1
|
da Cunha Menezes E, de Abreu FF, Davis JB, Maurer SV, Roshko VC, Richardson A, Dowell J, Cassella SN, Stevens HE. Effects of gestational hypothyroidism on mouse brain development: Gabaergic systems and oxidative stress. Dev Biol 2024; 515:112-120. [PMID: 39048051 PMCID: PMC11330572 DOI: 10.1016/j.ydbio.2024.07.010] [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: 10/20/2023] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Hormonal imbalance during pregnancy is a risk factor for neuropsychiatric impairment in the offspring. It has been suggested that hypothyroidism leads to dysfunction of cortical GABAergic interneurons and inhibitory system development that in turn underlies impairment of the central nervous system. Here we investigated how gestational hypothyroidism affected offspring GABAergic system development as well as redox regulation parameters, because of previous links identified between the two. Experimental Gestational Hypothyroidism (EGH) was induced in CD-1 mice with 0.02% methimazole (MMI) in drinking water from embryonic day 9 (E9) until tissue collection at embryonic day 14 (E14) or E18. We examined GABAergic cell distribution and inhibitory system development gene expression as well as redox relevant gene expression and direct measures across all embryos regardless of sex. Intrauterine restriction of maternal thyroid hormones significantly impacted both of these outcomes in brain, as well as altering redox regulation in the placenta. GAD67+ neuronal migration was reduced, accompanied by a disruption in gene expression influencing GABAergic cell migration and cortical inhibitory neural system development. EGH also altered embryonic brain gene expression of Gpx1, Nfe2l2, Cat levels in the dorsal E14 brains. Additionally, EGH resulted in elevated TBARS, Gpx1 and Nfe2l2 in the ventral E18 brains. Furthermore, EGH downregulated placental Gpx1 gene expression at E14 and increased protein oxidation at E18. These findings support the hypothesis that sufficient maternal thyroid hormone supply to the fetus influences central nervous system development, including processes of GABAergic system development and redox equilibrium.
Collapse
Affiliation(s)
- Edênia da Cunha Menezes
- Psychiatry Department, Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States; Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Fabiula Francisca de Abreu
- Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Federal de Sergipe, São Cristóvão, Brazil
| | - Jada B Davis
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sara V Maurer
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Venezia C Roshko
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | | | - Jonathan Dowell
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sarah N Cassella
- Neuroscience Department, Loras College, Dubuque, IA, United States
| | - Hanna E Stevens
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
2
|
Hernández-Frausto M, Vivar C. Entorhinal cortex-hippocampal circuit connectivity in health and disease. Front Hum Neurosci 2024; 18:1448791. [PMID: 39372192 PMCID: PMC11449717 DOI: 10.3389/fnhum.2024.1448791] [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: 06/13/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
The entorhinal cortex (EC) and hippocampal (HC) connectivity is the main source of episodic memory formation and consolidation. The entorhinal-hippocampal (EC-HC) connection is classified as canonically glutamatergic and, more recently, has been characterized as a non-canonical GABAergic connection. Recent evidence shows that both EC and HC receive inputs from dopaminergic, cholinergic, and noradrenergic projections that modulate the mnemonic processes linked to the encoding and consolidation of memories. In the present review, we address the latest findings on the EC-HC connectivity and the role of neuromodulations during the mnemonic mechanisms of encoding and consolidation of memories and highlight the value of the cross-species approach to unravel the underlying cellular mechanisms known. Furthermore, we discuss how EC-HC connectivity early neurodegeneration may contribute to the dysfunction of episodic memories observed in aging and Alzheimer's disease (AD). Finally, we described how exercise may be a fundamental tool to prevent or decrease neurodegeneration.
Collapse
Affiliation(s)
- Melissa Hernández-Frausto
- NYU Neuroscience Institute, Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York University Langone Medical Center, New York, NY, United States
| | - Carmen Vivar
- Laboratory of Neurogenesis and Neuroplasticity, Department of Physiology, Biophysics and Neuroscience, Centro de Investigacion y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
3
|
Teixeira-Silva B, de Mattos GVRM, Carvalho VDF, Campello-Costa P. Caffeine intake during lactation has a sex-dependent effect on the hippocampal excitatory/inhibitory balance and pups' behavior. Brain Res 2024; 1846:149247. [PMID: 39304106 DOI: 10.1016/j.brainres.2024.149247] [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: 07/03/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
During early life, disruptions in glutamatergic and GABAergic synapse development in the hippocampus may contribute to several neurodevelopmental disorders, including cognitive deficits and psychiatric disorders. Caffeine is the most consumed psychoactive drug in the world, and previous work from our group has shown that caffeine disrupts visual system connections at different stages of development. This work aimed to investigate the effects of caffeine consumption during lactation in the glutamatergic and GABAergic synaptic markers in the hippocampus and on the behavior of rat offspring. We found that maternal caffeine intake significantly reduced GluN1 subunits of the NMDA receptor, increased the GluA1/GluA2 ratio of AMPA receptor in the dorsal hippocampus, and decreased GAD content in female pups' ventral hippocampus. On the other hand, an increase in GluN1/GluN2b subunits, a decrease in GAD content in the dorsal hippocampus, and a reduction of the GluA1 content in the ventral hippocampus were observed in male pups. In addition, changes in the behavior of the offspring submitted to indirect caffeine consumption were also sex-dependent, with females developing anxiety-like behavior and males showing anxiety-like behavior and hyper-locomotion. These results highlight that maternal caffeine intake promotes changes in the hippocampal excitatory and inhibitory balance and offspring behavior in a sex-dependent manner, suggesting that the population should be alerted to reduced caffeine consumption by breastfeeding mothers.
Collapse
Affiliation(s)
- Bruna Teixeira-Silva
- Programa de Pós-graduação em Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | | | - Vinicius de Frias Carvalho
- Programa de Pós-graduação em Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Laboratório de Inflamação, Centro de Pesquisa, Inovação e Vigilância em Covid-19 e Emergências Sanitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n° 4036, Manguinhos, CEP 21041-361 Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação (INCT-NIM), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n° 4365, Manguinhos, CEP 21045-900 Rio de Janeiro, Brazil
| | - Paula Campello-Costa
- Programa de Pós-graduação em Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| |
Collapse
|
4
|
Liu M, Wang L, Zhang Y, Dong H, Wang C, Chen Y, Qian Q, Zhang N, Wang S, Zhao G, Zhang Z, Lei M, Wang S, Zhao Q, Liu F. Investigating the shared genetic architecture between depression and subcortical volumes. Nat Commun 2024; 15:7647. [PMID: 39223129 PMCID: PMC11368965 DOI: 10.1038/s41467-024-52121-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Depression, a widespread and highly heritable mental health condition, profoundly affects millions of individuals worldwide. Neuroimaging studies have consistently revealed volumetric abnormalities in subcortical structures associated with depression. However, the genetic underpinnings shared between depression and subcortical volumes remain inadequately understood. Here, we investigate the extent of polygenic overlap using the bivariate causal mixture model (MiXeR), leveraging summary statistics from the largest genome-wide association studies for depression (N = 674,452) and 14 subcortical volumetric phenotypes (N = 33,224). Additionally, we identify shared genomic loci through conditional/conjunctional FDR analyses. MiXeR shows that subcortical volumetric traits share a substantial proportion of genetic variants with depression, with 44 distinct shared loci identified by subsequent conjunctional FDR analysis. These shared loci are predominantly located in intronic regions (58.7%) and non-coding RNA intronic regions (25.4%). The 269 protein-coding genes mapped by these shared loci exhibit specific developmental trajectories, with the expression level of 55 genes linked to both depression and subcortical volumes, and 30 genes linked to cognitive abilities and behavioral symptoms. These findings highlight a shared genetic architecture between depression and subcortical volumetric phenotypes, enriching our understanding of the neurobiological underpinnings of depression.
Collapse
Affiliation(s)
- Mengge Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Wang
- Department of Geriatrics and Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujie Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Haoyang Dong
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Caihong Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yayuan Chen
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qian Qian
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Nannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaoying Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guoshu Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhihui Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Minghuan Lei
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Sijia Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Qiyu Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
5
|
Cheng TW, Mills KL, Pfeifer JH. Revisiting adolescence as a sensitive period for sociocultural processing. Neurosci Biobehav Rev 2024; 164:105820. [PMID: 39032845 PMCID: PMC11407824 DOI: 10.1016/j.neubiorev.2024.105820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Waves of research and public discourse have characterized adolescence as periods of developmental risk and opportunity. Underlying this discussion is the recognition that adolescence is a period of major biological and social transition when experience may have an outsized effect on development. This article updates and expands upon prior work suggesting that adolescence may be a sensitive period for sociocultural processing specifically. By integrating evidence from developmental psychology and neuroscience, we identify how trajectories of social and neurobiological development may relate to adolescents' ability to adapt to and learn from their social environments. However, we also highlight gaps in the literature, including challenges in attributing developmental change to adolescent experiences. We discuss the importance of better understanding variability in biology (e.g., pubertal development) and cultural environments, as well as distinguishing between sensitive periods and periods of heightened sensitivity. Finally, we look toward future directions and translational implications of this research.
Collapse
Affiliation(s)
- Theresa W Cheng
- Department of Psychology, University of Oregon, 1227 University of Oregon, Eugene, OR 97403-1227, USA; Department of Psychology, Harvard University, 33 Kirkland St., Cambridge, MA 02138, USA.
| | - Kathryn L Mills
- Department of Psychology, University of Oregon, 1227 University of Oregon, Eugene, OR 97403-1227, USA.
| | - Jennifer H Pfeifer
- Department of Psychology, University of Oregon, 1227 University of Oregon, Eugene, OR 97403-1227, USA.
| |
Collapse
|
6
|
Chen C, Lan Z, Tang X, Chen W, Zhou X, Su H, Su R, Chen Z, Chen H, Guo Y, Deng W. Human-Derived Induced GABAergic Progenitor Cells Improve Cognitive Function in Mice and Inhibit Astrocyte Activation with Anti-Inflammatory Exosomes. Ann Neurol 2024; 96:488-507. [PMID: 38860520 DOI: 10.1002/ana.27001] [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: 05/24/2023] [Revised: 04/17/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVE The role of gamma-aminobutyric acid-ergic (GABAergic) neuron impairment in Alzheimer's disease (AD), and if and how transplantation of healthy GABAergic neurons can improve AD, remain unknown. METHODS Human-derived medial ganglionic eminence progenitors (hiMGEs) differentiated from programmed induced neural precursor cells (hiNPCs) were injected into the dentate gyrus region of the hippocampus (HIP). RESULTS We showed that grafts migrate to the whole brain and form functional synaptic connections in amyloid precursor protein gene/ presenilin-1 (APP/PS1) chimeric mice. Following transplantation of hiMGEs, behavioral deficits and AD-related pathology were alleviated and defective neurons were repaired. Notably, exosomes secreted from hiMGEs, which are rich in anti-inflammatory miRNA, inhibited astrocyte activation invitro and in vivo, and the mechanism was related to regulation of CD4+ Th1 cells mediated tumor necrosis factor (TNF) pathway. INTERPRETATION Taken together, these findings support the hypothesis that hiMGEs transplantation is an alternative treatment for neuronal loss in AD and demonstrate that exosomes with anti-inflammatory activity derived from hiMGEs are important factors for graft survival. ANN NEUROL 2024;96:488-507.
Collapse
Affiliation(s)
- Chunxia Chen
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, P. R. China
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China
| | - Zhaohui Lan
- Center for Brain Health and Brain Technology, Global Institute of Future Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Xihe Tang
- Department of Neurosurgery, Aviation General Hospital, Beijing, P. R. China
- Department of Neurosurgery, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, P. R. China
| | - Wan Chen
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, P. R. China
| | - Xing Zhou
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, P. R. China
| | - Hua Su
- Department of Pharmacology, Guangxi Institute of Chinese Medicine & Pharmaceutical Science, Nanning, P. R. China
| | - Rixiang Su
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, P. R. China
| | - Zhaolin Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China
| | - Ying Guo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, P. R. China
| |
Collapse
|
7
|
Islam KUS, Blaess S. The impact of the mesoprefrontal dopaminergic system on the maturation of interneurons in the murine prefrontal cortex. Front Neurosci 2024; 18:1403402. [PMID: 39035778 PMCID: PMC11257905 DOI: 10.3389/fnins.2024.1403402] [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: 03/19/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
The prefrontal cortex (PFC) undergoes a protracted maturation process. This is true both for local interneurons and for innervation from midbrain dopaminergic (mDA) neurons. In the striatum, dopaminergic (DA) neurotransmission is required for the maturation of medium spiny neurons during a critical developmental period. To investigate whether DA innervation influences the maturation of interneurons in the PFC, we used a conditional knockout (cKO) mouse model in which innervation from mDA neurons to the mPFC (mesoprefrontal innnervation) is not established during development. In this mouse model, the maturation of parvalbumin (PV) and calbindin (CB) interneuron populations in the PFC is dysregulated during a critical period in adolescence with changes persisting into adulthood. PV interneurons are particularly vulnerable to lack of mesoprefrontal input, showing an inability to maintain adequate PV expression with a concomitant decrease in Gad1 expression levels. Interestingly, lack of mesoprefrontal innervation does not appear to induce compensatory changes such as upregulation of DA receptor expression in PFC neurons or increased innervation density of other neuromodulatory (serotonergic and noradrenergic) innervation. In conclusion, our study shows that adolescence is a sensitive period during which mesoprefrontal input plays a critical role in promoting the maturation of specific interneuron subgroups. The results of this study will help to understand how a dysregulated mesoprefrontal DA system contributes to the pathophysiology of neurodevelopmental disorders.
Collapse
Affiliation(s)
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
8
|
Pilotto F, Diab R, Al Qassab Z, Saxena S. Generation and enrichment of cerebellar GABAergic interneurons from human induced pluripotent stem cells and intracellular calcium measurements. STAR Protoc 2024; 5:102936. [PMID: 38735042 PMCID: PMC11101973 DOI: 10.1016/j.xpro.2024.102936] [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: 09/07/2023] [Revised: 01/09/2024] [Accepted: 02/20/2024] [Indexed: 05/14/2024] Open
Abstract
GABAergic interneurons are inhibitory neurons of the CNS, playing a fundamental role in neural circuitry and activity. Here, we provide a robust protocol for the successful enrichment of human cerebellar GABAergic interneurons from human induced pluripotent stem cells (iPSCs) and measuring intracellular calcium transients. We describe in detail steps for culturing iPSCs; generating embryoid bodies; and differentiating and enriching for cerebellar GABAergic neurons (cGNs), with precise steps for their molecular characterization. We then detail the procedure for adeno-associated virus-mediated transduction of cGNs with genetically encoded calcium indicators, followed by intracellular calcium imaging and analyses. For complete details on the use and execution of this protocol, please refer to Pilotto et al.1.
Collapse
Affiliation(s)
- Federica Pilotto
- Institut Neuromyogenè, Inserm Lyon, Lyon, France; CNRS Université Claude Bernard Lyon I, Lyon, France
| | - Rim Diab
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Zahraa Al Qassab
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland; Department of Physical Medicine & Rehabilitation, University of Missouri, Columbia, MO, USA; NextGen Precision Health, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
9
|
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] [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
|
10
|
Magalhães DM, Stewart NA, Mampay M, Rolle SO, Hall CM, Moeendarbary E, Flint MS, Sebastião AM, Valente CA, Dymond MK, Sheridan GK. The sphingosine 1-phosphate analogue, FTY720, modulates the lipidomic signature of the mouse hippocampus. J Neurochem 2024; 168:1113-1142. [PMID: 38339785 DOI: 10.1111/jnc.16073] [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: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
The small-molecule drug, FTY720 (fingolimod), is a synthetic sphingosine 1-phosphate (S1P) analogue currently used to treat relapsing-remitting multiple sclerosis in both adults and children. FTY720 can cross the blood-brain barrier (BBB) and, over time, accumulate in lipid-rich areas of the central nervous system (CNS) by incorporating into phospholipid membranes. FTY720 has been shown to enhance cell membrane fluidity, which can modulate the functions of glial cells and neuronal populations involved in regulating behaviour. Moreover, direct modulation of S1P receptor-mediated lipid signalling by FTY720 can impact homeostatic CNS physiology, including neurotransmitter release probability, the biophysical properties of synaptic membranes, ion channel and transmembrane receptor kinetics, and synaptic plasticity mechanisms. The aim of this study was to investigate how chronic FTY720 treatment alters the lipid composition of CNS tissue in adolescent mice at a key stage of brain maturation. We focused on the hippocampus, a brain region known to be important for learning, memory, and the processing of sensory and emotional stimuli. Using mass spectrometry-based lipidomics, we discovered that FTY720 increases the fatty acid chain length of hydroxy-phosphatidylcholine (PCOH) lipids in the mouse hippocampus. It also decreases PCOH monounsaturated fatty acids (MUFAs) and increases PCOH polyunsaturated fatty acids (PUFAs). A total of 99 lipid species were up-regulated in the mouse hippocampus following 3 weeks of oral FTY720 exposure, whereas only 3 lipid species were down-regulated. FTY720 also modulated anxiety-like behaviours in young mice but did not affect spatial learning or memory formation. Our study presents a comprehensive overview of the lipid classes and lipid species that are altered in the hippocampus following chronic FTY720 exposure and provides novel insight into cellular and molecular mechanisms that may underlie the therapeutic or adverse effects of FTY720 in the central nervous system.
Collapse
Affiliation(s)
- Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | | - Myrthe Mampay
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Sara O Rolle
- Green Templeton College, University of Oxford, Oxford, UK
| | - Chloe M Hall
- School of Applied Sciences, University of Brighton, Brighton, UK
- Department of Mechanical Engineering, University College London, London, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd, London, UK
| | - Melanie S Flint
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Marcus K Dymond
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | |
Collapse
|
11
|
Arutiunian V, Santhosh M, Neuhaus E, Borland H, Tompkins C, Bernier RA, Bookheimer SY, Dapretto M, Gupta AR, Jack A, Jeste S, McPartland JC, Naples A, Van Horn JD, Pelphrey KA, Webb SJ. The relationship between gamma-band neural oscillations and language skills in youth with Autism Spectrum Disorder and their first-degree relatives. Mol Autism 2024; 15:19. [PMID: 38711098 PMCID: PMC11075235 DOI: 10.1186/s13229-024-00598-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Most children with Autism Spectrum Disorder (ASD) have co-occurring language impairments and some of these autism-specific language difficulties are also present in their non-autistic first-degree relatives. One of the possible neural mechanisms associated with variability in language functioning is alterations in cortical gamma-band oscillations, hypothesized to be related to neural excitation and inhibition balance. METHODS We used a high-density 128-channel electroencephalography (EEG) to register brain response to speech stimuli in a large sex-balanced sample of participants: 125 youth with ASD, 121 typically developing (TD) youth, and 40 unaffected siblings (US) of youth with ASD. Language skills were assessed with Clinical Evaluation of Language Fundamentals. RESULTS First, during speech processing, we identified significantly elevated gamma power in ASD participants compared to TD controls. Second, across all youth, higher gamma power was associated with lower language skills. Finally, the US group demonstrated an intermediate profile in both language and gamma power, with nonverbal IQ mediating the relationship between gamma power and language skills. LIMITATIONS We only focused on one of the possible neural contributors to variability in language functioning. Also, the US group consisted of a smaller number of participants in comparison to the ASD or TD groups. Finally, due to the timing issue in EEG system we have provided only non-phase-locked analysis. CONCLUSIONS Autistic youth showed elevated gamma power, suggesting higher excitation in the brain in response to speech stimuli and elevated gamma power was related to lower language skills. The US group showed an intermediate pattern of gamma activity, suggesting that the broader autism phenotype extends to neural profiles.
Collapse
Affiliation(s)
- Vardan Arutiunian
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Megha Santhosh
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Emily Neuhaus
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Heather Borland
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Chris Tompkins
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
| | - Susan Y Bookheimer
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Mirella Dapretto
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Abha R Gupta
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Allison Jack
- Department of Psychology, George Mason University, Fairfax, VA, USA
| | - Shafali Jeste
- Department of Neurology, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | | | - Adam Naples
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - John D Van Horn
- School of Data Science, University of Virginia, Charlottesville, VA, USA
| | - Kevin A Pelphrey
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Sara Jane Webb
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA.
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA.
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA.
| |
Collapse
|
12
|
Komori T, Okamura K, Ikehara M, Yamamuro K, Endo N, Okumura K, Yamauchi T, Ikawa D, Ouji-Sageshima N, Toritsuka M, Takada R, Kayashima Y, Ishida R, Mori Y, Kamikawa K, Noriyama Y, Nishi Y, Ito T, Saito Y, Nishi M, Kishimoto T, Tanaka KF, Hiroi N, Makinodan M. Brain-derived neurotrophic factor from microglia regulates neuronal development in the medial prefrontal cortex and its associated social behavior. Mol Psychiatry 2024; 29:1338-1349. [PMID: 38243072 PMCID: PMC11189755 DOI: 10.1038/s41380-024-02413-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024]
Abstract
Microglia and brain-derived neurotrophic factor (BDNF) are essential for the neuroplasticity that characterizes critical developmental periods. The experience-dependent development of social behaviors-associated with the medial prefrontal cortex (mPFC)-has a critical period during the juvenile period in mice. However, whether microglia and BDNF affect social development remains unclear. Herein, we aimed to elucidate the effects of microglia-derived BDNF on social behaviors and mPFC development. Mice that underwent social isolation during p21-p35 had increased Bdnf in the microglia accompanied by reduced adulthood sociability. Additionally, transgenic mice overexpressing microglial Bdnf-regulated using doxycycline at different time points-underwent behavioral, electrophysiological, and gene expression analyses. In these mice, long-term overexpression of microglial BDNF impaired sociability and excessive mPFC inhibitory neuronal circuit activity. However, administering doxycycline to normalize BDNF from p21 normalized sociability and electrophysiological function in the mPFC, whereas normalizing BDNF from later ages (p45-p50) did not normalize electrophysiological abnormalities in the mPFC, despite the improved sociability. To evaluate the possible role of BDNF in human sociability, we analyzed the relationship between adverse childhood experiences and BDNF expression in human macrophages, a possible proxy for microglia. Results show that adverse childhood experiences positively correlated with BDNF expression in M2 but not M1 macrophages. In summary, our study demonstrated the influence of microglial BDNF on the development of experience-dependent social behaviors in mice, emphasizing its specific impact on the maturation of mPFC function, particularly during the juvenile period. Furthermore, our results propose a translational implication by suggesting a potential link between BDNF secretion from macrophages and childhood experiences in humans.
Collapse
Affiliation(s)
- Takashi Komori
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuya Okamura
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Minobu Ikehara
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Nozomi Endo
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Daisuke Ikawa
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | | | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Ryohei Takada
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yoshinori Kayashima
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Rio Ishida
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Mori
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kohei Kamikawa
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Noriyama
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Nishi
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Mayumi Nishi
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Noboru Hiroi
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan.
| |
Collapse
|
13
|
Di Berardino C, Mainardi M, Brusco S, Benvenuto E, Broccoli V, Colasante G. Temporal manipulation of the Scn1a gene reveals its essential role in adult brain function. Brain 2024; 147:1216-1230. [PMID: 37812819 PMCID: PMC10994529 DOI: 10.1093/brain/awad350] [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: 05/23/2023] [Revised: 09/23/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023] Open
Abstract
Dravet syndrome is a severe epileptic encephalopathy, characterized by drug-resistant epilepsy, severe cognitive and behavioural deficits, with increased risk of sudden unexpected death (SUDEP). It is caused by haploinsufficiency of SCN1A gene encoding for the α-subunit of the voltage-gated sodium channel Nav1.1. Therapeutic approaches aiming to upregulate the healthy copy of SCN1A gene to restore its normal expression levels are being developed. However, whether Scn1a gene function is required only during a specific developmental time-window or, alternatively, if its physiological expression is necessary in adulthood is untested up to now. We induced Scn1a gene haploinsufficiency at two ages spanning postnatal brain development (P30 and P60) and compared the phenotypes of those mice to Scn1a perinatally induced mice (P2), recapitulating all deficits of Dravet mice. Induction of heterozygous Nav1.1 mutation at P30 and P60 elicited susceptibility to the development of both spontaneous and hyperthermia-induced seizures and SUDEP rates comparable to P2-induced mice, with symptom onset accompanied by the characteristic GABAergic interneuron dysfunction. Finally, delayed Scn1a haploinsufficiency induction provoked hyperactivity, anxiety and social attitude impairment at levels comparable to age matched P2-induced mice, while it was associated with a better cognitive performance, with P60-induced mice behaving like the control group. Our data show that maintenance of physiological levels of Nav1.1 during brain development is not sufficient to prevent Dravet symptoms and that long-lasting restoration of Scn1a gene expression would be required to grant optimal clinical benefit in patients with Dravet syndrome.
Collapse
Affiliation(s)
- Claudia Di Berardino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Martina Mainardi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Simone Brusco
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- National Research Council (CNR), Institute of Neuroscience, 20129 Milan, Italy
| | - Elena Benvenuto
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Gene and Cell Therapy PhD Program, Vita- Salute San Raffaele University, 20132 Milan, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- National Research Council (CNR), Institute of Neuroscience, 20129 Milan, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
14
|
Jia G, Sun Y, An P, Wu W, Shen Y, Liu H, Shan Y, Wang J, Lai CSW, Schreiner CE, He H, Zhou X. Auditory training remodels hippocampus-related memory in adult rats. Cereb Cortex 2024; 34:bhae045. [PMID: 38367612 DOI: 10.1093/cercor/bhae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/19/2024] Open
Abstract
Consequences of perceptual training, such as improvements in discriminative ability, are highly stimulus and task specific. Therefore, most studies on auditory training-induced plasticity in adult brain have focused on the sensory aspects, particularly on functional and structural effects in the auditory cortex. Auditory training often involves, other than auditory demands, significant cognitive components. Yet, how auditory training affects cognition-related brain regions, such as the hippocampus, remains unclear. Here, we found in female rats that auditory cue-based go/no-go training significantly improved the memory-guided behaviors associated with hippocampus. The long-term potentiations of the trained rats recorded in vivo in the hippocampus were also enhanced compared with the naïve rats. In parallel, the phosphorylation level of calcium/calmodulin-dependent protein kinase II and the expression of parvalbumin-positive interneurons in the hippocampus were both upregulated. These findings demonstrate that auditory training substantially remodels the processing and function of brain regions beyond the auditory system, which are associated with task demands.
Collapse
Affiliation(s)
- Guoqiang Jia
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- New York University-East China Normal University (NYU-ECNU) Institute of Brain and Cognitive Science, NYU Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Yutian Sun
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- New York University-East China Normal University (NYU-ECNU) Institute of Brain and Cognitive Science, NYU Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Pengying An
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- New York University-East China Normal University (NYU-ECNU) Institute of Brain and Cognitive Science, NYU Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Weiwei Wu
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- New York University-East China Normal University (NYU-ECNU) Institute of Brain and Cognitive Science, NYU Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Yang Shen
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- New York University-East China Normal University (NYU-ECNU) Institute of Brain and Cognitive Science, NYU Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Hui Liu
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- New York University-East China Normal University (NYU-ECNU) Institute of Brain and Cognitive Science, NYU Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Ye Shan
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Wuhu Hospital, East China Normal University, 259 Middle Jiuhua Road, Wuhu 241000, China
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, China
| | - Christoph E Schreiner
- Coleman Memorial Laboratory, Department of Otolaryngology-Head and Neck Surgery, University of California, 675 Nelson Rising Lane, San Francisco, CA 94158, United States
| | - Hua He
- Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, 225 Changhai Road, Shanghai 200438, China
| | - Xiaoming Zhou
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- New York University-East China Normal University (NYU-ECNU) Institute of Brain and Cognitive Science, NYU Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| |
Collapse
|
15
|
Vandael D, Vints K, Baatsen P, Śliwińska MA, Gabarre S, De Groef L, Moons L, Rybakin V, Gounko NV. Cdk5-dependent rapid formation and stabilization of dendritic spines by corticotropin-releasing factor. Transl Psychiatry 2024; 14:29. [PMID: 38233378 PMCID: PMC10794228 DOI: 10.1038/s41398-024-02749-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/24/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024] Open
Abstract
The neuropeptide corticotropin-releasing factor (CRF) exerts a pivotal role in modulating neuronal activity in the mammalian brain. The effects of CRF exhibit notable variations, depending on factors such as duration of exposure, concentration, and anatomical location. In the CA1 region of the hippocampus, the impact of CRF is dichotomous: chronic exposure to CRF impairs synapse formation and dendritic integrity, whereas brief exposure enhances synapse formation and plasticity. In the current study, we demonstrate long-term effects of acute CRF on the density and stability of mature mushroom spines ex vivo. We establish that both CRF receptors are present in this hippocampal region, and we pinpoint their precise subcellular localization within synapses by electron microscopy. Furthermore, both in vivo and ex vivo data collectively demonstrate that a transient surge of CRF in the CA1 activates the cyclin-dependent kinase 5 (Cdk5)-pathway. This activation leads to a notable augmentation in CRF-dependent spine formation. Overall, these data suggest that upon acute release of CRF in the CA1-SR synapse, both CRF-Rs can be activated and promote synaptic plasticity via activating different downstream signaling pathways, such as the Cdk5-pathway.
Collapse
Affiliation(s)
- Dorien Vandael
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Katlijn Vints
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Pieter Baatsen
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Małgorzata A Śliwińska
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Sergio Gabarre
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Lies De Groef
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Naamsestraat 61 box 2464, 3000, Leuven, Belgium
| | - Lieve Moons
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Naamsestraat 61 box 2464, 3000, Leuven, Belgium
| | - Vasily Rybakin
- National University of Singapore, Department of Microbiology and Immunology, Yng Loo Lin School of Medicine, and Immunology Program, 5 Science Drive 2, Blk MD4, 117545, Singapore, Singapore
| | - Natalia V Gounko
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium.
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium.
| |
Collapse
|
16
|
Gillespie B, Panthi S, Sundram S, Hill RA. The impact of maternal immune activation on GABAergic interneuron development: A systematic review of rodent studies and their translational implications. Neurosci Biobehav Rev 2024; 156:105488. [PMID: 38042358 DOI: 10.1016/j.neubiorev.2023.105488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Mothers exposed to infections during pregnancy disproportionally birth children who develop autism and schizophrenia, disorders associated with altered GABAergic function. The maternal immune activation (MIA) model recapitulates this risk factor, with many studies also reporting disruptions to GABAergic interneuron expression, protein, cellular density and function. However, it is unclear if there are species, sex, age, region, or GABAergic subtype specific vulnerabilities to MIA. Furthermore, to fully comprehend the impact of MIA on the GABAergic system a synthesised account of molecular, cellular, electrophysiological and behavioural findings was required. To this end we conducted a systematic review of GABAergic interneuron changes in the MIA model, focusing on the prefrontal cortex and hippocampus. We reviewed 102 articles that revealed robust changes in a number of GABAergic markers that present as gestationally-specific, region-specific and sometimes sex-specific. Disruptions to GABAergic markers coincided with distinct behavioural phenotypes, including memory, sensorimotor gating, anxiety, and sociability. Findings suggest the MIA model is a valid tool for testing novel therapeutics designed to recover GABAergic function and associated behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Sandesh Panthi
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
17
|
Ni P, Fan L, Jiang Y, Zhou C, Chung S. From cells to insights: the power of human pluripotent stem cell-derived cortical interneurons in psychiatric disorder modeling. Front Psychiatry 2023; 14:1336085. [PMID: 38188058 PMCID: PMC10768008 DOI: 10.3389/fpsyt.2023.1336085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Psychiatric disorders, such as schizophrenia (SCZ) and autism spectrum disorders (ASD), represent a global health challenge with their poorly understood and complex etiologies. Cortical interneurons (cINs) are the primary inhibitory neurons in the cortex and their subtypes, especially those that are generated from the medial ganglionic emission (MGE) region, have been shown to play an important role in the pathogenesis of these psychiatric disorders. Recent advances in induced pluripotent stem cell (iPSC) technologies provide exciting opportunities to model and study these disorders using human iPSC-derived cINs. In this review, we present a comprehensive overview of various methods employed to generate MGE-type cINs from human iPSCs, which are mainly categorized into induction by signaling molecules vs. direct genetic manipulation. We discuss their advantages, limitations, and potential applications in psychiatric disorder modeling to aid researchers in choosing the appropriate methods based on their research goals. We also provide examples of how these methods have been applied to study the pathogenesis of psychiatric disorders. In addition, we discuss ongoing challenges and future directions in the field. Overall, iPSC-derived cINs provide a powerful tool to model the developmental pathogenesis of psychiatric disorders, thus aiding in uncovering disease mechanisms and potential therapeutic targets. This review article will provide valuable resources for researchers seeking to navigate the complexities of cIN generation methods and their applications in the study of psychiatric disorders.
Collapse
Affiliation(s)
- Peiyan Ni
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, State Key Laboratory of Brain-Machine Intelligence, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Lingyi Fan
- The Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Youhui Jiang
- The Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Chuqing Zhou
- The Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Sangmi Chung
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
18
|
Forsyth JK, Bearden CE. Rethinking the First Episode of Schizophrenia: Identifying Convergent Mechanisms During Development and Moving Toward Prediction. Am J Psychiatry 2023; 180:792-804. [PMID: 37908094 DOI: 10.1176/appi.ajp.20230736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Affiliation(s)
- Jennifer K Forsyth
- Department of Psychology, University of Washington, Seattle (Forsyth); Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Behavioral Sciences, and Department of Psychology, University of California, Los Angeles (Bearden)
| | - Carrie E Bearden
- Department of Psychology, University of Washington, Seattle (Forsyth); Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Behavioral Sciences, and Department of Psychology, University of California, Los Angeles (Bearden)
| |
Collapse
|
19
|
Kameyama T, Miyata M, Shiotani H, Adachi J, Kakuta S, Uchiyama Y, Mizutani K, Takai Y. Heterogeneity of perivascular astrocyte endfeet depending on vascular regions in the mouse brain. iScience 2023; 26:108010. [PMID: 37829206 PMCID: PMC10565786 DOI: 10.1016/j.isci.2023.108010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/14/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Astrocytes interact with not only synapses but also brain blood vessels through perivascular astrocyte endfeet (PV-AEF) to form the neurovascular unit (NVU). However, PV-AEF components have not been fully identified. Here, we biochemically isolated blood vessels from mouse brain homogenates and purified PV-AEF. The purified PV-AEF were observed in different sizes, similar to PV-AEF on brain blood vessels. Mass spectrometry analysis identified 9,762 proteins in the purified PV-AEF, including cell adhesion molecules, nectin-2δ, Kirrel2, and podoplanin. Immunofluorescence microscopic analysis revealed that nectin-2δ and podoplanin were concentrated mainly in arteries/arterioles and veins/venules of the mouse brain, whereas Kirrel2 was mainly in arteries/arterioles. Nectin-2α/δ, Kirrel2, and podoplanin were preferentially observed in large sizes of the purified PV-AEF. Furthermore, Kirrel2 potentially has cell adhesion activity of cultured astrocytes. Collectively, these results indicate that PV-AEF have heterogeneity in sizes and molecular components, implying different roles of PV-AEF in NVU function depending on vascular regions.
Collapse
Affiliation(s)
- Takeshi Kameyama
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima 770-8503, Japan
| | - Muneaki Miyata
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Hajime Shiotani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory of Clinical and Analytical Chemistry, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Department of Cellular Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yasuo Uchiyama
- Department of Cellular Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Kiyohito Mizutani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
| |
Collapse
|
20
|
van Bijnen S, Muotka J, Parviainen T. Divergent auditory activation in relation to inhibition task performance in children and adults. Hum Brain Mapp 2023; 44:4972-4985. [PMID: 37493309 PMCID: PMC10502686 DOI: 10.1002/hbm.26418] [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: 01/27/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Adults and children show remarkable differences in cortical auditory activation which, in children, have shown relevance for cognitive performance, specifically inhibitory control. However, it has not been tested whether these differences translate to functional differences in response inhibition between adults and children. We recorded auditory responses of adults and school-aged children (6-14 years) using combined magneto- and electroencephalography (M/EEG) during passive listening conditions and an auditory Go/No-go task. The associations between auditory cortical responses and inhibition performance measures diverge between adults and children; while in children the brain-behavior associations are not significant, or stronger responses are beneficial, adults show negative associations between auditory cortical responses and inhibitory performance. Furthermore, we found differences in brain responses between adults and children; the late (~200 ms post stimulation) adult peak activation shifts from auditory to frontomedial areas. In contrast, children show prolonged obligatory responses in the auditory cortex. Together this likely translates to a functional difference between adults and children in the cortical resources for performance consistency in auditory-based cognitive tasks.
Collapse
Affiliation(s)
- Sam van Bijnen
- Centre for Interdisciplinary Brain Research, Department of PsychologyUniversity of JyväskyläJyväskyläFinland
- Faculty of Social and Behavioural ScienceUtrecht UniversityThe Netherlands
| | - Joona Muotka
- Centre for Interdisciplinary Brain Research, Department of PsychologyUniversity of JyväskyläJyväskyläFinland
| | - Tiina Parviainen
- Centre for Interdisciplinary Brain Research, Department of PsychologyUniversity of JyväskyläJyväskyläFinland
| |
Collapse
|
21
|
Larsen B, Sydnor VJ, Keller AS, Yeo BTT, Satterthwaite TD. A critical period plasticity framework for the sensorimotor-association axis of cortical neurodevelopment. Trends Neurosci 2023; 46:847-862. [PMID: 37643932 PMCID: PMC10530452 DOI: 10.1016/j.tins.2023.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023]
Abstract
To understand human brain development it is necessary to describe not only the spatiotemporal patterns of neurodevelopment but also the neurobiological mechanisms that underlie them. Human neuroimaging studies have provided evidence for a hierarchical sensorimotor-to-association (S-A) axis of cortical neurodevelopment. Understanding the biological mechanisms that underlie this program of development using traditional neuroimaging approaches has been challenging. Animal models have been used to identify periods of enhanced experience-dependent plasticity - 'critical periods' - that progress along cortical hierarchies and are governed by a conserved set of neurobiological mechanisms that promote and then restrict plasticity. In this review we hypothesize that the S-A axis of cortical development in humans is partly driven by the cascading maturation of critical period plasticity mechanisms. We then describe how recent advances in in vivo neuroimaging approaches provide a promising path toward testing this hypothesis by linking signals derived from non-invasive imaging to critical period mechanisms.
Collapse
Affiliation(s)
- Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie J Sydnor
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arielle S Keller
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - B T Thomas Yeo
- Centre for Sleep and Cognition (CSC), and Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Electrical and Computer Engineering, National University of Singapore, Singapore; N.1 Institute for Health and Institute for Digital Medicine (WisDM), National University of Singapore, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
| | - Theodore D Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn-CHOP Lifespan Brain Institute, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Hernández-Frausto M, Bilash OM, Masurkar AV, Basu J. Local and long-range GABAergic circuits in hippocampal area CA1 and their link to Alzheimer's disease. Front Neural Circuits 2023; 17:1223891. [PMID: 37841892 PMCID: PMC10570439 DOI: 10.3389/fncir.2023.1223891] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
GABAergic inhibitory neurons are the principal source of inhibition in the brain. Traditionally, their role in maintaining the balance of excitation-inhibition has been emphasized. Beyond homeostatic functions, recent circuit mapping and functional manipulation studies have revealed a wide range of specific roles that GABAergic circuits play in dynamically tilting excitation-inhibition coupling across spatio-temporal scales. These span from gating of compartment- and input-specific signaling, gain modulation, shaping input-output functions and synaptic plasticity, to generating signal-to-noise contrast, defining temporal windows for integration and rate codes, as well as organizing neural assemblies, and coordinating inter-regional synchrony. GABAergic circuits are thus instrumental in controlling single-neuron computations and behaviorally-linked network activity. The activity dependent modulation of sensory and mnemonic information processing by GABAergic circuits is pivotal for the formation and maintenance of episodic memories in the hippocampus. Here, we present an overview of the local and long-range GABAergic circuits that modulate the dynamics of excitation-inhibition and disinhibition in the main output area of the hippocampus CA1, which is crucial for episodic memory. Specifically, we link recent findings pertaining to GABAergic neuron molecular markers, electrophysiological properties, and synaptic wiring with their function at the circuit level. Lastly, given that area CA1 is particularly impaired during early stages of Alzheimer's disease, we emphasize how these GABAergic circuits may contribute to and be involved in the pathophysiology.
Collapse
Affiliation(s)
- Melissa Hernández-Frausto
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Olesia M. Bilash
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Arjun V. Masurkar
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Jayeeta Basu
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Center for Neural Science, New York University, New York, NY, United States
| |
Collapse
|
23
|
Noguchi A, Matsumoto N, Ikegaya Y. Postnatal Maturation of Membrane Potential Dynamics during in Vivo Hippocampal Ripples. J Neurosci 2023; 43:6126-6140. [PMID: 37400254 PMCID: PMC10476637 DOI: 10.1523/jneurosci.0125-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023] Open
Abstract
Sharp-wave ripples (SWRs) are transient high-frequency oscillations of local field potentials (LFPs) in the hippocampus and play a critical role in memory consolidation. During SWRs, CA1 pyramidal cells exhibit rapid spike sequences that often replay the sequential activity that occurred during behavior. This temporally organized firing activity gradually emerges during 2 weeks after the eye opening; however, it remains unclear how the organized spikes during SWRs mature at the intracellular membrane potential (Vm) level. Here, we recorded Vm of CA1 pyramidal cells simultaneously with hippocampal LFPs from anesthetized immature mice of either sex after the developmental emergence of SWRs. On postnatal days 16 and 17, Vm dynamics around SWRs were premature, characterized by prolonged depolarizations without either pre- or post-SWR hyperpolarizations. The biphasic hyperpolarizations, features typical of adult SWR-relevant Vm, formed by approximately postnatal day 30. This Vm maturation was associated with an increase in SWR-associated inhibitory inputs to pyramidal cells. Thus, the development of SWR-relevant inhibition restricts the temporal windows for spikes of pyramidal cells and allows CA1 pyramidal cells to organize their spike sequences during SWRs.SIGNIFICANCE STATEMENT Sharp-wave ripples (SWRs) are prominent hippocampal oscillations and play a critical role in memory consolidation. During SWRs, hippocampal neurons synchronously emit spikes with organized temporal patterns. This temporal structure of spikes during SWRs develops during the third and fourth postnatal weeks, but the underlying mechanisms are not well understood. Here, we recorded in vivo membrane potentials from hippocampal neurons in premature mice and suggest that the maturation of SWR-associated inhibition enables hippocampal neurons to produce precisely controlled spike times during SWRs.
Collapse
Affiliation(s)
- Asako Noguchi
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
- Institute for AI and Beyond, University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
- Institute for AI and Beyond, University of Tokyo, Tokyo, 113-0033, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| |
Collapse
|
24
|
Zhang L, Shi W, Liu J, Chen K, Zhang G, Zhang S, Cong B, Li Y. Interleukin 6 (IL-6) Regulates GABAA Receptors in the Dorsomedial Hypothalamus Nucleus (DMH) through Activation of the JAK/STAT Pathway to Affect Heart Rate Variability in Stressed Rats. Int J Mol Sci 2023; 24:12985. [PMID: 37629166 PMCID: PMC10455568 DOI: 10.3390/ijms241612985] [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: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The dorsomedial hypothalamus nucleus (DMH) is an important component of the autonomic nervous system and plays a critical role in regulating the sympathetic outputs of the heart. Stress alters the neuronal activity of the DMH, affecting sympathetic outputs and triggering heart rate variability. However, the specific molecular mechanisms behind stress leading to abnormal DMH neuronal activity have still not been fully elucidated. Therefore, in the present study, we successfully constructed a stressed rat model and used it to investigate the potential molecular mechanisms by which IL-6 regulates GABAA receptors in the DMH through activation of the JAK/STAT pathway and thus affects heart rate variability in rats. By detecting the c-Fos expression of neurons in the DMH and electrocardiogram (ECG) changes in rats, we clarified the relationship between abnormal DMH neuronal activity and heart rate variability in stressed rats. Then, using ELISA, immunohistochemical staining, Western blotting, RT-qPCR, and RNAscope, we further explored the correlation between the IL-6/JAK/STAT signaling pathway and GABAA receptors. The data showed that an increase in IL-6 induced by stress inhibited GABAA receptors in DMH neurons by activating the JAK/STAT signaling pathway, while specific inhibition of the JAK/STAT signaling pathway using AG490 obviously reduced DMH neuronal activity and improved heart rate variability in rats. These findings suggest that IL-6 regulates the expression of GABAA receptors via the activation of the JAK/STAT pathway in the DMH, which may be an important cause of heart rate variability in stressed rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Department of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; (L.Z.); (W.S.); (J.L.); (K.C.); (G.Z.); (S.Z.)
| | - Yingmin Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Department of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; (L.Z.); (W.S.); (J.L.); (K.C.); (G.Z.); (S.Z.)
| |
Collapse
|
25
|
Hostetler RE, Hu H, Agmon A. Genetically Defined Subtypes of Somatostatin-Containing Cortical Interneurons. eNeuro 2023; 10:ENEURO.0204-23.2023. [PMID: 37463742 PMCID: PMC10414551 DOI: 10.1523/eneuro.0204-23.2023] [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: 06/14/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023] Open
Abstract
Inhibitory interneurons play a crucial role in proper development and function of the mammalian cerebral cortex. Of the different inhibitory subclasses, dendritic-targeting, somatostatin-containing (SOM) interneurons may be the most diverse. Earlier studies used GFP-expressing and recombinase-expressing mouse lines to characterize genetically defined subtypes of SOM interneurons by morphologic, electrophysiological, and neurochemical properties. More recently, large-scale studies classified SOM interneurons into 13 morpho-electric transcriptomic (MET) types. It remains unclear, however, how these various classification schemes relate to each other, and experimental access to MET types has been limited by the scarcity of specific mouse driver lines. To address these issues, we crossed Flp and Cre driver lines with a dual-color intersectional reporter, allowing experimental access to several combinatorially defined SOM subsets. Brains from adult mice of both sexes were retrogradely dye labeled from the pial surface to identify layer 1-projecting neurons and immunostained against several marker proteins, revealing correlations between genetic label, axonal target, and marker protein expression in the same neurons. Lastly, using whole-cell recordings ex vivo, we analyzed and compared electrophysiological properties between different intersectional subsets. We identified two layer 1-targeting subtypes with nonoverlapping marker protein expression and electrophysiological properties, which, together with a previously characterized layer 4-targeting subtype, account for >50% of all layer 5 SOM cells and >40% of all SOM cells, and appear to map onto 5 of the 13 MET types. Genetic access to these subtypes will allow researchers to determine their synaptic inputs and outputs and uncover their roles in cortical computations and animal behavior.
Collapse
Affiliation(s)
- Rachel E Hostetler
- Department of Neuroscience, West Virginia University Rockefeller Neuroscience Institute, Morgantown, WV 26506
| | - Hang Hu
- Department of Neuroscience, West Virginia University Rockefeller Neuroscience Institute, Morgantown, WV 26506
| | - Ariel Agmon
- Department of Neuroscience, West Virginia University Rockefeller Neuroscience Institute, Morgantown, WV 26506
| |
Collapse
|
26
|
Peerboom C, de Kater S, Jonker N, Rieter MPJM, Wijne T, Wierenga CJ. Delaying the GABA Shift Indirectly Affects Membrane Properties in the Developing Hippocampus. J Neurosci 2023; 43:5483-5500. [PMID: 37438107 PMCID: PMC10376938 DOI: 10.1523/jneurosci.0251-23.2023] [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: 02/10/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023] Open
Abstract
During the first two postnatal weeks, intraneuronal chloride concentrations in rodents gradually decrease, causing a shift from depolarizing to hyperpolarizing GABA responses. The postnatal GABA shift is delayed in rodent models for neurodevelopmental disorders and in human patients, but the impact of a delayed GABA shift on the developing brain remains obscure. Here we examine the direct and indirect consequences of a delayed postnatal GABA shift on network development in organotypic hippocampal cultures made from 6- to 7-d-old mice by treating the cultures for 1 week with VU0463271, a specific inhibitor of the chloride exporter KCC2. We verified that VU treatment delayed the GABA shift and kept GABA signaling depolarizing until DIV9. We found that the structural and functional development of excitatory and inhibitory synapses at DIV9 was not affected after VU treatment. In line with previous studies, we observed that GABA signaling was already inhibitory in control and VU-treated postnatal slices. Surprisingly, 14 d after the VU treatment had ended (DIV21), we observed an increased frequency of spontaneous inhibitory postsynaptic currents in CA1 pyramidal cells, while excitatory currents were not changed. Synapse numbers and release probability were unaffected. We found that dendrite-targeting interneurons in the stratum radiatum had an elevated resting membrane potential, while pyramidal cells were less excitable compared with control slices. Our results show that depolarizing GABA signaling does not promote synapse formation after P7, and suggest that postnatal intracellular chloride levels indirectly affect membrane properties in a cell-specific manner.SIGNIFICANCE STATEMENT During brain development, the action of neurotransmitter GABA shifts from depolarizing to hyperpolarizing. This shift is a thought to play a critical role in synapse formation. A delayed shift is common in rodent models for neurodevelopmental disorders and in human patients, but its consequences for synaptic development remain obscure. Here, we delayed the GABA shift by 1 week in organotypic hippocampal cultures and carefully examined the consequences for circuit development. We find that delaying the shift has no direct effects on synaptic development, but instead leads to indirect, cell type-specific changes in membrane properties. Our data call for careful assessment of alterations in cellular excitability in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Carlijn Peerboom
- Cell Biology, Neurobiology and Biophysics, Biology Department, Utrecht University, Utrecht, 3584 CH, The Netherlands
| | - Sam de Kater
- Cell Biology, Neurobiology and Biophysics, Biology Department, Utrecht University, Utrecht, 3584 CH, The Netherlands
| | - Nikki Jonker
- Cell Biology, Neurobiology and Biophysics, Biology Department, Utrecht University, Utrecht, 3584 CH, The Netherlands
| | - Marijn P J M Rieter
- Cell Biology, Neurobiology and Biophysics, Biology Department, Utrecht University, Utrecht, 3584 CH, The Netherlands
| | - Tessel Wijne
- Cell Biology, Neurobiology and Biophysics, Biology Department, Utrecht University, Utrecht, 3584 CH, The Netherlands
| | - Corette J Wierenga
- Cell Biology, Neurobiology and Biophysics, Biology Department, Utrecht University, Utrecht, 3584 CH, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, 6525 AJ, The Netherlands
| |
Collapse
|
27
|
Ibrahim LA, Wamsley B, Alghamdi N, Yusuf N, Sevier E, Hairston A, Sherer M, Jaglin XH, Xu Q, Guo L, Khodadadi-Jamayran A, Favuzzi E, Yuan Y, Dimidschstein J, Darnell RB, Fishell G. Nova proteins direct synaptic integration of somatostatin interneurons through activity-dependent alternative splicing. eLife 2023; 12:e86842. [PMID: 37347149 PMCID: PMC10287156 DOI: 10.7554/elife.86842] [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: 02/08/2023] [Accepted: 04/17/2023] [Indexed: 06/23/2023] Open
Abstract
Somatostatin interneurons are the earliest born population of cortical inhibitory cells. They are crucial to support normal brain development and function; however, the mechanisms underlying their integration into nascent cortical circuitry are not well understood. In this study, we begin by demonstrating that the maturation of somatostatin interneurons in mouse somatosensory cortex is activity dependent. We then investigated the relationship between activity, alternative splicing, and synapse formation within this population. Specifically, we discovered that the Nova family of RNA-binding proteins are activity-dependent and are essential for the maturation of somatostatin interneurons, as well as their afferent and efferent connectivity. Within this population, Nova2 preferentially mediates the alternative splicing of genes required for axonal formation and synaptic function independently from its effect on gene expression. Hence, our work demonstrates that the Nova family of proteins through alternative splicing are centrally involved in coupling developmental neuronal activity to cortical circuit formation.
Collapse
Affiliation(s)
- Leena Ali Ibrahim
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST)ThuwalSaudi Arabia
- Stanley Center at the BroadCambridgeUnited States
| | - Brie Wamsley
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University School of MedicineNew YorkUnited States
| | - Norah Alghamdi
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST)ThuwalSaudi Arabia
| | - Nusrath Yusuf
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
- Stanley Center at the BroadCambridgeUnited States
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University School of MedicineNew YorkUnited States
| | - Elaine Sevier
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
- Stanley Center at the BroadCambridgeUnited States
| | - Ariel Hairston
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mia Sherer
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
- Stanley Center at the BroadCambridgeUnited States
| | - Xavier Hubert Jaglin
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University School of MedicineNew YorkUnited States
| | - Qing Xu
- Center for Genomics & Systems Biology, New York UniversityAbu DhabiUnited Arab Emirates
| | - Lihua Guo
- Center for Genomics & Systems Biology, New York UniversityAbu DhabiUnited Arab Emirates
| | - Alireza Khodadadi-Jamayran
- Genome Technology Center, Applied Bioinformatics Laboratories, NYU Langone Medical CenterNew YorkUnited States
| | - Emilia Favuzzi
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
- Stanley Center at the BroadCambridgeUnited States
| | - Yuan Yuan
- Laboratory of Molecular Neuro-Oncology, The Rockefeller UniversityNew YorkUnited States
| | | | - Robert B Darnell
- Laboratory of Molecular Neuro-Oncology, The Rockefeller UniversityNew YorkUnited States
| | - Gordon Fishell
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
- Stanley Center at the BroadCambridgeUnited States
| |
Collapse
|
28
|
Gainutdinov A, Shipkov D, Sintsov M, Fabrizi L, Nasretdinov A, Khazipov R, Valeeva G. Somatosensory-Evoked Early Sharp Waves in the Neonatal Rat Hippocampus. Int J Mol Sci 2023; 24:8721. [PMID: 37240066 PMCID: PMC10217913 DOI: 10.3390/ijms24108721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
The developing entorhinal-hippocampal system is embedded within a large-scale bottom-up network, where spontaneous myoclonic movements, presumably via somatosensory feedback, trigger hippocampal early sharp waves (eSPWs). The hypothesis, that somatosensory feedback links myoclonic movements with eSPWs, implies that direct somatosensory stimulation should also be capable of evoking eSPWs. In this study, we examined hippocampal responses to electrical stimulation of the somatosensory periphery in urethane-anesthetized, immobilized neonatal rat pups using silicone probe recordings. We found that somatosensory stimulation in ~33% of the trials evoked local field potential (LFP) and multiple unit activity (MUA) responses identical to spontaneous eSPWs. The somatosensory-evoked eSPWs were delayed from the stimulus, on average, by 188 ms. Both spontaneous and somatosensory-evoked eSPWs (i) had similar amplitude of ~0.5 mV and half-duration of ~40 ms, (ii) had similar current-source density (CSD) profiles, with current sinks in CA1 strata radiatum, lacunosum-moleculare and DG molecular layer and (iii) were associated with MUA increase in CA1 and DG. Our results indicate that eSPWs can be triggered by direct somatosensory stimulations and support the hypothesis that sensory feedback from movements is involved in the association of eSPWs with myoclonic movements in neonatal rats.
Collapse
Affiliation(s)
- Azat Gainutdinov
- Institut de Neurobiologie de la Méditerranée (INMED U1249), Aix-Marseille University, 13273 Marseille, France;
| | - Dmitrii Shipkov
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia (M.S.); (L.F.); (G.V.)
| | - Mikhail Sintsov
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia (M.S.); (L.F.); (G.V.)
| | - Lorenzo Fabrizi
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia (M.S.); (L.F.); (G.V.)
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Azat Nasretdinov
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia (M.S.); (L.F.); (G.V.)
| | - Roustem Khazipov
- Institut de Neurobiologie de la Méditerranée (INMED U1249), Aix-Marseille University, 13273 Marseille, France;
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia (M.S.); (L.F.); (G.V.)
| | - Guzel Valeeva
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia (M.S.); (L.F.); (G.V.)
| |
Collapse
|
29
|
Karst H, Droogers WJ, van der Weerd N, Damsteegt R, van Kroonenburg N, Sarabdjitsingh RA, Joëls M. Acceleration of GABA-switch after early life stress changes mouse prefrontal glutamatergic transmission. Neuropharmacology 2023; 234:109543. [PMID: 37061088 DOI: 10.1016/j.neuropharm.2023.109543] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 04/17/2023]
Abstract
Early life stress (ELS) alters the excitation-inhibition-balance (EI-balance) in various rodent brain areas and may be responsible for behavioral impairment later in life. The EI-balance is (amongst others) influenced by the switch of GABAergic transmission from excitatory to inhibitory, the so-called "GABA-switch". Here, we investigated how ELS affects the GABA-switch in mouse infralimbic Prefrontal Cortex layer 2/3 neurons, using the limited-nesting-and-bedding model. In ELS mice, the GABA-switch occurred already between postnatal day (P) 6 and P9, as opposed to P15-P21 in controls. This was associated with increased expression of the inward chloride transporter NKCC1, compared to the outward chloride transporter KCC2, both of which are important for the intracellular chloride concentration and, hence, the GABA reversal potential (Erev). Chloride transporters are not only important for regulating chloride concentration postsynaptically, but also presynaptically. Depending on the Erev of GABA, presynaptic GABAA receptor stimulation causes a depolarization or hyperpolarization, and thereby enhanced or reduced fusion of glutamate vesicles respectively, in turn changing the frequency of miniature postsynaptic currents (mEPSCs). In accordance, bumetanide, a blocker of NKCC1, shifted the Erev GABA towards more hyperpolarized levels in P9 control mice and reduced the mEPSC frequency. Other modulators of chloride transporters, e.g. VU0463271 (a KCC2 antagonist) and aldosterone -which increases NKCC1 expression-did not affect postsynaptic Erev in ELS P9 mice, but did increase the mEPSC frequency. We conclude that the mouse GABA-switch is accelerated after ELS, affecting both the pre- and postsynaptic chloride homeostasis, the former altering glutamatergic transmission. This may considerably affect brain development.
Collapse
Affiliation(s)
- Henk Karst
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Wouter J Droogers
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Nelleke van der Weerd
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Ruth Damsteegt
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Nicky van Kroonenburg
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - R Angela Sarabdjitsingh
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Marian Joëls
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands; University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
30
|
Vaquero-Rodríguez A, Ortuzar N, Lafuente JV, Bengoetxea H. Enriched environment as a nonpharmacological neuroprotective strategy. Exp Biol Med (Maywood) 2023; 248:553-560. [PMID: 37309729 PMCID: PMC10350798 DOI: 10.1177/15353702231171915] [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] [Indexed: 06/14/2023] Open
Abstract
The structure and functions of the central nervous system are influenced by environmental stimuli, which also play an important role in brain diseases. Enriched environment (EE) consists of producing modifications in the environment of standard laboratory animals to induce an improvement in their biological conditions. This paradigm promotes transcriptional and translational effects that result in ameliorated motor, sensory, and cognitive stimulation. EE has been shown to enhance experience-dependent cellular plasticity and cognitive performance in animals housed under these conditions compared with animals housed under standard conditions. In addition, several studies claim that EE induces nerve repair by restoring functional activities through morphological, cellular, and molecular adaptations in the brain that have clinical relevance in neurological and psychiatric disorders. In fact, the effects of EE have been studied in different animal models of psychiatric and neurological diseases, such as Alzheimer's disease, Parkinson's disease, schizophrenia, ischemic brain injury, or traumatic brain injury, delaying the onset and progression of a wide variety of symptoms of these disorders. In this review, we analyze the action of EE focused on diseases of the central nervous system and the translation to humans to develop a bridge to its application.
Collapse
Affiliation(s)
- Andrea Vaquero-Rodríguez
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Naiara Ortuzar
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - José Vicente Lafuente
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Harkaitz Bengoetxea
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
31
|
Arutiunian V, Arcara G, Buyanova I, Davydova E, Pereverzeva D, Sorokin A, Tyushkevich S, Mamokhina U, Danilina K, Dragoy O. Neuromagnetic 40 Hz Auditory Steady-State Response in the left auditory cortex is related to language comprehension in children with Autism Spectrum Disorder. Prog Neuropsychopharmacol Biol Psychiatry 2023; 122:110690. [PMID: 36470421 DOI: 10.1016/j.pnpbp.2022.110690] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 11/06/2022] [Accepted: 11/29/2022] [Indexed: 12/08/2022]
Abstract
Language impairment is comorbid in most children with Autism Spectrum Disorder (ASD), but its neural mechanisms are still poorly understood. Some studies hypothesize that the atypical low-level sensory perception in the auditory cortex accounts for the abnormal language development in these children. One of the potential non-invasive measures of such low-level perception can be the cortical gamma-band oscillations registered with magnetoencephalography (MEG), and 40 Hz Auditory Steady-State Response (40 Hz ASSR) is a reliable paradigm for eliciting auditory gamma response. Although there is research in children with and without ASD using 40 Hz ASSR, nothing is known about the relationship between this auditory response in children with ASD and their language abilities measured directly in formal assessment. In the present study, we used MEG and individual brain models to investigate 40 Hz ASSR in primary-school-aged children with and without ASD. It was also used to assess how the strength of the auditory response is related to language abilities of children with ASD, their non-verbal IQ, and social functioning. A total of 40 children were included in the study. The results demonstrated that 40 Hz ASSR was reduced in the right auditory cortex in children with ASD when comparing them to typically developing controls. Importantly, our study provides the first evidence of the association between 40 Hz ASSR in the language-dominant left auditory cortex and language comprehension in children with ASD. This link was domain-specific because the other brain-behavior correlations were non-significant.
Collapse
Affiliation(s)
| | | | - Irina Buyanova
- Center for Language and Brain, HSE University, Moscow, Russia
| | - Elizaveta Davydova
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia; Chair of Differential Psychology and Psychophysiology, Moscow State University of Psychology and Education, Moscow, Russia
| | - Darya Pereverzeva
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Alexander Sorokin
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia; Haskins Laboratories, New Haven, CT, United States of America
| | - Svetlana Tyushkevich
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Uliana Mamokhina
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Kamilla Danilina
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Olga Dragoy
- Center for Language and Brain, HSE University, Moscow, Russia; Institute of Linguistics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
32
|
Yu Y, Tian D, Ri S, Kim T, Ju K, Zhang J, Teng S, Zhang W, Shi W, Liu G. Gamma-aminobutyric acid (GABA) suppresses hemocyte phagocytosis by binding to GABA receptors and modulating corresponding downstream pathways in blood clam, Tegillarca granosa. FISH & SHELLFISH IMMUNOLOGY 2023; 134:108608. [PMID: 36764632 DOI: 10.1016/j.fsi.2023.108608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
Although accumulating data demonstrated that gamma-aminobutyric acid (GABA), an inhibitory neurotransmitter, plays an important regulatory role in immunity of vertebrates, its immunomodulatory function and mechanisms of action remain poorly understood in invertebrates such as bivalve mollusks. In this study, the effect of GABA on phagocytic activity of hemocytes was evaluated in a commercial bivalve species, Tegillarca granosa. Furthermore, the potential regulatory mechanism underpinning was investigated by assessing potential downstream targets. Data obtained demonstrated that in vitro GABA incubation significantly constrained the phagocytic activity of hemocytes. In addition, the GABA-induced suppression of phagocytosis was markedly relieved by blocking of GABAA and GABAB receptors using corresponding antagonists. Hemocytes incubated with lipopolysaccharides (LPS) and GABA had significant higher K+-Cl- cotransporter 2 (KCC2) content compared to the control. In addition, GABA treatment led to an elevation in intracellular Cl-, which was shown to be leveled down to normal by blocking the ionotropic GABAA receptor. Treatment with GABAA receptor antagonist also rescued the suppression of GABAA receptor-associated protein (GABARAP), KCC, TNF receptor associated factor 6 (TRAF6), inhibitor of nuclear factor kappa-B kinase subunit alpha (IKKα), and nuclear factor kappa B subunit 1 (NFκB) caused by GABA incubation. Furthermore, incubation of hemocytes with GABA resulted in a decrease in cAMP content, an increase in intracellular Ca2+, and downregulation of cAMP-dependent protein kinase (PKA), calmodulin kinase II (CAMK2), calmodulin (CaM), calcineurin (CaN), TRAF6, IKKα, and NFκB. All these above-mentioned changes were found to be evidently relieved by blocking the metabotropic G-protein-coupled GABAB receptor. Our results suggest GABA may play an inhibitory role on phagocytosis through binding to both GABAA and GABAB receptors, and subsequently regulating corresponding downstream pathways in bivalve invertebrates.
Collapse
Affiliation(s)
- Yihan Yu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Dandan Tian
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Sanghyok Ri
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China; College of Life Science, Kim Hyong Jik University of Education, Pyongyang, 99903, North Korea
| | - Tongchol Kim
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China; College of Life Science, Kim Hyong Jik University of Education, Pyongyang, 99903, North Korea
| | - Kwangjin Ju
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China; College of Aquaculture, Wonsan Fisheries University, Wonsan, 999093, North Korea
| | - Jiongming Zhang
- Zhejiang Mariculture Research Institute, Wenzhou, 325005, PR China
| | | | - Weixia Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
33
|
Ryner RF, Derera ID, Armbruster M, Kansara A, Sommer ME, Pirone A, Noubary F, Jacob M, Dulla CG. Cortical Parvalbumin-Positive Interneuron Development and Function Are Altered in the APC Conditional Knockout Mouse Model of Infantile and Epileptic Spasms Syndrome. J Neurosci 2023; 43:1422-1440. [PMID: 36717229 PMCID: PMC9987578 DOI: 10.1523/jneurosci.0572-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/14/2022] [Accepted: 12/22/2022] [Indexed: 02/01/2023] Open
Abstract
Infantile and epileptic spasms syndrome (IESS) is a childhood epilepsy syndrome characterized by infantile or late-onset spasms, abnormal neonatal EEG, and epilepsy. Few treatments exist for IESS, clinical outcomes are poor, and the molecular and circuit-level etiologies of IESS are not well understood. Multiple human IESS risk genes are linked to Wnt/β-catenin signaling, a pathway that controls developmental transcriptional programs and promotes glutamatergic excitation via β-catenin's role as a synaptic scaffold. We previously showed that deleting adenomatous polyposis coli (APC), a component of the β-catenin destruction complex, in excitatory neurons (APC cKO mice, APCfl/fl x CaMKIIαCre) increased β-catenin levels in developing glutamatergic neurons and led to infantile behavioral spasms, abnormal neonatal EEG, and adult epilepsy. Here, we tested the hypothesis that the development of GABAergic interneurons (INs) is disrupted in APC cKO male and female mice. IN dysfunction is implicated in human IESS, is a feature of other rodent models of IESS, and may contribute to the manifestation of spasms and seizures. We found that parvalbumin-positive INs (PV+ INs), an important source of cortical inhibition, were decreased in number, underwent disproportionate developmental apoptosis, and had altered dendrite morphology at P9, the peak of behavioral spasms. PV+ INs received excessive excitatory input, and their intrinsic ability to fire action potentials was reduced at all time points examined (P9, P14, P60). Subsequently, GABAergic transmission onto pyramidal neurons was uniquely altered in the somatosensory cortex of APC cKO mice at all ages, with both decreased IPSC input at P14 and enhanced IPSC input at P9 and P60. These results indicate that inhibitory circuit dysfunction occurs in APC cKOs and, along with known changes in excitation, may contribute to IESS-related phenotypes.SIGNIFICANCE STATEMENT Infantile and epileptic spasms syndrome (IESS) is a devastating epilepsy with limited treatment options and poor clinical outcomes. The molecular, cellular, and circuit disruptions that cause infantile spasms and seizures are largely unknown, but inhibitory GABAergic interneuron dysfunction has been implicated in rodent models of IESS and may contribute to human IESS. Here, we use a rodent model of IESS, the APC cKO mouse, in which β-catenin signaling is increased in excitatory neurons. This results in altered parvalbumin-positive GABAergic interneuron development and GABAergic synaptic dysfunction throughout life, showing that pathology arising in excitatory neurons can initiate long-term interneuron dysfunction. Our findings further implicate GABAergic dysfunction in IESS, even when pathology is initiated in other neuronal types.
Collapse
Affiliation(s)
- Rachael F Ryner
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
- Cell, Molecular, and Developmental Biology Graduate Program, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts 02111
| | - Isabel D Derera
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Anar Kansara
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Mary E Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Antonella Pirone
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Farzad Noubary
- Department of Health Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115
| | - Michele Jacob
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
34
|
Wildenberg G, Li H, Kasthuri N. The Development of Synapses in Mouse and Macaque Primary Sensory Cortices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528564. [PMID: 36824798 PMCID: PMC9949058 DOI: 10.1101/2023.02.15.528564] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
We report that the rate of synapse development in primary sensory cortices of mice and macaques is unrelated to lifespan, as was previously thought. We analyzed 28,084 synapses over multiple developmental time points in both species and find, instead, that net excitatory synapse development of mouse and macaque neurons primarily increased at similar rates in the first few postnatal months, and then decreased over a span of 1-1.5 years of age. The development of inhibitory synapses differed qualitatively across species. In macaques, net inhibitory synapses first increase and then decrease on excitatory soma at similar ages as excitatory synapses. In mice, however, such synapses are added throughout life. These findings contradict the long-held belief that the cycle of synapse formation and pruning occurs earlier in shorter-lived animals. Instead, our results suggest more nuanced rules, with the development of different types of synapses following different timing rules or different trajectories across species.
Collapse
Affiliation(s)
- Gregg Wildenberg
- Department of Neurobiology, The University of Chicago
- Argonne National Laboratory, Biosciences Division
| | - Hanyu Li
- Department of Neurobiology, The University of Chicago
- Argonne National Laboratory, Biosciences Division
| | - Narayanan Kasthuri
- Department of Neurobiology, The University of Chicago
- Argonne National Laboratory, Biosciences Division
| |
Collapse
|
35
|
Yang P, Davidson JO, Zhou KQ, Wilson R, Wassink G, Prasad JD, Bennet L, Gunn AJ, Dean JM. Therapeutic Hypothermia Attenuates Cortical Interneuron Loss after Cerebral Ischemia in Near-Term Fetal Sheep. Int J Mol Sci 2023; 24:ijms24043706. [PMID: 36835117 PMCID: PMC9962824 DOI: 10.3390/ijms24043706] [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: 12/22/2022] [Revised: 01/26/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Therapeutic hypothermia significantly improves outcomes after neonatal hypoxic-ischemic (HI) encephalopathy but is only partially protective. There is evidence that cortical inhibitory interneuron circuits are particularly vulnerable to HI and that loss of interneurons may be an important contributor to long-term neurological dysfunction in these infants. In the present study, we examined the hypothesis that the duration of hypothermia has differential effects on interneuron survival after HI. Near-term fetal sheep received sham ischemia or cerebral ischemia for 30 min, followed by cerebral hypothermia from 3 h after ischemia end and continued up to 48 h, 72 h, or 120 h recovery. Sheep were euthanized after 7 days for histology. Hypothermia up to 48 h recovery resulted in moderate neuroprotection of glutamate decarboxylase (GAD)+ and parvalbumin+ interneurons but did not improve survival of calbindin+ cells. Hypothermia up to 72 h recovery was associated with significantly increased survival of all three interneuron phenotypes compared with sham controls. By contrast, while hypothermia up to 120 h recovery did not further improve (or impair) GAD+ or parvalbumin+ neuronal survival compared with hypothermia up to 72 h, it was associated with decreased survival of calbindin+ interneurons. Finally, protection of parvalbumin+ and GAD+ interneurons, but not calbindin+ interneurons, with hypothermia was associated with improved recovery of electroencephalographic (EEG) power and frequency by day 7 after HI. The present study demonstrates differential effects of increasing the duration of hypothermia on interneuron survival after HI in near-term fetal sheep. These findings may contribute to the apparent preclinical and clinical lack of benefit of very prolonged hypothermia.
Collapse
|
36
|
Mueller-Buehl C, Wegrzyn D, Bauch J, Faissner A. Regulation of the E/I-balance by the neural matrisome. Front Mol Neurosci 2023; 16:1102334. [PMID: 37143468 PMCID: PMC10151766 DOI: 10.3389/fnmol.2023.1102334] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
In the mammalian cortex a proper excitatory/inhibitory (E/I) balance is fundamental for cognitive functions. Especially γ-aminobutyric acid (GABA)-releasing interneurons regulate the activity of excitatory projection neurons which form the second main class of neurons in the cortex. During development, the maturation of fast-spiking parvalbumin-expressing interneurons goes along with the formation of net-like structures covering their soma and proximal dendrites. These so-called perineuronal nets (PNNs) represent a specialized form of the extracellular matrix (ECM, also designated as matrisome) that stabilize structural synapses but prevent the formation of new connections. Consequently, PNNs are highly involved in the regulation of the synaptic balance. Previous studies revealed that the formation of perineuronal nets is accompanied by an establishment of mature neuronal circuits and by a closure of critical windows of synaptic plasticity. Furthermore, it has been shown that PNNs differentially impinge the integrity of excitatory and inhibitory synapses. In various neurological and neuropsychiatric disorders alterations of PNNs were described and aroused more attention in the last years. The following review gives an update about the role of PNNs for the maturation of parvalbumin-expressing interneurons and summarizes recent findings about the impact of PNNs in different neurological and neuropsychiatric disorders like schizophrenia or epilepsy. A targeted manipulation of PNNs might provide an interesting new possibility to indirectly modulate the synaptic balance and the E/I ratio in pathological conditions.
Collapse
|
37
|
Canetta SE, Holt ES, Benoit LJ, Teboul E, Sahyoun GM, Ogden RT, Harris AZ, Kellendonk C. Mature parvalbumin interneuron function in prefrontal cortex requires activity during a postnatal sensitive period. eLife 2022; 11:80324. [PMID: 36576777 PMCID: PMC9797185 DOI: 10.7554/elife.80324] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022] Open
Abstract
In their seminal findings, Hubel and Wiesel identified sensitive periods in which experience can exert lasting effects on adult visual cortical functioning and behavior via transient changes in neuronal activity during development. Whether comparable sensitive periods exist for non-sensory cortices, such as the prefrontal cortex, in which alterations in activity determine adult circuit function and behavior is still an active area of research. Here, using mice we demonstrate that inhibition of prefrontal parvalbumin (PV)-expressing interneurons during the juvenile and adolescent period, results in persistent impairments in adult prefrontal circuit connectivity, in vivo network function, and behavioral flexibility that can be reversed by targeted activation of PV interneurons in adulthood. In contrast, reversible suppression of PV interneuron activity in adulthood produces no lasting effects. These findings identify an activity-dependent sensitive period for prefrontal circuit maturation and highlight how abnormal PV interneuron activity during development alters adult prefrontal circuit function and cognitive behavior.
Collapse
Affiliation(s)
- Sarah E Canetta
- Department of Psychiatry, Columbia University Medical Center, New York, United States.,Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, United States
| | - Emma S Holt
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, United States
| | - Laura J Benoit
- Department of Psychiatry, Columbia University Medical Center, New York, United States.,Division of Molecular Therapeutics, New York Psychiatric Institute, New York, United States
| | - Eric Teboul
- Division of Molecular Therapeutics, New York Psychiatric Institute, New York, United States
| | - Gabriella M Sahyoun
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, United States
| | - R Todd Ogden
- Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center, New York, United States
| | - Alexander Z Harris
- Department of Psychiatry, Columbia University Medical Center, New York, United States.,Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, United States
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University Medical Center, New York, United States.,Division of Molecular Therapeutics, New York Psychiatric Institute, New York, United States.,Department of Molecular Pharmacology & Therapeutics, Columbia University Medical Center, New York, United States
| |
Collapse
|
38
|
Tran The J, Magistretti PJ, Ansermet F. The critical periods of cerebral plasticity: A key aspect in a dialog between psychoanalysis and neuroscience centered on the psychopathology of schizophrenia. Front Mol Neurosci 2022; 15:1057539. [PMID: 36590919 PMCID: PMC9795046 DOI: 10.3389/fnmol.2022.1057539] [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: 10/06/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022] Open
Abstract
Through research into the molecular and cellular mechanisms that occur during critical periods, recent experimental neurobiological data have brought to light the importance of early childhood. These have demonstrated that childhood and early environmental stimuli play a part not only in our subjective construction, but also in brain development; thus, confirming Freud's intuition regarding the central role of childhood and early experiences of the environment in our psychological development and our subjective outcomes. "Critical periods" of cerebral development represent temporal windows that mark favorable, but also circumscribed, moments in developmental cerebral plasticity. They also vary between different cortical areas. There are, therefore, strictly defined temporal periods for learning language, music, etc., after which this learning becomes more difficult, or even impossible, to acquire. Now, research into these critical periods can be seen as having a significant part to play in the interdisciplinary dialog between psychoanalysis and neurosciences with regard to the role of early experiences in the etiology of some psychopathological conditions. Research into the cellular and molecular mechanisms controlling the onset and end of these critical periods, notably controlled by the maturation of parvalbumin-expressing basket cells, have brought to light the presence of anomalies in the maturation of these neurons in patients with schizophrenia. Starting from these findings we propose revisiting the psychoanalytic theories on the etiology of psychosis from an interdisciplinary perspective. Our study works from the observation, common to both psychoanalysis and neurosciences, that experience leaves a trace; be it a "psychic" or a "synaptic" trace. Thus, we develop a hypothesis for an "absence of trace" in psychosis; reexamining psychosis through the prism of the biological theory of critical periods in plasticity.
Collapse
Affiliation(s)
- Jessica Tran The
- INSERM U1077 Neuropsychologie et Imagerie de la Mémoire Humaine, Caen, France,Ecole Pratique des Hautes Etudes, Université Paris Sciences et Lettres, Paris, France,UFR de Psychologie, Université de Caen Normandie, Caen, France,Centre Hospitalier Universitaire de Caen, Caen, France,Cyceron, Caen, France,Agalma Foundation Geneva, Chemin des Mines, Switzerland,*Correspondence: Jessica Tran The,
| | - Pierre J. Magistretti
- Agalma Foundation Geneva, Chemin des Mines, Switzerland,Brain Mind Institute, Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland,Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Francois Ansermet
- Agalma Foundation Geneva, Chemin des Mines, Switzerland,Département de Psychiatrie, Faculté de Médecine, Université de Genève, Geneva, Switzerland
| |
Collapse
|
39
|
Liaci C, Camera M, Zamboni V, Sarò G, Ammoni A, Parmigiani E, Ponzoni L, Hidisoglu E, Chiantia G, Marcantoni A, Giustetto M, Tomagra G, Carabelli V, Torelli F, Sala M, Yanagawa Y, Obata K, Hirsch E, Merlo GR. Loss of ARHGAP15 affects the directional control of migrating interneurons in the embryonic cortex and increases susceptibility to epilepsy. Front Cell Dev Biol 2022; 10:875468. [PMID: 36568982 PMCID: PMC9774038 DOI: 10.3389/fcell.2022.875468] [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: 02/14/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022] Open
Abstract
GTPases of the Rho family are components of signaling pathways linking extracellular signals to the control of cytoskeleton dynamics. Among these, RAC1 plays key roles during brain development, ranging from neuronal migration to neuritogenesis, synaptogenesis, and plasticity. RAC1 activity is positively and negatively controlled by guanine nucleotide exchange factors (GEFs), guanosine nucleotide dissociation inhibitors (GDIs), and GTPase-activating proteins (GAPs), but the specific role of each regulator in vivo is poorly known. ARHGAP15 is a RAC1-specific GAP expressed during development in a fraction of migrating cortical interneurons (CINs) and in the majority of adult CINs. During development, loss of ARHGAP15 causes altered directionality of the leading process of tangentially migrating CINs, along with altered morphology in vitro. Likewise, time-lapse imaging of embryonic CINs revealed a poorly coordinated directional control during radial migration, possibly due to a hyper-exploratory behavior. In the adult cortex, the observed defects lead to subtle alteration in the distribution of CALB2-, SST-, and VIP-positive interneurons. Adult Arhgap15-knock-out mice also show reduced CINs intrinsic excitability, spontaneous subclinical seizures, and increased susceptibility to the pro-epileptic drug pilocarpine. These results indicate that ARHGAP15 imposes a fine negative regulation on RAC1 that is required for morphological maturation and directional control during CIN migration, with consequences on their laminar distribution and inhibitory function.
Collapse
Affiliation(s)
- Carla Liaci
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Mattia Camera
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Valentina Zamboni
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Gabriella Sarò
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Alessandra Ammoni
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | | | - Luisa Ponzoni
- Neuroscience Institute, Consiglio Nazionale Ricerche, Milan, Italy
| | - Enis Hidisoglu
- Department of Drug Science, NIS Center, University of Turin, Turin, Italy
| | - Giuseppe Chiantia
- Department of Neuroscience and National Institute of Neuroscience, University of Turin, Turin, Italy
| | - Andrea Marcantoni
- Department of Drug Science, NIS Center, University of Turin, Turin, Italy
| | - Maurizio Giustetto
- Department of Neuroscience and National Institute of Neuroscience, University of Turin, Turin, Italy
| | - Giulia Tomagra
- Department of Drug Science, NIS Center, University of Turin, Turin, Italy
| | | | - Federico Torelli
- Institute for Physiology I, Medical Faculty, Albert-Ludwigs-University Freiburg, Freiburg, Germany,Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Mariaelvina Sala
- Neuroscience Institute, Consiglio Nazionale Ricerche, Milan, Italy
| | - Yuchio Yanagawa
- Department of Genetic Behavioral Neuroscience, Gunma University, Maebashi, Japan
| | | | - Emilio Hirsch
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Giorgio R. Merlo
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy,*Correspondence: Giorgio R. Merlo,
| |
Collapse
|
40
|
Ota M, Maki Y, Xu LY, Makino T. Prolonging effects of Valeriana fauriei root extract on pentobarbital-induced sleep in caffeine-induced insomnia model mice and the pharmacokinetics of its active ingredients under conditions of glycerol fatty acid ester as emulsifiers. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115625. [PMID: 35970315 DOI: 10.1016/j.jep.2022.115625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/01/2022] [Accepted: 08/06/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Valeriana plant roots have traditionally been used to treat central nervous system-related disorders in European countries. Among this genus, the Japanese Pharmacopoeia registers the dried roots of V. fauriei Briq. (VF). However, insufficient pharmacological data are available for this species. AIM OF THE STUDY We investigated the sedative effects of VF extract in a murine caffeine-induced insomnia model as well as the active ingredients and their pharmacokinetics to determine its basic pharmacological action mechanisms under conditions glycerol fatty acid ester is used as emulsifiers. MATERIALS AND METHODS A murine insomnia model was created by caffeine. Samples derived from the ethanol extract of VF were administered per oral (p.o.), and caffeine was injected intraperitoneally (i.p.). Pentobarbital was injected i.p. and the sleep latency and duration were measured. To confirm the mechanism of action of VF, flumazenil, a specific γ-aminobutyric acid receptor type A (GABAA receptor) antagonist, was administered (i.p.) immediately prior to the sample administration. We examined the pharmacokinetic profiles of the active ingredients in the plasma, brain, urine, and feces of mice after the administration (p.o and intravenous (i.v.)) of VF samples. RESULTS VF extract (5 g as VF/kg, p.o.) significantly shorten sleep latency and prolonged pentobarbital-induced sleep in caffeine-induced insomnia mice, partially mediated via the GABAergic nervous system, although a higher dose (10 g as VF/kg, p.o.) was required to exhibit the significant effects in normal mice. Kessyl glycol diacetate (KGD), the main constitutive compound in VF, did not shorten sleep latency but exhibited the same sleep prolonged effect at a dose related to VF extract. The concentration of kessyl glycol 8-acetate (KG8) in the plasma was higher than that of KGD in mice treated (p.o.) with VF extract. The profiles of brain concentrations of KGD and KG8 were similar to those in the plasma, and approximately 20% of those in the plasma were distributed throughout the brain. The excretions of KGD and KG8 in urine and feces was slightly detected, and an unknown large peak related to KG8 was detected in the urine of mice administered with VF extract by HPLC-MS/MS analysis. CONCLUSIONS VF exhibits more sedative effects under stressed conditions, such as insomnia, and the major active ingredients are KGD and its metabolite KG8, which are distributed from the blood circulation into the brain by simple diffusion. KG8 is further metabolized into other metabolites that are easily excreted in the urine.
Collapse
Affiliation(s)
- Misato Ota
- Department of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-Dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan; Kuki Sangyo Co., Ltd., 11 Onoe-cho, Yokkaichi-shi, Mie, 510-0059, Japan.
| | - Yasuhito Maki
- Department of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-Dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan; Kuki Sangyo Co., Ltd., 11 Onoe-cho, Yokkaichi-shi, Mie, 510-0059, Japan.
| | - Ling-Yu Xu
- Department of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-Dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan.
| | - Toshiaki Makino
- Department of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-Dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
41
|
Li X, Sun H, Zhu Y, Wang F, Wang X, Han L, Cui D, Luo D, Zhai Y, Zhuo L, Xu X, Yang J, Li Y. Dysregulation of prefrontal parvalbumin interneurons leads to adult aggression induced by social isolation stress during adolescence. Front Mol Neurosci 2022; 15:1010152. [PMID: 36267698 PMCID: PMC9577330 DOI: 10.3389/fnmol.2022.1010152] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Social isolation during the juvenile stage results in structural and functional impairment of the brain and deviant adult aggression. However, the specific subregions and cell types that underpin this deviant behavior are still largely unknown. Here, we found that adolescent social isolation led to a shortened latency to attack onset and extended the average attack time, accompanied by anxiety-like behavior and deficits in social preference in adult mice. However, when exposed to social isolation during adulthood, the mice did not show these phenotypes. We also found that the structural plasticity of prefrontal pyramidal neurons, including the dendritic complexity and spine ratio, was impaired in mice exposed to adolescent social isolation. The parvalbumin (PV) interneurons in the prefrontal infralimbic cortex (IL) are highly vulnerable to juvenile social isolation and exhibit decreased cell numbers and reduced activation in adulthood. Moreover, chemogenetic inactivation of IL-PV interneurons can mimic juvenile social isolation-induced deviant aggression and social preference. Conversely, artificial activation of IL-PV interneurons significantly attenuated deviant aggression and rescued social preference during adulthood in mice exposed to adolescent social isolation. These findings implicate juvenile social isolation-induced damage to IL-PV interneurons in long-term aggressive behavior in adulthood.
Collapse
Affiliation(s)
- Xinyang Li
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Huan Sun
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuanyuan Zhu
- Department of Neurobiology, Institute of Neurosciences, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Feidi Wang
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaodan Wang
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lin Han
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Dongqi Cui
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Danlei Luo
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yifang Zhai
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lixia Zhuo
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiangzhao Xu
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jian Yang
- Department of Diagnostic Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Jian Yang,
| | - Yan Li
- Department of Anesthesiology and Perioperative Medicine & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Yan Li,
| |
Collapse
|
42
|
Döhne N, Falck A, Janach GMS, Byvaltcev E, Strauss U. Interferon-γ augments GABA release in the developing neocortex via nitric oxide synthase/soluble guanylate cyclase and constrains network activity. Front Cell Neurosci 2022; 16:913299. [PMID: 36035261 PMCID: PMC9401097 DOI: 10.3389/fncel.2022.913299] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Interferon-γ (IFN-γ), a cytokine with neuromodulatory properties, has been shown to enhance inhibitory transmission. Because early inhibitory neurotransmission sculpts functional neuronal circuits, its developmental alteration may have grave consequences. Here, we investigated the acute effects of IFN-γ on γ-amino-butyric acid (GABA)ergic currents in layer 5 pyramidal neurons of the somatosensory cortex of rats at the end of the first postnatal week, a period of GABA-dependent cortical maturation. IFN-γ acutely increased the frequency and amplitude of spontaneous/miniature inhibitory postsynaptic currents (s/mIPSC), and this could not be reversed within 30 min. Neither the increase in amplitude nor frequency of IPSCs was due to upregulated interneuron excitability as revealed by current clamp recordings of layer 5 interneurons labeled with VGAT-Venus in transgenic rats. As we previously reported in more mature animals, IPSC amplitude increase upon IFN-γ activity was dependent on postsynaptic protein kinase C (PKC), indicating a similar activating mechanism. Unlike augmented IPSC amplitude, however, we did not consistently observe an increased IPSC frequency in our previous studies on more mature animals. Focusing on increased IPSC frequency, we have now identified a different activating mechanism-one that is independent of postsynaptic PKC but is dependent on inducible nitric oxide synthase (iNOS) and soluble guanylate cyclase (sGC). In addition, IFN-γ shifted short-term synaptic plasticity toward facilitation as revealed by a paired-pulse paradigm. The latter change in presynaptic function was not reproduced by the application of a nitric oxide donor. Functionally, IFN-γ-mediated alterations in GABAergic transmission overall constrained early neocortical activity in a partly nitric oxide-dependent manner as revealed by microelectrode array field recordings in brain slices analyzed with a spike-sorting algorithm. In summary, with IFN-γ-induced, NO-dependent augmentation of spontaneous GABA release, we have here identified a mechanism by which inflammation in the central nervous system (CNS) plausibly modulates neuronal development.
Collapse
Affiliation(s)
- Noah Döhne
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alice Falck
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gabriel M. S. Janach
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Egor Byvaltcev
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Neuroscience, Lobachevsky State, University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ulf Strauss
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
43
|
Altered Development of Prefrontal GABAergic Functions and Anxiety-like Behavior in Adolescent Offspring Induced by Prenatal Stress. Brain Sci 2022; 12:brainsci12081015. [PMID: 36009078 PMCID: PMC9406165 DOI: 10.3390/brainsci12081015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/18/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
Maternal stress can afflict fetal brain development, putting the offspring at risk of cognitive deficits, including anxiety. The prefrontal cortex (PFC), a protracted maturing region, is notably affected by prenatal stress (PS). However, it remains unclear how PS interferes with the maturation of the GABAergic system, considering its functional adjustment in the PFC during adolescence. The present study thus investigated the long-lasting consequences of PS on the prefrontal GABAergic functions of adolescent offspring. Pregnant Sprague–Dawley rats were divided into controls and the PS group, which underwent restraint stress during the last week of gestation. Male pups from postnatal days (PND) 40–42 were submitted to the elevated plus maze (EPM) test. Proteins essentially involved in GABAergic signaling were then examined in PFC tissues, including the K+-Cl− cotransporter (KCC2), Na+-K+-Cl− cotransporter (NKCC1), α1 and α5 subunits of GABA type A receptors (GABAA receptors), and parvalbumin (PV), along with cAMP response element-binding protein phosphorylation (pCREB), which reacts in the plasticity regulation of PV-positive interneurons. The results revealed that the higher anxiety-like behavior of PS adolescent rats concurred with the significant decreases of the KCC2 and α1 subunits, with PV- and pCREB-lowered levels. The findings suggested that PS disrupts the continuance of PFC maturity by reducing the essential elements of GABAergic functions. These changes likely underlie the anxiety emerging in adolescence, possibly progressing to mental disorders.
Collapse
|
44
|
Arutiunian V, Arcara G, Buyanova I, Gomozova M, Dragoy O. The age-related changes in 40 Hz Auditory Steady-State Response and sustained Event-Related Fields to the same amplitude-modulated tones in typically developing children: A magnetoencephalography study. Hum Brain Mapp 2022; 43:5370-5383. [PMID: 35833318 PMCID: PMC9812253 DOI: 10.1002/hbm.26013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 01/15/2023] Open
Abstract
Recent studies have revealed that gamma-band oscillatory and transient evoked potentials may change with age during childhood. It is hypothesized that these changes can be associated with a maturation of GABAergic neurotransmission and, subsequently, the age-related changes of excitation-inhibition balance in the neural circuits. One of the reliable paradigms for investigating these effects in the auditory cortex is 40 Hz Auditory Steady-State Response (ASSR), where participants are presented with the periodic auditory stimuli. It is known that such stimuli evoke two types of responses in magnetoencephalography (MEG)-40 Hz steady-state gamma response (or 40 Hz ASSR) and auditory evoked response called sustained Event-Related Field (ERF). Although several studies have been conducted in children, focusing on the changes of 40 Hz ASSR with age, almost nothing is known about the age-related changes of the sustained ERF to the same periodic stimuli and their relationships with changes in the gamma strength. Using MEG, we investigated the association between 40 Hz steady-state gamma response and sustained ERF response to the same stimuli and also their age-related changes in the group of 30 typically developing 7-to-12-year-old children. The results revealed a tight relationship between 40 Hz ASSR and ERF, indicating that the age-related increase in strength of 40 Hz ASSR was associated with the age-related decrease of the amplitude of ERF. These effects were discussed in the light of the maturation of the GABAergic system and excitation-inhibition balance development, which may contribute to the changes in ASSR and ERF.
Collapse
Affiliation(s)
| | | | | | | | - Olga Dragoy
- Center for Language and BrainHSE UniversityMoscowRussia,Institute of LinguisticsRussian Academy of SciencesMoscowRussia
| |
Collapse
|
45
|
Utagawa EC, Moreno DG, Schafernak KT, Arva NC, Malek-Ahmadi MH, Mufson EJ, Perez SE. Neurogenesis and neuronal differentiation in the postnatal frontal cortex in Down syndrome. Acta Neuropathol Commun 2022; 10:86. [PMID: 35676735 PMCID: PMC9175369 DOI: 10.1186/s40478-022-01385-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/17/2022] [Indexed: 12/17/2022] Open
Abstract
Although Down syndrome (DS), the most common developmental genetic cause of intellectual disability, displays proliferation and migration deficits in the prenatal frontal cortex (FC), a knowledge gap exists on the effects of trisomy 21 upon postnatal cortical development. Here, we examined cortical neurogenesis and differentiation in the FC supragranular (SG, II/III) and infragranular (IG, V/VI) layers applying antibodies to doublecortin (DCX), non-phosphorylated heavy-molecular neurofilament protein (NHF, SMI-32), calbindin D-28K (Calb), calretinin (Calr), and parvalbumin (Parv), as well as β-amyloid (APP/Aβ and Aβ1-42) and phospho-tau (CP13 and PHF-1) in autopsy tissue from age-matched DS and neurotypical (NTD) subjects ranging from 28-weeks (wk)-gestation to 3 years of age. Thionin, which stains Nissl substance, revealed disorganized cortical cellular lamination including a delayed appearance of pyramidal cells until 44 wk of age in DS compared to 28 wk in NTD. SG and IG DCX-immunoreactive (-ir) cells were only visualized in the youngest cases until 83 wk in NTD and 57 wk DS. Strong SMI-32 immunoreactivity was observed in layers III and V pyramidal cells in the oldest NTD and DS cases with few appearing as early as 28 wk of age in layer V in NTD. Small Calb-ir interneurons were seen in younger NTD and DS cases compared to Calb-ir pyramidal cells in older subjects. Overall, a greater number of Calb-ir cells were detected in NTD, however, the number of Calr-ir cells were comparable between groups. Diffuse APP/Aβ immunoreactivity was found at all ages in both groups. Few young cases from both groups presented non-neuronal granular CP13 immunoreactivity in layer I. Stronger correlations between brain weight, age, thionin, DCX, and SMI-32 counts were found in NTD. These findings suggest that trisomy 21 affects postnatal FC lamination, neuronal migration/neurogenesis and differentiation of projection neurons and interneurons that likely contribute to cognitive impairment in DS.
Collapse
Affiliation(s)
- Emma C Utagawa
- Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | - David G Moreno
- Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | - Kristian T Schafernak
- Department of Pathology and Laboratory Medicine, Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| | - Nicoleta C Arva
- Department of Pathology and Laboratory Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave, Chicago, IL, 60611, USA
| | | | - Elliott J Mufson
- Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | - Sylvia E Perez
- Department of Translational Neuroscience, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
46
|
Cellular and genetic drivers of RNA editing variation in the human brain. Nat Commun 2022; 13:2997. [PMID: 35637184 PMCID: PMC9151768 DOI: 10.1038/s41467-022-30531-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 05/02/2022] [Indexed: 11/08/2022] Open
Abstract
Posttranscriptional adenosine-to-inosine modifications amplify the functionality of RNA molecules in the brain, yet the cellular and genetic regulation of RNA editing is poorly described. We quantify base-specific RNA editing across three major cell populations from the human prefrontal cortex: glutamatergic neurons, medial ganglionic eminence-derived GABAergic neurons, and oligodendrocytes. We identify more selective editing and hyper-editing in neurons relative to oligodendrocytes. RNA editing patterns are highly cell type-specific, with 189,229 cell type-associated sites. The cellular specificity for thousands of sites is confirmed by single nucleus RNA-sequencing. Importantly, cell type-associated sites are enriched in GTEx RNA-sequencing data, edited ~twentyfold higher than all other sites, and variation in RNA editing is largely explained by neuronal proportions in bulk brain tissue. Finally, we uncover 661,791 cis-editing quantitative trait loci across thirteen brain regions, including hundreds with cell type-associated features. These data reveal an expansive repertoire of highly regulated RNA editing sites across human brain cell types and provide a resolved atlas linking cell types to editing variation and genetic regulatory effects. Here the authors provide a deep catalogue of cell-specific A-to-I editing sites in the human cortex. Thousands of sites are enriched and elevated in neurons relative to glial cells, and are genetically regulated across multiple brain regions.
Collapse
|
47
|
Neonatal Oxidative Stress Impairs Cortical Synapse Formation and GABA Homeostasis in Parvalbumin-Expressing Interneurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8469756. [PMID: 35663195 PMCID: PMC9159830 DOI: 10.1155/2022/8469756] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/08/2022] [Indexed: 11/28/2022]
Abstract
Neonatal brain injury is often caused by preterm birth. Brain development is vulnerable to increased environmental stress, including oxidative stress challenges. Due to a premature change of the fetal living environment from low oxygen in utero into postnatal high-oxygen room air conditions ex utero, the immature preterm brain is exposed to a relative hyperoxia, which can induce oxidative stress and impair neuronal cell development. To simulate the drastic increase of oxygen exposure in the immature brain, 5-day-old C57BL/6 mice were exposed to hyperoxia (80% oxygen) for 48 hours or kept in room air (normoxia, 21% oxygen) and mice were analyzed for maturational alterations of cortical GABAergic interneurons. As a result, oxidative stress was indicated by elevated tyrosine nitration of proteins. We found perturbation of perineuronal net formation in line with decreased density of parvalbumin-expressing (PVALB) cortical interneurons in hyperoxic mice. Moreover, maturational deficits of cortical PVALB+ interneurons were obtained by decreased glutamate decarboxylase 67 (GAD67) protein expression in Western blot analysis and lower gamma-aminobutyric acid (GABA) fluorescence intensity in immunostaining. Hyperoxia-induced oxidative stress affected cortical synaptogenesis by decreasing synapsin 1, synapsin 2, and synaptophysin expression. Developmental delay of synaptic marker expression was demonstrated together with decreased PI3K-signaling as a pathway being involved in synaptogenesis. These results elucidate that neonatal oxidative stress caused by increased oxygen exposure can lead to GABAergic interneuron damage which may serve as an explanation for the high incidence of psychiatric and behavioral alterations found in preterm infants.
Collapse
|
48
|
Manyukhina VO, Prokofyev AO, Galuta IA, Goiaeva DE, Obukhova TS, Schneiderman JF, Altukhov DI, Stroganova TA, Orekhova EV. Globally elevated excitation-inhibition ratio in children with autism spectrum disorder and below-average intelligence. Mol Autism 2022; 13:20. [PMID: 35550191 PMCID: PMC9102291 DOI: 10.1186/s13229-022-00498-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/25/2022] [Indexed: 12/04/2022] Open
Abstract
Background Altered neuronal excitation–inhibition (E–I) balance is strongly implicated in ASD. However, it is not known whether the direction and degree of changes in the E–I ratio in individuals with ASD correlates with intellectual disability often associated with this developmental disorder. The spectral slope of the aperiodic 1/f activity reflects the E–I balance at the scale of large neuronal populations and may uncover its putative alternations in individuals with ASD with and without intellectual disability. Methods Herein, we used magnetoencephalography (MEG) to test whether the 1/f slope would differentiate ASD children with average and below–average (< 85) IQ. MEG was recorded at rest with eyes open/closed in 49 boys with ASD aged 6–15 years with IQ ranging from 54 to 128, and in 49 age-matched typically developing (TD) boys. The cortical source activity was estimated using the beamformer approach and individual brain models. We then extracted the 1/f slope by fitting a linear function to the log–log-scale power spectra in the high-frequency range. Results The global 1/f slope averaged over all cortical sources demonstrated high rank-order stability between the two conditions. Consistent with previous research, it was steeper in the eyes-closed than in the eyes-open condition and flattened with age. Regardless of condition, children with ASD and below-average IQ had flatter slopes than either TD or ASD children with average or above-average IQ. These group differences could not be explained by differences in signal-to-noise ratio or periodic (alpha and beta) activity. Limitations Further research is needed to find out whether the observed changes in E–I ratios are characteristic of children with below-average IQ of other diagnostic groups. Conclusions The atypically flattened spectral slope of aperiodic activity in children with ASD and below-average IQ suggests a shift of the global E–I balance toward hyper-excitation. The spectral slope can provide an accessible noninvasive biomarker of the E–I ratio for making objective judgments about treatment effectiveness in people with ASD and comorbid intellectual disability. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-022-00498-2.
Collapse
Affiliation(s)
- Viktoriya O Manyukhina
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation.,Department of Psychology, National Research University Higher School of Economics, Moscow, Russian Federation
| | - Andrey O Prokofyev
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Ilia A Galuta
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Dzerassa E Goiaeva
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Tatiana S Obukhova
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Justin F Schneiderman
- MedTech West and the Institute of Neuroscience and Physiology, Sahlgrenska Academy, The University of Gothenburg, Gothenburg, Sweden
| | - Dmitrii I Altukhov
- Department of Psychology, National Research University Higher School of Economics, Moscow, Russian Federation
| | - Tatiana A Stroganova
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Elena V Orekhova
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation.
| |
Collapse
|
49
|
Cattane N, Vernon AC, Borsini A, Scassellati C, Endres D, Capuron L, Tamouza R, Benros ME, Leza JC, Pariante CM, Riva MA, Cattaneo A. Preclinical animal models of mental illnesses to translate findings from the bench to the bedside: Molecular brain mechanisms and peripheral biomarkers associated to early life stress or immune challenges. Eur Neuropsychopharmacol 2022; 58:55-79. [PMID: 35235897 DOI: 10.1016/j.euroneuro.2022.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
Animal models are useful preclinical tools for studying the pathogenesis of mental disorders and the effectiveness of their treatment. While it is not possible to mimic all symptoms occurring in humans, it is however possible to investigate the behavioral, physiological and neuroanatomical alterations relevant for these complex disorders in controlled conditions and in genetically homogeneous populations. Stressful and infection-related exposures represent the most employed environmental risk factors able to trigger or to unmask a psychopathological phenotype in animals. Indeed, when occurring during sensitive periods of brain maturation, including pre, postnatal life and adolescence, they can affect the offspring's neurodevelopmental trajectories, increasing the risk for mental disorders. Not all stressed or immune challenged animals, however, develop behavioral alterations and preclinical animal models can explain differences between vulnerable or resilient phenotypes. Our review focuses on different paradigms of stress (prenatal stress, maternal separation, social isolation and social defeat stress) and immune challenges (immune activation in pregnancy) and investigates the subsequent alterations in several biological and behavioral domains at different time points of animals' life. It also discusses the "double-hit" hypothesis where an initial early adverse event can prime the response to a second negative challenge. Interestingly, stress and infections early in life induce the activation of the hypothalamic-pituitary-adrenal (HPA) axis, alter the levels of neurotransmitters, neurotrophins and pro-inflammatory cytokines and affect the functions of microglia and oxidative stress. In conclusion, animal models allow shedding light on the pathophysiology of human mental illnesses and discovering novel molecular drug targets for personalized treatments.
Collapse
Affiliation(s)
- Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, United Kingdom
| | - Alessandra Borsini
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King's College London, United Kingdom
| | - Catia Scassellati
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Dominique Endres
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lucile Capuron
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Ryad Tamouza
- Département Medico-Universitaire de Psychiatrie et d'Addictologie (DMU ADAPT), Laboratoire Neuro-psychiatrie translationnelle, AP-HP, UniversitéParis Est Créteil, INSERM U955, IMRB, Hôpital Henri Mondor, Fondation FondaMental, F-94010 Créteil, France
| | - Michael Eriksen Benros
- Biological and Precision Psychiatry, Copenhagen Research Centre for Mental Health, Copenhagen University Hospital, Gentofte Hospitalsvej 15, 4th floor, 2900 Hellerup, Denmark; Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Juan C Leza
- Department of Pharmacology & Toxicology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Hospital 12 de Octubre (i+12), IUIN-UCM. Spain
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King's College London, United Kingdom
| | - Marco A Riva
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy.
| |
Collapse
|
50
|
Neurocan regulates vulnerability to stress and the anti-depressant effect of ketamine in adolescent rats. Mol Psychiatry 2022; 27:2522-2532. [PMID: 35264728 DOI: 10.1038/s41380-022-01495-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 12/13/2022]
Abstract
Depression is more prevalent among adolescents than adults, but the underlying mechanisms remain largely unknown. Using a subthreshold chronic stress model, here we show that developmentally regulated expressions of the perineuronal nets (PNNs), and one of the components, Neurocan in the prelimbic cortex (PrL) are important for the vulnerability to stress and depressive-like behaviors in both adolescent and adult rats. Reduction of PNNs or Neurocan with pharmacological or viral methods to mimic the expression of PNNs in the PrL during adolescence compromised resilience to stress in adult rats, while virally mediated overexpression of Neurocan reversed vulnerability to stress in adolescent rats. Ketamine, a recent-approved drug for treatment-resistant depression rescued impaired function of Parvalbumin-positive neurons function, increased expression of PNNs in the PrL, and reversed depressive-like behaviors in adolescent rats. Furthermore, we show that Neurocan mediates the anti-depressant effect of ketamine, virally mediated reduction of Neurocan in the PrL abolished the anti-depressant effect of ketamine in adolescent rats. Our findings show an important role of Neurocan in depression in adolescence, and suggest a novel mechanism for the anti-depressant effect of ketamine.
Collapse
|