1
|
Cornelissen L, Coffman S, Kim I, Underwood E, Tao A, Maloney MG, Donado C, Lobo K, Nelson CA, Hensch TK, Gabard-Durnam LJ, Berde CB. Neurodevelopment at 10 months and 2-3 years old after early and prolonged anaesthesia in infancy: General Anaesthesia & Brain Activity study (GABA) secondary analysis. BJA OPEN 2025; 14:100383. [PMID: 40129613 PMCID: PMC11931308 DOI: 10.1016/j.bjao.2025.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/09/2025] [Indexed: 03/26/2025]
Abstract
Background Effects of early and prolonged exposure to general anaesthesia on the developing brain are unclear. The study objective was to examine developmental outcomes at 10 months and 2-3 yr of age after general anaesthesia planned for >2 h in the first 2 months of life. Methods This is a secondary analysis of the General Anaesthesia & Brain Activity (GABA) study-a prospective, single-centre, longitudinal observational study. The final dataset included 59 children who were unexposed and 31 children who were exposed to early prolonged general anaesthesia who completed the primary outcome measure, Bayley Scales of Infant and Toddler Development-III (BSID) assessments at 10 months, at 2-3 yr old, or both. Analyses used adjusted Welch's t-tests, linear regression, and linear mixed effects models. Results BSID composite scores for cognition were similar between general anaesthesia and unexposed cohorts at 10 months (P adj=0.566, standardised mean difference [SMD]=0.27) and at 2-3 yr (P adj=0.651, SMD=0.25). Motor and language scores were similar between cohorts at 10 months (motor: P adj=1, SMD=0.13; language: P adj=0.806, SMD=0.19) and fell within typical reference ranges. Linear regression analysis found no association between BSID cognition scores and cumulative hours of general anaesthesia at 10 months (R=0.06, P=0.635) or at 2-3 yr (R=-0.13, P=0.293). Conclusions Children with early prolonged general anaesthesia showed BSID scores comparable to age-matched controls. This analysis provides additional preliminary support for the safety of general anaesthesia on the developing brain even when general anaesthesia is prolonged, repeated, or administered very early in life.
Collapse
Affiliation(s)
- Laura Cornelissen
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Siobhan Coffman
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabelle Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ellen Underwood
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alice Tao
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria G. Maloney
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carolina Donado
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kimberly Lobo
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Charles A. Nelson
- Laboratories of Cognitive Neurosciences, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Graduate School of Education, Cambridge, MA, USA
| | - Takao K. Hensch
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Charles B. Berde
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Lesch KP, Gorbunov N. Antisocial personality disorder:Failure to balance excitation/inhibition? Neuropharmacology 2025; 268:110321. [PMID: 39855295 DOI: 10.1016/j.neuropharm.2025.110321] [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: 08/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
While healthy brain function relies on a dynamic but tightly regulated interaction between excitation (E) and inhibition (I), a spectrum of social cognition disorders, including antisocial behavior and antisocial personality disorder (ASPD), frequently ensuing from irregular neurodevelopment, may be associated with E/I imbalance and concomitant alterations in neural connectivity. Technological advances in the evaluation of structural and functional E/I balance proxies in clinical settings and in human cell culture models provide a general basis for identification of biomarkers providing a powerful concept for prevention and intervention across different dimensions of mental health and disease. In this perspective we outline a framework for research to characterize neurodevelopmental pathways to antisocial behavior and ASPD driven by (epi)genetic factors across life, and to identify molecular targets for preventing the detrimental effects of cognitive dysfunction and maladaptive social behavior, considering psychosocial experience; to validate signatures of E/I imbalance and altered myelination proxies as biomarkers of pathogenic neural circuitry mechanisms to determine etiological processes in the transition from mental health to antisocial behavior and ASPD and in the switch from prevention to treatment; to develop a neurobiologically-grounded integrative model of antisocial behavior and ASPD resultant of disrupted E/I balance, allowing to establish objective diagnoses and monitoring tools, to personalize prevention and therapeutic decisions, to predict treatment response, and thus counteract relapse; and finally, to promote transformation of dimensional disorder taxonomy and to enhance societal awareness and reception of the neurobiological basis of antisocial behavior and ASPD.
Collapse
Affiliation(s)
- Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Child- and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
3
|
Beltrá P, Viudes-Sarrión N, Giner MJ, Tomás-Muñoz E, Pérez-Cervera L, Martín-San Agustín R, Ortega FJ, Valdesuso R, Suso-Martí L, Binshtok A, Delicado-Miralles M, Velasco E. Electrical Nerve Stimulation Induces Synaptic Plasticity in the Brain and the Spinal Cord: A Systematic Review. Neuromodulation 2025:S1094-7159(25)00053-4. [PMID: 40196976 DOI: 10.1016/j.neurom.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/22/2025] [Accepted: 02/25/2025] [Indexed: 04/09/2025]
Abstract
OBJECTIVES This review aimed to compile the literature on synaptic plasticity induced by electrical nerve stimulation (ENS) in nociceptive and somatosensory circuits within the central nervous system, with a particular focus on its effects on both the brain and spinal cord. Understanding the mechanisms underlying synaptic changes, enhances our comprehension of how ENS contributes to both pain relief and the development of experimental pain models. MATERIALS AND METHODS We conducted a systematic search of PubMed, Scopus, PEDro, SciELO, and Cochrane databases, adhering to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, and evaluated the quality of evidence using SYRCLE's risk of bias tool. The inclusion criteria were application of ENS to peripheral nerves, reporting of a detailed methodology, providing direct physiological measurements of synaptic activity (eg, field potentials or intracellular recordings), and publication in English or Spanish. From 8094 results, 85 studies met the inclusion criteria. RESULTS ENS was found to induce synaptic potentiation in 70 studies, depression in 7, and both effects in 8. These outcomes were determined by specific stimulation parameters and individual characteristics, with distinct molecular mechanisms involved in each case. Notably, most research focused on long-term potentiation in nociceptive pathways to create experimental pain models, with most studies conducted in the spinal cord. Few studies explored the link between ENS-induced synaptic plasticity and its analgesic effects or the role of plasticity in supraspinal brain regions, suggesting promising areas for future research. CONCLUSIONS ENS-induced synaptic plasticity presents a valuable opportunity for both pain management and the development of experimental pain models. Further research is needed to explore the connections between plasticity, analgesia, and higher brain regions.
Collapse
Affiliation(s)
- Patricia Beltrá
- Neuroscience in Physiotherapy, Independent Research Group, Elche, Spain; Department of Physiotherapy, Valencia University, Valencia, Spain; Department of Nursing and Nutrition, Faculty of Health Sciences, European University of Valencia, Valencia, Spain
| | - Nuria Viudes-Sarrión
- Skeletal Biology and Engineering Research Center, Catholic University of Leuven (KU Leuven), Leuven, Belgium; Human Movement Biomechanics Research Group, Department of Movement Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - María José Giner
- Neuroscience in Physiotherapy, Independent Research Group, Elche, Spain; Neuroscience Institute of Alicante, Miguel Hernández University-Superior Council for Scientific Research, San Juan de Alicante, Spain; Laboratory of Ion Channel Research, Flemish Institute for Biotechnology-Catholic University of Leuven (VIB-KU Leuven) Center for Brain & Disease Research, Leuven, Belgium
| | | | - Laura Pérez-Cervera
- Neuroscience in Physiotherapy, Independent Research Group, Elche, Spain; Faculty of Health Sciences, European University of Valencia, Valencia, Spain
| | | | - Francisco Javier Ortega
- Physical Therapy and Advanced Rehabilitation Clinic RehAv Elche, Elche, Spain; Department of Physical Therapy, University Studies Center-Cardenal Herrera University, Elche, Spain
| | - Raúl Valdesuso
- Department of Physical Therapy, University Studies Center-Cardenal Herrera University, Elche, Spain
| | - Luis Suso-Martí
- Neuroscience in Physiotherapy, Independent Research Group, Elche, Spain; Department of Physiotherapy, Valencia University, Valencia, Spain
| | - Alexander Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, Israel; Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miguel Delicado-Miralles
- Neuroscience in Physiotherapy, Independent Research Group, Elche, Spain; Department of Pathology and Surgery, Physiotherapy Area, Faculty of Medicine, Miguel Hernández University of Elche, Elche, Spain
| | - Enrique Velasco
- Neuroscience in Physiotherapy, Independent Research Group, Elche, Spain; Laboratory of Ion Channel Research, Flemish Institute for Biotechnology-Catholic University of Leuven (VIB-KU Leuven) Center for Brain & Disease Research, Leuven, Belgium.
| |
Collapse
|
4
|
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
|
5
|
Wagner L, Cakar ME, Banchik M, Chiem E, Glynn SS, Than AH, Green SA, Dapretto M. Beyond motor learning: Insights from infant magnetic resonance imaging on the critical role of the cerebellum in behavioral development. Dev Cogn Neurosci 2025; 72:101514. [PMID: 39919679 PMCID: PMC11848473 DOI: 10.1016/j.dcn.2025.101514] [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: 08/15/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 02/09/2025] Open
Abstract
Although the cerebellum is now recognized for its crucial role in non-motor functions such as language, perceptual processes, social communication, and executive function in adults, it is often overlooked in studies of non-motor behavioral development in infancy. Recent magnetic resonance imaging (MRI) research increasingly shows the cerebellum is key to understanding the emergence of complex human behaviors and neurodevelopmental conditions. This review summarizes studies from diverse MRI modalities that link early cerebellar development from birth to age two with emerging non-motor behaviors and psychiatric symptomatology. Our focus centered on both term and preterm infants, excluding studies of perinatal injury and cerebellar pathology. We conclude that the cerebellum is implicated in many non-motor behaviors and implicit learning mechanisms in infancy. The field's current limitations include inconsistencies in study design, a paucity of gold-standard infant neuroimaging tools, and treatment of the cerebellum as a uniform structure. Moving forward, the cerebellum should be considered a structure of greater interest to the developmental neuroimaging community. Studies should test developmental hypotheses about the behavioral roles of specific cerebro-cerebellar circuits, and theoretical frameworks such as Olson's "model switch" hypothesis of cerebellar learning. Large-scale, longitudinal, well-powered neuroimaging studies of typical and preterm development will be key.
Collapse
Affiliation(s)
- Lauren Wagner
- Neuroscience Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Melis E Cakar
- Neuroscience Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Megan Banchik
- Department of Psychiatry and Biobehavioral Science, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Emily Chiem
- Molecular, Cellular, Integrative Physiology Program, University of California Los Angeles, Los Angeles, CA, 90095, United States
| | - Siobhan Sive Glynn
- Department of Psychology, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Amy H Than
- Neuroscience Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Shulamite A Green
- Department of Psychiatry and Biobehavioral Science, University of California Los Angeles, Los Angeles, CA 90095, United States; Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Mirella Dapretto
- Department of Psychiatry and Biobehavioral Science, University of California Los Angeles, Los Angeles, CA 90095, United States; Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
6
|
Nishino D, Haginouchi T, Shimogiri T, Muroya S, Kawabata K, Urasoko S, Oshima I, Yasuo S, Gotoh T. A Pilot Study: Maternal Undernutrition Programs Energy Metabolism and Alters Metabolic Profile and Morphological Characteristics of Skeletal Muscle in Postnatal Beef Cattle. Metabolites 2025; 15:209. [PMID: 40137173 PMCID: PMC11944182 DOI: 10.3390/metabo15030209] [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: 01/11/2025] [Revised: 02/17/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
Objectives: This study investigated the long-term effects of maternal undernutrition on overall muscle metabolism, growth performance, and muscle characteristics in postnatal offspring of Wagyu (Japanese Black) cattle. Methods: Wagyu cows were divided into nutrient-adequate (control, CNT; n = 4, 120% of requirements) and nutrient-restricted groups (NR; n = 4; 60% of requirements), and treated from day 35 of gestation until parturition. Diets were delivered on the basis of crude protein requirements, meeting 100% and 80% of dry matter requirements in CNT and NR groups, respectively. All offspring were provided with the same diet from birth to 300 days of age (d). Longissimus thoracis muscle (LM) samples were collected from the postnatal offspring. Results: The NR offspring had lower birth body weight, but their body weight caught up before weaning. These offspring showed enhanced efficiency in nutrient utilization during the post-weaning growth period. Comprehensive analyses of metabolites and transcripts revealed the accumulation of proteinogenic amino acid, asparagine, in NR offspring LM at 300 d, while the abundance of nicotinamide adenine dinucleotide (NADH) and succinate were reduced. These changes were accompanied by decreased gene expression of nicotinamide phosphoribosyltransferase (NAMPT), NADH: ubiquinone oxidoreductase subunit A12 (NDUFA12), and NADH dehydrogenase subunit 5 (ND5), which are essential for mitochondrial energy production. Additionally, NR offspring LM exhibited decreased abundance of neurotransmitter, along with a higher proportion of slow-oxidative myofibers and a lower proportion of fast-oxidative myofibers at 300 d. Conclusions: Offspring from nutrient-restricted cows might suppress muscle energy production, primarily in the mitochondria, and conserve energy expenditure for muscle protein synthesis. These findings suggest that maternal undernutrition programs a thrifty metabolism in offspring muscle, with long-term effects.
Collapse
Affiliation(s)
- Daichi Nishino
- Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (D.N.); (S.Y.)
| | - Taketo Haginouchi
- Field Science Center for Northern Biosphere, Hokkaido University, Kita 8, Nishi 5, Kita-ku, Sapporo 060-0811, Japan;
| | - Takeshi Shimogiri
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-8580, Japan; (T.S.); (S.M.); (I.O.)
| | - Susumu Muroya
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-8580, Japan; (T.S.); (S.M.); (I.O.)
| | - Kenji Kawabata
- Livestock Experiment Station, Kagoshima Prefectural Institute for Agricultural Development, 2440 Kokubuuenodan, Kirishima 899-4461, Japan; (K.K.); (S.U.)
| | - Saki Urasoko
- Livestock Experiment Station, Kagoshima Prefectural Institute for Agricultural Development, 2440 Kokubuuenodan, Kirishima 899-4461, Japan; (K.K.); (S.U.)
| | - Ichiro Oshima
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-8580, Japan; (T.S.); (S.M.); (I.O.)
| | - Shinobu Yasuo
- Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (D.N.); (S.Y.)
| | - Takafumi Gotoh
- Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (D.N.); (S.Y.)
- Field Science Center for Northern Biosphere, Hokkaido University, Kita 8, Nishi 5, Kita-ku, Sapporo 060-0811, Japan;
| |
Collapse
|
7
|
Kopalli SR, Behl T, Kyada A, Rekha MM, Kundlas M, Rani P, Nathiya D, Satyam Naidu K, Gulati M, Bhise M, Gupta P, Wal P, Fareed M, Ramniwas S, Koppula S, Gasmi A. Synaptic plasticity and neuroprotection: The molecular impact of flavonoids on neurodegenerative disease progression. Neuroscience 2025; 569:161-183. [PMID: 39922366 DOI: 10.1016/j.neuroscience.2025.02.007] [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: 01/11/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Flavonoids are a broad family of polyphenolic chemicals that are present in a wide variety of fruits, vegetables, and medicinal plants. Because of their neuroprotective qualities, flavonoids have attracted a lot of interest. The potential of flavonoids to control synaptic plasticity-a crucial process underlying memory, learning, and cognitive function-is becoming more and more clear. Dysregulation of synaptic plasticity is a feature of neurodegenerative diseases such as amyotrophic lateral sclerosis (0.4 %), Parkinson's (1-2 %), Alzheimer's (5-7 %), and Huntington's ((0.2 %)). This review discusses the molecular mechanisms via which flavonoids influence synaptic plasticity as well as their therapeutic potential in neurodegenerative diseases. Flavonoids modulate key signaling pathways such as MAPK/ERK and PI3K/Akt/mTOR to support neuroprotection, synaptic plasticity, and neuronal health, while also influencing neurotrophic factors (BDNF, NGF) and their receptors (TrkB, TrkA). They regulate neurotransmitter receptors like GABA, AMPA, and NMDA to balance excitatory and inhibitory transmission, and exert antioxidant effects via the Nrf2-ARE pathway and anti-inflammatory actions by inhibiting NF-κB signaling, highlighting their potential for treating neurodegenerative diseases. These varied reactions support the preservation of synapse function and neuronal integrity in the face of neurodegenerative insults. Flavonoids can reduce the symptoms of neurodegeneration, prevent synaptic loss, and enhance cognitive function, according to experimental studies. However, there are still obstacles to using these findings in clinical settings, such as limited bioavailability and the need for consistent dose. The focus of future research should be on improving flavonoid delivery systems and combining them with conventional medications.
Collapse
Affiliation(s)
- Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006 Republic of Korea
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab 140306, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401 Punjab, India
| | - Pooja Rani
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | | | | | - Pranay Wal
- PSIT- Pranveer Singh Institute of Technology, Pharmacy Kanpur UP, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 13713, Saudi Arabia
| | - Seema Ramniwas
- University Centre for Research and Development, Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413 Punjab, India
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| | - Amin Gasmi
- Societe Francophone de Nutritherapie et de Nutrigenetique Appliquee, Villeurbanne, France; International Institute of Nutrition and Micronutrient Sciences, Saint-Etienne, France
| |
Collapse
|
8
|
Hayano Y, Miyoshi G, Paul A, Taniguchi H. Editorial: Cellular and molecular mechanisms that govern assembly, plasticity, and function of GABAergic inhibitory circuits in the mammalian brain. Front Cell Neurosci 2025; 19:1568845. [PMID: 40007758 PMCID: PMC11850328 DOI: 10.3389/fncel.2025.1568845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Affiliation(s)
- Yasufumi Hayano
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Goichi Miyoshi
- Department of Developmental Genetics and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Anirban Paul
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Hiroki Taniguchi
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
9
|
Lakhera S, Herbert E, Gjorgjieva J. Modeling the Emergence of Circuit Organization and Function during Development. Cold Spring Harb Perspect Biol 2025; 17:a041511. [PMID: 38858072 PMCID: PMC11864115 DOI: 10.1101/cshperspect.a041511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Developing neural circuits show unique patterns of spontaneous activity and structured network connectivity shaped by diverse activity-dependent plasticity mechanisms. Based on extensive experimental work characterizing patterns of spontaneous activity in different brain regions over development, theoretical and computational models have played an important role in delineating the generation and function of individual features of spontaneous activity and their role in the plasticity-driven formation of circuit connectivity. Here, we review recent modeling efforts that explore how the developing cortex and hippocampus generate spontaneous activity, focusing on specific connectivity profiles and the gradual strengthening of inhibition as the key drivers behind the observed developmental changes in spontaneous activity. We then discuss computational models that mechanistically explore how different plasticity mechanisms use this spontaneous activity to instruct the formation and refinement of circuit connectivity, from the formation of single neuron receptive fields to sensory feature maps and recurrent architectures. We end by highlighting several open challenges regarding the functional implications of the discussed circuit changes, wherein models could provide the missing step linking immature developmental and mature adult information processing capabilities.
Collapse
Affiliation(s)
- Shreya Lakhera
- School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Elizabeth Herbert
- School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Julijana Gjorgjieva
- School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
10
|
Hochstetler A, Courtney Y, Oloko P, Baskin B, Ding-Su A, Stinson T, McGuone D, Haynes R, Lehtinen MK, Costine-Bartell B. Acute temporal, regional, and cell-type specific NKCC1 disruption following severe TBI in the developing gyrencephalic brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633889. [PMID: 39896526 PMCID: PMC11785025 DOI: 10.1101/2025.01.20.633889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Traumatic brain injury (TBI) in children is a leading cause of morbidity and mortality, with no effective treatment and limited clinical management. We developed a multifactorial traumatic brain injury model in piglets which mirrors the evolving pathophysiology of severe pediatric TBI, showing age-dependent hypoxic-ischemic cerebral cortical injury and matrix metalloproteinase-driven vasogenic edema, with infant piglets experiencing less tissue damage than toddler piglets. Extracellular matrix breakdown can precipitate neuronal dysfunction, disrupting chloride homeostasis and the reversal potential for GABA. We hypothesized that ongoing tissue damage might be related to markers of "immature GABA", evaluated by changes to the expression and phosphorylation of sodium-potassium-2-chloride cotransporter 1 (NKCC1), potassium-chloride cotransporter 2 (KCC2), and a regulatory kinase, (STE20/SPS1-related proline-alanine-rich protein kinase) SPAK. We mapped these markers in developing swine and infant human brain, identifying a postnatal pNKCC1 decrease in human infant hippocampus, and a perinatal cortical and hippocampal GABA switch in pigs, with no change in the thalamus. In infant piglets with severe TBI, upregulation of neuronal pNKCC1 correlated with hypoxic-ischemic injury and seizure duration. We also observed dysregulation of NKCC1, KCC2, and SPAK in cortex and hippocampus in infant and toddler piglets with severe TBI, with thalamus unchanged. We noted ectopic, non-apical localization of pNKCC1 signal in choroid plexus epithelium across ages in piglets and humans with severe TBI, indicating acute dysregulation of the CSF chloride milieu. These findings position swine as a useful model for pediatric TBI research and suggest that SPAK or NKCC1 inhibition in infants may be therapeutic. Significance statement Severe TBI in early childhood, the majority of which is due to abuse, remains an understudied area of neurotrauma. Our piglet model effectively replicates the pathophysiology of severe pediatric TBI, capturing age-dependent injury patterns and mechanisms of spreading hypoxic-ischemic injury throughout the cortical ribbon. We identified upregulation of neuronal-pNKCC1 in infant piglets, but not toddler piglets with severe TBI, and found this correlates with injury severity, seizure duration, and subarachnoid hemorrhage. Our findings indicate that treatments targeted to inhibit neuronal NKCC1 might alleviate evolving brain injury in infants with severe TBI. This model provides a valuable platform for studying mechanisms of TBI and testing new interventions, potentially advancing therapeutic strategies for pediatric brain injury where stopping traumatic seizures is difficult.
Collapse
|
11
|
Feichtenbiner AB, Sytsma K, O'Boyle RP, Mittenzwei R, Maioli H, Scherpelz KP, Child DD, Li N, Ariza Torres J, Keene L, Kirkland A, Howard K, Latimer C, Keene CD, Ransom C, Nolan AL. Satellite microglia: marker of traumatic brain injury and regulator of neuronal excitability. J Neuroinflammation 2025; 22:9. [PMID: 39819341 PMCID: PMC11740464 DOI: 10.1186/s12974-024-03328-9] [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: 10/18/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
Traumatic brain injury is a leading cause of chronic neurologic disability and a risk factor for development of neurodegenerative disease. However, little is known regarding the pathophysiology of human traumatic brain injury, especially in the window after acute injury and the later life development of progressive neurodegenerative disease. Given the proposed mechanisms of toxic protein production and neuroinflammation as possible initiators or contributors to progressive pathology, we examined phosphorylated tau accumulation, microgliosis and astrogliosis using immunostaining in the orbitofrontal cortex, a region often vulnerable across traumatic brain injury exposures, in an age and sex-matched cohort of community traumatic brain injury including both mild and severe cases in midlife. We found that microglial response is most prominent after chronic traumatic brain injury, and interactions with neurons in the form of satellite microglia are increased, even after mild traumatic brain injury. Taking our investigation into a mouse model, we identified that these satellite microglia suppress neuronal excitability in control conditions but lose this ability with chronic traumatic brain injury. At the same time, network hyperexcitability is present in both mouse and human orbitofrontal cortex. Our findings support a role for loss of homeostatic control by satellite microglia in the maladaptive circuit changes that occur after traumatic brain injury.
Collapse
Affiliation(s)
- Alicia B Feichtenbiner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Karinn Sytsma
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Ryan P O'Boyle
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Rhonda Mittenzwei
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
- King County Office of the Medical Examiner, Seattle, WA, 98104, USA
| | - Heather Maioli
- Office of Chief Medical Examiner of the City of New York, New York, NY, 10016, USA
| | - Kathryn P Scherpelz
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Daniel D Child
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Ning Li
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA
| | | | - Lisa Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Amanda Kirkland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Kimberly Howard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Caitlin Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Christopher Ransom
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA
| | - Amber L Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA.
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA.
| |
Collapse
|
12
|
Cepeda C, Holley SM, Barry J, Oikonomou KD, Yazon VW, Peng A, Argueta D, Levine MS. Corticostriatal maldevelopment in the R6/2 mouse model of juvenile Huntington's disease. Neurobiol Dis 2025; 204:106752. [PMID: 39644979 DOI: 10.1016/j.nbd.2024.106752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024] Open
Abstract
There is a growing consensus that brain development in Huntington's disease (HD) is abnormal, leading to the idea that HD is not only a neurodegenerative but also a neurodevelopmental disorder. Indeed, structural and functional abnormalities have been observed during brain development in both humans and animal models of HD. However, a concurrent study of cortical and striatal development in a genetic model of HD is still lacking. Here we report significant alterations of corticostriatal development in the R6/2 mouse model of juvenile HD. We examined wildtype (WT) and R6/2 mice at postnatal (P) days 7, 14, and 21. Morphological examination demonstrated early structural and cellular alterations reminiscent of malformations of cortical development, and ex vivo electrophysiological recordings of cortical pyramidal neurons (CPNs) demonstrated significant age- and genotype-dependent changes of intrinsic membrane and synaptic properties. In general, R6/2 CPNs had reduced cell membrane capacitance and increased input resistance (P7 and P14), along with reduced frequency of spontaneous excitatory and inhibitory synaptic events during early development (P7), suggesting delayed cortical maturation. This was confirmed by increased occurrence of GABAA receptor-mediated giant depolarizing potentials at P7. At P14, the rheobase of CPNs was significantly reduced, along with increased excitability. Altered membrane and synaptic properties of R6/2 CPNs recovered progressively, and by P21 they were similar to WT CPNs. In striatal medium-sized spiny neurons (MSNs), a different picture emerged. Intrinsic membrane properties were relatively normal throughout development, except for a transient increase in membrane capacitance at P14. The first alterations in MSNs synaptic activity were observed at P14 and consisted of significant deficits in GABAergic inputs, however, these also were normalized by P21. In contrast, excitatory inputs began to decrease at this age. We conclude that the developing HD brain is capable of compensating for early developmental abnormalities and that cortical alterations precede and are a main contributor of striatal changes. Addressing cortical maldevelopment could help prevent or delay disease manifestations.
Collapse
Affiliation(s)
- Carlos Cepeda
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA.
| | - Sandra M Holley
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Joshua Barry
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Katerina D Oikonomou
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Vannah-Wila Yazon
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Allison Peng
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Deneen Argueta
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Michael S Levine
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
13
|
Margolis ET, Gabard‐Durnam LJ. Prenatal influences on postnatal neuroplasticity: Integrating DOHaD and sensitive/critical period frameworks to understand biological embedding in early development. INFANCY 2025; 30:e12588. [PMID: 38449347 PMCID: PMC11647198 DOI: 10.1111/infa.12588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Early environments can have significant and lasting effects on brain, body, and behavior across the lifecourse. Here, we address current research efforts to understand how experiences impact neurodevelopment with a new perspective integrating two well-known conceptual frameworks - the Developmental Origins of Health and Disease (DOHaD) and sensitive/critical period frameworks. Specifically, we consider how prenatal experiences characterized in the DOHaD model impact two key neurobiological mechanisms of sensitive/critical periods for adapting to and learning from the postnatal environment. We draw from both animal and human research to summarize the current state of knowledge on how particular prenatal substance exposures (psychoactive substances and heavy metals) and nutritional profiles (protein-energy malnutrition and iron deficiency) each differentially impact brain circuits' excitation/GABAergic inhibition balance and myelination. Finally, we highlight new research directions that emerge from this integrated framework, including testing how prenatal environments alter sensitive/critical period timing and learning and identifying potential promotional/buffering prenatal exposures to impact postnatal sensitive/critical periods. We hope this integrative framework considering prenatal influences on postnatal neuroplasticity will stimulate new research to understand how early environments have lasting consequences on our brains, behavior, and health.
Collapse
Affiliation(s)
- Emma T. Margolis
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
| | | |
Collapse
|
14
|
Sicher AR, Crowley NA. Adolescent Alcohol Exposure Dysregulates Developing Cortical GABA Circuits. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:159-177. [PMID: 40128479 DOI: 10.1007/978-3-031-81908-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Adolescence is a critical developmental period during which physical, behavioral, and neurobiological maturation occurs. Within the brain, the prefrontal cortex is one of the last brain regions to undergo remodeling, often into adulthood. These relatively late developmental changes leave the prefrontal cortex uniquely vulnerable to insults beginning in adolescence-including alcohol exposure. Adolescents initiate alcohol consumption at a high rate, increasing the risk of lasting consequences through impairing the typical development of the prefrontal cortex. In this chapter, we discuss the development of prefrontal circuitry and the current literature investigating how alcohol influences prefrontal development. We primarily focus on preclinical studies in rodent models, which allow for the study of specific populations of neurons in the prefrontal cortex. We identify several future directions for adolescent alcohol research, including greater focus on neuropeptides and stronger understanding of sex differences in brain maturation and alcohol consumption.
Collapse
Affiliation(s)
- Avery R Sicher
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Nicole A Crowley
- Department of Biology, The Pennsylvania State University, University Park, PA, USA.
- Neuroscience Graduate Program, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, USA.
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
15
|
Szpręgiel I, Bysiek A. Psilocybin and the glutamatergic pathway: implications for the treatment of neuropsychiatric diseases. Pharmacol Rep 2024; 76:1297-1304. [PMID: 39412581 PMCID: PMC11582295 DOI: 10.1007/s43440-024-00660-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: 07/07/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 11/22/2024]
Abstract
In recent decades, psilocybin has gained attention as a potential drug for several mental disorders. Clinical and preclinical studies have provided evidence that psilocybin can be used as a fast-acting antidepressant. However, the exact mechanisms of action of psilocybin have not been clearly defined. Data show that psilocybin as an agonist of 5-HT2A receptors located in cortical pyramidal cells exerted a significant effect on glutamate (GLU) extracellular levels in both the frontal cortex and hippocampus. Increased GLU release from pyramidal cells in the prefrontal cortex results in increased activity of γ-aminobutyric acid (GABA)ergic interneurons and, consequently, increased release of the GABA neurotransmitter. It seems that this mechanism appears to promote the antidepressant effects of psilocybin. By interacting with the glutamatergic pathway, psilocybin seems to participate also in the process of neuroplasticity. Therefore, the aim of this mini-review is to discuss the available literature data indicating the impact of psilocybin on glutamatergic neurotransmission and its therapeutic effects in the treatment of depression and other diseases of the nervous system.
Collapse
Affiliation(s)
- Izabela Szpręgiel
- Department of Pharmacology and Brain Biostructure, Unit II, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland.
| | - Agnieszka Bysiek
- Department of Pharmacology and Brain Biostructure, Unit II, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland
| |
Collapse
|
16
|
de Groot ER, Dudink J, Austin T. Sleep as a driver of pre- and postnatal brain development. Pediatr Res 2024; 96:1503-1509. [PMID: 38956219 PMCID: PMC11624135 DOI: 10.1038/s41390-024-03371-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
In 1966, Howard Roffwarg proposed the ontogenic sleep hypothesis, relating neural plasticity and development to rapid eye movement (REM) sleep, a hypothesis that current fetal and neonatal sleep research is still exploring. Recently, technological advances have enabled researchers to automatically quantify neonatal sleep architecture, which has caused a resurgence of research in this field as attempts are made to further elucidate the important role of sleep in pre- and postnatal brain development. This article will review our current understanding of the role of sleep as a driver of brain development and identify possible areas for future research. IMPACT: The evidence to date suggests that Roffwarg's ontogenesis hypothesis of sleep and brain development is correct. A better understanding of the relationship between sleep and the development of functional connectivity is needed. Reliable, non-invasive tools to assess sleep in the NICU and at home need to be tested in a real-world environment and the best way to promote healthy sleep needs to be understood before clinical trials promoting and optimizing sleep quality in neonates could be undertaken.
Collapse
Affiliation(s)
- Eline R de Groot
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
- Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Topun Austin
- NeoLab, Evelyn Perinatal Imaging Centre, The Rosie Hospital, Cambridge University Hospitals, Cambridge, UK.
| |
Collapse
|
17
|
Gonda X, Tarazi FI, Dome P. The emergence of antidepressant drugs targeting GABA A receptors: A concise review. Biochem Pharmacol 2024; 228:116481. [PMID: 39147329 DOI: 10.1016/j.bcp.2024.116481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Depression is among the most common psychiatric illnesses, which imposes a major socioeconomic burden on patients, caregivers, and the public health system. Treatment with classical antidepressants (e.g. tricyclic antidepressants and selective serotonine reuptake inhibitors), which primarily affect monoaminergic systems has several limitations, such as delayed onset of action and moderate efficacy in a relatively large proportion of depressed patients. Furthermore, depression is highly heterogeneus, and its different subtypes, including post-partum depression, involve distinct neurobiology, warranting a differential approach to pharmacotherapy. Given these shortcomings, the need for novel antidepressants that are superior in efficacy and faster in onset of action is fully justified. The development and market introduction of rapid-acting antidepressants has accelerated in recent years. Some of these new antidepressants act through the GABAergic system. In this review, we discuss the discovery, efficacy, and limitations of treatment with classic antidepressants. We provide a detailed discussion of GABAergic neurotransmission, with a special focus on GABAA receptors, and possible explanations for the mood-enhancing effects of GABAergic medications (in particular neurosteroids acting at GABAA receptors), and, ultimately, we present the most promising molecules belonging to this family which are currently used in clinical practice or are in late phases of clinical development.
Collapse
Affiliation(s)
- Xenia Gonda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary.
| | - Frank I Tarazi
- Department of Psychiatry and Neurology, Harvard Medical School and McLean Hospital, Boston, MA, USA
| | - Peter Dome
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Nyiro Gyula National Institute of Psychiatry and Addictology, Budapest, Hungary
| |
Collapse
|
18
|
Aroniadou-Anderjaska V, Figueiredo TH, De Araujo Furtado M, Pidoplichko VI, Lumley LA, Braga MFM. Alterations in GABA A receptor-mediated inhibition triggered by status epilepticus and their role in epileptogenesis and increased anxiety. Neurobiol Dis 2024; 200:106633. [PMID: 39117119 DOI: 10.1016/j.nbd.2024.106633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
The triggers of status epilepticus (SE) in non-epileptic patients can vary widely, from idiopathic causes to exposure to chemoconvulsants. Regardless of its etiology, prolonged SE can cause significant brain damage, commonly resulting in the development of epilepsy, which is often accompanied by increased anxiety. GABAA receptor (GABAAR)-mediated inhibition has a central role among the mechanisms underlying brain damage and the ensuing epilepsy and anxiety. During SE, calcium influx primarily via ionotropic glutamate receptors activates signaling cascades which trigger a rapid internalization of synaptic GABAARs; this weakens inhibition, exacerbating seizures and excitotoxicity. GABAergic interneurons are more susceptible to excitotoxic death than principal neurons. During the latent period of epileptogenesis, the aberrant reorganization in synaptic interactions that follow interneuronal loss in injured brain regions, leads to the formation of hyperexcitable, seizurogenic neuronal circuits, along with disturbances in brain oscillatory rhythms. Reduction in the spontaneous, rhythmic "bursts" of IPSCs in basolateral amygdala neurons is likely to play a central role in anxiogenesis. Protecting interneurons during SE is key to preventing both epilepsy and anxiety. Antiglutamatergic treatments, including antagonism of calcium-permeable AMPA receptors, can be expected to control seizures and reduce excitotoxicity not only by directly suppressing hyperexcitation, but also by counteracting the internalization of synaptic GABAARs. Benzodiazepines, as delayed treatment of SE, have low efficacy due to the reduction and dispersion of their targets (the synaptic GABAARs), but also because themselves contribute to further reduction of available GABAARs at the synapse; furthermore, benzodiazepines may be completely ineffective in the immature brain.
Collapse
Affiliation(s)
- Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Marcio De Araujo Furtado
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Volodymyr I Pidoplichko
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Lucille A Lumley
- U.S. Army Medical Research Institute of Chemical Defense, Aberdeen, Proving Ground, MD, USA.
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
19
|
Capilla-López J, Hernández RG, Carrero-Rojas G, Calvo PM, Alvarez FJ, de la Cruz RR, Pastor AM. VEGF, but Not BDNF, Prevents the Downregulation of KCC2 Induced by Axotomy in Extraocular Motoneurons. Int J Mol Sci 2024; 25:9942. [PMID: 39337430 PMCID: PMC11432591 DOI: 10.3390/ijms25189942] [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: 08/14/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
The potassium-chloride cotransporter KCC2 is the main extruder of Cl- in neurons. It plays a fundamental role in the activity of the inhibitory neurotransmitters (GABA and glycine) since low levels of KCC2 promote intracellular Cl- accumulation, leading to the depolarizing activity of GABA and glycine. The downregulation of this cotransporter occurs in neurological disorders characterized by hyperexcitability, such as epilepsy, neuropathic pain, and spasticity. KCC2 is also downregulated after axotomy. If muscle reinnervation is allowed, the KCC2 levels recover in motoneurons. Therefore, we argued that target-derived neurotrophic factors might be involved in the regulation of KCC2 expression. For this purpose, we performed the axotomy of extraocular motoneurons via the monocular enucleation of adult rats, and a pellet containing either VEGF or BDNF was chronically implanted in the orbit. Double confocal immunofluorescence of choline acetyl-transferase (ChAT) and KCC2 was carried out in the brainstem sections. Axotomy led to a KCC2 decrease in the neuropil and somata of extraocular motoneurons, peaking at 15 days post-lesion, with the exception of the abducens motoneuron somata. VEGF administration prevented the axotomy-induced KCC2 downregulation. By contrast, BDNF either maintained or reduced the KCC2 levels following axotomy, suggesting that BDNF is involved in the axotomy-induced KCC2 downregulation in extraocular motoneurons. The finding that VEGF prevents KCC2 decrease opens up new possibilities for the treatment of neurological disorders coursing with neuronal hyperactivity due to KCC2 downregulation.
Collapse
Affiliation(s)
- Jaime Capilla-López
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Rosendo G Hernández
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Génova Carrero-Rojas
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University Vienna, 1090 Vienna, Austria
| | - Paula M Calvo
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | | | - Rosa R de la Cruz
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| |
Collapse
|
20
|
Guha A, Hunter SK, Legget KT, McHugo M, Hoffman MC, Tregellas JR. Intrinsic Infant Hippocampal Function Supports Inhibitory Processing. Dev Psychobiol 2024; 66:e22529. [PMID: 39010701 PMCID: PMC11254329 DOI: 10.1002/dev.22529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/22/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024]
Abstract
Impaired cerebral inhibition is commonly observed in neurodevelopmental disorders and may represent a vulnerability factor for their development. The hippocampus plays a key role in inhibition among adults and undergoes significant and rapid changes during early brain development. Therefore, the structure represents an important candidate region for early identification of pathology that is relevant to inhibitory dysfunction. To determine whether hippocampal function corresponds to inhibition in the early postnatal period, the present study evaluated relationships between hippocampal activity and sensory gating in infants 4-20 weeks of age (N = 18). Resting-state functional magnetic resonance imaging was used to measure hippocampal activity, including the amplitude of low-frequency fluctuations (ALFFs) and fractional ALFF. Electroencephalography during a paired-stimulus paradigm was used to measure sensory gating (P50). Higher activity of the right hippocampus was associated with better sensory gating (P50 ratio), driven by a reduction in response to the second stimulus. These findings suggest that meaningful effects of hippocampal function can be detected early in infancy. Specifically, higher intrinsic hippocampal activity in the early postnatal period may support effective inhibitory processing. Future work will benefit from longitudinal analysis to clarify the trajectory of hippocampal function, alterations of which may contribute to the risk of neurodevelopmental disorders and represent an intervention target.
Collapse
Affiliation(s)
- Anika Guha
- Department of Psychiatry, University of Colorado Anschutz Medical Campus
| | - Sharon K. Hunter
- Department of Psychiatry, University of Colorado Anschutz Medical Campus
| | - Kristina T. Legget
- Department of Psychiatry, University of Colorado Anschutz Medical Campus
- Research Service, Rocky Mountain Regional VA Medical Center
| | - Maureen McHugo
- Department of Psychiatry, University of Colorado Anschutz Medical Campus
| | - M. Camille Hoffman
- Department of Psychiatry, University of Colorado Anschutz Medical Campus
| | - Jason R. Tregellas
- Department of Psychiatry, University of Colorado Anschutz Medical Campus
- Research Service, Rocky Mountain Regional VA Medical Center
| |
Collapse
|
21
|
Kapr J, Scharkin I, Ramachandran H, Westhoff P, Pollet M, Dangeleit S, Brockerhoff G, Rossi A, Koch K, Krutmann J, Fritsche E. HiPSC-derived 3D neural models reveal neurodevelopmental pathomechanisms of the Cockayne Syndrome B. Cell Mol Life Sci 2024; 81:368. [PMID: 39179905 PMCID: PMC11343962 DOI: 10.1007/s00018-024-05406-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: 01/08/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
Cockayne Syndrome B (CSB) is a hereditary multiorgan syndrome which-through largely unknown mechanisms-can affect the brain where it clinically presents with microcephaly, intellectual disability and demyelination. Using human induced pluripotent stem cell (hiPSC)-derived neural 3D models generated from CSB patient-derived and isogenic control lines, we here provide explanations for these three major neuropathological phenotypes. In our models, CSB deficiency is associated with (i) impaired cellular migration due to defective autophagy as an explanation for clinical microcephaly; (ii) altered neuronal network functionality and neurotransmitter GABA levels, which is suggestive of a disturbed GABA switch that likely impairs brain circuit formation and ultimately causes intellectual disability; and (iii) impaired oligodendrocyte maturation as a possible cause of the demyelination observed in children with CSB. Of note, the impaired migration and oligodendrocyte maturation could both be partially rescued by pharmacological HDAC inhibition.
Collapse
Affiliation(s)
- Julia Kapr
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Ilka Scharkin
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | | | - Philipp Westhoff
- CEPLAS Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Marius Pollet
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Selina Dangeleit
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | | | - Andrea Rossi
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Katharina Koch
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany.
- DNTOX GmbH, Duesseldorf, Germany.
| | - Jean Krutmann
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
- Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Ellen Fritsche
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
- Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- DNTOX GmbH, Duesseldorf, Germany
- SCAHT, Swiss Centre for Applied Human Toxicology, University of Basel, Basel, Switzerland
| |
Collapse
|
22
|
Campbell BFN, Cruz-Ochoa N, Otomo K, Lukacsovich D, Espinosa P, Abegg A, Luo W, Bellone C, Földy C, Tyagarajan SK. Gephyrin phosphorylation facilitates sexually dimorphic development and function of parvalbumin interneurons in the mouse hippocampus. Mol Psychiatry 2024; 29:2510-2526. [PMID: 38503929 PMCID: PMC11412903 DOI: 10.1038/s41380-024-02517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 03/21/2024]
Abstract
The precise function of specialized GABAergic interneuron subtypes is required to provide appropriate synaptic inhibition for regulating principal neuron excitability and synchronization within brain circuits. Of these, parvalbumin-type (PV neuron) dysfunction is a feature of several sex-biased psychiatric and brain disorders, although, the underlying developmental mechanisms are unclear. While the transcriptional action of sex hormones generates sexual dimorphism during brain development, whether kinase signaling contributes to sex differences in PV neuron function remains unexplored. In the hippocampus, we report that gephyrin, the main inhibitory post-synaptic scaffolding protein, is phosphorylated at serine S268 and S270 in a developmentally-dependent manner in both males and females. When examining GphnS268A/S270A mice in which site-specific phosphorylation is constitutively blocked, we found that sex differences in PV neuron density in the hippocampal CA1 present in WT mice were abolished, coincident with a female-specific increase in PV neuron-derived terminals and increased inhibitory input onto principal cells. Electrophysiological analysis of CA1 PV neurons indicated that gephyrin phosphorylation is required for sexually dimorphic function. Moreover, while male and female WT mice showed no difference in hippocampus-dependent memory tasks, GphnS268A/S270A mice exhibited sex- and task-specific deficits, indicating that gephyrin phosphorylation is differentially required by males and females for convergent cognitive function. In fate mapping experiments, we uncovered that gephyrin phosphorylation at S268 and S270 establishes sex differences in putative PV neuron density during early postnatal development. Furthermore, patch-sequencing of putative PV neurons at postnatal day 4 revealed that gephyrin phosphorylation contributes to sex differences in the transcriptomic profile of developing interneurons. Therefore, these early shifts in male-female interneuron development may drive adult sex differences in PV neuron function and connectivity. Our results identify gephyrin phosphorylation as a new substrate organizing PV neuron development at the anatomical, functional, and transcriptional levels in a sex-dependent manner, thus implicating kinase signaling disruption as a new mechanism contributing to the sex-dependent etiology of brain disorders.
Collapse
Affiliation(s)
- Benjamin F N Campbell
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
| | - Natalia Cruz-Ochoa
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, 8057, Zürich, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, 8057, Zürich, Switzerland
| | - Kanako Otomo
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, 8057, Zürich, Switzerland
| | - Pedro Espinosa
- Department of Basic Neuroscience, University of Geneva, 1211, Geneva, Switzerland
| | - Andrin Abegg
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
| | - Wenshu Luo
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, 8057, Zürich, Switzerland
| | - Camilla Bellone
- Department of Basic Neuroscience, University of Geneva, 1211, Geneva, Switzerland
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, 8057, Zürich, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, 8057, Zürich, Switzerland
| | - Shiva K Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland.
| |
Collapse
|
23
|
Schill DJ, Attili D, DeLong CJ, McInnis MG, Johnson CN, Murphy GG, O’Shea KS. Human-Induced Pluripotent Stem Cell (iPSC)-Derived GABAergic Neuron Differentiation in Bipolar Disorder. Cells 2024; 13:1194. [PMID: 39056776 PMCID: PMC11275104 DOI: 10.3390/cells13141194] [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: 05/23/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Bipolar disorder (BP) is a recurring psychiatric condition characterized by alternating episodes of low energy (depressions) followed by manias (high energy). Cortical network activity produced by GABAergic interneurons may be critical in maintaining the balance in excitatory/inhibitory activity in the brain during development. Initially, GABAergic signaling is excitatory; with maturation, these cells undergo a functional switch that converts GABAA channels from depolarizing (excitatory) to hyperpolarizing (inhibitory), which is controlled by the intracellular concentration of two chloride transporters. The earliest, NKCC1, promotes chloride entry into the cell and depolarization, while the second (KCC2) stimulates movement of chloride from the neuron, hyperpolarizing it. Perturbations in the timing or expression of NKCC1/KCC2 may affect essential morphogenetic events including cell proliferation, migration, synaptogenesis and plasticity, and thereby the structure and function of the cortex. We derived induced pluripotent stem cells (iPSC) from BP patients and undiagnosed control (C) individuals, then modified a differentiation protocol to form GABAergic interneurons, harvesting cells at sequential stages of differentiation. qRT-PCR and RNA sequencing indicated that after six weeks of differentiation, controls transiently expressed high levels of NKCC1. Using multi-electrode array (MEA) analysis, we observed that BP neurons exhibit increased firing, network bursting and decreased synchrony compared to C. Understanding GABA signaling in differentiation may identify novel approaches and new targets for treatment of neuropsychiatric disorders such as BP.
Collapse
Affiliation(s)
- Daniel J. Schill
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA; (D.A.); (C.J.D.); (C.N.J.); (K.S.O.)
| | - Durga Attili
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA; (D.A.); (C.J.D.); (C.N.J.); (K.S.O.)
| | - Cynthia J. DeLong
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA; (D.A.); (C.J.D.); (C.N.J.); (K.S.O.)
| | - Melvin G. McInnis
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI 48109, USA;
| | - Craig N. Johnson
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA; (D.A.); (C.J.D.); (C.N.J.); (K.S.O.)
| | - Geoffrey G. Murphy
- Department of Molecular and Integrative Physiology, The University of Michigan, Ann Arbor, MI 48109, USA;
| | - K. Sue O’Shea
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA; (D.A.); (C.J.D.); (C.N.J.); (K.S.O.)
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
24
|
Johnston GAR, Beart PM. Milestone review: GABA, from chemistry, conformations, ionotropic receptors, modulators, epilepsy, flavonoids, and stress to neuro-nutraceuticals. J Neurochem 2024; 168:1179-1192. [PMID: 38383146 DOI: 10.1111/jnc.16087] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Arising out of a PhD project more than 50 years ago to synthesise analogues of the neurotransmitter GABA, a series of new chemical entities were found to have selective actions on ionotropic GABA receptors. Several of these neurochemicals are now commercially available. A new subtype of these receptors was discovered that could be a target for the treatment of myopia, the facilitation of learning and memory, and the improvement of post-stroke motor recovery. The development of these new chemical entities over many years demonstrates the importance of neurochemicals with which to investigate selective aspects of GABA receptors and illustrates the significance of collaboration between chemists and biologists in neurochemistry. Vital were the improvements in synthetic organic chemistry and the use of functional human receptors expressed in oocytes. Current interest in ionotropic GABA receptors includes the clinical development of subtype-specific agents and the role of gain-of-function receptor variants in epilepsy. Dietary flavonoids were found to cross the blood-brain barrier to influence brain function. Natural and synthetic flavonoids had a range of effects on GABA receptors, ranging from positive, silent, and negative allosteric modulators, to even second-order modulation of first-order modulators. Flavonoids have been called "a new family of benzodiazepines." Like benzodiazepines, flavonoids reduce stress. Stress produces changes in GABA receptors in the brain that may be because of changes in endogenous modulators, such as neurosteroids and corticosteroids. GABA also occurs naturally in the diet leading to studies of the effects of oral GABA on brain function. This finding has resulted in studies of GABA and related neurochemicals as neuro-nutraceuticals. GABA systems in the gut microbiome are essential to such studies. The actions of oral GABA and of GABA-enriched beverages and foodstuffs are now an area of considerable scientific and commercial interest. GABA is a deceptively simple chemical that can take up many shapes, which may underlie its complex functions. The need for new chemical entities with selective actions for further studies highlights the need for continuing collaboration between chemists and biologists.
Collapse
Affiliation(s)
- Graham A R Johnston
- Faculty of Medicine and Health, Pharmacology, Sydney Pharmacy School, The University of Sydney, Sydney, New South Wales, Australia
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
25
|
Ionescu MI, Grigoras IF, Ionescu RB, Chitimus DM, Haret RM, Ianosi B, Ceanga M, Zagrean AM. Oxytocin Exhibits Neuroprotective Effects on Hippocampal Cultures under Severe Oxygen-Glucose Deprivation Conditions. Curr Issues Mol Biol 2024; 46:6223-6236. [PMID: 38921042 PMCID: PMC11202210 DOI: 10.3390/cimb46060371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/12/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
Perinatal asphyxia (PA) and hypoxic-ischemic encephalopathy can result in severe, long-lasting neurological deficits. In vitro models, such as oxygen-glucose deprivation (OGD), are used experimentally to investigate neuronal response to metabolic stress. However, multiple variables can affect the severity level of OGD/PA and may confound any measured treatment effect. Oxytocin (OXT) has emerged as a potential neuroprotective agent against the deleterious effects of PA. Previous studies have demonstrated OXT's potential to enhance neuronal survival in immature hippocampal cultures exposed to OGD, possibly by modulating gamma-aminobutyric acid-A receptor activity. Moreover, OXT's precise impact on developing hippocampal neurons under different severities of OGD/PA remains uncertain. In this study, we investigated the effects of OXT (0.1 µM and 1 µM) on 7-day-old primary rat hippocampal cultures subjected to 2 h OGD/sham normoxic conditions. Cell culture viability was determined using the resazurin assay. Our results indicate that the efficacy of 1 µM OXT treatment varied according to the severity of the OGD-induced lesion, exhibiting a protective effect (p = 0.022) only when cellular viability dropped below 49.41% in non-treated OGD cultures compared to normoxic ones. Furthermore, administration of 0.1 µM OXT did not yield significant effects, irrespective of lesion severity (p > 0.05). These findings suggest that 1 µM OXT treatment during OGD confers neuroprotection exclusively in severe lesions in hippocampal neurons after 7 days in vitro. Further research is warranted to elucidate the mechanisms involved in OXT-mediated neuroprotection.
Collapse
Affiliation(s)
- Mara Ioana Ionescu
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
| | - Ioana-Florentina Grigoras
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Wellcome Centre for Integrative Neuroimaging, Functional MRI of the Brain, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Rosana-Bristena Ionescu
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
- NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Diana Maria Chitimus
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
| | - Robert Mihai Haret
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Department of Ophthalmology, University Medical Center Gottingen, 37075 Gottingen, Germany
| | - Bogdan Ianosi
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Department of Neurology, Stroke Unit, Neuromed Campus, Kepler University Hospital, 4020 Linz, Austria
| | - Mihai Ceanga
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Ana-Maria Zagrean
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
| |
Collapse
|
26
|
Goussakov I, Synowiec S, Fabres RB, Almeida GD, Takada SH, Aksenov D, Drobyshevsky A. Abnormal local cortical functional connectivity due to interneuron dysmaturation after neonatal intermittent hypoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.596449. [PMID: 38895332 PMCID: PMC11185617 DOI: 10.1101/2024.06.04.596449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Premature infants often experience frequent hypoxic episodes due to immaturity of respiratory control that may result in disturbances of gray and white matter development and long-term cognitive and behavioral abnormalities. We hypothesize that neonatal intermittent hypoxia alters cortical maturation of excitatory and inhibitory circuits that can be detected early with functional MRI. Methods C57BL/6 mouse pups were exposed to an intermittent hypoxia (IH) regimen consisting of 12 to 20 daily hypoxic episodes of 5% oxygen exposure for 2 min at 37C from P3 to P7, followed by MRI at P12 and electrophysiological recordings in cortical slices and in vivo at several time points between P7 and P13. Behavioral tests were conducted at P41-P50 to assess animal activity and motor learning. Results Adult mice after neonatal IH exhibited hyperactivity in open field test and impaired motor learning in complex wheel tasks. Patch clamp and evoked field potential electrophysiology revealed increased glutamatergic transmission accompanied by elevation of tonic inhibition. A decreased synaptic inhibitory drive was evidenced by miniature IPSC frequency on pyramidal cells, multi-unit activity recording in vivo in the motor cortex with selective GABA A receptor inhibitor picrotoxin injection, as well as by the decreased interneuron density at P13. There was also an increased tonic depolarizing effect of picrotoxin after IH on principal cells' membrane potential on patch clamp and direct current potential in extracellular recordings. The amplitude of low-frequency fluctuation on resting-state fMRI was larger, with a larger increase after picrotoxin injection in the IH group. Conclusions Increased excitatory glutamatergic transmission, decreased numbers, and activity of inhibitory interneurons after neonatal IH may affect the maturation of connectivity in cortical networks, resulting in long-term cognitive and behavioral changes, including impaired motor learning and hyperactivity. Functional MRI reveals increased intrinsic connectivity in the sensorimotor cortex, suggesting neuronal dysfunction in cortical maturation after neonatal IH. The increased tonic inhibition, presumably due to tonic extrasynaptic GABA receptor drive, may be compensatory to the elevated excitatory glutamatergic transmission.
Collapse
|
27
|
Zavalin K, Hassan A, Zhang Y, Khera Z, Lagrange AH. Region and layer-specific expression of GABA A receptor isoforms and KCC2 in developing cortex. Front Cell Neurosci 2024; 18:1390742. [PMID: 38894703 PMCID: PMC11184147 DOI: 10.3389/fncel.2024.1390742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction γ-Aminobutyric acid (GABA) type A receptors (GABAARs) are ligand-gated Cl-channels that mediate the bulk of inhibitory neurotransmission in the mature CNS and are targets of many drugs. During cortical development, GABAAR-mediated signals are significantly modulated by changing subunit composition and expression of Cl-transporters as part of developmental processes and early network activity. To date, this developmental evolution has remained understudied, particularly at the level of cortical layer-specific changes. In this study, we characterized the expression of nine major GABAAR subunits and K-Cl transporter 2 (KCC2) in mouse somatosensory cortex from embryonic development to postweaning maturity. Methods We evaluated expression of α1-5, β2-3, γ2, and δ GABAAR subunits using immunohistochemistry and Western blot techniques, and expression of KCC2 using immunohistochemistry in cortices from E13.5 to P25 mice. Results We found that embryonic cortex expresses mainly α3, α5, β3, and γ2, while expression of α1, α2, α4, β2, δ, and KCC2 begins at later points in development; however, many patterns of nuanced expression can be found in specific lamina, cortical regions, and cells and structures. Discussion While the general pattern of expression of each subunit and KCC2 is similar to previous studies, we found a number of unique temporal, regional, and laminar patterns that were previously unknown. These findings provide much needed knowledge of the intricate developmental evolution in GABAAR composition and KCC2 expression to accommodate developmental signals that transition to mature neurotransmission.
Collapse
Affiliation(s)
- Kirill Zavalin
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Anjana Hassan
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Yueli Zhang
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Zain Khera
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Andre H. Lagrange
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Neurology, TVH VA Medical Center, Nashville, TN, United States
| |
Collapse
|
28
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [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: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Ng ACH, Chahine M, Scantlebury MH, Appendino JP. Channelopathies in epilepsy: an overview of clinical presentations, pathogenic mechanisms, and therapeutic insights. J Neurol 2024; 271:3063-3094. [PMID: 38607431 DOI: 10.1007/s00415-024-12352-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: 03/17/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Pathogenic variants in genes encoding ion channels are causal for various pediatric and adult neurological conditions. In particular, several epilepsy syndromes have been identified to be caused by specific channelopathies. These encompass a spectrum from self-limited epilepsies to developmental and epileptic encephalopathies spanning genetic and acquired causes. Several of these channelopathies have exquisite responses to specific antiseizure medications (ASMs), while others ASMs may prove ineffective or even worsen seizures. Some channelopathies demonstrate phenotypic pleiotropy and can cause other neurological conditions outside of epilepsy. This review aims to provide a comprehensive exploration of the pathophysiology of seizure generation, ion channels implicated in epilepsy, and several genetic epilepsies due to ion channel dysfunction. We outline the clinical presentation, pathogenesis, and the current state of basic science and clinical research for these channelopathies. In addition, we briefly look at potential precision therapy approaches emerging for these disorders.
Collapse
Affiliation(s)
- Andy Cheuk-Him Ng
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Division of Neurology, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta and Stollery Children's Hospital, Edmonton, AB, Canada
| | - Mohamed Chahine
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- CERVO, Brain Research Centre, Quebec City, Canada
| | - Morris H Scantlebury
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Canada
| | - Juan P Appendino
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada.
| |
Collapse
|
30
|
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
|
31
|
Hamad MIK, Rabaya O, Jbara A, Daoud S, Petrova P, Ali BR, Allouh MZ, Herz J, Förster E. Reelin Regulates Developmental Desynchronization Transition of Neocortical Network Activity. Biomolecules 2024; 14:593. [PMID: 38786001 PMCID: PMC11118507 DOI: 10.3390/biom14050593] [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: 03/12/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
During the first and second stages of postnatal development, neocortical neurons exhibit a wide range of spontaneous synchronous activity (SSA). Towards the end of the second postnatal week, the SSA is replaced by a more sparse and desynchronized firing pattern. The developmental desynchronization of neocortical spontaneous neuronal activity is thought to be intrinsically generated, since sensory deprivation from the periphery does not affect the time course of this transition. The extracellular protein reelin controls various aspects of neuronal development through multimodular signaling. However, so far it is unclear whether reelin contributes to the developmental desynchronization transition of neocortical neurons. The present study aims to investigate the role of reelin in postnatal cortical developmental desynchronization using a conditional reelin knockout (RelncKO) mouse model. Conditional reelin deficiency was induced during early postnatal development, and Ca2+ recordings were conducted from organotypic cultures (OTCs) of the somatosensory cortex. Our results show that both wild type (wt) and RelncKO exhibited an SSA pattern during the early postnatal week. However, at the end of the second postnatal week, wt OTCs underwent a transition to a desynchronized network activity pattern, while RelncKO activity remained synchronous. This changing activity pattern suggests that reelin is involved in regulating the developmental desynchronization of cortical neuronal network activity. Moreover, the developmental desynchronization impairment observed in RelncKO was rescued when RelncKO OTCs were co-cultured with wt OTCs. Finally, we show that the developmental transition to a desynchronized state at the end of the second postnatal week is not dependent on glutamatergic signaling. Instead, the transition is dependent on GABAAR and GABABR signaling. The results suggest that reelin controls developmental desynchronization through GABAAR and GABABR signaling.
Collapse
Affiliation(s)
- Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Obada Rabaya
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Abdalrahim Jbara
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Solieman Daoud
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Petya Petrova
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Mohammed Z. Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 5323, USA
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| |
Collapse
|
32
|
Sakaguchi K, Tawata S. Giftedness and atypical sexual differentiation: enhanced perceptual functioning through estrogen deficiency instead of androgen excess. Front Endocrinol (Lausanne) 2024; 15:1343759. [PMID: 38752176 PMCID: PMC11094242 DOI: 10.3389/fendo.2024.1343759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Syndromic autism spectrum conditions (ASC), such as Klinefelter syndrome, also manifest hypogonadism. Compared to the popular Extreme Male Brain theory, the Enhanced Perceptual Functioning model explains the connection between ASC, savant traits, and giftedness more seamlessly, and their co-emergence with atypical sexual differentiation. Overexcitability of primary sensory inputs generates a relative enhancement of local to global processing of stimuli, hindering the abstraction of communication signals, in contrast to the extraordinary local information processing skills in some individuals. Weaker inhibitory function through gamma-aminobutyric acid type A (GABAA) receptors and the atypicality of synapse formation lead to this difference, and the formation of unique neural circuits that process external information. Additionally, deficiency in monitoring inner sensory information leads to alexithymia (inability to distinguish one's own emotions), which can be caused by hypoactivity of estrogen and oxytocin in the interoceptive neural circuits, comprising the anterior insular and cingulate gyri. These areas are also part of the Salience Network, which switches between the Central Executive Network for external tasks and the Default Mode Network for self-referential mind wandering. Exploring the possibility that estrogen deficiency since early development interrupts GABA shift, causing sensory processing atypicality, it helps to evaluate the co-occurrence of ASC with attention deficit hyperactivity disorder, dyslexia, and schizophrenia based on phenotypic and physiological bases. It also provides clues for understanding the common underpinnings of these neurodevelopmental disorders and gifted populations.
Collapse
Affiliation(s)
- Kikue Sakaguchi
- Research Department, National Institution for Academic Degrees and Quality Enhancement of Higher Education (NIAD-QE), Kodaira-shi, Tokyo, Japan
| | - Shintaro Tawata
- Graduate School of Human Sciences, Sophia University, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
33
|
Nascimento AA, Pereira-Figueiredo D, Borges-Martins VP, Kubrusly RC, Calaza KC. GABAergic system and chloride cotransporters as potential therapeutic targets to mitigate cell death in ischemia. J Neurosci Res 2024; 102:e25355. [PMID: 38808645 DOI: 10.1002/jnr.25355] [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: 08/25/2023] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Gamma aminobutyric acid (GABA) is a critical inhibitory neurotransmitter in the central nervous system that plays a vital role in modulating neuronal excitability. Dysregulation of GABAergic signaling, particularly involving the cotransporters NKCC1 and KCC2, has been implicated in various pathologies, including epilepsy, schizophrenia, autism spectrum disorder, Down syndrome, and ischemia. NKCC1 facilitates chloride influx, whereas KCC2 mediates chloride efflux via potassium gradient. Altered expression and function of these cotransporters have been associated with excitotoxicity, inflammation, and cellular death in ischemic events characterized by reduced cerebral blood flow, leading to compromised tissue metabolism and subsequent cell death. NKCC1 inhibition has emerged as a potential therapeutic approach to attenuate intracellular chloride accumulation and mitigate neuronal damage during ischemic events. Similarly, targeting KCC2, which regulates chloride efflux, holds promise for improving outcomes and reducing neuronal damage under ischemic conditions. This review emphasizes the critical roles of GABA, NKCC1, and KCC2 in ischemic pathologies and their potential as therapeutic targets. Inhibiting or modulating the activity of these cotransporters represents a promising strategy for reducing neuronal damage, preventing excitotoxicity, and improving neurological outcomes following ischemic events. Furthermore, exploring the interactions between natural compounds and NKCC1/KCC2 provides additional avenues for potential therapeutic interventions for ischemic injury.
Collapse
Affiliation(s)
- A A Nascimento
- Neurobiology of the Retina Laboratory, Department of Neurobiology and Graduate Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - D Pereira-Figueiredo
- Graduate Program in Biomedical Sciences (Physiology and Pharmacology), Fluminense Federal University, Niterói, Brazil
| | - V P Borges-Martins
- Laboratory of Neuropharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - R C Kubrusly
- Laboratory of Neuropharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - K C Calaza
- Neurobiology of the Retina Laboratory, Department of Neurobiology and Graduate Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
- Graduate Program in Biomedical Sciences (Physiology and Pharmacology), Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
34
|
Xia M, Xia Y, Sun Y, Wang J, Lu J, Wang X, Xia D, Xu X, Sun B. Gut microbiome is associated with personality traits of free-ranging Tibetan macaques ( Macaca thibetana). Front Microbiol 2024; 15:1381372. [PMID: 38711972 PMCID: PMC11070476 DOI: 10.3389/fmicb.2024.1381372] [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: 02/03/2024] [Accepted: 04/03/2024] [Indexed: 05/08/2024] Open
Abstract
Recent studies have emphasized that there is a strong link between the gut microbiome and the brain that affects social behavior and personality in animals. However, the interface between personality and the gut microbiome in wild primates remains poorly understood. Here, we used high-throughput sequencing and ethological methods in primate behavioral ecology to investigate the relationship between gut microbiome and personality in Tibetan macaques (Macaca thibetana). The behavioral assessment results indicated three personality dimensions including socialization, shyness, and anxiety. There was significant variation in alpha diversity only for shyness, with a significantly lower alpha diversity indices (including Shannon, Chao1, and PD) for bold individuals than for shy individuals. Using regression models to control for possible confounding factors, we found that the relative abundance of three genera, Akkermansia, Dialister, and Asteroleplasma, was significantly and positively correlated with the sociability scores in the macaques. In addition, Oscillospiraceae exhibited a positive correlation with scores for Shy Dimension. Furthermore, we found that the predicted functional genes for propionate and pyruvate, porphyrin and chlorophyll metabolic pathways related to animal behavior, were significant enriched in shyness group. We propose that the gut microbiome may play an important role in the formation of personality of Tibetan macaques.
Collapse
Affiliation(s)
- Mengyi Xia
- School of Resources and Environmental Engineering, Anhui University, Hefei, China
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
| | - Yingna Xia
- School of Resources and Environmental Engineering, Anhui University, Hefei, China
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
| | - Yu Sun
- School of Resources and Environmental Engineering, Anhui University, Hefei, China
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
| | - Jingjing Wang
- School of Resources and Environmental Engineering, Anhui University, Hefei, China
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
| | - Jiakai Lu
- School of Resources and Environmental Engineering, Anhui University, Hefei, China
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
| | - Xi Wang
- School of Resources and Environmental Engineering, Anhui University, Hefei, China
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
| | - Dongpo Xia
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
- School of Life Sciences, Anhui University, Hefei, China
| | - Xiaojuan Xu
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
- School of Biology and Food Engineering, Hefei Normal University, Hefei, China
| | - Binghua Sun
- School of Resources and Environmental Engineering, Anhui University, Hefei, China
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral Ecology, Anhui University, Hefei, China
| |
Collapse
|
35
|
Neuwirth LS, Emenike BU. Comment on "Neurotoxicity and Outcomes from Developmental Lead Exposure: Persistent or Permanent?". ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:48001. [PMID: 38607984 PMCID: PMC11014073 DOI: 10.1289/ehp14809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024]
Affiliation(s)
- Lorenz S. Neuwirth
- Department of Psychology, State University of New York at Old Westbury (SUNY Old Westbury), Old Westbury, New York, USA
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, New York, USA
| | - Bright U. Emenike
- Department of Chemistry and Physics, SUNY Old Westbury, Old Westbury, New York, USA
| |
Collapse
|
36
|
Zinchenko VP, Dolgacheva LP, Tuleukhanov ST. Calcium-permeable AMPA and kainate receptors of GABAergic neurons. Biophys Rev 2024; 16:165-171. [PMID: 38737208 PMCID: PMC11078900 DOI: 10.1007/s12551-024-01184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/16/2024] [Indexed: 05/14/2024] Open
Abstract
This Commentary presents a brief discussion of the action of glutamate calcium permeable receptors present with neurons on the release of the neurotransmitter gamma-aminobutyric acid (GABA). In particular, Glutamate sensitive Kainic Acid Receptors (KARs) and α-Amino-3-hydroxy-5-Methyl-4-isoxazole Propionic Acid Receptor (AMPARs) are Na+ channels that typically cause neuronal cells to depolarize and release GABA. Some of these receptors are also permeable to Ca2+ and are hence involved in the calcium-dependent release of GABA neurotransmitters. Calcium-permeable kainate and AMPA receptors (CP-KARs and CP-AMPARs) are predominantly located in GABAergic neurons in the mature brain and their primary role is to regulate GABA release. AMPARs which do not contain the GluA2 subunit are mainly localized in the postsynaptic membrane. CP-KAR receptors are located mainly in the presynapse. GABAergic neurons expressing CP-KARs and CP-AMPARs respond to excitation earlier and faster, suppressing hyperexcitation of other neurons by the advanced GABA release due to an early rapid [Ca2+]i increase. CP-AMPARs have demonstrated a more pronounced impact on plasticity compared to NMDARs because of their capacity to elevate intracellular Ca2+ levels independently of voltage. GABAergic neurons that express CP-AMPARs contribute to the disinhibition of glutamatergic neurons by suppressing GABAergic neurons that express CP-KARs. Hence, the presence of glutamate CP-KARs and CP-AMPARs is crucial in governing hyperexcitation and synaptic plasticity in GABAergic neurons.
Collapse
Affiliation(s)
- V. P. Zinchenko
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Institutskaya 3, Pushchino, Russia 142290
| | - L. P. Dolgacheva
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Institutskaya 3, Pushchino, Russia 142290
| | - S. T. Tuleukhanov
- Al-Farabi Kazakh National University, 050040 Al-Farabi Avenue 71, Almaty, Republic of Kazakhstan
| |
Collapse
|
37
|
Snyder HE, Jain P, RamachandranNair R, Jones KC, Whitney R. Genetic Advancements in Infantile Epileptic Spasms Syndrome and Opportunities for Precision Medicine. Genes (Basel) 2024; 15:266. [PMID: 38540325 PMCID: PMC10970414 DOI: 10.3390/genes15030266] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 06/15/2024] Open
Abstract
Infantile epileptic spasms syndrome (IESS) is a devastating developmental epileptic encephalopathy (DEE) consisting of epileptic spasms, as well as one or both of developmental regression or stagnation and hypsarrhythmia on EEG. A myriad of aetiologies are associated with the development of IESS; broadly, 60% of cases are thought to be structural, metabolic or infectious in nature, with the remainder genetic or of unknown cause. Epilepsy genetics is a growing field, and over 28 copy number variants and 70 single gene pathogenic variants related to IESS have been discovered to date. While not exhaustive, some of the most commonly reported genetic aetiologies include trisomy 21 and pathogenic variants in genes such as TSC1, TSC2, CDKL5, ARX, KCNQ2, STXBP1 and SCN2A. Understanding the genetic mechanisms of IESS may provide the opportunity to better discern IESS pathophysiology and improve treatments for this condition. This narrative review presents an overview of our current understanding of IESS genetics, with an emphasis on animal models of IESS pathogenesis, the spectrum of genetic aetiologies of IESS (i.e., chromosomal disorders, single-gene disorders, trinucleotide repeat disorders and mitochondrial disorders), as well as available genetic testing methods and their respective diagnostic yields. Future opportunities as they relate to precision medicine and epilepsy genetics in the treatment of IESS are also explored.
Collapse
Affiliation(s)
- Hannah E. Snyder
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Puneet Jain
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1E8, Canada
| | - Rajesh RamachandranNair
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Kevin C. Jones
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| | - Robyn Whitney
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada (R.R.)
| |
Collapse
|
38
|
Napoli AJ, Laderwager S, Zoodsma JD, Biju B, Mucollari O, Schubel SK, Aprea C, Sayed A, Morgan K, Napoli A, Flanagan S, Wollmuth LP, Sirotkin HI. Developmental loss of NMDA receptors results in supernumerary forebrain neurons through delayed maturation of transit-amplifying neuroblasts. Sci Rep 2024; 14:3395. [PMID: 38336823 PMCID: PMC10858180 DOI: 10.1038/s41598-024-53910-7] [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: 10/06/2023] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
Developmental neurogenesis is a tightly regulated spatiotemporal process with its dysregulation implicated in neurodevelopmental disorders. NMDA receptors are glutamate-gated ion channels that are widely expressed in the early nervous system, yet their contribution to neurogenesis is poorly understood. Notably, a variety of mutations in genes encoding NMDA receptor subunits are associated with neurodevelopmental disorders. To rigorously define the role of NMDA receptors in developmental neurogenesis, we used a mutant zebrafish line (grin1-/-) that lacks all NMDA receptors yet survives to 10 days post-fertilization, offering the opportunity to study post-embryonic neurodevelopment in the absence of NMDA receptors. Focusing on the forebrain, we find that these fish have a progressive supernumerary neuron phenotype confined to the telencephalon at the end of embryonic neurogenesis, but which extends to all forebrain regions during postembryonic neurogenesis. This enhanced neuron population does not arise directly from increased numbers or mitotic activity of radial glia cells, the principal neural stem cells. Rather, it stems from a lack of timely maturation of transit-amplifying neuroblasts into post-mitotic neurons, as indicated by a decrease in expression of the ontogenetically-expressed chloride transporter, KCC2. Pharmacological blockade with MK-801 recapitulates the grin1-/- supernumerary neuron phenotype, indicating a requirement for ionotropic signaling. Thus, NMDA receptors are required for suppression of indirect, transit amplifying cell-driven neurogenesis by promoting maturational termination of mitosis. Loss of suppression results in neuronal overpopulation that can fundamentally change brain circuitry and may be a key factor in pathogenesis of neurodevelopmental disorders caused by NMDA receptor dysfunction.
Collapse
Affiliation(s)
- Amalia J Napoli
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Stephanie Laderwager
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Josiah D Zoodsma
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Bismi Biju
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Olgerta Mucollari
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Sarah K Schubel
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Christieann Aprea
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Aaliya Sayed
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Kiele Morgan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Annelysia Napoli
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Stephanie Flanagan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Lonnie P Wollmuth
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5215, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Howard I Sirotkin
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
| |
Collapse
|
39
|
Kim HR, Martina M. Bidirectional Regulation of GABA A Reversal Potential in the Adult Brain: Physiological and Pathological Implications. Life (Basel) 2024; 14:143. [PMID: 38276272 PMCID: PMC10817304 DOI: 10.3390/life14010143] [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: 12/08/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
In physiological conditions, the intracellular chloride concentration is much lower than the extracellular. As GABAA channels are permeable to anions, the reversal potential of GABAA is very close to that of Cl-, which is the most abundant free anion in the intra- and extracellular spaces. Intracellular chloride is regulated by the activity ratio of NKCC1 and KCC2, two chloride-cation cotransporters that import and export Cl-, respectively. Due to the closeness between GABAA reversal potential and the value of the resting membrane potential in most neurons, small changes in intracellular chloride have a major functional impact, which makes GABAA a uniquely flexible signaling system. In most neurons of the adult brain, the GABAA reversal potential is slightly more negative than the resting membrane potential, which makes GABAA hyperpolarizing. Alterations in GABAA reversal potential are a common feature in numerous conditions as they are the consequence of an imbalance in the NKCC1-KCC2 activity ratio. In most conditions (including Alzheimer's disease, schizophrenia, and Down's syndrome), GABAA becomes depolarizing, which causes network desynchronization and behavioral impairment. In other conditions (neonatal inflammation and neuropathic pain), however, GABAA reversal potential becomes hypernegative, which affects behavior through a potent circuit deactivation.
Collapse
Affiliation(s)
- Haram R. Kim
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, 300 E. Superior, Chicago, IL 60611, USA;
| | - Marco Martina
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, 300 E. Superior, Chicago, IL 60611, USA;
- Department of Psychiatry, Feinberg School of Medicine, Northwestern University, 300 E. Superior, Chicago, IL 60611, USA
| |
Collapse
|
40
|
Xue J, Brawner AT, Thompson JR, Yelhekar TD, Newmaster KT, Qiu Q, Cooper YA, Yu CR, Ahmed-Braima YH, Kim Y, Lin Y. Spatiotemporal Mapping and Molecular Basis of Whole-brain Circuit Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.572456. [PMID: 38260331 PMCID: PMC10802351 DOI: 10.1101/2024.01.03.572456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Brain development is highly dynamic and asynchronous, marked by the sequential maturation of functional circuits across the brain. The timing and mechanisms driving circuit maturation remain elusive due to an inability to identify and map maturing neuronal populations. Here we create DevATLAS (Developmental Activation Timing-based Longitudinal Acquisition System) to overcome this obstacle. We develop whole-brain mapping methods to construct the first longitudinal, spatiotemporal map of circuit maturation in early postnatal mouse brains. Moreover, we uncover dramatic impairments within the deep cortical layers in a neurodevelopmental disorders (NDDs) model, demonstrating the utility of this resource to pinpoint when and where circuit maturation is disrupted. Using DevATLAS, we reveal that early experiences accelerate the development of hippocampus-dependent learning by increasing the synaptically mature granule cell population in the dentate gyrus. Finally, DevATLAS enables the discovery of molecular mechanisms driving activity-dependent circuit maturation.
Collapse
Affiliation(s)
- Jian Xue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew T. Brawner
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Equal contribution
| | - Jacqueline R. Thompson
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Equal contribution
| | - Tushar D. Yelhekar
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kyra T. Newmaster
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Qiang Qiu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, MO 66160, USA
| | - Yonatan A. Cooper
- Current address: Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, MO 66160, USA
| | | | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Yingxi Lin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Lead contact
| |
Collapse
|
41
|
Zhang M, Chen Y, Xu T, Jiang J, Zhang D, Huang H, Kurth CD, Yuan I, Wang R, Liu J, Zhu T, Zhou C. γ-Aminobutyric Acid-Ergic Development Contributes to the Enhancement of Electroencephalogram Slow-Delta Oscillations Under Volatile Anesthesia in Neonatal Rats. Anesth Analg 2024; 138:198-209. [PMID: 36753442 DOI: 10.1213/ane.0000000000006396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
BACKGROUND General anesthetics (eg, propofol and volatile anesthetics) enhance the slow-delta oscillations of the cortical electroencephalogram (EEG), which partly results from the enhancement of (γ-aminobutyric acid [GABA]) γ-aminobutyric acid-ergic (GABAergic) transmission. There is a GABAergic excitatory-inhibitory shift during postnatal development. Whether general anesthetics can enhance slow-delta oscillations in the immature brain has not yet been unequivocally determined. METHODS Perforated patch-clamp recording was used to confirm the reversal potential of GABAergic currents throughout GABAergic development in acute brain slices of neonatal rats. The power density of the electrocorticogram and the minimum alveolar concentrations (MAC) of isoflurane and/or sevoflurane were measured in P4-P21 rats. Then, the effects of bumetanide, an inhibitor of the Na + -K + -2Cl - cotransporter (NKCC1) and K + -Cl - cotransporter (KCC2) knockdown on the potency of volatile anesthetics and the power density of the EEG were determined in vivo. RESULTS Reversal potential of GABAergic currents were gradually hyperpolarized from P4 to P21 in cortical pyramidal neurons. Bumetanide enhanced the hypnotic effects of volatile anesthetics at P5 (for MAC LORR , isoflurane: 0.63% ± 0.07% vs 0.81% ± 0.05%, 95% confidence interval [CI], -0.257 to -0.103, P < .001; sevoflurane: 1.46% ± 0.12% vs 1.66% ± 0.09%, 95% CI, -0.319 to -0.081, P < .001); while knockdown of KCC2 weakened their hypnotic effects at P21 in rats (for MAC LORR , isoflurane: 0.58% ± 0.05% to 0.77% ± 0.20%, 95% CI, 0.013-0.357, P = .003; sevoflurane: 1.17% ± 0.04% to 1.33% ± 0.04%, 95% CI, 0.078-0.244, P < .001). For cortical EEG, slow-delta oscillations were the predominant components of the EEG spectrum in neonatal rats. Isoflurane and/or sevoflurane suppressed the power density of slow-delta oscillations rather than enhancement of it until GABAergic maturity. Enhancement of slow-delta oscillations under volatile anesthetics was simulated by preinjection of bumetanide at P5 (isoflurane: slow-delta changed ratio from -0.31 ± 0.22 to 1.57 ± 1.15, 95% CI, 0.67-3.08, P = .007; sevoflurane: slow-delta changed ratio from -0.46 ± 0.25 to 0.95 ± 0.97, 95% CI, 0.38-2.45, P = .014); and suppressed by KCC2-siRNA at P21 (isoflurane: slow-delta changed ratio from 16.13 ± 5.69 to 3.98 ± 2.35, 95% CI, -18.50 to -5.80, P = .002; sevoflurane: slow-delta changed ratio from 0.13 ± 2.82 to 3.23 ± 2.49, 95% CI, 3.02-10.79, P = .003). CONCLUSIONS Enhancement of cortical EEG slow-delta oscillations by volatile anesthetics may require mature GABAergic inhibitory transmission during neonatal development.
Collapse
Affiliation(s)
- Mingyue Zhang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yali Chen
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Ting Xu
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingyao Jiang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Donghang Zhang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, West China Second Hospital of Sichuan University, Chengdu, China
| | - Charles D Kurth
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ian Yuan
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Rurong Wang
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jin Liu
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Zhu
- From the Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
42
|
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
|
43
|
Pitzer EM, Shafer TJ, Herr DW. Identification of neurotoxicology (NT)/developmental neurotoxicology (DNT) adverse outcome pathways and key event linkages with in vitro DNT screening assays. Neurotoxicology 2023; 99:184-194. [PMID: 37866692 PMCID: PMC12117389 DOI: 10.1016/j.neuro.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/14/2023] [Accepted: 10/13/2023] [Indexed: 10/24/2023]
Abstract
There is a need to assess compounds reliably and quickly for neurotoxicity (NT) and developmental neurotoxicity (DNT). Adverse outcome pathways (AOPs) enable the mapping of molecular events to an apical endpoint in a chemical agnostic manner and have begun to be applied in NT and DNT testing frameworks. We assessed the status of NT/DNT AOPs in the AOP-Wiki (ca. 2/1/23; https://aopwiki.org/), to characterize the state of AOP development, identify strengths and knowledge gaps, elucidate areas for improvement, and describe areas for future focus. AOPs in the Wiki database were assessed for inclusion of NT/DNT molecular events and endpoints, AOP development and endorsement, as well as the linkages of key neurodevelopmental processes with in vitro new approach methods (NAMs). This review found that 41 AOPs have been proposed detailing NT/DNT, of which eight were endorsed by working parties in OECD. Further, this review determined that learning and memory is included as an adverse outcome in eight NT/DNT AOPS, often without distinction regarding the varying forms of learning and memory, regional specification, temporal dynamics, or acquisition mechanisms involved. There is also an overlap with key events (KEs) and in vitro NAMs, which synaptogenesis appeared as a common process. Overall, progress on NT/DNT AOPs could be expanded, adding in modes of action that are missing, improvement in defining apical endpoints, as well as utilizing NAMs further to develop AOPs and identify gaps in current knowledge.
Collapse
Affiliation(s)
- Emily M Pitzer
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - David W Herr
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| |
Collapse
|
44
|
Yasir M, Park J, Lee Y, Han ET, Park WS, Han JH, Kwon YS, Lee HJ, Chun W. Discovery of GABA Aminotransferase Inhibitors via Molecular Docking, Molecular Dynamic Simulation, and Biological Evaluation. Int J Mol Sci 2023; 24:16990. [PMID: 38069313 PMCID: PMC10707509 DOI: 10.3390/ijms242316990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
γ-Aminobutyric acid aminotransferase (GABA-AT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that degrades γ-aminobutyric (GABA) in the brain. GABA is an important inhibitory neurotransmitter that plays important neurological roles in the brain. Therefore, GABA-AT is an important drug target that regulates GABA levels. Novel and potent drug development to inhibit GABA-AT is still a very challenging task. In this study, we aimed to devise novel and potent inhibitors against GABA-AT using computer-aided drug design (CADD) tools. Since the crystal structure of human GABA-AT was not yet available, we utilized a homologous structure derived from our previously published paper. To identify highly potent compounds relative to vigabatrin, an FDA-approved drug against human GABA-AT, we developed a pharmacophore analysis protocol for 530,000 Korea Chemical Bank (KCB) compounds and selected the top 50 compounds for further screening. Preliminary biological analysis was carried out for these 50 compounds and 16 compounds were further assessed. Subsequently, molecular docking, molecular dynamics (MD) simulations, and binding free energy calculations were carried out. In the results, four predicted compounds, A07, B07, D08, and H08, were found to be highly potent and were further evaluated by a biological activity assay to confirm the results of the GABA-AT activity inhibition assay.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| | - Yuno Lee
- Drug Information Platform Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea;
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (E.-T.H.); (J.-H.H.)
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea;
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (E.-T.H.); (J.-H.H.)
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Hee-Jae Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| |
Collapse
|
45
|
Boerma T, Ter Haar S, Ganga R, Wijnen F, Blom E, Wierenga CJ. What risk factors for Developmental Language Disorder can tell us about the neurobiological mechanisms of language development. Neurosci Biobehav Rev 2023; 154:105398. [PMID: 37741516 DOI: 10.1016/j.neubiorev.2023.105398] [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: 04/21/2023] [Revised: 07/03/2023] [Accepted: 09/17/2023] [Indexed: 09/25/2023]
Abstract
Language is a complex multidimensional cognitive system that is connected to many neurocognitive capacities. The development of language is therefore strongly intertwined with the development of these capacities and their neurobiological substrates. Consequently, language problems, for example those of children with Developmental Language Disorder (DLD), are explained by a variety of etiological pathways and each of these pathways will be associated with specific risk factors. In this review, we attempt to link previously described factors that may interfere with language development to putative underlying neurobiological mechanisms of language development, hoping to uncover openings for future therapeutical approaches or interventions that can help children to optimally develop their language skills.
Collapse
Affiliation(s)
- Tessel Boerma
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Sita Ter Haar
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands; Cognitive Neurobiology and Helmholtz Institute, Department of Psychology, Utrecht University/Translational Neuroscience, University Medical Center Utrecht, the Netherlands
| | - Rachida Ganga
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Frank Wijnen
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Elma Blom
- Department of Development and Education of youth in Diverse Societies (DEEDS), Utrecht University, Utrecht, the Netherlands; Department of Language and Culture, The Arctic University of Norway UiT, Tromsø, Norway.
| | - Corette J Wierenga
- Biology Department, Faculty of Science, Utrecht University, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
46
|
López-Merino E, Cuartero MI, Esteban JA, Briz V. Perinatal exposure to pesticides alters synaptic plasticity signaling and induces behavioral deficits associated with neurodevelopmental disorders. Cell Biol Toxicol 2023; 39:2089-2111. [PMID: 35137321 PMCID: PMC10547633 DOI: 10.1007/s10565-022-09697-2] [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: 09/23/2021] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
Increasing evidence from animal and epidemiological studies indicates that perinatal exposure to pesticides cause developmental neurotoxicity and may increase the risk for psychiatric disorders such as autism and intellectual disability. However, the underlying pathogenic mechanisms remain largely elusive. This work was aimed at testing the hypothesis that developmental exposure to different classes of pesticides hijacks intracellular neuronal signaling contributing to synaptic and behavioral alterations associated with neurodevelopmental disorders (NDD). Low concentrations of organochlorine (dieldrin, endosulfan, and chlordane) and organophosphate (chlorpyrifos and its oxon metabolite) pesticides were chronically dosed ex vivo (organotypic rat hippocampal slices) or in vivo (perinatal exposure in rats), and then biochemical, electrophysiological, behavioral, and proteomic studies were performed. All the pesticides tested caused prolonged activation of MAPK/ERK pathway in a concentration-dependent manner. Additionally, some of them impaired metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD). In the case of the pesticide chlordane, the effect was attributed to chronic modulation of MAPK/ERK signaling. These synaptic alterations were reproduced following developmental in vivo exposure to chlordane and chlorpyrifos-oxon, and were also associated with prototypical behavioral phenotypes of NDD, including impaired motor development, increased anxiety, and social and memory deficits. Lastly, proteomic analysis revealed that these pesticides differentially regulate the expression of proteins in the hippocampus with pivotal roles in brain development and synaptic signaling, some of which are associated with NDD. Based on these results, we propose a novel mechanism of synaptic dysfunction, involving chronic overactivation of MAPK and impaired mGluR-LTD, shared by different pesticides which may have important implications for NDD.
Collapse
Affiliation(s)
| | - María I Cuartero
- Neurovascular Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José A Esteban
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
| | - Víctor Briz
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
47
|
Rao S, Farhat A, Rakshasbhuvankar A, Athikarisamy S, Ghosh S, Nagarajan L. Effects of bumetanide on neonatal seizures: A systematic review of animal and human studies. Seizure 2023; 111:206-214. [PMID: 37690372 DOI: 10.1016/j.seizure.2023.09.007] [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: 06/22/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Bumetanide, an inhibitor of the sodium-potassium-chloride cotransporter-1, has been suggested as an adjunct to phenobarbital for treating neonatal seizures. METHODS A systematic review of animal and human studies was conducted to evaluate the efficacy and safety of bumetanide for neonatal seizures. PubMed, Embase, CINAHL and Cochrane databases were searched in March 2023. RESULTS 26 animal (rat or mice) studies describing 38 experiments (28 in-vivo and ten in-vitro) and two human studies (one RCT and one open-label dose-finding) were included. The study designs, methods to induce seizures, bumetanide dose, and outcome measures were heterogeneous, with only 4/38 experiments being in animal hypoxia/ischaemia models. Among 38 animal experiments, bumetanide was reported to have antiseizure effects in 21, pro-seizure in six and ineffective in 11. The two human studies (n = 57) did not show the benefits of bumetanide as an add-on agent to phenobarbital in their primary analyses, but one study reported benefit on post-hoc analysis. Overall, hearing impairment was detected in 5/37 surviving infants in the bumetanide group vs. 0/13 in controls. Four of the five infants with hearing impairment had received aminoglycosides concurrently. Other adverse effects reported were diuresis, mild-to-moderate dehydration, hypotension, and electrolyte disturbances. The studies did not report on long-term neurodevelopment. The certainty of the evidence was very low. CONCLUSION Animal data suggest that bumetanide has inconsistent effects as an antiseizure medication in neonates. Data from human studies are scarce and raise some concerns regarding ototoxicity when given with aminoglycosides. Well conducted studies in animal models of hypoxic-ischaemic encephalopathy are urgently needed. Future RCTs, if conducted in human neonates, should have an adequate sample size, assess neurodevelopment, minimize using aminoglycosides, be transparent about the potential ototoxicity in the parent information sheet, conduct early hearing tests and have trial-stopping rules that include hearing impairment as an outcome.
Collapse
Affiliation(s)
- Shripada Rao
- Neonatal Intensive Care Unit, King Edward Memorial and Perth Children's Hospitals, Perth, Australia; Paediatric Division, Medical School, University of Western Australia, Perth, Australia.
| | - Asifa Farhat
- General Paediatrics, Perth Children's Hospital, Perth, Australia
| | - Abhijeet Rakshasbhuvankar
- Neonatal Intensive Care Unit, King Edward Memorial and Perth Children's Hospitals, Perth, Australia; Paediatric Division, Medical School, University of Western Australia, Perth, Australia
| | - Sam Athikarisamy
- Neonatal Intensive Care Unit, King Edward Memorial and Perth Children's Hospitals, Perth, Australia; Paediatric Division, Medical School, University of Western Australia, Perth, Australia
| | - Soumya Ghosh
- Children's Neuroscience Service, Department of Neurology, Perth Children's Hospital, Perth, Australia; Centre for Neuromuscular and Neurological Disorders, Perron Institute, University of Western Australia, Perth, Australia
| | - Lakshmi Nagarajan
- Paediatric Division, Medical School, University of Western Australia, Perth, Australia; Children's Neuroscience Service, Department of Neurology, Perth Children's Hospital, Perth, Australia
| |
Collapse
|
48
|
Woodman R, Miller C, Student J, Freeman K, Perl D, Lockette W. Alpha-methyltyrosine reduces the acute cardiovascular and behavioral sequelae in a murine model of traumatic brain injury. J Trauma Acute Care Surg 2023; 95:542-548. [PMID: 37165479 PMCID: PMC10545058 DOI: 10.1097/ta.0000000000004023] [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: 01/10/2023] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Increased catecholamines contribute to heightened cardiovascular reactivity and behavioral deficits after traumatic brain injury (TBI); adrenergic receptor blockade has limited success in reducing adverse sequelae of TBI. Injury-induced increases in the synthesis of catecholamines could contribute to adverse outcomes in TBI. Inhibition of catecholamine synthesis with alpha-methyltyrosine (αMT) could offer a benefit after TBI. METHODS Original research trial in mice randomized to αMT (50 mg·kg -1 ·d -1 ) or vehicle for 1 week after TBI induced by controlled cortical impact. Primary outcomes of cardiovascular reactivity and behavioral deficits were assessed after 1 week. Secondary outcomes included blood brain barrier permeability and quantification of gene transcription whose products determine intraneuronal chloride concentrations, the release of catecholamines, and activation of the sympathetic nervous system. These genes were the alpha-2 adrenergic receptor ("Adra2c"), the sodium-potassium-chloride cotransporter ("Nkcc1"), and the potassium chloride cotransporter ("Kcc2"). We also assessed the effect of TBI and αMT on the neuronal chloride/bicarbonate exchanger ("Ae3"). RESULTS Traumatic brain injury-induced increases in blood pressure and cardiac reactivity were blocked by αMT. Inhibition of catecholamine synthesis decreased blood brain barrier leakage and improved behavioral outcomes after TBI. Traumatic brain injury diminished the transcription of Adra2c and enhanced expression of Nkcc1 while reducing Kcc2 transcription; αMT prevented the induction of the Nkcc1 by TBI without reversing the effects of TBI on Kcc2 expression; αMT also diminished Ae3 transcription. CONCLUSION Traumatic brain injury acutely increases cardiovascular reactivity and induces behavioral deficits in an αMT-sensitive manner, most likely by inducing Nkcc1 gene transcription. Alpha-methyltyrosine may prove salutary in the treatment of TBI by attenuating the enhanced expression of Nkcc1, minimizing blood brain barrier leakage, and diminishing central catecholamine and sympathetic output. We also found an unreported relationship between Kcc2 and the chloride/bicarbonate exchanger, which should be considered in the design of trials planned to manipulate central intraneuronal chloride concentrations following acute brain injury.
Collapse
|
49
|
Napoli AJ, Laderwager S, Zoodsma JD, Biju B, Mucollari O, Schubel SK, Aprea C, Sayed A, Morgan K, Napoli A, Flanagan S, Wollmuth LP, Sirotkin HI. Loss of NMDA receptor function during development results in decreased KCC2 expression and increased neurons in the zebrafish forebrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554812. [PMID: 37786708 PMCID: PMC10541604 DOI: 10.1101/2023.08.25.554812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Developmental neurogenesis is a tightly regulated spatiotemporal process with its dysregulation implicated in neurodevelopmental disorders. NMDA receptors are glutamate-gated ion channels that are widely expressed in the early nervous system, yet their contribution to neurogenesis is poorly understood. Notably, a variety of mutations in genes encoding NMDA receptor subunits are associated with neurodevelopmental disorders. To rigorously define the role of NMDA receptors in developmental neurogenesis, we used a mutant zebrafish line ( grin1 -/- ) that lacks all NMDA receptors yet survives to 10 days post-fertilization, offering the opportunity to study post-embryonic neurodevelopment in the absence of NMDA receptors. Focusing on the forebrain, we find that these fish have a progressive supernumerary neuron phenotype confined to the telencephalon at the end of embryonic neurogenesis, but which extends to all forebrain regions during postembryonic neurogenesis. This enhanced neuron population does not arise directly from increased numbers or mitotic activity of radial glia cells, the principal neural stem cells. Rather, it stems from a lack of timely maturation of transit-amplifying neuroblasts into post-mitotic neurons, as indicated by a decrease in expression of the ontogenetically-expressed chloride transporter, KCC2. Pharmacological blockade with MK-801 recapitulates the grin1 -/- supernumerary neuron phenotype, indicating a requirement for ionotropic signaling. Thus, NMDA receptors are required for suppression of indirect, transit amplifying cell-driven neurogenesis by promoting maturational termination of mitosis. Loss of suppression results in neuronal overpopulation that can fundamentally change brain circuitry and may be a key factor in pathogenesis of neurodevelopmental disorders caused by NMDA receptor dysfunction.
Collapse
|
50
|
Shan X, Contreras MP, Sawangjit A, Dimitrov S, Born J, Inostroza M. Rearing is critical for forming spatial representations in pre-weanling rats. Behav Brain Res 2023; 452:114545. [PMID: 37321311 DOI: 10.1016/j.bbr.2023.114545] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/24/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Rearing, i.e., standing on the hind limbs in an upright posture, is part of a rat's innate exploratory motor program. Here, we examined in developing rats whether rearing is critical for the pup's capability to form spatial representations based on distal environmental cues. Pups (male) were tested at PD18, i.e., the first day they typically exhibit stable rearing, on a spatial habituation paradigm comprising a Familiarization session (with the pup exposed to an arena with a specific configuration of distal cues) followed, 3 h later, by a Test session where the pups were either re-exposed to the identical distal cue configuration (NoChange) or a changed configuration (DistalChange). In Experiment 1, rearing activity (rearing events, duration) decreased from Familiarization to Test in the NoChange pups but, remained elevated in the DistalChange group indicating that these pups recognized the distal novelty. Recognition of distal novelty was associated with increased c-Fos expression in hippocampal and medial prefrontal cortex (mPFC) areas, compared with NoChange pups. Analysis of GAD67+ cells suggested a parallel increase in excitation and inhibition specifically in prelimbic mPFC networks in response to distal cue changes. In Experiment 2, the pups were mechanically prevented from rearing while still seeing the distal cues during Familiarization. Rearing activity in the Test session of these pups did not differ between groups that were or were not exposed to a changed distal cue configuration at Test. The findings evidence a critical role of rearing for the emergence of allocentric representations integrating distal space during early development.
Collapse
Affiliation(s)
- Xia Shan
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; Graduate School of Neural & Behavioural Science, International Max Planck Research School, Tübingen, Germany
| | - María Paz Contreras
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; Graduate School of Neural & Behavioural Science, International Max Planck Research School, Tübingen, Germany
| | - Anuck Sawangjit
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), Institute for Diabetes Research & Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Germany; Werner Reichert Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany; German Center for Mental Health (DZPG), Germany.
| | - Marion Inostroza
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|