Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Psychiatry ; 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38499656

ABSTRACT

Autism spectrum disorder (ASD) is a major neurodevelopmental disorder affecting 1 in 36 children in the United States. While neurons have been the focus of understanding ASD, an altered neuro-immune response in the brain may be closely associated with ASD, and a neuro-immune interaction could play a role in the disease progression. As the resident immune cells of the brain, microglia regulate brain development and homeostasis via core functions including phagocytosis of synapses. While ASD has been traditionally considered a polygenic disorder, recent large-scale human genetic studies have identified SCN2A deficiency as a leading monogenic cause of ASD and intellectual disability. We generated a Scn2a-deficient mouse model, which displays major behavioral and neuronal phenotypes. However, the role of microglia in this disease model is unknown. Here, we reported that Scn2a-deficient mice have impaired learning and memory, accompanied by reduced synaptic transmission and lower spine density in neurons of the hippocampus. Microglia in Scn2a-deficient mice are partially activated, exerting excessive phagocytic pruning of post-synapses related to the complement C3 cascades during selective developmental stages. The ablation of microglia using PLX3397 partially restores synaptic transmission and spine density. To extend our findings from rodents to human cells, we established a microglia-incorporated human cerebral organoid model carrying an SCN2A protein-truncating mutation identified in children with ASD. We found that human microglia display increased elimination of post-synapse in cerebral organoids carrying the SCN2A mutation. Our study establishes a key role of microglia in multi-species autism-associated models of SCN2A deficiency from mouse to human cells.

2.
bioRxiv ; 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37961213

ABSTRACT

Neuronal hyperexcitability is a hallmark of seizures. It has been recently shown in rodent models of seizures that microglia, the brain's resident immune cells, can respond to and modulate neuronal excitability. However, how human microglia interacts with human neurons to regulate hyperexcitability mediated by epilepsy-causing genetic mutation found in human patients remains unknown. The SCN2A genetic locus is responsible for encoding the voltage-gated sodium channel Nav1.2, recognized as one of the leading contributors to monogenic epilepsies. Previously, we demonstrated that the recurring Nav1.2-L1342P mutation identified in patients with epilepsy leads to hyperexcitability in a hiPSC-derived cortical neuron model from a male donor. While microglia play an important role in the brain, these cells originate from a different lineage (yolk sac) and thus are not naturally present in hiPSCs-derived neuronal culture. To study how microglia respond to diseased neurons and influence neuronal excitability, we established a co-culture model comprising hiPSC-derived neurons and microglia. We found that microglia display altered morphology with increased branch length and enhanced calcium signal when co-cultured with neurons carrying the Nav1.2-L1342P mutation. Moreover, the presence of microglia significantly lowers the action potential firing of neurons carrying the mutation. Interestingly, we further demonstrated that the current density of sodium channels in neurons carrying the epilepsy-associated mutation was reduced in the presence of microglia. Taken together, our work reveals a critical role of human iPSCs-derived microglia in sensing and dampening hyperexcitability mediated by an epilepsy-causing mutation present in human neurons, highlighting the importance of neuron-microglia interactions in human pathophysiology.

3.
Int J Cardiol ; 346: 47-52, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34801613

ABSTRACT

BACKGROUND: While ST-Elevation Myocardial Infarction (STEMI) door-to-balloon times are often below 90 min, symptom to door times remain long at 2.5-h, due at least in part to a delay in diagnosis. OBJECTIVES: To develop and validate a machine learning-guided algorithm which uses a single­lead electrocardiogram (ECG) for STEMI detection to speed diagnosis. METHODS: Data was extracted from the Latin America Telemedicine Infarct Network (LATIN), a population-based Acute Myocardial Infarction (AMI) program that provides care to patients in Brazil, Colombia, Mexico, and Argentina through telemedicine. SAMPLE: the first dataset was comprised of 8511 ECGs that were used for various machine learning experiments to test our Deep Learning approach for STEMI diagnosis. The second dataset of 2542 confirmed STEMI diagnosis EKG records, including specific ischemic heart wall information (anterior, inferior, and lateral), was derived from the previous dataset to test the STEMI localization model. Preprocessing: Detection of QRS complexes by wavelet system, segmentation of each EKG record into individual heartbeats with fixed window of 0.4 s to the left and 0.9 s to the right of main. Training & Testing: 90% and 10% of the total dataset, respectively, were used for both models. CLASSIFICATION: two 1-D convolutional neural networks were implemented, two classes were considered for first models (STEMI/Not-STEMI) and three classes for the second model (Anterior/Inferior/Lateral) each corresponding to the heart wall affected. These individual probabilities were aggregated to generate the final label for each model. RESULTS: The single­lead ECG strategy was able to provide an accuracy of 90.5% for STEMI detection with Lead V2, which also yielded the best results overall among individual leads. STEMI Localization model provided promising results for anterior and inferior wall STEMIs but remained suboptimal for Lateral STEMI. CONCLUSIONS: An Artificial Intelligence-enhanced single­lead ECG is a promising screening tool. This technology provides an autonomous and accurate STEMI diagnostic alternative that can be incorporated into wearable devices, potentially providing patients reliable means to seek treatment early and offers the potential to thereby improve STEMI outcomes in the long run.


Subject(s)
Deep Learning , Myocardial Infarction , ST Elevation Myocardial Infarction , Artificial Intelligence , Electrocardiography , Humans , ST Elevation Myocardial Infarction/diagnosis
4.
J Neurosci ; 41(49): 10194-10208, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34716231

ABSTRACT

With the wide adoption of genomic sequencing in children having seizures, an increasing number of SCN2A genetic variants have been revealed as genetic causes of epilepsy. Voltage-gated sodium channel Nav1.2, encoded by gene SCN2A, is predominantly expressed in the pyramidal excitatory neurons and supports action potential (AP) firing. One recurrent SCN2A genetic variant is L1342P, which was identified in multiple patients with epileptic encephalopathy and intractable seizures. However, the mechanism underlying L1342P-mediated seizures and the pharmacogenetics of this variant in human neurons remain unknown. To understand the core phenotypes of the L1342P variant in human neurons, we took advantage of a reference human-induced pluripotent stem cell (hiPSC) line from a male donor, in which L1342P was introduced by CRISPR/Cas9-mediated genome editing. Using patch-clamping and microelectrode array (MEA) recordings, we revealed that cortical neurons derived from hiPSCs carrying heterozygous L1342P variant have significantly increased intrinsic excitability, higher sodium current density, and enhanced bursting and synchronous network firing, suggesting hyperexcitability phenotypes. Interestingly, L1342P neuronal culture displayed a degree of resistance to the anticonvulsant medication phenytoin, which recapitulated aspects of clinical observation of patients carrying the L1342P variant. In contrast, phrixotoxin-3 (PTx3), a Nav1.2 isoform-specific blocker, can potently alleviate spontaneous and chemically-induced hyperexcitability of neurons carrying the L1342P variant. Our results reveal a possible pathogenic underpinning of Nav1.2-L1342P mediated epileptic seizures and demonstrate the utility of genome-edited hiPSCs as an in vitro platform to advance personalized phenotyping and drug discovery.SIGNIFICANCE STATEMENT A mounting number of SCN2A genetic variants have been identified from patients with epilepsy, but how SCN2A variants affect the function of human neurons contributing to seizures is still elusive. This study investigated the functional consequences of a recurring SCN2A variant (L1342P) using human iPSC-derived neurons and revealed both intrinsic and network hyperexcitability of neurons carrying a mutant Nav1.2 channel. Importantly, this study recapitulated elements of clinical observations of drug-resistant features of the L1342P variant, and provided a platform for in vitro drug testing. Our study sheds light on cellular mechanism of seizures resulting from a recurring Nav1.2 variant, and helps to advance personalized drug discovery to treat patients carrying pathogenic SCN2A variant.


Subject(s)
Epilepsy/genetics , Epilepsy/physiopathology , Gene Editing/methods , NAV1.2 Voltage-Gated Sodium Channel/genetics , Neurons/pathology , Cerebral Cortex/physiopathology , Humans , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/physiology , Mutation
5.
Cell Rep ; 36(5): 109495, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348148

ABSTRACT

Scn2a encodes the voltage-gated sodium channel NaV1.2, a main mediator of neuronal action potential firing. The current paradigm suggests that NaV1.2 gain-of-function variants enhance neuronal excitability, resulting in epilepsy, whereas NaV1.2 deficiency impairs neuronal excitability, contributing to autism. However, this paradigm does not explain why ∼20%-30% of individuals with NaV1.2 deficiency still develop seizures. Here, we report the counterintuitive finding that severe NaV1.2 deficiency results in increased neuronal excitability. Using a NaV1.2-deficient mouse model, we show enhanced intrinsic excitability of principal neurons in the prefrontal cortex and striatum, brain regions known to be involved in Scn2a-related seizures. This increased excitability is autonomous and reversible by genetic restoration of Scn2a expression in adult mice. RNA sequencing reveals downregulation of multiple potassium channels, including KV1.1. Correspondingly, KV channel openers alleviate the hyperexcitability of NaV1.2-deficient neurons. This unexpected neuronal hyperexcitability may serve as a cellular basis underlying NaV1.2 deficiency-related seizures.


Subject(s)
Aging/physiology , NAV1.2 Voltage-Gated Sodium Channel/deficiency , Neurons/physiology , Action Potentials , Animals , Down-Regulation , Ion Channel Gating , Mice, Inbred C57BL , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Potassium Channels/metabolism
6.
Sci Rep ; 8(1): 1410, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362425

ABSTRACT

The regulation of Rac1 by HACE1-mediated ubiquitination and proteasomal degradation is emerging as an essential element in the maintenance of cell homeostasis. However, how the E3 ubiquitin ligase activity of HACE1 is regulated remains undetermined. Using a proteomic approach, we identified serine 385 as a target of group-I PAK kinases downstream Rac1 activation by CNF1 toxin from pathogenic E. coli. Moreover, cell treatment with VEGF also promotes Ser-385 phosphorylation of HACE1. We have established in vitro that HACE1 is a direct target of PAK1 kinase activity. Mechanistically, we found that the phospho-mimetic mutant HACE1(S385E), as opposed to HACE1(S385A), displays a lower capacity to ubiquitinate Rac1 in cells. Concomitantly, phosphorylation of Ser-385 plays a pivotal role in controlling the oligomerization state of HACE1. Finally, Ser-385 phosphorylated form of HACE1 localizes in the cytosol away from its target Rac1. Together, our data point to a feedback inhibition of HACE1 ubiquitination activity on Rac1 by group-I PAK kinases.


Subject(s)
Serine/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Bacterial Toxins/pharmacology , Cell Line , Escherichia coli Proteins/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Phosphorylation , Protein Multimerization , Proteomics , Ubiquitination , Vascular Endothelial Growth Factor A/pharmacology
7.
Cytoskeleton (Hoboken) ; 72(10): 542-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26403219

ABSTRACT

It remains a challenge to decode the molecular basis of the long-term actin cytoskeleton rearrangements that are governed by the reprogramming of gene expression. Bacillus anthracis lethal toxin (LT) inhibits mitogen-activated protein kinase (MAPK) signaling, thereby modulating gene expression, with major consequences for actin cytoskeleton organization and the loss of endothelial barrier function. Using a laser ablation approach, we characterized the contractile and tensile mechanical properties of LT-induced stress fibers. These actin cables resist pulling forces that are transmitted at cell-matrix interfaces and at cell-cell discontinuous adherens junctions. We report that treating the cells with trichostatin A (TSA), a broad range inhibitor of histone deacetylases (HDACs), or with MS-275, which targets HDAC1, 2 and 3, induces stress fibers. LT decreased the cellular levels of HDAC1, 2 and 3 and reduced the global HDAC activity in the nucleus. Both the LT and TSA treatments induced Rnd3 expression, which is required for the LT-mediated induction of actin stress fibers. Furthermore, we reveal that treating the LT-intoxicated cells with garcinol, an inhibitor of histone acetyl-transferases (HATs), disrupts the stress fibers and limits the monolayer barrier dysfunctions. These data demonstrate the importance of modulating the flux of protein acetylation in order to control actin cytoskeleton organization and the endothelial cell monolayer barrier.


Subject(s)
Actins/chemistry , Antigens, Bacterial/chemistry , Bacillus anthracis/chemistry , Bacterial Toxins/chemistry , Histones/chemistry , Stress Fibers/chemistry , Acetylation , Adherens Junctions , Cell Communication , Cell Nucleus/metabolism , Endothelial Cells/cytology , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Hydroxamic Acids/chemistry , Light , Microscopy, Fluorescence , Tensile Strength
SELECTION OF CITATIONS
SEARCH DETAIL
...