Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
1.
Nat Commun ; 15(1): 6534, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095390

ABSTRACT

Huntington's disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.


Subject(s)
Huntington Disease , Organoids , Phenotype , Telencephalon , Huntington Disease/pathology , Huntington Disease/genetics , Huntington Disease/metabolism , Organoids/pathology , Organoids/metabolism , Animals , Telencephalon/pathology , Telencephalon/cytology , Telencephalon/metabolism , Humans , Mice , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Single-Cell Analysis , Cell Communication , Mosaicism , Neurons/metabolism , Neurons/pathology
2.
Nat Commun ; 15(1): 5524, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951485

ABSTRACT

The three-dimensional genome structure organized by CTCF is required for development. Clinically identified mutations in CTCF have been linked to adverse developmental outcomes. Nevertheless, the underlying mechanism remains elusive. In this investigation, we explore the regulatory roles of a clinically relevant R567W point mutation, located within the 11th zinc finger of CTCF, by introducing this mutation into both murine models and human embryonic stem cell-derived cortical organoid models. Mice with homozygous CTCFR567W mutation exhibit growth impediments, resulting in postnatal mortality, and deviations in brain, heart, and lung development at the pathological and single-cell transcriptome levels. This mutation induces premature stem-like cell exhaustion, accelerates the maturation of GABAergic neurons, and disrupts neurodevelopmental and synaptic pathways. Additionally, it specifically hinders CTCF binding to peripheral motifs upstream to the core consensus site, causing alterations in local chromatin structure and gene expression, particularly at the clustered protocadherin locus. Comparative analysis using human cortical organoids mirrors the consequences induced by this mutation. In summary, this study elucidates the influence of the CTCFR567W mutation on human neurodevelopmental disorders, paving the way for potential therapeutic interventions.


Subject(s)
CCCTC-Binding Factor , Neurodevelopmental Disorders , Organoids , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Humans , Animals , Mice , Neurodevelopmental Disorders/genetics , Organoids/metabolism , Mutation , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Male , Chromatin/metabolism , Chromatin/genetics , Female , Brain/metabolism , Brain/pathology , Point Mutation , Human Embryonic Stem Cells/metabolism
3.
Eur J Pharmacol ; 977: 176756, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38897021

ABSTRACT

Repeated exposure to propofol during early brain development is associated with anxiety disorders in adulthood, yet the mechanisms underlying propofol-induced susceptibility to anxiety disorders remain elusive. The lateral septum (LS), primarily composed of γ-aminobutyric acidergic (GABAergic) neurons, serves as a key brain region in the regulation of anxiety. However, it remains unclear whether LS GABAergic neurons are implicated in propofol-induced anxiety. Therefore, we conducted c-Fos immunostaining of whole-brain slices from mice exposed to propofol during early life. Our findings indicate that propofol exposure activates GABAergic neurons in the LS. Selective activation of LS GABAergic neurons resulted in increased anxiety-like behavior, while selective inhibition of these neurons reduced such behaviors. These results suggest that the LS is a critical brain region involved in propofol-induced anxiety. Furthermore, we investigated the molecular mechanism of propofol-induced anxiety in the LS. Microglia activation underlies the development of anxiety. Immunofluorescence staining and Western blot analysis of LS revealed activated microglia and significantly elevated levels of phospho-NF-κB p65 protein. Additionally, a decrease in the number of neuronal spines was observed. Our study highlights the crucial role of the LS in the development of anxiety-like behavior in adulthood following childhood propofol exposure, accompanied by the activation of inflammatory pathways.


Subject(s)
Anxiety , Behavior, Animal , GABAergic Neurons , Microglia , Propofol , Propofol/pharmacology , Animals , Anxiety/chemically induced , Mice , Male , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Behavior, Animal/drug effects , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Proto-Oncogene Proteins c-fos/metabolism , Mice, Inbred C57BL , Transcription Factor RelA/metabolism , Dendritic Spines/drug effects , Dendritic Spines/pathology , Dendritic Spines/metabolism
4.
Proc Natl Acad Sci U S A ; 121(22): e2316176121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38771878

ABSTRACT

The striato-nigral (Str-SN) circuit is composed of medium spiny neuronal projections that are mainly sent from the striatum to the midbrain substantial nigra (SN), which is essential for regulating motor behaviors. Dysfunction of the Str-SN circuitry may cause a series of motor disabilities that are associated with neurodegenerative disorders, such as Huntington's disease (HD). Although the etiology of HD is known as abnormally expanded CAG repeats of the huntingtin gene, treatment of HD remains tremendously challenging. One possible reason is the lack of effective HD model that resembles Str-SN circuitry deficits for pharmacological studies. Here, we first differentiated striatum-like organoids from human pluripotent stem cells (hPSCs), containing functional medium spiny neurons (MSNs). We then generated 3D Str-SN assembloids by assembling striatum-like organoids with midbrain SN-like organoids. With AAV-hSYN-GFP-mediated viral tracing, extensive MSN projections from the striatum to the SN are established, which formed synaptic connection with GABAergic neurons in SN organoids and showed the optically evoked inhibitory postsynaptic currents and electronic field potentials by labeling the striatum-like organoids with optogenetic virus. Furthermore, these Str-SN assembloids exhibited enhanced calcium activity compared to that of individual striatal organoids. Importantly, we further demonstrated the reciprocal projection defects in HD iPSC-derived assembloids, which could be ameliorated by treatment of brain-derived neurotrophic factor. Taken together, these findings suggest that Str-SN assembloids could be used for identifying MSN projection defects and could be applied as potential drug test platforms for HD.


Subject(s)
Huntington Disease , Organoids , Humans , Huntington Disease/pathology , Huntington Disease/metabolism , Organoids/pathology , Organoids/metabolism , Substantia Nigra/pathology , Substantia Nigra/metabolism , Corpus Striatum/pathology , Corpus Striatum/metabolism , Neurons/metabolism , Neurons/pathology , Cell Differentiation , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Pluripotent Stem Cells/metabolism , Optogenetics
5.
Acta Neuropathol ; 147(1): 80, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714540

ABSTRACT

GABAergic interneurons play a critical role in maintaining neural circuit balance, excitation-inhibition regulation, and cognitive function modulation. In tuberous sclerosis complex (TSC), GABAergic neuron dysfunction contributes to disrupted network activity and associated neurological symptoms, assumingly in a cell type-specific manner. This GABAergic centric study focuses on identifying specific interneuron subpopulations within TSC, emphasizing the unique characteristics of medial ganglionic eminence (MGE)- and caudal ganglionic eminence (CGE)-derived interneurons. Using single-nuclei RNA sequencing in TSC patient material, we identify somatostatin-expressing (SST+) interneurons as a unique and immature subpopulation in TSC. The disrupted maturation of SST+ interneurons may undergo an incomplete switch from excitatory to inhibitory GABAergic signaling during development, resulting in reduced inhibitory properties. Notably, this study reveals markers of immaturity specifically in SST+ interneurons, including an abnormal NKCC1/KCC2 ratio, indicating an imbalance in chloride homeostasis crucial for the postsynaptic consequences of GABAergic signaling as well as the downregulation of GABAA receptor subunits, GABRA1, and upregulation of GABRA2. Further exploration of SST+ interneurons revealed altered localization patterns of SST+ interneurons in TSC brain tissue, concentrated in deeper cortical layers, possibly linked to cortical dyslamination. In the epilepsy context, our research underscores the diverse cell type-specific roles of GABAergic interneurons in shaping seizures, advocating for precise therapeutic considerations. Moreover, this study illuminates the potential contribution of SST+ interneurons to TSC pathophysiology, offering insights for targeted therapeutic interventions.


Subject(s)
GABAergic Neurons , Interneurons , Tuberous Sclerosis , Humans , GABAergic Neurons/pathology , GABAergic Neurons/metabolism , Ganglionic Eminence , Interneurons/pathology , Interneurons/metabolism , Median Eminence/pathology , Median Eminence/metabolism , Receptors, GABA-A/metabolism , Somatostatin/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis/metabolism , Animals
6.
Neuroscience ; 546: 63-74, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38537894

ABSTRACT

GABAergic interneurons and perineuronal nets (PNNs) are important regulators of plasticity throughout life and their dysfunction has been implicated in the pathogenesis of several neuropsychiatric conditions, including autism spectrum disorders (ASD). PNNs are condensed portions of the extracellular matrix (ECM) that are crucial for neural development and proper formation of synaptic connections. We previously showed a reduced expression of GABAergic interneuron markers in the hippocampus and somatosensory cortex of adult mice lacking the Engrailed2 gene (En2-/- mice), a mouse model of ASD. Since alterations in PNNs have been proposed as a possible pathogenic mechanism in ASD, we hypothesized that the PNN dysfunction may contribute to the neural and behavioral abnormalities of En2-/- mice. Here, we show an increase in the PNN fluorescence intensity, evaluated by Wisteria floribunda agglutinin, in brain regions involved in social behavior and somatosensory processing. In addition, we found that En2-/- mice exhibit altered texture discrimination through whiskers and display a marked decrease in the preference for social novelty. Our results raise the possibility that altered expression of PNNs, together with defects of GABAergic interneurons, might contribute to the pathogenesis of social and sensory behavioral abnormalities.


Subject(s)
Homeodomain Proteins , Mice, Knockout , Nerve Tissue Proteins , Plant Lectins , Social Behavior , Vibrissae , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Male , Mice, Inbred C57BL , Extracellular Matrix/metabolism , Interneurons/metabolism , Disease Models, Animal , Mice , Somatosensory Cortex/metabolism , Discrimination, Psychological/physiology , Receptors, N-Acetylglucosamine/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Brain/metabolism , Brain/pathology
7.
Stem Cell Res Ther ; 14(1): 170, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37365654

ABSTRACT

BACKGROUND: Brainstem stroke causes severe and persistent neurological impairment. Due to the limited spontaneous recovery and regeneration of the disrupted neural circuits, transplantation of exogenous neural stem cells (NSCs) was an alternative, while there were limitations for primitive NSCs. METHODS: We established a mouse model of brainstem stroke by injecting endothelin in the right pons. Brain-derived neurotrophic factor (BDNF)- and distal-less homeobox 2 (Dlx2)-modified NSCs were transplanted to treat brainstem stroke. Transsynaptic viral tracking, immunostaining, magnetic resonance imaging, behavioral testing, and whole-cell patch clamp recordings were applied to probe the pathophysiology and therapeutic prospects of BDNF- and Dlx2-modified NSCs. RESULTS: GABAergic neurons were predominantly lost after the brainstem stroke. No endogenous NSCs were generated in situ or migrated from the neurogenesis niches within the brainstem infarct region. Co-overexpressions of BDNF and Dlx2 not only promoted the survival of NSCs, but also boosted the differentiation of NSCs into GABAergic neurons. Results from transsynaptic virus tracking, immunostaining, and evidence from whole-cell patch clamping revealed the morphological and functional integration of the grafted BDNF- and Dlx2-modified NSCs-derived neurons with the host neural circuits. Neurological function was improved by transplantation of BDNF- and Dlx2-modified NSCs in brainstem stroke. CONCLUSIONS: These findings demonstrated that BDNF- and Dlx2-modified NSCs differentiated into GABAergic neurons, integrated into and reconstituted the host neural networks, and alleviated the ischemic injury. It thus provided a potential therapeutic strategy for brainstem stroke.


Subject(s)
Neural Stem Cells , Stroke , Mice , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Differentiation , Disease Models, Animal , GABAergic Neurons/pathology , Stroke/therapy , Stroke/pathology
8.
Am J Psychiatry ; 180(7): 495-507, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37073488

ABSTRACT

OBJECTIVE: In schizophrenia, somatostatin (SST) and parvalbumin (PV) mRNA levels are lower in the dorsolateral prefrontal cortex (DLPFC), but it remains unclear whether these findings reflect lower transcript levels per neuron, fewer neurons, or both. Distinguishing among these alternatives has implications for understanding the pathogenesis of, and developing new treatments for, DLPFC dysfunction in schizophrenia. METHODS: To identify SST and PV neurons in postmortem human DLPFC, the authors used fluorescent in situ hybridization to label cells expressing two transcripts not altered in schizophrenia: vesicular GABA transporter (VGAT; a marker of all GABA neurons) and SOX6 (a marker of only SST and PV neurons). In cortical layers 2 and 4, where SST and PV neurons, respectively, are differentially enriched, levels of SST and PV mRNA per neuron and the relative densities of SST-, PV-, and VGAT/SOX6-positive neurons were quantified. RESULTS: In individuals with schizophrenia, mRNA levels per positive neuron were markedly and significantly lower for SST in both layers (effect sizes >1.48) and for PV only in layer 4 (effect size=1.14) relative to matched unaffected individuals. In contrast, the relative densities of all SST-, PV-, or VGAT/SOX6-positive neurons were unaltered in schizophrenia. CONCLUSIONS: Novel multiplex fluorescent in situ hybridization techniques permit definitive distinction between cellular levels of transcripts and the presence of neurons expressing those transcripts. In schizophrenia, pronounced SST and PV mRNA deficits are attributable to lower levels of each transcript per neuron, not fewer neurons, arguing against death or abnormal migration of these neurons. Instead, these neurons appear to be functionally altered and thus amenable to therapeutic interventions.


Subject(s)
Schizophrenia , Humans , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Expression/genetics , In Situ Hybridization, Fluorescence , Parvalbumins/genetics , Parvalbumins/metabolism , Prefrontal Cortex , RNA, Messenger/genetics , RNA, Messenger/metabolism , Somatostatin/genetics , Somatostatin/metabolism
9.
Cells ; 12(2)2023 01 04.
Article in English | MEDLINE | ID: mdl-36672158

ABSTRACT

Multiple system atrophy of the parkinsonian type (MSA-P) is a rare, fatal neurodegenerative disease with sporadic onset. It is still unknown if MSA-P is a primary oligodendropathy or caused by neuronal pathophysiology leading to severe, α-synuclein-associated neurodegeneration, mainly in the striatum. In this study, we generated and differentiated induced pluripotent stem cells (iPSCs) from patients with the clinical diagnosis of probable MSA-P (n = 3) and from three matched healthy controls into GABAergic striatal medium spiny neurons (MSNs). We found a significantly elevated release and neuronal distribution for α-synuclein, as well as hypoexcitability in the MSNs derived from the MSA-P patients compared to the healthy controls. These data suggest that the striatal hypoexcitable neurons of MSA-P patients contribute to a pathological α-synuclein burden which is likely to spread to neighboring cells and projection targets, facilitating disease progression.


Subject(s)
Induced Pluripotent Stem Cells , Multiple System Atrophy , Humans , Multiple System Atrophy/pathology , alpha-Synuclein , Induced Pluripotent Stem Cells/pathology , Medium Spiny Neurons , GABAergic Neurons/pathology
10.
Nature ; 602(7896): 268-273, 2022 02.
Article in English | MEDLINE | ID: mdl-35110736

ABSTRACT

Genetic risk for autism spectrum disorder (ASD) is associated with hundreds of genes spanning a wide range of biological functions1-6. The alterations in the human brain resulting from mutations in these genes remain unclear. Furthermore, their phenotypic manifestation varies across individuals7,8. Here we used organoid models of the human cerebral cortex to identify cell-type-specific developmental abnormalities that result from haploinsufficiency in three ASD risk genes-SUV420H1 (also known as KMT5B), ARID1B and CHD8-in multiple cell lines from different donors, using single-cell RNA-sequencing (scRNA-seq) analysis of more than 745,000 cells and proteomic analysis of individual organoids, to identify phenotypic convergence. Each of the three mutations confers asynchronous development of two main cortical neuronal lineages-γ-aminobutyric-acid-releasing (GABAergic) neurons and deep-layer excitatory projection neurons-but acts through largely distinct molecular pathways. Although these phenotypes are consistent across cell lines, their expressivity is influenced by the individual genomic context, in a manner that is dependent on both the risk gene and the developmental defect. Calcium imaging in intact organoids shows that these early-stage developmental changes are followed by abnormal circuit activity. This research uncovers cell-type-specific neurodevelopmental abnormalities that are shared across ASD risk genes and are finely modulated by human genomic context, finding convergence in the neurobiological basis of how different risk genes contribute to ASD pathology.


Subject(s)
Autism Spectrum Disorder , Genetic Predisposition to Disease , Neurons , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Cerebral Cortex/cytology , DNA-Binding Proteins/genetics , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Histone-Lysine N-Methyltransferase/genetics , Humans , Neurons/classification , Neurons/metabolism , Neurons/pathology , Organoids/cytology , Proteomics , RNA-Seq , Single-Cell Analysis , Transcription Factors/genetics
11.
J Neural Transm (Vienna) ; 129(5-6): 643-647, 2022 06.
Article in English | MEDLINE | ID: mdl-34935080

ABSTRACT

Research in Peter Riederer's lab in Vienna in the late 1970's came from a strong tradition in post-mortem neurochemical studies, at that time a relatively niche approach in neuroscience research. He was also early to recognise the value of post-mortem brain tissue in elucidating pharmacological mechanisms of neuropsychiatric treatments. I was fortunate to have Peter Riederer as a mentor in my early post-doctoral career; his generous support and the opportunities to use post-mortem brain tissue provided an invaluable grounding on which much of my future research was based. In this paper, I shall provide a brief overview of one trajectory of my research into the neurobiology of schizophrenia that started in the Riederer lab in Vienna investigating dopamine and the D2 receptor. Subsequent research to understand findings of increased dopamine resulted in the identification of reduced GABAergic innervation, culminating in the finding of a deficit in the parvalbumin-containing subtype of GABAergic neurons. Most recent work has been studying how changes in DNA methylation of the parvalbumin gene may relate to these findings in psychotic illness and its animal models.


Subject(s)
Parvalbumins , Schizophrenia , Animals , Dopamine , GABAergic Neurons/pathology , Male , Receptors, Dopamine D2/genetics , Schizophrenia/pathology
12.
Toxicology ; 465: 153012, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34718030

ABSTRACT

Rare earth elements (REEs) are widely used in the industry, agriculture, biomedicine, aerospace, etc, and have been shown to pose toxic effects on animals, as such, studies focusing on their biomedical properties are gaining wide attention. However, environmental and population health risks of REEs are still not very clear. Also, the REEs damage to the nervous system and related molecular mechanisms needs further research. In this study, the L1 and L4 stages of the model organism Caenorhabditis elegans were used to evaluate the effects and possible neurotoxic mechanism of lanthanum(III) nitrate hexahydrate (La(NO3)3·6H2O). For the L1 and L4 stage worms, the 48-h median lethal concentrations (LC50s) of La(NO3)3·6H2O were 93.163 and 648.0 mg/L respectively. Our results show that La(NO3)3·6H2O induces growth inhibition and defects in behavior such as body length, body width, body bending frequency, head thrashing frequency and pharyngeal pumping frequency at the L1 and L4 stages in C. elegans. The L1 stage is more sensitive to the toxicity of lanthanum than the L4 stage worms. Using transgenic strains (BZ555, EG1285 and NL5901), we found that La(NO3)3·6H2O caused the loss or break of soma and dendrite neurons in L1 and L4 stages; and α-synuclein aggregation in L1 stage, indicating that Lanthanum can cause toxic damage to dopaminergic and GABAergic neurons. Mechanistically, La(NO3)3·6H2O exposure inhibited or activated the neurotransmitter transporters and receptors (glutamate, serotonin and dopamine) in C. elegans, which regulate behavior and movement functions. Furthermore, significant increase in the production of reactive oxygen species (ROS) was found in the L4 stage C. elegans exposed to La(NO3)3·6H2O. Altogether, our data show that exposure to lanthanum can cause neuronal toxic damage and behavioral defects in C. elegans, and provide basic information for understanding the neurotoxic effect mechanism and environmental health risks of rare earth elements.


Subject(s)
Behavior, Animal/drug effects , Caenorhabditis elegans/drug effects , Dopaminergic Neurons/drug effects , GABAergic Neurons/drug effects , Gene Expression Regulation, Developmental/drug effects , Lanthanum/toxicity , Neurotoxicity Syndromes/etiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Lethal Dose 50 , Movement/drug effects , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Reactive Oxygen Species/metabolism , Risk Assessment , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
13.
Cell Rep ; 37(13): 110177, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965426

ABSTRACT

The hippocampus is a temporal lobe structure critical for cognition, such as learning, memory, and attention, as well as emotional responses. Hippocampal dysfunction can lead to persistent anxiety and/or depression. However, how millions of neurons in the hippocampus are molecularly and structurally organized to engage their divergent functions remains unknown. Here, we genetically target a subset of neurons expressing the coagulation factor c homolog (COCH) gene. COCH-expressing neurons or COCH neurons are topographically segregated in the distal region of the ventral CA3 hippocampus and express Mtf1 and Cacna1h. MTF1 activation of Cacna1h transcription in COCH neurons encodes the ability of COCH neurons to burst action potentials and cause social-stress-induced anxiety-like behaviors by synapsing directly with a subset of GABAergic inhibitory neurons in the lateral septum. Together, this study provides a molecular and circuitry-based framework for understanding how COCH neurons in the hippocampus are assembled to engage social behavior.


Subject(s)
Action Potentials , Anxiety/pathology , CA3 Region, Hippocampal/pathology , Extracellular Matrix Proteins/metabolism , GABAergic Neurons/pathology , Social Behavior , Stress, Psychological , Animals , Anxiety/etiology , Anxiety/metabolism , CA3 Region, Hippocampal/metabolism , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Emotions , Extracellular Matrix Proteins/genetics , Fear , GABAergic Neurons/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factor MTF-1
14.
Cells ; 10(11)2021 11 03.
Article in English | MEDLINE | ID: mdl-34831225

ABSTRACT

Understanding seizure development requires an integrated knowledge of different scales of organization of epileptic networks. We developed a model of "epilepsy-in-a-dish" based on dissociated primary neuronal cells from neonatal rat hippocampus. We demonstrate how a single application of glutamate stimulated neurons to generate spontaneous synchronous spiking activity with further progression into spontaneous seizure-like events after a distinct latency period. By computational analysis, we compared the observed neuronal activity in vitro with intracranial electroencephalography (EEG) data recorded from epilepsy patients and identified strong similarities, including a related sequence of events with defined onset, progression, and termination. Next, a link between the neurophysiological changes with network composition and cellular structure down to molecular changes was established. Temporal development of epileptiform network activity correlated with increased neurite outgrowth and altered branching, increased ratio of glutamatergic over GABAergic synapses, and loss of calbindin-positive interneurons, as well as genome-wide alterations in DNA methylation. Differentially methylated genes were engaged in various cellular activities related to cellular structure, intracellular signaling, and regulation of gene expression. Our data provide evidence that a single short-term excess of glutamate is sufficient to induce a cascade of events covering different scales from molecule- to network-level, all of which jointly contribute to seizure development.


Subject(s)
Brain/pathology , Epilepsy/pathology , Models, Biological , Neurons/pathology , Animals , Calbindins/metabolism , Calcium/metabolism , Cells, Cultured , DNA Methylation/genetics , Epigenesis, Genetic , Epilepsy/genetics , GABAergic Neurons/pathology , Gene Regulatory Networks , Neurons/metabolism , Principal Component Analysis , Rats , Time Factors
15.
J Clin Invest ; 131(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34491914

ABSTRACT

Spreading depolarizations (SDs) are involved in migraine, epilepsy, stroke, traumatic brain injury, and subarachnoid hemorrhage. However, the cellular origin and specific differential mechanisms are not clear. Increased glutamatergic activity is thought to be the key factor for generating cortical spreading depression (CSD), a pathological mechanism of migraine. Here, we show that acute pharmacological activation of NaV1.1 (the main Na+ channel of interneurons) or optogenetic-induced hyperactivity of GABAergic interneurons is sufficient to ignite CSD in the neocortex by spiking-generated extracellular K+ build-up. Neither GABAergic nor glutamatergic synaptic transmission were required for CSD initiation. CSD was not generated in other brain areas, suggesting that this is a neocortex-specific mechanism of CSD initiation. Gain-of-function mutations of NaV1.1 (SCN1A) cause familial hemiplegic migraine type-3 (FHM3), a subtype of migraine with aura, of which CSD is the neurophysiological correlate. Our results provide the mechanism linking NaV1.1 gain of function to CSD generation in FHM3. Thus, we reveal the key role of hyperactivity of GABAergic interneurons in a mechanism of CSD initiation, which is relevant as a pathological mechanism of Nav1.1 FHM3 mutations, and possibly also for other types of migraine and diseases in which SDs are involved.


Subject(s)
Cortical Spreading Depression , GABAergic Neurons/metabolism , Interneurons/metabolism , Migraine Disorders/metabolism , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Neocortex/metabolism , Animals , GABAergic Neurons/pathology , Interneurons/pathology , Mice , Mice, Transgenic , Migraine Disorders/genetics , Migraine Disorders/pathology , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neocortex/pathology
16.
Neurochem Int ; 150: 105179, 2021 11.
Article in English | MEDLINE | ID: mdl-34500023

ABSTRACT

Schizophrenia is a major psychiatric disorder, but the molecular mechanisms leading to its initiation or progression remain unclear. To elucidate the pathophysiology of schizophrenia, we used an in vitro neuronal cell culture model involving human induced pluripotent stem cells (hiPSCs) derived from a monozygotic-twin discordant schizophrenia pair. The cultured neurons differentiated from hiPSCs were composed of a mixture of glutamatergic excitatory neurons and gamma aminobutyric acid (GABA)ergic inhibitory neurons. In the electrophysiological analysis, a different pattern of spontaneous neuronal activity was observed under the condition without any stimulants. The frequency of spontaneous excitatory post-synaptic currents (sEPSCs) was significantly higher in the hiPSC-derived neurons of the patient with schizophrenia than in the control sibling at day-in-vitro 30. However, the synaptic formation was not different between the patient with schizophrenia and the control sibling during the same culture period. To explain underlying mechanisms of higher excitability of presynaptic cells, we focused on the potassium-chloride co-transporter KCC2, which contributes to excitatory-to-inhibitory GABA polarity switch in developing neurons. We also revealed the altered expression pattern of KCC2 in hiPSC-derived neurons from the patient with schizophrenia, which could contribute to understanding the pathology of schizophrenia in the developing nervous system.


Subject(s)
GABAergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Schizophrenia/metabolism , Symporters/biosynthesis , Twins, Monozygotic , Cell Differentiation/physiology , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Fibroblasts/metabolism , Fibroblasts/pathology , GABAergic Neurons/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Male , Neural Inhibition/physiology , Neurons/pathology , Schizophrenia/genetics , Schizophrenia/pathology , Symporters/genetics , Twins, Monozygotic/genetics , Young Adult
17.
Eur J Med Genet ; 64(9): 104282, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34284163

ABSTRACT

The Forkhead transcription factor FOXG1 is a prerequisite for telencephalon development in mammals and is an essential factor controlling expansion of the dorsal telencephalon by promoting neuron and interneuron production. Heterozygous FOXG1 gene mutations cause FOXG1 syndrome characterized by severe intellectual disability, motor delay, dyskinetic movements and epilepsy. Neuroimaging studies in patients disclose constant features including microcephaly, corpus callosum dysgenesis and delayed myelination. Currently, investigative research on the underlying pathophysiology relies on mouse models only and indicates that de-repression of FOXG1 target genes may cause premature neuronal differentiation at the expense of the progenitor pool, patterning and migration defects with impaired formation of cortico-cortical projections. It remains an open question to which extent this recapitulates the neurodevelopmental pathophysiology in FOXG1-haploinsufficient patients. To close this gap, we performed neuropathological analyses in two foetal cases with FOXG1 premature stop codon mutations interrupted during the third trimester of the pregnancy for microcephaly and corpus callosum dysgenesis. In these foetuses, we observed cortical lamination defects and decreased neuronal density mainly affecting layers II, III and V that normally give rise to cortico-cortical and inter-hemispheric axonal projections. GABAergic interneurons were also reduced in number in the cortical plate and persisting germinative zones. Additionally, we observed more numerous PDGFRα-positive oligodendrocyte precursor cells and fewer Olig2-positive pre-oligodendrocytes compared to age-matched control brains, arguing for delayed production and differentiation of oligodendrocyte lineage leading to delayed myelination. These findings provide key insights into the human pathophysiology of FOXG1 syndrome.


Subject(s)
Agenesis of Corpus Callosum/genetics , Axons/pathology , Forkhead Transcription Factors/genetics , Microcephaly/genetics , Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/genetics , Neurogenesis , Oligodendroglia/pathology , Aborted Fetus/metabolism , Aborted Fetus/pathology , Adult , Agenesis of Corpus Callosum/pathology , Axons/metabolism , Brain/embryology , Brain/metabolism , Brain/pathology , Codon, Nonsense , Female , Forkhead Transcription Factors/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Humans , Interneurons/metabolism , Interneurons/pathology , Microcephaly/pathology , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , Neurodevelopmental Disorders/pathology , Oligodendroglia/metabolism , Pedigree , Pregnancy , Syndrome
18.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299211

ABSTRACT

Glaucoma is a leading cause of irreversible blindness worldwide, and increased intraocular pressure (IOP) is a major risk factor. We aimed to determine if early functional and molecular differences in the glaucomatous retina manifest before significant retinal ganglion cell (RGC) loss is apparent. Adenoviral vectors expressing a pathogenic form of myocilin (Ad5.MYOC) were used to induce IOP elevation in C57BL/6 mice. IOP and pattern electroretinograms (pERG) were recorded, and retinas were prepared for RNA sequencing, immunohistochemistry, or to determine RGC loss. Ocular injection of Ad5.MYOC leads to reliable IOP elevation, resulting in significant loss of RGC after nine weeks. A significant decrease in the pERG amplitude was evident in eyes three weeks after IOP elevation. Retinal gene expression analysis revealed increased expression for 291 genes related to complement cascade, inflammation, and antigen presentation in hypertensive eyes. Decreased expression was found for 378 genes associated with the γ-aminobutyric acid (GABA)ergic and glutamatergic systems and axon guidance. These data suggest that early functional changes in RGC might be due to reduced GABAA receptor signaling and neuroinflammation that precedes RGC loss in this glaucoma model. These initial changes may offer new targets for early detection of glaucoma and the development of new interventions.


Subject(s)
GABAergic Neurons/metabolism , Glaucoma/pathology , Retinal Ganglion Cells/pathology , gamma-Aminobutyric Acid/metabolism , Animals , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/metabolism , Female , GABAergic Neurons/pathology , Gene Expression Regulation , Glaucoma/etiology , Glaucoma/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Inflammation/metabolism , Inflammation/pathology , Intraocular Pressure , Male , Mice , Mice, Inbred C57BL , Retinal Ganglion Cells/metabolism
19.
Neurobiol Dis ; 157: 105442, 2021 09.
Article in English | MEDLINE | ID: mdl-34246770

ABSTRACT

Neuregulin 1 (NRG1) and its receptor ERBB4 are schizophrenia (SZ) risk genes that control the development of both excitatory and inhibitory cortical circuits. Most studies focused on the characterization ErbB4 deficient mice. However, ErbB4 deletion concurrently perturbs the signaling of Nrg1 and Neuregulin 3 (Nrg3), another ligand expressed in the cortex. In addition, NRG1 polymorphisms linked to SZ locate mainly in non-coding regions and they may partially reduce Nrg1 expression. Here, to study the relevance of Nrg1 partial loss-of-function in cortical circuits we characterized a recently developed haploinsufficient mouse model of Nrg1 (Nrg1tm1Lex). These mice display SZ-like behavioral deficits. The cellular and molecular underpinnings of the behavioral deficits in Nrg1tm1Lex mice remain to be established. With multiple approaches including Magnetic Resonance Spectroscopy (MRS), electrophysiology, quantitative imaging and molecular analysis we found that Nrg1 haploinsufficiency impairs the inhibitory cortical circuits. We observed changes in the expression of molecules involved in GABAergic neurotransmission, decreased density of Vglut1 excitatory buttons onto Parvalbumin interneurons and decreased frequency of spontaneous inhibitory postsynaptic currents. Moreover, we found a decreased number of Parvalbumin positive interneurons in the cortex and altered expression of Calretinin. Interestingly, we failed to detect other alterations in excitatory neurons that were previously reported in ErbB4 null mice suggesting that the Nrg1 haploinsufficiency does not entirely phenocopies ErbB4 deletions. Altogether, this study suggests that Nrg1 haploinsufficiency primarily affects the cortical inhibitory circuits in the cortex and provides new insights into the structural and molecular synaptic impairment caused by NRG1 hypofunction in a preclinical model of SZ.


Subject(s)
Cerebral Cortex/metabolism , GABAergic Neurons/metabolism , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials/genetics , Interneurons/metabolism , Neural Inhibition/genetics , Neuregulin-1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Animals , Calbindin 2/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , GABAergic Neurons/pathology , Gene Expression , Haploinsufficiency , Hippocampus/diagnostic imaging , Hippocampus/pathology , Hippocampus/physiopathology , Interneurons/pathology , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Mice , Parvalbumins/metabolism , RNA, Messenger/metabolism , Receptor, ErbB-4/genetics , gamma-Aminobutyric Acid/metabolism
20.
Mol Brain ; 14(1): 96, 2021 06 26.
Article in English | MEDLINE | ID: mdl-34174930

ABSTRACT

Reductions in the GABAergic neurotransmitter system exist across multiple brain regions in schizophrenia and encompass both pre- and postsynaptic components. While reduced midbrain GABAergic inhibitory neurotransmission may contribute to the hyperdopaminergia thought to underpin psychosis in schizophrenia, molecular changes consistent with this have not been reported. We hypothesised that reduced GABA-related molecular markers would be found in the midbrain of people with schizophrenia and that these would correlate with dopaminergic molecular changes. We hypothesised that downregulation of inhibitory neuron markers would be exacerbated in schizophrenia cases with high levels of neuroinflammation. Eight GABAergic-related transcripts were measured with quantitative PCR, and glutamate decarboxylase (GAD) 65/67 and GABAA alpha 3 (α3) (GABRA3) protein were measured with immunoblotting, in post-mortem midbrain (28/28 and 28/26 control/schizophrenia cases for mRNA and protein, respectively), and analysed by both diagnosis and inflammatory subgroups (as previously defined by higher levels of four pro-inflammatory cytokine transcripts). We found reductions (21 - 44%) in mRNA encoding both presynaptic and postsynaptic proteins, vesicular GABA transporter (VGAT), GAD1, and parvalbumin (PV) mRNAs and four alpha subunits (α1, α2, α3, α5) of the GABAA receptor in people with schizophrenia compared to controls (p < 0.05). Gene expression of somatostatin (SST) was unchanged (p = 0.485). We confirmed the reduction in GAD at the protein level (34%, p < 0.05). When stratifying by inflammation, only GABRA3 mRNA exhibited more pronounced changes in high compared to low inflammatory subgroups in schizophrenia. GABRA3 protein was expressed by 98% of tyrosine hydroxylase-positive neurons and was 23% lower in schizophrenia, though this did not reach statistical significance (p > 0.05). Expression of transcripts for GABAA receptor alpha subunits 2 and 3 (GABRA2, GABRA3) were positively correlated with tyrosine hydroxylase (TH) and dopamine transporter (DAT) transcripts in schizophrenia cases (GABRA2; r > 0.630, GABRA3; r > 0.762, all p < 0.001) but not controls (GABRA2; r < - 0.200, GABRA3; r < 0.310, all p > 0.05). Taken together, our results support a profound disruption to inhibitory neurotransmission in the substantia nigra regardless of inflammatory status, which provides a potential mechanism for disinhibition of nigrostriatal dopamine neurotransmission.


Subject(s)
Biomarkers/metabolism , Dopaminergic Neurons/pathology , GABAergic Neurons/pathology , Mesencephalon/pathology , Schizophrenia/pathology , Adult , Aged , Cohort Studies , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Gene Expression Regulation , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Male , Middle Aged , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/pathology , Parvalbumins/metabolism , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Schizophrenia/genetics , Somatostatin/genetics , Somatostatin/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Young Adult , gamma-Aminobutyric Acid
SELECTION OF CITATIONS
SEARCH DETAIL