1
|
Chen X, Wang YJ, Mu TW. Proteostasis regulation of GABA A receptors in neuronal function and disease. Biomed Pharmacother 2025; 186:117992. [PMID: 40112516 DOI: 10.1016/j.biopha.2025.117992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
The γ-aminobutyric acid type A receptors (GABAARs) are ligand-gated anion channels that mediate fast inhibitory neurotransmission in the mammalian central nervous system. GABAARs form heteropentameric assemblies comprising two α1, two β2, and one γ2 subunits as the most common subtype in mammalian brains. Proteostasis regulation of GABAARs involves subunit folding within the endoplasmic reticulum, assembling into heteropentamers, receptor trafficking to the cell surface, and degradation of terminally misfolded subunits. As GABAARs are surface proteins, their trafficking to the plasma membrane is critical for proper receptor function. Thus, variants in the genes encoding GABAARs that disrupt proteostasis result in various neurodevelopmental disorders, ranging from intellectual disability to idiopathic generalized epilepsy. This review summarizes recent progress about how the proteostasis network regulates protein folding, assembly, degradation, trafficking, and synaptic clustering of GABAARs. Additionally, emerging pharmacological approaches that restore proteostasis of pathogenic GABAAR variants are presented, providing a promising strategy to treat related neurological diseases.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
2
|
Lu MQ, Shi ZG, Shang J, Gao L, Gao L, Gao WJ. ChangPu YuJin Tang improves Tourette disorder symptoms by modulating amino acid neurotransmitters in IDPN model rats. Metab Brain Dis 2024; 39:1543-1558. [PMID: 39312065 DOI: 10.1007/s11011-024-01411-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 08/09/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Changpu Yujin Tang(CPYJT), a Chinese herbal compound, is an effective therapeutic strategy for pediatric patients with Tourette disorder (TD). Therefore, this work aims to investigate the therapeutic mechanisms of CPYJT. METHODS Behavioral and cellular ultrastructural evaluation of the therapeutic effects of CPYJT in TD model rats. Colorimetric methods, reverse transcription‑quantitative PCR, and Western Blot were used to measure the altered levels of GLU, GABA, and the levels of VGLUT1, GLUD1, GABRA3, and GAD65 in the cortex, striatum, and thalamus of the TD model rats after 7, 14, 21, and 28 days of CPYJT administration. RESULTS CPYJT significantly reduced stereotypic behavior and motor behavior scores in TD model rats. CPYJT ameliorates myelin structural damage in TD model rat neuronal cells. CPYJT decreased GLU content, elevated GABA content, decreased GLUD1 and VGLUT1 levels, and elevated GAD65 and GABRA3 levels in TD model rats' cortex, striatum, and thalamus. CPYJT has different regulatory time points in the cortex, striatum, and thalamus for critical factors of amino acid-based neurotransmission. CONCLUSION CPYJT protects behavioral and structural damage of neuronal cells in multiple brain regions in TD model rats.
Collapse
Affiliation(s)
- Man-Qi Lu
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, shanghai, 200000, China
| | - Zheng-Gang Shi
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China.
| | - Jing Shang
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| | - Lü Gao
- Shanxi University Of Chinese Medicine Third Clinical Medical College Pediatric Teaching and Research Department, Taiyuan, 140100, Shanxi, China
| | - Lei Gao
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| | - Wei-Jiao Gao
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| |
Collapse
|
3
|
Ji J, Chao H, Chen H, Liao J, Shi W, Ye Y, Wang T, You Y, Liu N, Ji J, Petretto E. Decoding frontotemporal and cell-type-specific vulnerabilities to neuropsychiatric disorders and psychoactive drugs. Open Biol 2024; 14:240063. [PMID: 38864245 DOI: 10.1098/rsob.240063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/29/2024] [Indexed: 06/13/2024] Open
Abstract
Frontotemporal lobe abnormalities are linked to neuropsychiatric disorders and cognition, but the role of cellular heterogeneity between temporal lobe (TL) and frontal lobe (FL) in the vulnerability to genetic risk factors remains to be elucidated. We integrated single-nucleus transcriptome analysis in 'fresh' human FL and TL with genetic susceptibility, gene dysregulation in neuropsychiatric disease and psychoactive drug response data. We show how intrinsic differences between TL and FL contribute to the vulnerability of specific cell types to both genetic risk factors and psychoactive drugs. Neuronal populations, specifically PVALB neurons, were most highly vulnerable to genetic risk factors for psychiatric disease. These psychiatric disease-associated genes were mostly upregulated in the TL, and dysregulated in the brain of patients with obsessive-compulsive disorder, bipolar disorder and schizophrenia. Among these genes, GRIN2A and SLC12A5, implicated in schizophrenia and bipolar disorder, were significantly upregulated in TL PVALB neurons and in psychiatric disease patients' brain. PVALB neurons from the TL were twofold more vulnerable to psychoactive drugs than to genetic risk factors, showing the influence and specificity of frontotemporal lobe differences on cell vulnerabilities. These studies provide a cell type resolved map of the impact of brain regional differences on cell type vulnerabilities in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jiatong Ji
- Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University (CPU), Nanjing, Jiangsu 211198, People's Republic of China
| | - Honglu Chao
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Huimei Chen
- Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University (CPU), Nanjing, Jiangsu 211198, People's Republic of China
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jun Liao
- High Performance Computing Center, School of Science, China Pharmaceutical University (CPU), Nanjing, Jiangsu 211198, People's Republic of China
| | - Wenqian Shi
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Yangfan Ye
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Tian Wang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Yongping You
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Ning Liu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Jing Ji
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
- Department of Neurosurgery, The Affiliated Kizilsu Kirghiz Autonomous Prefecture People's Hospital of Nanjing Medical University, Xinjiang, Artux 845350, People's Republic of China
- Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Enrico Petretto
- Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University (CPU), Nanjing, Jiangsu 211198, People's Republic of China
- Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
4
|
Lichtenfeld MJ, Mulvey AG, Nejat H, Xiong YS, Carlson BM, Mitchell BA, Mendoza-Halliday D, Westerberg JA, Desimone R, Maier A, Kaas JH, Bastos AM. The laminar organization of cell types in macaque cortex and its relationship to neuronal oscillations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587084. [PMID: 38585801 PMCID: PMC10996711 DOI: 10.1101/2024.03.27.587084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The canonical microcircuit (CMC) has been hypothesized to be the fundamental unit of information processing in cortex. Each CMC unit is thought to be an interconnected column of neurons with specific connections between excitatory and inhibitory neurons across layers. Recently, we identified a conserved spectrolaminar motif of oscillatory activity across the primate cortex that may be the physiological consequence of the CMC. The spectrolaminar motif consists of local field potential (LFP) gamma-band power (40-150 Hz) peaking in superficial layers 2 and 3 and alpha/beta-band power (8-30 Hz) peaking in deep layers 5 and 6. Here, we investigate whether specific conserved cell types may produce the spectrolaminar motif. We collected laminar histological and electrophysiological data in 11 distinct cortical areas spanning the visual hierarchy: V1, V2, V3, V4, TEO, MT, MST, LIP, 8A/FEF, PMD, and LPFC (area 46), and anatomical data in DP and 7A. We stained representative slices for the three main inhibitory subtypes, Parvalbumin (PV), Calbindin (CB), and Calretinin (CR) positive neurons, as well as pyramidal cells marked with Neurogranin (NRGN). We found a conserved laminar structure of PV, CB, CR, and pyramidal cells. We also found a consistent relationship between the laminar distribution of inhibitory subtypes with power in the local field potential. PV interneuron density positively correlated with gamma (40-150 Hz) power. CR and CB density negatively correlated with alpha (8-12 Hz) and beta (13-30 Hz) oscillations. The conserved, layer-specific pattern of inhibition and excitation across layers is therefore likely the anatomical substrate of the spectrolaminar motif. Significance Statement Neuronal oscillations emerge as an interplay between excitatory and inhibitory neurons and underlie cognitive functions and conscious states. These oscillations have distinct expression patterns across cortical layers. Does cellular anatomy enable these oscillations to emerge in specific cortical layers? We present a comprehensive analysis of the laminar distribution of the three main inhibitory cell types in primate cortex (Parvalbumin, Calbindin, and Calretinin positive) and excitatory pyramidal cells. We found a canonical relationship between the laminar anatomy and electrophysiology in 11 distinct primate areas spanning from primary visual to prefrontal cortex. The laminar anatomy explained the expression patterns of neuronal oscillations in different frequencies. Our work provides insight into the cortex-wide cellular mechanisms that generate neuronal oscillations in primates.
Collapse
|
5
|
Zhang WJ, Shi HZ, Guo MN, Xu LF, Zhai HR, Liu ZZ, Zhu YQ, Zhang WN, Wang J. PGC-1α regulates critical period onset/closure, mediating cortical plasticity. Front Mol Neurosci 2023; 16:1149906. [PMID: 37822967 PMCID: PMC10563514 DOI: 10.3389/fnmol.2023.1149906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
Peroxisome proliferator-activated receptor PPARγ coactivator-α (PGC-1α) is concentrated in inhibitory interneurons and plays a vital role in neuropsychiatric diseases. We previously reported some characteristic features of schizophrenia (SZ) in GABAergic neuron-specific Pgc-1alpha knockout (KO) mice (Dlx5/6-Cre: Pgc-1alphaf/f). However, there is a fundamental gap in the molecular mechanism by which the Pgc-1alpha gene is involved in the neurobehavioral abnormalities of SZ. The loss of critical period (CP) triggers-maturations of parvalbumin interneurons (PVIs) and brakes-and the formation of perineuronal nets (PNNs) implicates mistimed trajectories during adult brain development. In this study, using the Pgc-1alpha KO mouse line, we investigated the association of Pgc-1alpha gene deletion with SZ-like behavioral deficits, PVI maturation, PNN integrity and synaptic ultrastructure. These findings suggest that Pgc-1alpha gene deletion resulted in a failure of CP onset and closure, thereby prolonging cortical plasticity timing. To determine whether the manipulation of the PNN structure is a potential method of altering neuronal plasticity, GM6001, a broad-spectrum matrix metalloproteinase (MMP)-inhibitor was applied. Here we confirmed that the treatment could effectively correct the CP plasticity window and ameliorate the synaptic ultrastructure in the Pgc-1alpha KO brain. Moreover, the intervention effect on neuronal plasticity was followed by the rescue of short-term habituation deficits and the mitigation of aberrant salience, which are some characteristic features of SZ. Taken collectively, these findings suggest that the role of PGC-1α in regulating cortical plasticity is mediated, at least partially, through the regulation of CP onset/closure. Strategically introduced reinforcement of molecular brakes may be a novel preventive therapy for psychiatric disorders associated with PGC-1α dysregulation.
Collapse
Affiliation(s)
- Wei-Jun Zhang
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hou-Zhen Shi
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mei-Na Guo
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Long-Fei Xu
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hong-Ru Zhai
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zi-Zhong Liu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yong-Qiang Zhu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei-Ning Zhang
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jia Wang
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Zhenjiang Jieshengrui Biotechnology Co., Ltd., Zhenjiang, Jiangsu, China
| |
Collapse
|
6
|
Kositsyn YM, de Abreu MS, Kolesnikova TO, Lagunin AA, Poroikov VV, Harutyunyan HS, Yenkoyan KB, Kalueff AV. Towards Novel Potential Molecular Targets for Antidepressant and Antipsychotic Pharmacotherapies. Int J Mol Sci 2023; 24:ijms24119482. [PMID: 37298431 DOI: 10.3390/ijms24119482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023] Open
Abstract
Depression and schizophrenia are two highly prevalent and severely debilitating neuropsychiatric disorders. Both conventional antidepressant and antipsychotic pharmacotherapies are often inefficient clinically, causing multiple side effects and serious patient compliance problems. Collectively, this calls for the development of novel drug targets for treating depressed and schizophrenic patients. Here, we discuss recent translational advances, research tools and approaches, aiming to facilitate innovative drug discovery in this field. Providing a comprehensive overview of current antidepressants and antipsychotic drugs, we also outline potential novel molecular targets for treating depression and schizophrenia. We also critically evaluate multiple translational challenges and summarize various open questions, in order to foster further integrative cross-discipline research into antidepressant and antipsychotic drug development.
Collapse
Affiliation(s)
- Yuriy M Kositsyn
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 197341, Russia
- Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory 354340, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny 197758, Russia
| | - Murilo S de Abreu
- Neuroscience Group, Moscow Institute of Physics and Technology, Moscow 115184, Russia
| | - Tatiana O Kolesnikova
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 197341, Russia
- Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory 354340, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Vivarium, Ural Federal University, Yekaterinburg 620049, Russia
| | - Alexey A Lagunin
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Vladimir V Poroikov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow 119121, Russia
| | - Hasmik S Harutyunyan
- Neuroscience Laboratory, COBRAIN Center, Yerevan State Medical University Named after M. Heratsi, Yerevan 0025, Armenia
- Department of Biochemistry, Yerevan State Medical University Named after M. Heratsi, Yerevan 0025, Armenia
| | - Konstantin B Yenkoyan
- Neuroscience Laboratory, COBRAIN Center, Yerevan State Medical University Named after M. Heratsi, Yerevan 0025, Armenia
- Department of Biochemistry, Yerevan State Medical University Named after M. Heratsi, Yerevan 0025, Armenia
| | - Allan V Kalueff
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 197341, Russia
- Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory 354340, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny 197758, Russia
- Neuroscience Group, Moscow Institute of Physics and Technology, Moscow 115184, Russia
- Vivarium, Ural Federal University, Yekaterinburg 620049, Russia
- Neuroscience Laboratory, COBRAIN Center, Yerevan State Medical University Named after M. Heratsi, Yerevan 0025, Armenia
| |
Collapse
|
7
|
Griego E, Segura-Villalobos D, Lamas M, Galván EJ. Maternal immune activation increases excitability via downregulation of A-type potassium channels and reduces dendritic complexity of hippocampal neurons of the offspring. Brain Behav Immun 2022; 105:67-81. [PMID: 35803480 DOI: 10.1016/j.bbi.2022.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 11/29/2022] Open
Abstract
The epidemiological association between bacterial or viral maternal infections during pregnancy and increased risk for developing psychiatric disorders in offspring is well documented. Numerous rodent and non-human primate studies of viral- or, to a lesser extent, bacterial-induced maternal immune activation (MIA) have documented a series of neurological alterations that may contribute to understanding the pathophysiology of schizophrenia and autism spectrum disorders. Long-term neuronal and behavioral alterations are now ascribed to the effect of maternal proinflammatory cytokines rather than the infection itself. However, detailed electrophysiological alterations in brain areas relevant to psychiatric disorders, such as the dorsal hippocampus, are lacking in response to bacterial-induced MIA. This study determined if electrophysiological and morphological alterations converge in CA1 pyramidal cells (CA1 PC) from the dorsal hippocampus in bacterial-induced MIA offspring. A series of changes in the functional expression of K+ and Na+ ion channels altered the passive and active membrane properties and triggered hyperexcitability of CA1 PC. Contributing to the hyperexcitability, the somatic A-type potassium current (IA) was decreased in MIA CA1 PC. Likewise, the spontaneous glutamatergic and GABAergic inputs were dysregulated and biased toward increased excitation, thereby reshaping the excitation-inhibition balance. Consistent with these findings, the dendritic branching complexity of MIA CA1 PC was reduced. Together, these morphophysiological alterations modify CA1 PC computational capabilities and contribute to explaining cellular alterations that may underlie the cognitive symptoms of MIA-associated psychiatric disorders.
Collapse
Affiliation(s)
- Ernesto Griego
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico
| | | | - Mónica Lamas
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico
| | - Emilio J Galván
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico.
| |
Collapse
|
8
|
Biazzo M, Allegra M, Deidda G. Clostridioides difficile and neurological disorders: New perspectives. Front Neurosci 2022; 16:946601. [PMID: 36203814 PMCID: PMC9530032 DOI: 10.3389/fnins.2022.946601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Despite brain physiological functions or pathological dysfunctions relying on the activity of neuronal/non-neuronal populations, over the last decades a plethora of evidence unraveled the essential contribution of the microbial populations living and residing within the gut, called gut microbiota. The gut microbiota plays a role in brain (dys)functions, and it will become a promising valuable therapeutic target for several brain pathologies. In the present mini-review, after a brief overview of the role of gut microbiota in normal brain physiology and pathology, we focus on the role of the bacterium Clostridioides difficile, a pathogen responsible for recurrent and refractory infections, in people with neurological diseases, summarizing recent correlative and causative evidence in the scientific literature and highlighting the potential of microbiota-based strategies targeting this pathogen to ameliorate not only gastrointestinal but also the neurological symptoms.
Collapse
Affiliation(s)
- Manuele Biazzo
- The BioArte Limited, Life Sciences Park, San Gwann, Malta
- SienabioACTIVE, University of Siena, Siena, Italy
| | - Manuela Allegra
- Neuroscience Institute, National Research Council (IN-CNR), Padua, Italy
| | - Gabriele Deidda
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- *Correspondence: Gabriele Deidda
| |
Collapse
|
9
|
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: 13] [Impact Index Per Article: 4.3] [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
|
10
|
Saad AK, Akour A, Mahboob A, AbuRuz S, Sadek B. Role of Brain Modulators in Neurodevelopment: Focus on Autism Spectrum Disorder and Associated Comorbidities. Pharmaceuticals (Basel) 2022; 15:612. [PMID: 35631438 PMCID: PMC9144645 DOI: 10.3390/ph15050612] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Autism spectrum disorder (ASD) and associated neurodevelopmental disorders share similar pathogenesis and clinical features. Pathophysiological changes in these diseases are rooted in early neuronal stem cells in the uterus. Several genetic and environmental factors potentially perturb neurogenesis and synaptogenesis processes causing incomplete or altered maturation of the brain that precedes the symptomology later in life. In this review, the impact of several endogenous neuromodulators and pharmacological agents on the foetus during pregnancy, manifested on numerous aspects of neurodevelopment is discussed. Within this context, some possible insults that may alter these modulators and therefore alter their role in neurodevelopment are high-lighted. Sometimes, a particular insult could influence several neuromodulator systems as is supported by recent research in the field of ASD and associated disorders. Dopaminergic hy-pothesis prevailed on the table for discussion of the pathogenesis of schizophrenia (SCH), atten-tion-deficit hyperactivity disorder (ADHD) and ASD for a long time. However, recent cumulative evidence suggests otherwise. Indeed, the neuromodulators that are dysregulated in ASD and comorbid disorders are as diverse as the causes and symptoms of this disease. Additionally, these neuromodulators have roles in brain development, further complicating their involvement in comorbidity. This review will survey the current understanding of the neuromodulating systems to serve the pharmacological field during pregnancy and to minimize drug-related insults in pa-tients with ASD and associated comorbidity disorders, e.g., SCH or ADHD.
Collapse
Affiliation(s)
- Ali K. Saad
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman P.O. Box 11942, Jordan
| | - Abdulla Mahboob
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates;
| | - Salahdein AbuRuz
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman P.O. Box 11942, Jordan
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| |
Collapse
|
11
|
What is the mechanism of loudness hyperacusis in autism? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2021.110759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
Deidda G, Biazzo M. Gut and Brain: Investigating Physiological and Pathological Interactions Between Microbiota and Brain to Gain New Therapeutic Avenues for Brain Diseases. Front Neurosci 2021; 15:753915. [PMID: 34712115 PMCID: PMC8545893 DOI: 10.3389/fnins.2021.753915] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Brain physiological functions or pathological dysfunctions do surely depend on the activity of both neuronal and non-neuronal populations. Nevertheless, over the last decades, compelling and fast accumulating evidence showed that the brain is not alone. Indeed, the so-called "gut brain," composed of the microbial populations living in the gut, forms a symbiotic superorganism weighing as the human brain and strongly communicating with the latter via the gut-brain axis. The gut brain does exert a control on brain (dys)functions and it will eventually become a promising valuable therapeutic target for a number of brain pathologies. In the present review, we will first describe the role of gut microbiota in normal brain physiology from neurodevelopment till adulthood, and thereafter we will discuss evidence from the literature showing how gut microbiota alterations are a signature in a number of brain pathologies ranging from neurodevelopmental to neurodegenerative disorders, and how pre/probiotic supplement interventions aimed to correct the altered dysbiosis in pathological conditions may represent a valuable future therapeutic strategy.
Collapse
Affiliation(s)
- Gabriele Deidda
- The BioArte Limited, Life Sciences Park, San Gwann, Malta
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Manuele Biazzo
- The BioArte Limited, Life Sciences Park, San Gwann, Malta
- SienabioACTIVE, University of Siena, Siena, Italy
| |
Collapse
|
13
|
Garcia-Lopez R, Pombero A, Estirado A, Geijo-Barrientos E, Martinez S. Interneuron Heterotopia in the Lis1 Mutant Mouse Cortex Underlies a Structural and Functional Schizophrenia-Like Phenotype. Front Cell Dev Biol 2021; 9:693919. [PMID: 34327202 PMCID: PMC8313859 DOI: 10.3389/fcell.2021.693919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/16/2021] [Indexed: 11/24/2022] Open
Abstract
LIS1 is one of the principal genes related to Type I lissencephaly, a severe human brain malformation characterized by an abnormal neuronal migration in the cortex during embryonic development. This is clinically associated with epilepsy and cerebral palsy in severe cases, as well as a predisposition to developing mental disorders, in cases with a mild phenotype. Although genetic variations in the LIS1 gene have been associated with the development of schizophrenia, little is known about the underlying neurobiological mechanisms. We have studied how the Lis1 gene might cause deficits associated with the pathophysiology of schizophrenia using the Lis1/sLis1 murine model, which involves the deletion of the first coding exon of the Lis1 gene. Homozygous mice are not viable, but heterozygous animals present abnormal neuronal morphology, cortical dysplasia, and enhanced cortical excitability. We have observed reduced number of cells expressing GABA-synthesizing enzyme glutamic acid decarboxylase 67 (GAD67) in the hippocampus and the anterior cingulate area, as well as fewer parvalbumin-expressing cells in the anterior cingulate cortex in Lis1/sLis1 mutants compared to control mice. The cFOS protein expression (indicative of neuronal activity) in Lis1/sLis1 mice was higher in the medial prefrontal (mPFC), perirhinal (PERI), entorhinal (ENT), ectorhinal (ECT) cortices, and hippocampus compared to control mice. Our results suggest that deleting the first coding exon of the Lis1 gene might cause cortical anomalies associated with the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
| | - Ana Pombero
- Instituto de Neurociencias, UMH-CSIC, Alicante, Spain
| | | | | | - Salvador Martinez
- Instituto de Neurociencias, UMH-CSIC, Alicante, Spain.,Centro de Investigación Biomédica En Red en Salud Mental-CIBERSAM-ISCIII, Valencia, Spain
| |
Collapse
|
14
|
De Gregorio R, Chen X, Petit EI, Dobrenis K, Sze JY. Disruption of Transient SERT Expression in Thalamic Glutamatergic Neurons Alters Trajectory of Postnatal Interneuron Development in the Mouse Cortex. Cereb Cortex 2021; 30:1623-1636. [PMID: 31504267 DOI: 10.1093/cercor/bhz191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/29/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
In mice, terminal differentiation of subpopulations of interneurons occurs in late postnatal stages, paralleling the emergence of the adult cortical architecture. Here, we investigated the effects of altered initial cortical architecture on later interneuron development. We identified that a class of somatostatin (SOM)-expressing GABAergic interneurons undergoes terminal differentiation between 2nd and 3rd postnatal week in the mouse somatosensory barrel cortex and upregulates Reelin expression during neurite outgrowth. Our previous work demonstrated that transient expression (E15-P10) of serotonin uptake transporter (SERT) in thalamocortical projection neurons regulates barrel elaboration during cortical map establishment. We show here that in thalamic neuron SERT knockout mice, these SOM-expressing interneurons develop at the right time, reach correct positions and express correct neurochemical markers, but only 70% of the neurons remain in the adult barrel cortex. Moreover, those neurons that remain display altered dendritic patterning. Our data indicate that a precise architecture at the cortical destination is not essential for specifying late-developing interneuron identities, their cortical deposition, and spatial organization, but dictates their number and dendritic structure ultimately integrated into the cortex. Our study illuminates how disruption of temporal-specific SERT function and related key regulators during cortical map establishment can alter interneuron development trajectory that persists to adult central nervous system.
Collapse
Affiliation(s)
- Roberto De Gregorio
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Xiaoning Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Emilie I Petit
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| |
Collapse
|
15
|
Antón-Toro LF, Bruña R, Suárez-Méndez I, Correas Á, García-Moreno LM, Maestú F. Abnormal organization of inhibitory control functional networks in future binge drinkers. Drug Alcohol Depend 2021; 218:108401. [PMID: 33246710 DOI: 10.1016/j.drugalcdep.2020.108401] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS Adolescent Binge drinking has become an increasing health and social concern, which cause several detrimental consequences for brain integrity. However, research on neurophysiological traits of vulnerability for binge drinking predisposition is limited at this time. In this work, we conducted a two-year longitudinal study with magnetoencephalography (MEG) over a cohort of initially alcohol-naive adolescents with the purpose of characterize inhibitory cortical networks' anomalies prior to alcohol consumption onset in those youths who will transit into binge drinkers years later. METHODS Sixty-seven participant's inhibitory functional networks, and dysexecutive/impulsivity traits were measured by means of inhibitory task (go/no-go) and questionnaires battery. After a follow-up period of two years, we evaluated their alcohol consumption habits, sub-dividing them in two groups according to their alcohol intake patterns: future binge drinkers (fBD): n = 22; future Light/non-drinkers (fLD): n = 17. We evaluated whole-brain and seed-based functional connectivity profiles, as well as its correlation with impulsive and dysexecutive behaviours, searching for early abnormalities before consumption onset. RESULTS For the first time, abnormalities in MEG functional networks and higher dysexecutive and impulsivity profiles were detected in alcohol-naïve adolescents who two years later became binge drinkers. Concretely, fBD exhibit a distinctive pattern of beta band hyperconnectivity among crucial regions of inhibitory control networks, positively correlated with behavioral traits and future alcohol intake rate. CONCLUSIONS These findings strongly support the idea of early neurobiological vulnerabilities for substances consumption initiation, with inhibitory functional networks' abnormalities as a relevant neurophysiological marker of subjects at risk- we hypothesize this profile is due to neurodevelopmental and neurobiological differences involving cognitive control networks and neurotransmission pathways.
Collapse
Affiliation(s)
- Luis F Antón-Toro
- Department of Experimental Psychology, Complutense University of Madrid (UCM), 28223, Madrid, Spain; Laboratory for Cognitive and Computational Neuroscience (UCM - UPM), Center for Biomedical Technology (CBT), 28223, Madrid, Spain.
| | - Ricardo Bruña
- Department of Experimental Psychology, Complutense University of Madrid (UCM), 28223, Madrid, Spain; Laboratory for Cognitive and Computational Neuroscience (UCM - UPM), Center for Biomedical Technology (CBT), 28223, Madrid, Spain; Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Isabel Suárez-Méndez
- Department of Experimental Psychology, Complutense University of Madrid (UCM), 28223, Madrid, Spain; Laboratory for Cognitive and Computational Neuroscience (UCM - UPM), Center for Biomedical Technology (CBT), 28223, Madrid, Spain; Department of Structure of Matter, Thermal Physics and Electronics, Complutense University of Madrid (UCM), 28223, Madrid, Spain
| | - Ángeles Correas
- Laboratory for Cognitive and Computational Neuroscience (UCM - UPM), Center for Biomedical Technology (CBT), 28223, Madrid, Spain; Department of Psychology, San Diego State University, 5500 Campanile Drive San Diego, CA, 92182-4611, USA
| | - Luis M García-Moreno
- Department of Psychobiology and Methodology in Behavioral Sciences, Complutense University of Madrid (UCM), 28040, Madrid, Spain
| | - Fernando Maestú
- Department of Experimental Psychology, Complutense University of Madrid (UCM), 28223, Madrid, Spain; Laboratory for Cognitive and Computational Neuroscience (UCM - UPM), Center for Biomedical Technology (CBT), 28223, Madrid, Spain; Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
16
|
Neuregulins 1, 2, and 3 Promote Early Neurite Outgrowth in ErbB4-Expressing Cortical GABAergic Interneurons. Mol Neurobiol 2020; 57:3568-3588. [PMID: 32542595 DOI: 10.1007/s12035-020-01966-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022]
Abstract
The neuregulins (Nrgs 1-4) are a family of signaling molecules that play diverse roles in the nervous system. Nrg1 has been implicated in the formation of synapses and in synaptic plasticity. Previous studies have shown Nrg1 can affect neurite outgrowth in several neuronal populations, while the role of Nrg2 and Nrg3 in this process has remained understudied. The Nrgs can bind and activate the ErbB4 receptor tyrosine kinase which is preferentially expressed in GABAergic interneurons in the rodent hippocampus and cerebral cortex. In the present study, we evaluated the effects of Nrgs 1, 2, and 3 on neurite outgrowth of dissociated rat cortical ErbB4-positive (+)/GABA+ interneurons in vitro. All three Nrgs were able to promote neurite outgrowth during the first 2 days in vitro, with increases detected for both the axon (116-120%) and other neurites (100-120%). Increases in the average number of primary and secondary neurites were also observed. Treatment with the Nrgs for an additional 3 days promoted an increase in axonal length (86-96%), with only minimal effects on the remaining neurites (8-13%). ErbB4 expression persisted throughout the dendritic arbor and cell soma at all stages examined, while its expression in the axon was transient and declined with cell maturation. ErbB4 overexpression in GABAergic neurons promoted neurite outgrowth, an effect that was potentiated by Nrg treatment. These results show that Nrgs 1, 2, and 3 are each capable of influencing dendritic and axonal growth at early developmental stages in GABAergic neurons grown in vitro.
Collapse
|
17
|
Bitanihirwe BKY, Woo TUW. A conceptualized model linking matrix metalloproteinase-9 to schizophrenia pathogenesis. Schizophr Res 2020; 218:28-35. [PMID: 32001079 DOI: 10.1016/j.schres.2019.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinase 9 (MMP-9) is an extracellularly operating zinc-dependent endopeptidase that is commonly expressed in the brain, other tissues. It is synthesized in a latent zymogen form known as pro-MMP-9 that is subsequently converted to the active MMP-9 enzyme following cleavage of the pro-domain. Within the central nervous system, MMP-9 is localized and released from neurons, astrocytes and microglia where its expression levels are modulated by cytokines and growth factors during both normal and pathological conditions as well as by reactive oxygen species generated during oxidative stress. MMP-9 is involved in a number of key neurodevelopmental processes that are thought to be affected in schizophrenia, including maturation of the inhibitory neurons that contain the calcium-binding protein parvalbumin, developmental formation of the specialized extracellular matrix structure perineuronal net, synaptic pruning, and myelination. In this context, the present article provides a narrative synthesis of the existing evidence linking MMP-9 dysregulation to schizophrenia pathogenesis. We start by providing an overview of MMP-9 involvement in brain development and physiology. We then discuss the potential mechanisms through which MMP-9 dysregulation may affect neural circuitry maturation as well as how these anomalies may contribute to the disease process of schizophrenia. We conclude by articulating a comprehensive, cogent, and experimentally testable hypothesis linking MMP-9 to the developmental pathophysiologic cascade that triggers the onset and sustains the chronicity of the illness.
Collapse
Affiliation(s)
| | - Tsung-Ung W Woo
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Program in Cellular Neuropathology, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Myers AK, Cunningham JG, Smith SE, Snow JP, Smoot CA, Tucker ES. JNK signaling is required for proper tangential migration and laminar allocation of cortical interneurons. Development 2020; 147:dev180646. [PMID: 31915148 PMCID: PMC6983726 DOI: 10.1242/dev.180646] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
The precise migration of cortical interneurons is essential for the formation and function of cortical circuits, and disruptions to this key developmental process are implicated in the etiology of complex neurodevelopmental disorders, including schizophrenia, autism and epilepsy. We have recently identified the Jun N-terminal kinase (JNK) pathway as an important mediator of cortical interneuron migration in mice, regulating the proper timing of interneuron arrival into the cortical rudiment. In the current study, we demonstrate a vital role for JNK signaling at later stages of corticogenesis, when interneurons transition from tangential to radial modes of migration. Pharmacological inhibition of JNK signaling in ex vivo slice cultures caused cortical interneurons to rapidly depart from migratory streams and prematurely enter the cortical plate. Similarly, genetic loss of JNK function led to precocious stream departure ex vivo, and stream disruption, morphological changes and abnormal allocation of cortical interneurons in vivo These data suggest that JNK signaling facilitates the tangential migration and laminar deposition of cortical interneurons, and further implicates the JNK pathway as an important regulator of cortical development.
Collapse
Affiliation(s)
- Abigail K Myers
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Neuroscience Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Jessica G Cunningham
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Neuroscience Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Skye E Smith
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Biochemistry Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - John P Snow
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Catherine A Smoot
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Neuroscience Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Eric S Tucker
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| |
Collapse
|
19
|
Lucchese G, Flöel A, Stahl B. A Peptide Link Between Human Cytomegalovirus Infection, Neuronal Migration, and Psychosis. Front Psychiatry 2020; 11:349. [PMID: 32457660 PMCID: PMC7225321 DOI: 10.3389/fpsyt.2020.00349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 04/06/2020] [Indexed: 01/28/2023] Open
Abstract
Alongside biological, psychological, and social risk factors, psychotic syndromes may be related to disturbances of neuronal migration. This highly complex process characterizes the developing brain of the fetus, the early postnatal brain, and the adult brain, as reflected by changes within the subventricular zone and the dentate gyrus of the hippocampus, where neurogenesis persists throughout life. Psychosis also appears to be linked to human cytomegalovirus (HCMV) infection. However, little is known about the connection between psychosis, HCMV infection, and disruption of neuronal migration. The present study addresses the hypothesis that HCMV infection may lead to mental disorders through mechanisms of autoimmune cross-reactivity. Searching for common peptides that underlie immune cross-reactions, the analyses focus on HCMV and human proteins involved in neuronal migration. Results demonstrate a large overlap of viral peptides with human proteins associated with neuronal migration, such as ventral anterior homeobox 1 and cell adhesion molecule 1 implicated in GABAergic and glutamatergic neurotransmission. The present findings support the possibility of immune cross-reactivity between HCMV and human proteins that-when altered, mutated, or improperly functioning-may disrupt normal neuronal migration. In addition, these findings are consistent with a molecular and mechanistic framework for pathological sequences of events, beginning with HCMV infection, followed by immune activation, cross-reactivity, and neuronal protein variations that may ultimately contribute to the emergence of mental disorders, including psychosis.
Collapse
Affiliation(s)
- Guglielmo Lucchese
- Department of Neurology, University of Greifswald, Greifswald, Germany.,Department of Computing, Goldsmiths, University of London, London, United Kingdom
| | - Agnes Flöel
- Department of Neurology, University of Greifswald, Greifswald, Germany.,Partner Site Rostock/Greifswald, German Center for Neurodegenerative Diseases, Greifswald, Germany
| | - Benjamin Stahl
- Department of Neurology, University of Greifswald, Greifswald, Germany.,Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.,Psychologische Hochschule Berlin, Berlin, Germany
| |
Collapse
|
20
|
Demin KA, Meshalkina DA, Volgin AD, Yakovlev OV, de Abreu MS, Alekseeva PA, Friend AJ, Lakstygal AM, Zabegalov K, Amstislavskaya TG, Strekalova T, Bao W, Kalueff AV. Developing zebrafish experimental animal models relevant to schizophrenia. Neurosci Biobehav Rev 2019; 105:126-133. [DOI: 10.1016/j.neubiorev.2019.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 07/20/2019] [Accepted: 07/27/2019] [Indexed: 12/18/2022]
|
21
|
Kataria H, Alizadeh A, Karimi-Abdolrezaee S. Neuregulin-1/ErbB network: An emerging modulator of nervous system injury and repair. Prog Neurobiol 2019; 180:101643. [PMID: 31229498 DOI: 10.1016/j.pneurobio.2019.101643] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022]
Abstract
Neuregulin-1 (Nrg-1) is a member of the Neuregulin family of growth factors with essential roles in the developing and adult nervous system. Six different types of Nrg-1 (Nrg-1 type I-VI) and over 30 isoforms have been discovered; however, their specific roles are not fully determined. Nrg-1 signals through a complex network of protein-tyrosine kinase receptors, ErbB2, ErbB3, ErbB4 and multiple intracellular pathways. Genetic and pharmacological studies of Nrg-1 and ErbB receptors have identified a critical role for Nrg-1/ErbB network in neurodevelopment including neuronal migration, neural differentiation, myelination as well as formation of synapses and neuromuscular junctions. Nrg-1 signaling is best known for its characterized role in development and repair of the peripheral nervous system (PNS) due to its essential role in Schwann cell development, survival and myelination. However, our knowledge of the impact of Nrg-1/ErbB on the central nervous system (CNS) has emerged in recent years. Ongoing efforts have uncovered a multi-faceted role for Nrg-1 in regulating CNS injury and repair processes. In this review, we provide a timely overview of the most recent updates on Nrg-1 signaling and its role in nervous system injury and diseases. We will specifically highlight the emerging role of Nrg-1 in modulating the glial and immune responses and its capacity to foster neuroprotection and remyelination in CNS injury. Nrg-1/ErbB network is a key regulatory pathway in the developing nervous system; therefore, unraveling its role in neuropathology and repair can aid in development of new therapeutic approaches for nervous system injuries and associated disorders.
Collapse
Affiliation(s)
- Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
22
|
Haijima A, Lesmana R, Shimokawa N, Amano I, Takatsuru Y, Koibuchi N. Differential neurotoxic effects of in utero and lactational exposure to hydroxylated polychlorinated biphenyl (OH-PCB 106) on spontaneous locomotor activity and motor coordination in young adult male mice. J Toxicol Sci 2017; 42:407-416. [PMID: 28717099 DOI: 10.2131/jts.42.407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
We investigated whether in utero or lactational exposure to 4-hydroxy-2',3,3',4',5'-pentachlorobiphenyl (OH-PCB 106) affects spontaneous locomotor activity and motor coordination in young adult male mice. For in utero exposure, pregnant C57BL/6J mice received 0.05 or 0.5 mg/kg body weight of OH-PCB 106 or corn oil vehicle via gavage every second day from gestational day 10 to 18. For lactational exposure, the different groups of dams received 0.05 or 0.5 mg/kg body weight of OH-PCB 106 or corn oil vehicle via gavage every second day from postpartum day 3 to 13. At 6-7 weeks of age, the spontaneous locomotor activities of male offspring were evaluated for a 24-hr continuous session in a home cage and in an open field for 30-min. Motor coordination function on an accelerating rotarod was also measured. Mice exposed prenatally to OH-PCB 106 showed increased spontaneous locomotor activities during the dark phase in the home cage and during the first 10-min in the open field compared with control mice. Mice exposed lactationally to OH-PCB 106, however, did not show a time-dependent decrease in locomotor activity in the open field. Instead, their locomotor activity increased significantly during the second 10-min block. In addition, mice exposed lactationally to OH-PCB 106 displayed impairments in motor coordination in the rotarod test. These results suggest that perinatal exposure to OH-PCB 106 affects motor behaviors in young adult male mice. Depending on the period of exposure, OH-PCB 106 may have different effects on neurobehavioral development.
Collapse
Affiliation(s)
- Asahi Haijima
- Center for Medical Education, Gunma University Graduate School of Medicine.,Department of Integrative Physiology, Gunma University Graduate School of Medicine
| | - Ronny Lesmana
- Department of Integrative Physiology, Gunma University Graduate School of Medicine.,Department of Physiology, Universitas Padjadjaran, Indonesia
| | - Noriaki Shimokawa
- Department of Integrative Physiology, Gunma University Graduate School of Medicine.,Department of Nutrition, Takasaki University of Health and Welfare
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine
| | - Yusuke Takatsuru
- Department of Integrative Physiology, Gunma University Graduate School of Medicine
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine
| |
Collapse
|
23
|
Sammali E, Alia C, Vegliante G, Colombo V, Giordano N, Pischiutta F, Boncoraglio GB, Barilani M, Lazzari L, Caleo M, De Simoni MG, Gaipa G, Citerio G, Zanier ER. Intravenous infusion of human bone marrow mesenchymal stromal cells promotes functional recovery and neuroplasticity after ischemic stroke in mice. Sci Rep 2017; 7:6962. [PMID: 28761170 PMCID: PMC5537246 DOI: 10.1038/s41598-017-07274-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/27/2017] [Indexed: 12/13/2022] Open
Abstract
Transplantation of human bone marrow mesenchymal stromal cells (hBM-MSC) promotes functional recovery after stroke in animal models, but the mechanisms underlying these effects remain incompletely understood. We tested the efficacy of Good Manufacturing Practices (GMP) compliant hBM-MSC, injected intravenously 3.5 hours after injury in mice subjected to transient middle cerebral artery occlusion (tMCAo). We addressed whether hBM-MSC are efficacious and if this efficacy is associated with cortical circuit reorganization using neuroanatomical analysis of GABAergic neurons (parvalbumin; PV-positive cells) and perineuronal nets (PNN), a specialized extracellular matrix structure which acts as an inhibitor of neural plasticity. tMCAo mice receiving hBM-MSC, showed early and lasting improvement of sensorimotor and cognitive functions compared to control tMCAo mice. Furthermore, 5 weeks post-tMCAo, hBM-MSC induced a significant rescue of ipsilateral cortical neurons; an increased proportion of PV-positive neurons in the perilesional cortex, suggesting GABAergic interneurons preservation; and a lower percentage of PV-positive cells surrounded by PNN, indicating an enhanced plastic potential of the perilesional cortex. These results show that hBM-MSC improve functional recovery and stimulate neuroprotection after stroke. Moreover, the downregulation of “plasticity brakes” such as PNN suggests that hBM-MSC treatment stimulates plasticity and formation of new connections in the perilesional cortex.
Collapse
Affiliation(s)
- Eliana Sammali
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy.,Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milano, Italy
| | - Claudia Alia
- Neuroscience Institute, CNR, Pisa, Italy.,Scuola Normale Superiore, Pisa, Italy
| | - Gloria Vegliante
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Valentina Colombo
- Laboratory for Cell and Gene Therapy "Stefano Verri", ASST-Monza, San Gerardo Hospital, Monza, Italy.,Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Nadia Giordano
- Neuroscience Institute, CNR, Pisa, Italy.,Scuola Normale Superiore, Pisa, Italy
| | - Francesca Pischiutta
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Giorgio B Boncoraglio
- Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milano, Italy
| | - Mario Barilani
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | - Lorenza Lazzari
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | | | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Giuseppe Gaipa
- Laboratory for Cell and Gene Therapy "Stefano Verri", ASST-Monza, San Gerardo Hospital, Monza, Italy.,Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.,Neurointensive Care, ASST-Monza, San Gerardo Hospital, Monza, Italy
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy.
| |
Collapse
|
24
|
Genetic polymorphisms and their association with brain and behavioural measures in heterogeneous stock mice. Sci Rep 2017; 7:41204. [PMID: 28145470 PMCID: PMC5286500 DOI: 10.1038/srep41204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 12/16/2016] [Indexed: 12/30/2022] Open
Abstract
Although the search for quantitative trait loci for behaviour remains a considerable challenge, the complicated genetic architecture of quantitative traits is beginning to be understood. The current project utilised heterogeneous stock (HS) male mice (n = 580) to investigate the genetic basis for brain weights, activity, anxiety and cognitive phenotypes. We identified 126 single nucleotide polymorphisms (SNPs) in genes involved in regulation of neurotransmitter systems, nerve growth/death and gene expression, and subsequently investigated their associations with changes in behaviour and/or brain weights in our sample. We found significant associations between four SNP-phenotype pairs, after controlling for multiple testing. Specificity protein 2 (Sp2, rs3708840), tryptophan hydroxylase 1 (Tph1, rs262731280) and serotonin receptor 3A (Htr3a, rs50670893) were associated with activity/anxiety behaviours, and microtubule-associated protein 2 (Map2, rs13475902) was associated with cognitive performance. All these genes except for Tph1 were expressed in the brain above the array median, and remained significantly associated with relevant behaviours after controlling for the family structure. Additionally, we found evidence for a correlation between Htr3a expression and activity. We discuss our findings in the light of the advantages and limitations of currently available mouse genetic tools, suggesting further directions for association studies in rodents.
Collapse
|
25
|
Alia C, Spalletti C, Lai S, Panarese A, Micera S, Caleo M. Reducing GABA A-mediated inhibition improves forelimb motor function after focal cortical stroke in mice. Sci Rep 2016; 6:37823. [PMID: 27897203 PMCID: PMC5126677 DOI: 10.1038/srep37823] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/21/2016] [Indexed: 11/25/2022] Open
Abstract
A deeper understanding of post-stroke plasticity is critical to devise more effective pharmacological and rehabilitative treatments. The GABAergic system is one of the key modulators of neuronal plasticity, and plays an important role in the control of “critical periods” during brain development. Here, we report a key role for GABAergic inhibition in functional restoration following ischemia in the adult mouse forelimb motor cortex. After stroke, the majority of cortical sites in peri-infarct areas evoked simultaneous movements of forelimb, hindlimb and tail, consistent with a loss of inhibitory signalling. Accordingly, we found a delayed decrease in several GABAergic markers that accompanied cortical reorganization. To test whether reductions in GABAergic signalling were causally involved in motor improvements, we treated animals during an early post-stroke period with a benzodiazepine inverse agonist, which impairs GABAA receptor function. We found that hampering GABAA signalling led to significant restoration of function in general motor tests (i.e., gridwalk and pellet reaching tasks), with no significant impact on the kinematics of reaching movements. Improvements were persistent as they remained detectable about three weeks after treatment. These data demonstrate a key role for GABAergic inhibition in limiting motor improvements after cortical stroke.
Collapse
Affiliation(s)
- Claudia Alia
- Scuola Normale Superiore, 56126, Pisa, Italy.,CNR Neuroscience Institute, 56124, Pisa, Italy
| | | | - Stefano Lai
- The BioRobotics Institute Scuola Superiore Sant'Anna, 56025, Pontedera (PI), Italy
| | - Alessandro Panarese
- The BioRobotics Institute Scuola Superiore Sant'Anna, 56025, Pontedera (PI), Italy
| | - Silvestro Micera
- The BioRobotics Institute Scuola Superiore Sant'Anna, 56025, Pontedera (PI), Italy.,Ecole Polytechnique Federale de Lausanne (EPFL), Bertarelli Foundation Chair in Translational NeuroEngineering Laboratory, Center for Neuroprosthetics and Institute of Bioengineering, CH-1015 Lausanne, Switzerland
| | | |
Collapse
|
26
|
Pizzamiglio L, Focchi E, Murru L, Tamborini M, Passafaro M, Menna E, Matteoli M, Antonucci F. New Role of ATM in Controlling GABAergic Tone During Development. Cereb Cortex 2016; 26:3879-88. [PMID: 27166172 DOI: 10.1093/cercor/bhw125] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The capacity to guarantee the proper excitatory/inhibitory balance is one of the most critical steps during early development responsible for the correct brain organization, function, and plasticity. GABAergic neurons guide this process leading to the right structural organization, brain circuitry, and neuronal firing. Here, we identified the ataxia telangiectasia mutated (ATM), a serine/threonine protein kinase linked to DNA damage response, as crucial in regulating neurotransmission. We found that reduced levels of ATM in the hippocampal neuronal cultures produce an excitatory/inhibitory unbalance toward inhibition as indicated by the higher frequency of miniature inhibitory postsynaptic current events and an increased number of GABAergic synapses. In vivo, the increased inhibition still persists and, even if a higher excitation is also present, a reduced neuronal excitability is found as indicated by the lower action potential frequency generated in response to high-current intensity stimuli. Finally, we found an elevated extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation in heterozygous hippocampi associated with lower expression levels of the ERK1/2 phosphatase PP1. Given that the neurodegenerative condition associated with genetic mutations in the Atm gene, ataxia telangiectasia, presents a variable phenotype with impairment in cognition, our molecular findings provide a logical frame for a more clear comprehension of cognitive defects in the pathology, opening to novel therapeutic strategies.
Collapse
Affiliation(s)
- Lara Pizzamiglio
- Department of Biology and Biotechnology, Lazzaro Spallanzani, University of Pavia, 27100 Pavia, Italy Department of Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Elisa Focchi
- Department of Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy Humanitas Clinical and Research Center, IRCCS Rozzano, Rozzano (Milan), Italy
| | - Luca Murru
- Institute of Neuroscience, C.N.R., 20129 Milan, Italy
| | - Matteo Tamborini
- Department of Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | | | - Elisabetta Menna
- Humanitas Clinical and Research Center, IRCCS Rozzano, Rozzano (Milan), Italy Institute of Neuroscience, C.N.R., 20129 Milan, Italy
| | - Michela Matteoli
- Humanitas Clinical and Research Center, IRCCS Rozzano, Rozzano (Milan), Italy Institute of Neuroscience, C.N.R., 20129 Milan, Italy
| | - Flavia Antonucci
- Department of Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy Institute of Neuroscience, C.N.R., 20129 Milan, Italy
| |
Collapse
|
27
|
Fragile X mental retardation protein knockdown in the developing Xenopus tadpole optic tectum results in enhanced feedforward inhibition and behavioral deficits. Neural Dev 2016; 11:14. [PMID: 27503008 PMCID: PMC4977860 DOI: 10.1186/s13064-016-0069-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/03/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Fragile X Syndrome is the leading monogenetic cause of autism and most common form of intellectual disability. Previous studies have implicated changes in dendritic spine architecture as the primary result of loss of Fragile X Mental Retardation Protein (FMRP), but recent work has shown that neural proliferation is decreased and cell death is increased with either loss of FMRP or overexpression of FMRP. The purpose of this study was to investigate the effects of loss of FMRP on behavior and cellular activity. METHODS We knocked down FMRP expression using morpholino oligos in the optic tectum of Xenopus laevis tadpoles and performed a series of behavioral and electrophysiological assays. We investigated visually guided collision avoidance, schooling, and seizure propensity. Using single cell electrophysiology, we assessed intrinsic excitability and synaptic connectivity of tectal neurons. RESULTS We found that FMRP knockdown results in decreased swimming speed, reduced schooling behavior and decreased seizure severity. In single cells, we found increased inhibition relative to excitation in response to sensory input. CONCLUSIONS Our results indicate that the electrophysiological development of single cells in the absence of FMRP is largely unaffected despite the large neural proliferation defect. The changes in behavior are consistent with an increase in inhibition, which could be due to either changes in cell number or altered inhibitory drive, and indicate that FMRP can play a significant role in neural development much earlier than previously thought.
Collapse
|
28
|
Asmus SE, Raghanti MA, Beyerle ER, Fleming-Beattie JC, Hawkins SM, McKernan CM, Rauh NA. Tyrosine hydroxylase-producing neurons in the human cerebral cortex do not colocalize with calcium-binding proteins or the serotonin 3A receptor. J Chem Neuroanat 2016; 78:1-9. [PMID: 27448941 DOI: 10.1016/j.jchemneu.2016.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 07/14/2016] [Accepted: 07/15/2016] [Indexed: 11/25/2022]
Abstract
Interneurons of the cerebral cortex play a significant role in cortical information processing and are of clinical interest due to their involvement in neurological disorders. In the human neocortex, three subsets of interneurons can be identified based on the production of the calcium-binding proteins parvalbumin, calretinin or calbindin. A subset of interneurons in the mouse cortex expresses the serotonin 3A receptor (5-HT3AR). Previous work in humans has also demonstrated the presence of a subgroup of cortical neurons that produces the catecholaminergic enzyme tyrosine hydroxylase (TH). Many TH-producing cells in the rat cortex coexpress calretinin and are adjacent to blood vessels. However, little is known about the phenotype of these TH interneurons in humans. Here we immunohistochemically examined the coexpression of TH with parvalbumin, calretinin, calbindin or 5-HT3AR in human Brodmann's areas 10 and 24, cortical regions with high densities of TH-containing neurons. Colocalization of TH with these calcium-binding proteins and with 5-HT3AR was not detected in either area. Cortical TH cells were rarely apposed to blood vessels, denoted by immunolabeling for the gliovascular marker aquaporin-4. Our results suggest that the TH-immunoreactive cells in the human cortex do not overlap with any known neurochemically-defined subsets of interneurons and provide further evidence of differences in the phenotype of these cells across species.
Collapse
Affiliation(s)
- Stephen E Asmus
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA.
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Eric R Beyerle
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Julia C Fleming-Beattie
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Sarah M Hawkins
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Courtney M McKernan
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Nicholas A Rauh
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| |
Collapse
|
29
|
Li JT, Su YA, Wang HL, Zhao YY, Liao XM, Wang XD, Si TM. Repeated Blockade of NMDA Receptors During Adolescence Impairs Reversal Learning and Disrupts GABAergic Interneurons in Rat Medial Prefrontal Cortex. Front Mol Neurosci 2016; 9:17. [PMID: 26973457 PMCID: PMC4776083 DOI: 10.3389/fnmol.2016.00017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/19/2016] [Indexed: 11/13/2022] Open
Abstract
Adolescence is of particular significance to schizophrenia, since psychosis onset typically occurs in this critical period. Based on the N-methyl-D-aspartate (NMDA) receptor hypofunction hypothesis of schizophrenia, in this study, we investigated whether and how repeated NMDA receptor blockade during adolescence would affect GABAergic interneurons in rat medial prefrontal cortex (mPFC) and mPFC-mediated cognitive functions. Specifically, adolescent rats were subjected to intraperitoneal administration of MK-801 (0.1, 0.2, 0.4 mg/kg), a non-competitive NMDA receptor antagonist, for 14 days and then tested for reference memory and reversal learning in the water maze. The density of parvabumin (PV)-, calbindin (CB)- and calretinin (CR)-positive neurons in mPFC was analyzed at either 24 h or 7 days after drug cessation. We found that MK-801 treatment delayed reversal learning in the water maze without affecting initial acquisition. Strikingly, MK-801 treatment also significantly reduced the density of PV+ and CB+ neurons, and this effect persisted for 7 days after drug cessation at the dose of 0.2 mg/kg. We further demonstrated that the reduction in PV+ and CB+ neuron densities was ascribed to a downregulation of the expression levels of PV and CB, but not to neuronal death. These results parallel the behavioral and neuropathological changes of schizophrenia and provide evidence that adolescent NMDA receptors antagonism offers a useful tool for unraveling the etiology of the disease.
Collapse
Affiliation(s)
- Ji-Tao Li
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Yun-Ai Su
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Hong-Li Wang
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Ying-Ying Zhao
- Depression Treatment Center, Beijing Anding Hospital of Capital Medical University Beijing, China
| | - Xue-Mei Liao
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine Hangzhou, China
| | - Tian-Mei Si
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| |
Collapse
|
30
|
Pépés SE, Draper A, Jackson GM, Jackson SR. Effects of age on motor excitability measures from children and adolescents with Tourette syndrome. Dev Cogn Neurosci 2016; 19:78-86. [PMID: 26934638 PMCID: PMC6988104 DOI: 10.1016/j.dcn.2016.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 01/01/2016] [Accepted: 02/12/2016] [Indexed: 12/27/2022] Open
Abstract
Tourette syndrome (TS) is a neurological disorder characterised by vocal and motor tics. It is associated with cortical-striatal-thalamic-cortical circuit [CSTC] dysfunction and hyper-excitability of cortical motor regions. TS follows a developmental time course, in which tics often become increasingly more controlled during adolescence. Importantly, however, a substantial minority of patients continue to have debilitating tics into adulthood. This indicates that there may be important differences between adult TS patients and children and adolescents with the disorder. We use TMS to examine cortical motor excitability in a sample of children, adolescents and young adults with TS. We demonstrate that, in contrast to studies of adult patients, resting motor threshold and the variability of MEP responses are increased in children with TS, while the gain of motor excitability in reduced. Importantly, we demonstrate that these differences normalise with age over adolescence. We conclude that these effects are likely due to a developmental delay in the maturation of key brain networks in TS, consistent with recent brain imaging studies of structural and functional brain connectivity. Importantly, these findings suggest that the alterations in brain network structure and function associated with TS may be quite different in children and adult patients with the condition.
Collapse
Affiliation(s)
| | | | - Georgina M Jackson
- Institute of Mental Health, School of Medicine, University of Nottingham, UK
| | | |
Collapse
|
31
|
Tavassoli T, Bellesheim K, Tommerdahl M, Holden JM, Kolevzon A, Buxbaum JD. Altered tactile processing in children with autism spectrum disorder. Autism Res 2015; 9:616-20. [PMID: 26568449 DOI: 10.1002/aur.1563] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 06/19/2015] [Accepted: 07/28/2015] [Indexed: 11/06/2022]
Abstract
Although tactile reactivity issues are commonly reported in children with autism spectrum disorder (ASD), the underlying mechanisms are poorly understood. Less feed-forward inhibition has been proposed as a potential mechanism for some symptoms of ASD. We tested static and dynamic tactile thresholds as a behavioral proxy of feed-forward inhibition in 42 children (21 children with ASD and 21 typically developing [TD] children). Subthreshold conditioning typically raises the dynamic detection threshold, thus comparison of the dynamic to the static threshold generates a metric that predicts gamma-aminobutyric acid (GABA) mediated feed-forward inhibition. Children with ASD had marginally higher static thresholds and a significantly lower ratio between thresholds as compared with TD children. The lower ratio, only seen in children with ASD, might be indicative of less inhibition. Static thresholds were correlated with autism spectrum quotient scores, indicating the higher the tactile threshold, the more ASD traits. The amount of feed-forward inhibition (ratio between dynamic/static) was negatively correlated with autism diagnostic observation schedule repetitive behavior scores, meaning the less inhibition the more ASD symptoms. In summary, children with ASD showed altered tactile processing compared with TD children; thus measuring static and dynamic thresholds could be a potential biomarker for ASD and might be useful for prediction of treatment response with therapeutics, including those that target the GABAergic system. Autism Res 2016, 9: 616-620. © 2015 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Teresa Tavassoli
- Department of Psychiatry, Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | | | - Mark Tommerdahl
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, 27599
| | - Jameson M Holden
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, 27599
| | - Alexander Kolevzon
- Department of Psychiatry, Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joseph D Buxbaum
- Department of Psychiatry, Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
32
|
Regional Specificity of GABAergic Regulation of Cross-Modal Plasticity in Mouse Visual Cortex after Unilateral Enucleation. J Neurosci 2015; 35:11174-89. [PMID: 26269628 DOI: 10.1523/jneurosci.3808-14.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
UNLABELLED In adult mice, monocular enucleation (ME) results in an immediate deactivation of the contralateral medial monocular visual cortex. An early restricted reactivation by open eye potentiation is followed by a late overt cross-modal reactivation by whiskers (Van Brussel et al., 2011). In adolescence (P45), extensive recovery of cortical activity after ME fails as a result of suppression or functional immaturity of the cross-modal mechanisms (Nys et al., 2014). Here, we show that dark exposure before ME in adulthood also prevents the late cross-modal reactivation component, thereby converting the outcome of long-term ME into a more P45-like response. Because dark exposure affects GABAergic synaptic transmission in binocular V1 and the plastic immunity observed at P45 is reminiscent of the refractory period for inhibitory plasticity reported by Huang et al. (2010), we molecularly examined whether GABAergic inhibition also regulates ME-induced cross-modal plasticity. Comparison of the adaptation of the medial monocular and binocular cortices to long-term ME or dark exposure or a combinatorial deprivation revealed striking differences. In the medial monocular cortex, cortical inhibition via the GABAA receptor α1 subunit restricts cross-modal plasticity in P45 mice but is relaxed in adults to allow the whisker-mediated reactivation. In line, in vivo pharmacological activation of α1 subunit-containing GABAA receptors in adult ME mice specifically reduces the cross-modal aspect of reactivation. Together with region-specific changes in glutamate acid decarboxylase (GAD) and vesicular GABA transporter expression, these findings put intracortical inhibition forward as an important regulator of the age-, experience-, and cortical region-dependent cross-modal response to unilateral visual deprivation. SIGNIFICANCE STATEMENT In adult mice, vision loss through one eye instantly reduces neuronal activity in the visual cortex. Strengthening of remaining eye inputs in the binocular cortex is followed by cross-modal adaptations in the monocular cortex, in which whiskers become a dominant nonvisual input source to attain extensive cortical reactivation. We show that the cross-modal component does not occur in adolescence because of increased intracortical inhibition, a phenotype that was mimicked in adult enucleated mice when treated with indiplon, a GABAA receptor α1 agonist. The cross-modal versus unimodal responses of the adult monocular and binocular cortices also mirror regional specificity in inhibitory alterations after visual deprivation. Understanding cross-modal plasticity in response to sensory loss is essential to maximize patient susceptibility to sensory prosthetics.
Collapse
|
33
|
Purves-Tyson TD, Allen K, Fung S, Rothmond D, Noble PL, Handelsman DJ, Shannon Weickert C. Adolescent testosterone influences BDNF and TrkB mRNA and neurotrophin-interneuron marker relationships in mammalian frontal cortex. Schizophr Res 2015; 168:661-70. [PMID: 26088421 DOI: 10.1016/j.schres.2015.05.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/11/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
Late adolescence in males is a period of increased susceptibility for the onset of schizophrenia, coinciding with increased circulating testosterone. The cognitive deficits prevalent in schizophrenia may be related to unhealthy cortical interneurons, which are trophically dependent on brain derived neurotrophic factor. We investigated, under conditions of depleted (monkey and rat) and replaced (rat) testosterone over adolescence, changes in gene expression of cortical BDNF and TrkB transcripts and interneuron markers and the relationships between these mRNAs and circulating testosterone. Testosterone removal by gonadectomy reduced gene expression of some BDNF transcripts in monkey and rat frontal cortices and the BDNF mRNA reduction was prevented by testosterone replacement. In rat, testosterone replacement increased the potential for classical TrkB signalling by increasing the full length to truncated TrkB mRNA ratio, whereas in the monkey cortex, circulating testosterone was negatively correlated with the TrkB full length/truncated mRNA ratio. We did not identify changes in interneuron gene expression in monkey frontal cortex in response to gonadectomy, and in rat, we showed that only somatostatin mRNA was decreased by gonadectomy but not restored by testosterone replacement. We identified complex and possibly species-specific, relationships between BDNF/TrkB gene expression and interneuron marker gene expression that appear to be dependent on the presence of testosterone at adolescence in rat and monkey frontal cortices. Taken together, our findings suggest there are dynamic relationships between BDNF/TrkB and interneuron markers that are dependent on the presence of testosterone but that this may not be a straightforward increase in testosterone leading to changes in BDNF/TrkB that contributes to interneuron health.
Collapse
Affiliation(s)
- Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Medical Sciences, University of New South Wales, Sydney 2031, Australia
| | - Katherine Allen
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | - Samantha Fung
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | - Debora Rothmond
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia
| | | | | | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| |
Collapse
|
34
|
Subcortical glutamate mediates the reduction of short-range functional connectivity with age in a developmental cohort. J Neurosci 2015; 35:8433-41. [PMID: 26041912 DOI: 10.1523/jneurosci.4375-14.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Marked changes in brain physiology and structure take place between childhood and adulthood, including changes in functional connectivity and changes in the balance between main excitatory and inhibitory neurotransmitters glutamate (Glu) and GABA. The balance of these neurotransmitters is thought to underlie neural activity in general and functional connectivity networks in particular, but so far no studies have investigated the relationship between human development related differences in these neurotransmitters and concomitant changes in functional connectivity. GABA+/H2O and Glu/H2O levels were acquired in a group of healthy children, adolescents, and adults in a subcortical (basal ganglia) region, as well as in a frontal region in adolescents and adults. Our results showed higher GABA+/Glu with age in both the subcortical and the frontal voxel, which were differentially associated with significantly lower Glu/H2O with age in the subcortical voxel and by significantly higher GABA+/H2O with age in the frontal voxel. Using a seed-to-voxel analysis, we were further able to show that functional connectivity between the putamen (seed) and other subcortical structures was lower with age. Lower subcortical Glu/H2O with age mediated the lower connectivity in the dorsal putamen. Based on these results, and the potential role of Glu in synaptic pruning, we suggest that lower Glu mediates a reduction of local connectivity during human development.
Collapse
|
35
|
Nys J, Scheyltjens I, Arckens L. Visual system plasticity in mammals: the story of monocular enucleation-induced vision loss. Front Syst Neurosci 2015; 9:60. [PMID: 25972788 PMCID: PMC4412011 DOI: 10.3389/fnsys.2015.00060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/30/2015] [Indexed: 11/30/2022] Open
Abstract
The groundbreaking work of Hubel and Wiesel in the 1960’s on ocular dominance plasticity instigated many studies of the visual system of mammals, enriching our understanding of how the development of its structure and function depends on high quality visual input through both eyes. These studies have mainly employed lid suturing, dark rearing and eye patching applied to different species to reduce or impair visual input, and have created extensive knowledge on binocular vision. However, not all aspects and types of plasticity in the visual cortex have been covered in full detail. In that regard, a more drastic deprivation method like enucleation, leading to complete vision loss appears useful as it has more widespread effects on the afferent visual pathway and even on non-visual brain regions. One-eyed vision due to monocular enucleation (ME) profoundly affects the contralateral retinorecipient subcortical and cortical structures thereby creating a powerful means to investigate cortical plasticity phenomena in which binocular competition has no vote.In this review, we will present current knowledge about the specific application of ME as an experimental tool to study visual and cross-modal brain plasticity and compare early postnatal stages up into adulthood. The structural and physiological consequences of this type of extensive sensory loss as documented and studied in several animal species and human patients will be discussed. We will summarize how ME studies have been instrumental to our current understanding of the differentiation of sensory systems and how the structure and function of cortical circuits in mammals are shaped in response to such an extensive alteration in experience. In conclusion, we will highlight future perspectives and the clinical relevance of adding ME to the list of more longstanding deprivation models in visual system research.
Collapse
Affiliation(s)
- Julie Nys
- Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven Leuven, Belgium
| | | | - Lutgarde Arckens
- Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven Leuven, Belgium
| |
Collapse
|
36
|
Benítez-Burraco A, Boeckx C. Approaching motor and language deficits in autism from below: a biolinguistic perspective. Front Integr Neurosci 2015; 9:25. [PMID: 25870545 PMCID: PMC4378279 DOI: 10.3389/fnint.2015.00025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/13/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Cedric Boeckx
- Catalan Institute for Advanced Studies and Research (ICREA) Barcelona, Spain ; Department of Linguistics, Universitat de Barcelona Barcelona, Spain
| |
Collapse
|
37
|
Roux C, Lesueur C, Aligny C, Brasse-Lagnel C, Genty D, Marret S, Laquerrière A, Bekri S, Gonzalez BJ. 3-MA Inhibits Autophagy and Favors Long-Term Integration of Grafted Gad67–GFP GABAergic Precursors in the Developing Neocortex by Preventing Apoptosis. Cell Transplant 2014; 23:1425-50. [DOI: 10.3727/096368913x670174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In human neonates, immature GABAergic interneurons are markedly affected by an excitotoxic insult. While in adults the interest of cell transplantation has been demonstrated in several neurological disorders, few data are available regarding the immature brain. The low survival rate constitutes a strong limitation in the capacity of transplanted neurons to integrate the host tissue. Because i) autophagy is an adaptive process to energetic/nutrient deprivation essential for cell survival and ii) literature describes cross-talks between autophagy and apoptosis, we hypothesized that regulation of autophagy would represent an original strategy to favor long-term survival of GABAergic precursors grafted in the immature neocortex. Morphological, neurochemical, and functional data showed that in control conditions, few grafted Gad67-GFP precursors survived. The first hours following transplantation were a critical period with intense apoptosis. Experiments performed on E15.5 ganglionic eminences revealed that Gad67-GFP precursors were highly sensitive to autophagy. Rapamycin and 3-MA impacted on LC3 cleavage, LC3II translocation, and autophagosome formation. Quantification of Bax, mitochondrial integrity, caspase-3 cleavage, and caspase-3 immunolocalization and activity showed that 3-MA induced a significant decrease of Gad67-GFP precursor apoptosis. In vivo, 3-MA induced, within the first 24 h, a diffuse LC3 pattern of grafted Gad67-GFP precursors, an increase of precursors with neurites, a reduction of the density of caspase-3 immunoreactive cells. A twofold increase in the survival rate occurred 15 days after the graft. Surviving neurons were localized in the cortical layers II–IV, which were still immature when the transplantation was done. Altogether, these data indicate that inhibition of autophagy represents an original strategy to allow GABAergic interneurons to overpass the first critical hours following transplantation and to increase their long-term survival in mice neonates.
Collapse
Affiliation(s)
- Christian Roux
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
| | - Céline Lesueur
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
- Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Caroline Aligny
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
| | - Carole Brasse-Lagnel
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
- Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Damien Genty
- Department of Pathology, Rouen University Hospital, Rouen, France
| | - Stéphane Marret
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
- Department of Neonatal Paediatrics and Intensive Care, Rouen Hospital, Rouen, France
| | - Annie Laquerrière
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
- Department of Pathology, Rouen University Hospital, Rouen, France
| | - Soumeya Bekri
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
- Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Bruno J. Gonzalez
- NeoVasc Laboratory, ERI28, Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
| |
Collapse
|
38
|
Calfa G, Li W, Rutherford JM, Pozzo-Miller L. Excitation/inhibition imbalance and impaired synaptic inhibition in hippocampal area CA3 of Mecp2 knockout mice. Hippocampus 2014; 25:159-68. [PMID: 25209930 DOI: 10.1002/hipo.22360] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2014] [Indexed: 01/01/2023]
Abstract
Rett syndrome (RTT) is a neurodevelopment disorder associated with intellectual disabilities and caused by loss-of-function mutations in the gene encoding the transcriptional regulator Methyl-CpG-binding Protein-2 (MeCP2). Neuronal dysfunction and changes in cortical excitability occur in RTT individuals and Mecp2-deficient mice, including hippocampal network hyperactivity and higher frequency of spontaneous multiunit spikes in the CA3 cell body layer. Here, we describe impaired synaptic inhibition and an excitation/inhibition (E/I) imbalance in area CA3 of acute slices from symptomatic Mecp2 knockout male mice (referred to as Mecp2(-/y) ). The amplitude of TTX-resistant miniature inhibitory postsynaptic currents (mIPSC) was smaller in CA3 pyramidal neurons of Mecp2(-/y) slices than in wildtype controls, while the amplitude of miniature excitatory postsynaptic currents (mEPSC) was significantly larger in Mecp2(-/y) neurons. Consistently, quantitative confocal immunohistochemistry revealed significantly lower intensity of the alpha-1 subunit of GABAA Rs in the CA3 cell body layer of Mecp2(-/y) mice, while GluA1 puncta intensities were significantly higher in the CA3 dendritic layers of Mecp2(-/y) mice. In addition, the input/output (I/O) relationship of evoked IPSCs had a shallower slope in CA3 pyramidal neurons Mecp2(-/y) neurons. Consistent with the absence of neuronal degeneration in RTT and MeCP2-based mouse models, the density of parvalbumin- and somatostatin-expressing interneurons in area CA3 was not affected in Mecp2(-/y) mice. Furthermore, the intrinsic membrane properties of several interneuron subtypes in area CA3 were not affected by Mecp2 loss. However, mEPSCs are smaller and less frequent in CA3 fast-spiking basket cells of Mecp2(-/y) mice, suggesting an impaired glutamatergic drive in this interneuron population. These results demonstrate that a loss-of-function mutation in Mecp2 causes impaired E/I balance onto CA3 pyramidal neurons, leading to a hyperactive hippocampal network, likely contributing to limbic seizures in Mecp2(-/y) mice and RTT individuals.
Collapse
Affiliation(s)
- Gaston Calfa
- Department of Neurobiology, Civitan International Research Center, The University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | |
Collapse
|
39
|
Crider A, Pandya CD, Peter D, Ahmed AO, Pillai A. Ubiquitin-proteasome dependent degradation of GABAAα1 in autism spectrum disorder. Mol Autism 2014; 5:45. [PMID: 25392730 PMCID: PMC4228821 DOI: 10.1186/2040-2392-5-45] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/11/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Although the neurobiological basis of autism spectrum disorder (ASD) is not fully understood, recent studies have indicated the potential role of GABAA receptors in the pathophysiology of ASD. GABAA receptors play a crucial role in various neurodevelopmental processes and adult neuroplasticity. However, the mechanism(s) of regulation of GABAA receptors in ASD remains poorly understood. METHODS Postmortem middle frontal gyrus tissues (13 ASD and 13 control subjects) were used. In vitro studies were performed in primary cortical neurons at days in vitro (DIV) 14. The protein levels were examined by western blotting. Immunofluorescence studies were employed for cellular localization. The gene expression was determined by RT-PCR array and qRT-PCR. RESULTS A significant decrease in GABAAα1 protein, but not mRNA levels was found in the middle frontal gyrus of ASD subjects indicating a post-translational regulation of GABAA receptors in ASD. At the cellular level, treatment with proteasomal inhibitor, MG132, or lactacystin significantly increased GABAAα1 protein levels and Lys48-linked polyubiquitination of GABAAα1, but reduced proteasome activity in mouse primary cortical neurons (DIV 14 from E16 embryos). Moreover, treatment with betulinic acid, a proteasome activator significantly decreased GABAAα1 protein levels in cortical neurons indicating the role of polyubiquitination of GABAAα1 proteins with their subsequent proteasomal degradation in cortical neurons. Ubiquitination specific RT-PCR array followed by western blot analysis revealed a significant increase in SYVN1, an endoplasmic reticulum (ER)-associated degradation (ERAD) E3 ubiquitin ligase in the middle frontal gyrus of ASD subjects. In addition, the inhibition of proteasomal activity by MG132 increased the expression of GABAAα1 in the ER. The siRNA knockdown of SYVN1 significantly increased GABAAα1 protein levels in cortical neurons. Moreover, reduced association between SYVN1 and GABAAα1 was found in the middle frontal gyrus of ASD subjects. CONCLUSIONS SYVN1 plays a critical role as an E3 ligase in the ubiquitin proteasome system (UPS)-mediated GABAAα1 degradation. Thus, inhibition of the ubiquitin-proteasome-mediated GABAAα1 degradation may be an important mechanism for preventing GABAAα1 turnover to maintain GABAAα1 levels and GABA signaling in ASD.
Collapse
Affiliation(s)
- Amanda Crider
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Georgia Regents University, 997 St. Sebastian Way, Augusta, GA 30912, USA
| | - Chirayu D Pandya
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Georgia Regents University, 997 St. Sebastian Way, Augusta, GA 30912, USA
| | - Diya Peter
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Georgia Regents University, 997 St. Sebastian Way, Augusta, GA 30912, USA
| | - Anthony O Ahmed
- Department of Psychiatry, Weill Cornell Medical College, 21 Bloomingdale Rd, White Plains, NY 10605, USA
| | - Anilkumar Pillai
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Georgia Regents University, 997 St. Sebastian Way, Augusta, GA 30912, USA
| |
Collapse
|
40
|
Abstract
The clinical symptoms and cognitive and functional deficits of schizophrenia typically begin to gradually emerge during late adolescence and early adulthood. Recent findings suggest that disturbances of a specific subset of inhibitory neurons that contain the calcium-binding protein parvalbumin (PV), which may regulate the course of postnatal developmental experience-dependent synaptic plasticity in the cerebral cortex, including the prefrontal cortex (PFC), may be involved in the pathogenesis of the onset of this illness. Specifically, converging lines of evidence suggest that oxidative stress, extracellular matrix (ECM) deficit and impaired glutamatergic innervation may contribute to the functional impairment of PV neurons, which may then lead to aberrant developmental synaptic pruning of pyramidal cell circuits during adolescence in the PFC. In addition to promoting the functional integrity of PV neurons, maturation of ECM may also play an instrumental role in the termination of developmental PFC synaptic pruning; thus, ECM deficit can directly lead to excessive loss of synapses by prolonging the course of pruning. Together, these mechanisms may contribute to the onset of schizophrenia by compromising the integrity, stability, and fidelity of PFC connectional architecture that is necessary for reliable and predictable information processing. As such, further characterization of these mechanisms will have implications for the conceptualization of rational strategies for the diagnosis, early intervention, and prevention of this debilitating disorder.
Collapse
Affiliation(s)
- Tsung-Ung W Woo
- Laboratory of Cellular Neuropathology, MRC303E, McLean Hospital, 115 Mill Street, Belmont, MA, 02478, USA,
| |
Collapse
|
41
|
Deidda G, Bozarth IF, Cancedda L. Modulation of GABAergic transmission in development and neurodevelopmental disorders: investigating physiology and pathology to gain therapeutic perspectives. Front Cell Neurosci 2014; 8:119. [PMID: 24904277 PMCID: PMC4033255 DOI: 10.3389/fncel.2014.00119] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/14/2014] [Indexed: 01/30/2023] Open
Abstract
During mammalian ontogenesis, the neurotransmitter GABA is a fundamental regulator of neuronal networks. In neuronal development, GABAergic signaling regulates neural proliferation, migration, differentiation, and neuronal-network wiring. In the adult, GABA orchestrates the activity of different neuronal cell-types largely interconnected, by powerfully modulating synaptic activity. GABA exerts these functions by binding to chloride-permeable ionotropic GABAA receptors and metabotropic GABAB receptors. According to its functional importance during development, GABA is implicated in a number of neurodevelopmental disorders such as autism, Fragile X, Rett syndrome, Down syndrome, schizophrenia, Tourette's syndrome and neurofibromatosis. The strength and polarity of GABAergic transmission is continuously modulated during physiological, but also pathological conditions. For GABAergic transmission through GABAA receptors, strength regulation is achieved by different mechanisms such as modulation of GABAA receptors themselves, variation of intracellular chloride concentration, and alteration in GABA metabolism. In the never-ending effort to find possible treatments for GABA-related neurological diseases, of great importance would be modulating GABAergic transmission in a safe and possibly physiological way, without the dangers of either silencing network activity or causing epileptic seizures. In this review, we will discuss the different ways to modulate GABAergic transmission normally at work both during physiological and pathological conditions. Our aim is to highlight new research perspectives for therapeutic treatments that reinstate natural and physiological brain functions in neuro-pathological conditions.
Collapse
Affiliation(s)
- Gabriele Deidda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| | - Ignacio F Bozarth
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| |
Collapse
|
42
|
Boeckx C, Benítez-Burraco A. The shape of the human language-ready brain. Front Psychol 2014; 5:282. [PMID: 24772099 PMCID: PMC3983487 DOI: 10.3389/fpsyg.2014.00282] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/17/2014] [Indexed: 12/14/2022] Open
Abstract
Our core hypothesis is that the emergence of our species-specific language-ready brain ought to be understood in light of the developmental changes expressed at the levels of brain morphology and neural connectivity that occurred in our species after the split from Neanderthals–Denisovans and that gave us a more globular braincase configuration. In addition to changes at the cortical level, we hypothesize that the anatomical shift that led to globularity also entailed significant changes at the subcortical level. We claim that the functional consequences of such changes must also be taken into account to gain a fuller understanding of our linguistic capacity. Here we focus on the thalamus, which we argue is central to language and human cognition, as it modulates fronto-parietal activity. With this new neurobiological perspective in place, we examine its possible molecular basis. We construct a candidate gene set whose members are involved in the development and connectivity of the thalamus, in the evolution of the human head, and are known to give rise to language-associated cognitive disorders. We submit that the new gene candidate set opens up new windows into our understanding of the genetic basis of our linguistic capacity. Thus, our hypothesis aims at generating new testing grounds concerning core aspects of language ontogeny and phylogeny.
Collapse
Affiliation(s)
- Cedric Boeckx
- Catalan Institute for Advanced Studies and Research (ICREA) Barcelona, Spain ; Department of Linguistics, Universitat de Barcelona Barcelona, Spain
| | | |
Collapse
|
43
|
Draper A, Jude L, Jackson GM, Jackson SR. Motor excitability during movement preparation in Tourette syndrome. J Neuropsychol 2013; 9:33-44. [PMID: 24283505 PMCID: PMC4374703 DOI: 10.1111/jnp.12033] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/18/2013] [Indexed: 11/28/2022]
Abstract
Tourette syndrome (TS) is a neurodevelopmental disorder characterized by the occurrence of motor and vocal tics. TS has been linked to the impaired operation of cortical-striatal-thalamic-cortical circuits that give rise to hyper-excitability of cortical motor areas, which may be exacerbated by dysfunctional intra-cortical inhibitory mechanisms. That said, many individuals gain control over their tics during adolescence and it has been suggested that this increased control arises as a result of the development of mechanisms that operate to suppress corticospinal excitability (CSE) ahead of volitional movements. Here we used single-pulse transcranial magnetic stimulation (TMS) in conjunction with a manual Go/NoGo task to investigate alterations in CSE ahead of volitional movements in a group of adolescents with TS (N = 10). Our study demonstrated that CSE, as measured by TMS-induced motor-evoked potentials (MEPs), was significantly reduced in the TS group in the period immediately preceding a finger movement. More specifically, we show that individuals with TS, unlike their age-matched controls, do not exhibit the predicted increase in mean MEP amplitude and decrease in MEP variability that immediately precede the execution of volitional movements in typically developing young adults. Finally, we report that the magnitude of the rise in MEP amplitude across the movement preparation period in TS is significantly negatively correlated with clinical measures of motor tic severity, suggesting that individuals with severe motor tics are least able to modulate motor cortical excitability.
Collapse
|
44
|
A murine model of variant late infantile ceroid lipofuscinosis recapitulates behavioral and pathological phenotypes of human disease. PLoS One 2013; 8:e78694. [PMID: 24223841 PMCID: PMC3815212 DOI: 10.1371/journal.pone.0078694] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 09/24/2013] [Indexed: 01/07/2023] Open
Abstract
Neuronal ceroid lipofuscinoses (NCLs; also known collectively as Batten Disease) are a family of autosomal recessive lysosomal storage disorders. Mutations in as many as 13 genes give rise to ∼10 variants of NCL, all with overlapping clinical symptomatology including visual impairment, motor and cognitive dysfunction, seizures, and premature death. Mutations in CLN6 result in both a variant late infantile onset neuronal ceroid lipofuscinosis (vLINCL) as well as an adult-onset form of the disease called Type A Kufs. CLN6 is a non-glycosylated membrane protein of unknown function localized to the endoplasmic reticulum (ER). In this study, we perform a detailed characterization of a naturally occurring Cln6 mutant (Cln6nclf) mouse line to validate its utility for translational research. We demonstrate that this Cln6nclf mutation leads to deficits in motor coordination, vision, memory, and learning. Pathologically, we demonstrate loss of neurons within specific subregions and lamina of the cortex that correlate to behavioral phenotypes. As in other NCL models, this model displays selective loss of GABAergic interneuron sub-populations in the cortex and the hippocampus with profound, early-onset glial activation. Finally, we demonstrate a novel deficit in memory and learning, including a dramatic reduction in dendritic spine density in the cerebral cortex, which suggests a reduction in synaptic strength following disruption in CLN6. Together, these findings highlight the behavioral and pathological similarities between the Cln6nclf mouse model and human NCL patients, validating this model as a reliable format for screening potential therapeutics.
Collapse
|
45
|
Katayama KI, Imai F, Campbell K, Lang RA, Zheng Y, Yoshida Y. RhoA and Cdc42 are required in pre-migratory progenitors of the medial ganglionic eminence ventricular zone for proper cortical interneuron migration. Development 2013; 140:3139-45. [PMID: 23861058 DOI: 10.1242/dev.092585] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cortical interneurons arise from the ganglionic eminences in the ventral telencephalon and migrate tangentially to the cortex. Although RhoA and Cdc42, members of the Rho family of small GTPases, have been implicated in regulating neuronal migration, their respective roles in the tangential migration of cortical interneurons remain unknown. Here we show that loss of RhoA and Cdc42 in the ventricular zone (VZ) of the medial ganglionic eminence (MGE) using Olig2-Cre mice causes moderate or severe defects in the migration of cortical interneurons, respectively. Furthermore, RhoA- or Cdc42-deleted MGE cells exhibit impaired migration in vitro. To determine whether RhoA and Cdc42 directly regulate the motility of cortical interneurons during migration, we deleted RhoA and Cdc42 in the subventricular zone (SVZ), where more fate-restricted progenitors are located within the ganglionic eminences, using Dlx5/6-Cre-ires-EGFP (Dlx5/6-CIE) mice. Deletion of either gene within the SVZ does not cause any obvious defects in cortical interneuron migration, indicating that cell motility is not dependent upon RhoA or Cdc42. These findings provide genetic evidence that RhoA and Cdc42 are required in progenitors of the MGE in the VZ, but not the SVZ, for proper cortical interneuron migration.
Collapse
Affiliation(s)
- Kei-ichi Katayama
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| | | | | | | | | | | |
Collapse
|
46
|
MPP3 is required for maintenance of the apical junctional complex, neuronal migration, and stratification in the developing cortex. J Neurosci 2013; 33:8518-27. [PMID: 23658188 DOI: 10.1523/jneurosci.5627-12.2013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During mammalian cortical development, division of progenitor cells occurs at the apical ventricular zone. Apical complex proteins and adherens junctions regulate the different modes of division. Here, we have identified the membrane-associated guanylate kinase protein membrane palmitoylated protein 3 (MPP3) as an essential protein for the maintenance of these complexes. MPP3 localizes at the apical membrane in which it shows partial colocalization with adherens junction proteins and apical proteins. We generated Mpp3 conditional knock-out mice and specifically ablated Mpp3 expression in cortical progenitor cells. Conditional deletion of Mpp3 during cortical development resulted in a gradual loss of the apical complex proteins and disrupted adherens junctions. Although there is cellular disorganization in the ventricular zone, gross morphology of the cortex was unaffected during loss of MPP3. However, in the ventricular zone, removal of MPP3 resulted in randomization of spindle orientation and ectopically localized mitotic cells. Loss of MPP3 in the developing cortex resulted in delayed migration of progenitor cells, whereas the rate of cell division and exit from the cell cycle was not affected. This resulted in defects in cortical stratification and ectopically localized layer II-IV pyramidal neurons and interneurons. These data show that MPP3 is required for maintenance of the apical protein complex and adherens junctions and for stratification and proper migration of neurons during the development of the cortex.
Collapse
|
47
|
Gilmore EC, Walsh CA. Genetic causes of microcephaly and lessons for neuronal development. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2013; 2:461-78. [PMID: 24014418 PMCID: PMC3767923 DOI: 10.1002/wdev.89] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The study of human developmental microcephaly is providing important insights into brain development. It has become clear that developmental microcephalies are associated with abnormalities in cellular production, and that the pathophysiology of microcephaly provides remarkable insights into how the brain generates the proper number of neurons that determine brain size. Most of the genetic causes of 'primary' developmental microcephaly (i.e., not associated with other syndromic features) are associated with centrosomal abnormalities. In addition to other functions, centrosomal proteins control the mitotic spindle, which is essential for normal cell proliferation during mitosis. However, the brain is often uniquely affected when microcephaly genes are mutated implying special centrosomal-related functions in neuronal production. Although models explaining how this could occur have some compelling data, they are not without controversy. Interestingly, some of the microcephaly genes show evidence that they were targets of evolutionary selection in primates and human ancestors, suggesting potential evolutionary roles in controlling neuronal number and brain volume across species. Mutations in DNA repair pathway genes also lead to microcephaly. Double-stranded DNA breaks appear to be a prominent type of damage that needs to be repaired during brain development, yet why defects in DNA repair affect the brain preferentially and if DNA repair relates to centrosome function, are not clearly understood.
Collapse
Affiliation(s)
- Edward C Gilmore
- Division of Pediatric Neurology, Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|
48
|
Pohlkamp T, Dávid C, Cauli B, Gallopin T, Bouché E, Karagiannis A, May P, Herz J, Frotscher M, Staiger JF, Bock HH. Characterization and distribution of Reelin-positive interneuron subtypes in the rat barrel cortex. ACTA ACUST UNITED AC 2013; 24:3046-58. [PMID: 23803971 DOI: 10.1093/cercor/bht161] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
GABAergic inhibitory interneurons (IN) represent a heterogeneous population with different electrophysiological, morphological, and molecular properties. The correct balance between interneuronal subtypes is important for brain function and is impaired in several neurological and psychiatric disorders. Here we show the data of 123 molecularly and electrophysiologically characterized neurons of juvenile rat barrel cortex acute slices, 48 of which expressed Reelin (Reln). Reln mRNA was exclusively detected in Gad65/67-positive cells but was found in interneuronal subtypes in different proportions: all cells of the adapting-Somatostatin (SST) cluster expressed Reln, whereas 63% of the adapting-neuropeptide Y (NPY, 50% of the fast-spiking Parvalbumin (PVALB), and 27% of the adapting/bursting-Vasoactive Intestinal Peptide (VIP) cluster were Reln-positive. Silhouette analysis revealed a high impact of the parameter Reln on cluster quality. By analyzing the co-localization of RELN immunoreactivity with those of different IN-markers, we found that RELN is produced layer-independently in SST-, NPY-, and NOS1-expressing INs, whereas co-localization of RELN and VIP was mostly absent. Of note, RELN co-localized with PVALB, predominantly in INs of layers IV/V (>30%). Our findings emphasize RELN's role as an important IN-marker protein and provide a basis for the functional characterization of Reln-expressing INs and its role in the regulation of inhibitory IN networks.
Collapse
Affiliation(s)
- Theresa Pohlkamp
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Csaba Dávid
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany Department of Human Morphology and Developmental Biology, Semmelweis University, H1094 Budapest, Hungary
| | - Bruno Cauli
- Neurobiologie des Processus Adaptatifs, CNRS UMR7102, Université Pierre et Marie Curie, F75005 Paris, France
| | - Thierry Gallopin
- Laboratoire de Neurobiologie, CNRS UMR7637, Ecole Supérieure de Physique et de Chimie Industrielles de Paris, F75005 Paris, France
| | - Elisabeth Bouché
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany
| | - Anastassios Karagiannis
- Neurobiologie des Processus Adaptatifs, CNRS UMR7102, Université Pierre et Marie Curie, F75005 Paris, France
| | - Petra May
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany Clinic for Gastroenterology, Hepatology and Infectiology, Heinrich-Heine-University, D40225 Düsseldorf, Germany
| | - Joachim Herz
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Michael Frotscher
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany Institute for Structural Neurobiology, Center for Molecular Neurobiology, D20251 Hamburg, Germany and
| | - Jochen F Staiger
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany Institute for Neuroanatomy, UMG, Georg-August-University, D37075 Göttingen, Germany
| | - Hans H Bock
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, D79104 Freiburg, Germany Clinic for Gastroenterology, Hepatology and Infectiology, Heinrich-Heine-University, D40225 Düsseldorf, Germany
| |
Collapse
|
49
|
Catts VS, Fung SJ, Long LE, Joshi D, Vercammen A, Allen KM, Fillman SG, Rothmond DA, Sinclair D, Tiwari Y, Tsai SY, Weickert TW, Shannon Weickert C. Rethinking schizophrenia in the context of normal neurodevelopment. Front Cell Neurosci 2013; 7:60. [PMID: 23720610 PMCID: PMC3654207 DOI: 10.3389/fncel.2013.00060] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 04/16/2013] [Indexed: 01/11/2023] Open
Abstract
The schizophrenia brain is differentiated from the normal brain by subtle changes, with significant overlap in measures between normal and disease states. For the past 25 years, schizophrenia has increasingly been considered a neurodevelopmental disorder. This frame of reference challenges biological researchers to consider how pathological changes identified in adult brain tissue can be accounted for by aberrant developmental processes occurring during fetal, childhood, or adolescent periods. To place schizophrenia neuropathology in a neurodevelopmental context requires solid, scrutinized evidence of changes occurring during normal development of the human brain, particularly in the cortex; however, too often data on normative developmental change are selectively referenced. This paper focuses on the development of the prefrontal cortex and charts major molecular, cellular, and behavioral events on a similar time line. We first consider the time at which human cognitive abilities such as selective attention, working memory, and inhibitory control mature, emphasizing that attainment of full adult potential is a process requiring decades. We review the timing of neurogenesis, neuronal migration, white matter changes (myelination), and synapse development. We consider how molecular changes in neurotransmitter signaling pathways are altered throughout life and how they may be concomitant with cellular and cognitive changes. We end with a consideration of how the response to drugs of abuse changes with age. We conclude that the concepts around the timing of cortical neuronal migration, interneuron maturation, and synaptic regression in humans may need revision and include greater emphasis on the protracted and dynamic changes occurring in adolescence. Updating our current understanding of post-natal neurodevelopment should aid researchers in interpreting gray matter changes and derailed neurodevelopmental processes that could underlie emergence of psychosis.
Collapse
Affiliation(s)
- Vibeke S. Catts
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Samantha J. Fung
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Leonora E. Long
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Medical Sciences, University of New South WalesSydney, NSW, Australia
| | - Dipesh Joshi
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Ans Vercammen
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
- School of Psychology, Australian Catholic UniversitySydney, NSW, Australia
| | - Katherine M. Allen
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Stu G. Fillman
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Debora A. Rothmond
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
| | - Duncan Sinclair
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Yash Tiwari
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Medical Sciences, University of New South WalesSydney, NSW, Australia
| | - Shan-Yuan Tsai
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Thomas W. Weickert
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Schizophrenia Research InstituteSydney, NSW, Australia
- Neuroscience Research AustraliaSydney, NSW, Australia
- School of Psychiatry, University of New South WalesSydney, NSW, Australia
| |
Collapse
|
50
|
Van den Boomen C, de Graaff JC, de Jong TPVM, Kalkman CJ, Kemner C. General anesthesia as a possible GABAergic modulator affects visual processing in children. Front Cell Neurosci 2013; 7:42. [PMID: 23630461 PMCID: PMC3632787 DOI: 10.3389/fncel.2013.00042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 03/28/2013] [Indexed: 11/13/2022] Open
Abstract
Gamma-Aminobutyric Acid (GABA) inhibitory interneurons play an important role in visual processing, as is revealed by studies administering drugs in human and monkey adults. Investigating this process in children requires different methodologies, due to ethical considerations. The current study aimed to investigate whether a new method, being general anesthesia using Sevoflurane, can be used to trace the effects of GABAergic modulation on visual brain functioning in children. To this aim, visual processing was investigated in children aged 4–12 years who were scheduled for minor urologic procedures under general anesthesia in day-care treatment. In a visual segmentation task, the difference in Event-Related Potential (ERP) response to homogeneous and textured stimuli was investigated. In addition, psychophysical performance on visual acuity and contrast sensitivity were measured. Results were compared between before and shortly after anesthesia. In two additional studies, effects at 1 day after anesthesia and possible effects of task-repetition were investigated. ERP results showed longer latency and lower amplitude of the Texture Negativity (TN) component shortly after compared to before anesthesia. No effects of anesthesia on psychophysical measurements were found. No effects at 1 day after anesthesia or of repetition were revealed either. These results show that GABAergic modulation through general anesthesia affects ERP reflections of visual segmentation in a similar way in children as benzodiazepine does in adults, but that effects are not permanent. This demonstrates that ERP measurement after anesthesia is a successful method to study effects of GABAergic modulation in children.
Collapse
Affiliation(s)
- C Van den Boomen
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University Utrecht, Netherlands ; Department of Developmental Psychology, Utrecht University Utrecht, Netherlands
| | | | | | | | | |
Collapse
|