
- Department of Research, Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| |
Collapse
|
44
|
Gillespie B, Panthi S, Sundram S, Hill RA. The impact of maternal immune activation on GABAergic interneuron development: A systematic review of rodent studies and their translational implications. Neurosci Biobehav Rev 2024; 156:105488. [PMID: 38042358 DOI: 10.1016/j.neubiorev.2023.105488] [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: 08/11/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Mothers exposed to infections during pregnancy disproportionally birth children who develop autism and schizophrenia, disorders associated with altered GABAergic function. The maternal immune activation (MIA) model recapitulates this risk factor, with many studies also reporting disruptions to GABAergic interneuron expression, protein, cellular density and function. However, it is unclear if there are species, sex, age, region, or GABAergic subtype specific vulnerabilities to MIA. Furthermore, to fully comprehend the impact of MIA on the GABAergic system a synthesised account of molecular, cellular, electrophysiological and behavioural findings was required. To this end we conducted a systematic review of GABAergic interneuron changes in the MIA model, focusing on the prefrontal cortex and hippocampus. We reviewed 102 articles that revealed robust changes in a number of GABAergic markers that present as gestationally-specific, region-specific and sometimes sex-specific. Disruptions to GABAergic markers coincided with distinct behavioural phenotypes, including memory, sensorimotor gating, anxiety, and sociability. Findings suggest the MIA model is a valid tool for testing novel therapeutics designed to recover GABAergic function and associated behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Sandesh Panthi
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
45
|
Havranek T, Bacova Z, Bakos J. Oxytocin, GABA, and dopamine interplay in autism. Endocr Regul 2024; 58:105-114. [PMID: 38656256 DOI: 10.2478/enr-2024-0012] [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] [Indexed: 04/26/2024] Open
Abstract
Oxytocin plays an important role in brain development and is associated with various neurotransmitter systems in the brain. Abnormalities in the production, secretion, and distribution of oxytocin in the brain, at least during some stages of the development, are critical for the pathogenesis of neuropsychiatric diseases, particularly in the autism spectrum disorder. The etiology of autism includes changes in local sensory and dopaminergic areas of the brain, which are also supplied by the hypothalamic sources of oxytocin. It is very important to understand their mutual relationship. In this review, the relationship of oxytocin with several components of the dopaminergic system, gamma-aminobutyric acid (GABA) inhibitory neurotransmission and their alterations in the autism spectrum disorder is discussed. Special attention has been paid to the results describing a reduced expression of inhibitory GABAergic markers in the brain in the context of dopaminergic areas in various models of autism. It is presumed that the altered GABAergic neurotransmission, due to the absence or dysfunction of oxytocin at certain developmental stages, disinhibits the dopaminergic signaling and contributes to the autism symptoms.
Collapse
Affiliation(s)
- Tomas Havranek
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Zuzana Bacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
46
|
D'Antoni S, Schiavi S, Buzzelli V, Giuffrida S, Feo A, Ascone F, Busceti CL, Nicoletti F, Trezza V, Catania MV. Group I and group II metabotropic glutamate receptors are upregulated in the synapses of infant rats prenatally exposed to valproic acid. Psychopharmacology (Berl) 2023; 240:2617-2629. [PMID: 37707611 PMCID: PMC10640443 DOI: 10.1007/s00213-023-06457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
RATIONALE Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social interaction and restricted/stereotyped behavior. Prenatal exposure to valproic acid (VPA) is associated with an increased risk of developing ASD in humans and autistic-like behaviors in rodents. Increasing evidence indicates that dysfunctions of glutamate receptors at synapses are associated with ASD. In the VPA rat model, an involvement of glutamate receptors in autism-like phenotypes has been suggested; however, few studies were carried out on metabotropic glutamate (mGlu) receptors. OBJECTIVES We examined the protein expression levels of group I (mGlu1 and mGlu5) and group II (mGlu2/3) mGlu receptors in rats prenatally exposed to VPA and evaluated the effect of mGlu receptor modulation on an early autism-like phenotype in these animals. METHODS We used western blotting analysis on synaptosomes obtained from forebrain of control and VPA rats at different ages (postnatal day P13, 35, 90) and carried out ultrasonic vocalization (USV) emission test in infant control and VPA rats. RESULTS The expression levels of all these receptors were significantly increased in infant VPA rats. No changes were detected in adolescent and adult rats. An acute treatment with the preferential mGlu2/3 antagonist, LY341495, attenuated the impairment in the USV emission in VPA rats. No effect was observed after a treatment with the mGlu5 selective antagonist, MTEP. CONCLUSIONS Our findings demonstrate that the expression of group I and group II mGlu receptors is upregulated at synapses of infant VPA rats and suggest that mGlu2/3 receptor modulation may have a therapeutic potential in ASD.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Samuele Giuffrida
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | - Alessandro Feo
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Fabrizio Ascone
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | | | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Vincenza Catania
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy.
| |
Collapse
|
47
|
Varian BJ, Weber KT, Erdman SE. Oxytocin and the microbiome. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2023; 16:100205. [PMID: 38108027 PMCID: PMC10724733 DOI: 10.1016/j.cpnec.2023.100205] [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: 05/04/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 12/19/2023] Open
Abstract
The mammalian host microbiome affects many targets throughout the body, at least in part through an integrated gut-brain-immune axis and neuropeptide hormone oxytocin. It was discovered in animal models that microbial symbionts, such as Lactobacillus reuteri, leverage perinatal niches to promote multigenerational good health and reproductive fitness. While roles for oxytocin were once limited to women, such as giving birth and nurturing offspring, oxytocin is now also proposed to have important roles linking microbial symbionts with overall host fitness and survival throughout the evolutionary journey.
Collapse
Affiliation(s)
- Bernard J. Varian
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Katherine T. Weber
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Susan E. Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
48
|
Becker L, Hausmann J, Hartmann AM. Both chloride-binding sites are required for KCC2-mediated transport. J Biol Chem 2023; 299:105190. [PMID: 37625593 PMCID: PMC10518353 DOI: 10.1016/j.jbc.2023.105190] [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: 01/25/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
The K+-Cl- cotransporter 2 (KCC2) plays an important role in inhibitory neurotransmission, and its impairment is associated with neurological and psychiatric disorders, including epilepsy, schizophrenia, and autism. Although KCCs transport K+ and Cl- in a 1:1 stoichiometry, two Cl- coordination sites were indicated via cryo-EM. In a comprehensive analysis, we analyzed the consequences of point mutations of residues coordinating Cl- in Cl1 and Cl2. Individual mutations of residues in Cl1 and Cl2 reduce or abolish KCC2WT function, indicating a crucial role of both Cl- coordination sites for KCC2 function. Structural changes in the extracellular loop 2 by inserting a 3xHA tag switches the K+ coordination site to another position. To investigate, whether the extension of the extracellular loop 2 with the 3xHA tag also affects the coordination of the two Cl- coordination sites, we carried out the analogous experiments for both Cl- coordinating sites in the KCC2HA construct. These analyses showed that most of the individual mutation of residues in Cl1 and Cl2 in the KCC2HA construct reduces or abolishes KCC2 function, indicating that the coordination of Cl- remains at the same position. However, the coupling of K+ and Cl- in Cl1 is still apparent in the KCC2HA construct, indicating a mutual dependence of both ions. In addition, the coordination residue Tyr569 in Cl2 shifted in KCC2HA. Thus, conformational changes in the extracellular domain affect K+ and Cl--binding sites. However, the effect on the Cl--binding sites is subtler.
Collapse
Affiliation(s)
- Lisa Becker
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jens Hausmann
- Division of Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Anna-Maria Hartmann
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany; Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| |
Collapse
|
49
|
Althammer F. Heralding a new era of oxytocinergic research: New tools, new problems? J Neuroendocrinol 2023; 35:e13333. [PMID: 37621199 DOI: 10.1111/jne.13333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
According to classic neuroendocrinology, hypothalamic oxytocin cells can be categorized into parvo- and magnocellular neurons. However, research in the last decade provided ample evidence that this black-and-white model of oxytocin neurons is most likely oversimplified. Novel genetic, functional and morphological studies indicate that oxytocin neurons might be organized in functional modules and suggest the existence of five or more distinct oxytocinergic subpopulations. However, many of these novel, automated high-throughput techniques might be inherently biased and interpretation of acquired data needs to be approached with caution to enable drawing sound and reliable conclusions. In addition, the recent finding that astrocytes in various brain regions express functional oxytocin receptors represents a paradigm shift and challenges the view that oxytocin primarily acts as a direct peptidergic neurotransmitter. This review highlights the latest technical advances in oxytocinergic research, puts recent studies on the oxytocin system into context and formulates various provocative ideas based on novel findings that challenges various prevailing hypotheses and dogmas about oxytocinergic modulation.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
50
|
Fuentes J, Parellada M, Georgoula C, Oliveira G, Marret S, Crutel V, Albarran C, Lambert E, Pénélaud PF, Ravel D, Ben Ari Y. Bumetanide oral solution for the treatment of children and adolescents with autism spectrum disorder: Results from two randomized phase III studies. Autism Res 2023; 16:2021-2034. [PMID: 37794745 DOI: 10.1002/aur.3005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 07/21/2023] [Indexed: 10/06/2023]
Abstract
The efficacy and safety of bumetanide oral solution for the treatment of autism spectrum disorder (ASD) in children and adolescents was evaluated in two international, multi-center, randomized, double-blind, placebo-controlled phase III trials; one enrolled patients aged 7-17 years (SIGN 1 trial) and the other enrolled younger patients aged 2-6 years (SIGN 2). In both studies, patients were randomized to receive bumetanide oral solution twice daily (BID) or placebo BID during a 6-month double-blind treatment period. The primary endpoint was change in Childhood Autism Rating Scale 2 (CARS2) total raw score from baseline to Week 26. Key secondary endpoints included changes in Social Responsiveness Scale-2, Clinical Global Impression Scale, and Vineland Adaptive Behavior Scale. Each study enrolled 211 patients (bumetanide, n = 107; placebo, n = 104). Both studies were terminated early due to absence of any significant difference between bumetanide and placebo in the overall studied populations. In both studies, CARS2 total raw score decreased from baseline to Week 26 in the bumetanide and placebo groups, with no statistically significant difference between groups. No differences were observed between treatment groups for any of the secondary efficacy endpoints in either study. In both studies, treatment-emergent adverse events that occurred more frequently with bumetanide than placebo included thirst, polyuria, hypokalemia, and dry mouth. These large phase III trials failed to demonstrate a benefit of bumetanide for the treatment of pediatric ASD compared with placebo. Consequently, the sponsor has discontinued the development of bumetanide for the treatment of this condition. Trial registration: https://clinicaltrials.gov: SIGN 1: NCT03715166; SIGN 2: NCT03715153.
Collapse
Affiliation(s)
- Joaquin Fuentes
- Child & Adolescent Psychiatry Service, Policlínica Gipuzkoa & GAUTENA Autism Society, San Sebastián, Spain
| | - Mara Parellada
- Servicio de Psiquiatría del Niño y del Adolescente Hospital, General Universitario Gregorio Marañón, CIBERSAM, IiSGM, Madrid, Spain
| | | | - Guiomar Oliveira
- Neurodevelopmental and Autism Unit from Child Developmental Center and Centro de Investigação e Formação Clínica, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Stéphane Marret
- Department of Neonatal Pediatrics, Intensive Care, and Neuropediatrics, Rouen University Hospital and INSERM U 1245 Team 4 Neovasc, School of Medicine, Normandy University, Rouen, France
| | - Véronique Crutel
- Neuro Immuno-Inflammation Therapeutic Area, Institut de Recherches Internationales Servier, Suresnes CEDEX, France
| | - Cristina Albarran
- Neuro Immuno-Inflammation Therapeutic Area, Institut de Recherches Internationales Servier, Suresnes CEDEX, France
| | - Estelle Lambert
- Neuro Immuno-Inflammation Therapeutic Area, Institut de Recherches Internationales Servier, Suresnes CEDEX, France
| | - Pierre-François Pénélaud
- Neuro Immuno-Inflammation Therapeutic Area, Institut de Recherches Internationales Servier, Suresnes CEDEX, France
| | | | | |
Collapse
|