Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Nat Cancer ; 5(1): 66-84, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38151625

RÉSUMÉ

Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.


Sujet(s)
Kinésine , Tumeurs du sein triple-négatives , Humains , Animaux , Souris , Kinésine/génétique , Kinésine/métabolisme , Mitose/génétique , Lignée cellulaire , Points de contrôle de la phase M du cycle cellulaire
2.
Biol Psychiatry ; 95(2): 187-198, 2024 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-37454787

RÉSUMÉ

BACKGROUND: Converging evidence from large-scale genetic and postmortem studies highlights the role of aberrant neurotransmission and genetic regulation in brain-related disorders. However, identifying neuronal activity-regulated transcriptional programs in the human brain and understanding how changes contribute to disease remain challenging. METHODS: To better understand how the activity-dependent regulome contributes to risk for brain-related disorders, we profiled the transcriptomic and epigenomic changes following neuronal depolarization in human induced pluripotent stem cell-derived glutamatergic neurons (NGN2) from 6 patients with schizophrenia and 5 control participants. RESULTS: Multiomic data integration associated global patterns of chromatin accessibility with gene expression and identified enhancer-promoter interactions in glutamatergic neurons. Within 1 hour of potassium chloride-induced depolarization, independent of diagnosis, glutamatergic neurons displayed substantial activity-dependent changes in the expression of genes regulating synaptic function. Depolarization-induced changes in the regulome revealed significant heritability enrichment for schizophrenia and Parkinson's disease, adding to mounting evidence that sequence variation within activation-dependent regulatory elements contributes to the genetic risk for brain-related disorders. Gene coexpression network analysis elucidated interactions among activity-dependent and disease-associated genes and pointed to a key driver (NAV3) that interacted with multiple genes involved in axon guidance. CONCLUSIONS: Overall, we demonstrated that deciphering the activity-dependent regulome in glutamatergic neurons reveals novel targets for advanced diagnosis and therapy.


Sujet(s)
Cellules souches pluripotentes induites , Schizophrénie , Humains , Cellules souches pluripotentes induites/métabolisme , Régulation de l'expression des gènes , Neurones/métabolisme , Encéphale
3.
Mol Psychiatry ; 28(5): 1970-1982, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-34493831

RÉSUMÉ

Dopaminergic neurons are critical to movement, mood, addiction, and stress. Current techniques for generating dopaminergic neurons from human induced pluripotent stem cells (hiPSCs) yield heterogenous cell populations with variable purity and inconsistent reproducibility between donors, hiPSC clones, and experiments. Here, we report the rapid (5 weeks) and efficient (~90%) induction of induced dopaminergic neurons (iDANs) through transient overexpression of lineage-promoting transcription factors combined with stringent selection across five donors. We observe maturation-dependent increase in dopamine synthesis and electrophysiological properties consistent with midbrain dopaminergic neuron identity, such as slow-rising after- hyperpolarization potentials, an action potential duration of ~3 ms, tonic sub-threshold oscillatory activity, and spontaneous burst firing at a frequency of ~1.0-1.75 Hz. Transcriptome analysis reveals robust expression of genes involved in fetal midbrain dopaminergic neuron identity. Specifically expressed genes in iDANs, as well as those from isogenic induced GABAergic and glutamatergic neurons, were enriched in loci conferring heritability for cannabis use disorder, schizophrenia, and bipolar disorder; however, each neuronal subtype demonstrated subtype-specific heritability enrichments in biologically relevant pathways, and iDANs alone were uniquely enriched in autism spectrum disorder risk loci. Therefore, iDANs provide a critical tool for modeling midbrain dopaminergic neuron development and dysfunction in psychiatric disease.


Sujet(s)
Trouble du spectre autistique , Cellules souches pluripotentes induites , Humains , Neurones dopaminergiques/métabolisme , Trouble du spectre autistique/métabolisme , Reproductibilité des résultats , Cellules souches pluripotentes induites/métabolisme , Mésencéphale/métabolisme
4.
STAR Protoc ; 2(2): 100580, 2021 06 18.
Article de Anglais | MEDLINE | ID: mdl-34151300

RÉSUMÉ

We describe a CRISPR inhibition (CRISPRi) protocol to repress endogenous gene expression (e.g., ATP6V1A) in human induced pluripotent stem cell-derived NGN2-induced glutamatergic neurons. CRISPRi enables efficient and precise gene repression of one or multiple target genes via delivering gRNA(s) to direct a dCas9-KRAB fusion protein to the gene(s) of interest. This protocol can also be adapted for gene activation and high-throughput gene manipulation, allowing assessment of the transcriptomic and phenotypic impact of candidate gene(s) associated with neurodevelopment or brain disease. For complete details on the use and execution of this protocol, please refer to Ho et al. (2017) and Wang et al. (2021).


Sujet(s)
Systèmes CRISPR-Cas , Régulation de l'expression des gènes , Cellules souches pluripotentes induites/métabolisme , Protéines de tissu nerveux/génétique , Neurones/métabolisme , Humains , Neurones/cytologie , Transcriptome
5.
Neuron ; 109(2): 257-272.e14, 2021 01 20.
Article de Anglais | MEDLINE | ID: mdl-33238137

RÉSUMÉ

To identify the molecular mechanisms and novel therapeutic targets of late-onset Alzheimer's Disease (LOAD), we performed an integrative network analysis of multi-omics profiling of four cortical areas across 364 donors with varying cognitive and neuropathological phenotypes. Our analyses revealed thousands of molecular changes and uncovered neuronal gene subnetworks as the most dysregulated in LOAD. ATP6V1A was identified as a key regulator of a top-ranked neuronal subnetwork, and its role in disease-related processes was evaluated through CRISPR-based manipulation in human induced pluripotent stem cell-derived neurons and RNAi-based knockdown in Drosophila models. Neuronal impairment and neurodegeneration caused by ATP6V1A deficit were improved by a repositioned compound, NCH-51. This study provides not only a global landscape but also detailed signaling circuits of complex molecular interactions in key brain regions affected by LOAD, and the resulting network models will serve as a blueprint for developing next-generation therapeutic agents against LOAD.


Sujet(s)
Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/thérapie , Encéphale/physiologie , Bases de données génétiques , Réseaux de régulation génique/physiologie , Transduction du signal/physiologie , Maladie d'Alzheimer/anatomopathologie , Animaux , Animal génétiquement modifié , Encéphale/anatomopathologie , Bases de données génétiques/tendances , Drosophila melanogaster , Femelle , Humains , Cellules souches pluripotentes induites/physiologie , Mâle , Analyse de séquence d'ARN/méthodes
6.
Mol Cell Neurosci ; 107: 103532, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32712198

RÉSUMÉ

Neuropsychiatric disorders are highly heritable polygenic disorders arising from the complex interplay of highly penetrant rare variants and common variants of small effect. There is a large index of comorbidity and shared genetic risk between disorders, reflecting the pleiotropy of individual variants as well as predicted downstream pathway-level convergence. Importantly, the mechanism(s) through which psychiatric disease-associated variants interact to contribute to disease risk remains unknown. Human induced pluripotent stem cell (hiPSC)-based models are increasingly useful for the systematic study of the complex genetics associated with brain diseases, particularly when combined with CRISPR-mediated genomic engineering, which together facilitate isogenic comparisons of defined neuronal cell types. In this review, we discuss the latest CRISPR technologies and consider how they can be successfully applied to the functional characterization of the growing list genetic variants linked to psychiatric disease.


Sujet(s)
Encéphalopathies/génétique , Clustered regularly interspaced short palindromic repeats/génétique , Cellules souches pluripotentes induites/métabolisme , Troubles mentaux/génétique , Animaux , Édition de gène/méthodes , Humains , Neurones/métabolisme
7.
J Neurosci Methods ; 334: 108548, 2020 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-32065989

RÉSUMÉ

BACKGROUND: Somatic cell reprogramming is routinely used to generate donor-specific human induced pluripotent stem cells (hiPSCs) to facilitate studies of disease in a human context. The directed differentiation of hiPSCs can generate large quantities of patient-derived cells; however, such methodologies frequently yield heterogeneous populations of neurons and glia that require extended timelines to achieve electrophysiological maturity. More recently, transcription factor-based induction protocols have been show to rapidly generate defined neuronal populations from hiPSCs. NEW METHOD: In a manner similar to our previous adaption of NGN2-glutamatergic neuronal induction from hiPSC-derived neural progenitor cells (NPCs), we now adapt an established protocol of lentiviral overexpression of ASCL1 and DLX2 to hiPSC-NPCs. RESULTS: We demonstrate induction of a robust and highly pure population of functional GABAergic neurons (iGANs). Importantly, we successfully applied this technique to hiPSC-NPCs derived from ten donors across two independent laboratories, finding it to be an efficient and highly reproducible approach to generate induced GABAergic neurons. Our results show that, like hiPSC-iGANs, NPC-iGANs exhibit increased GABAergic marker expression, electrophysiological maturity, and have distinct transcriptional profiles that distinguish them from other cell-types of the brain. Nonetheless, until donor-matched hiPSCs-iGANs and NPC-iGANs are directly compared, we cannot rule out the possibility that subtle differences in patterning or maturity may exist between these populations; one should always control for cell source in all iGAN experiments. CONCLUSIONS: This methodology, relying upon an easily cultured starting population of hiPSC-NPCs, makes possible the generation of large-scale defined co-cultures of induced glutamatergic and GABAergic neurons for hiPSC-based disease models and precision drug screening.

8.
Nat Genet ; 51(10): 1475-1485, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31548722

RÉSUMÉ

The mechanisms by which common risk variants of small effect interact to contribute to complex genetic disorders are unclear. Here, we apply a genetic approach, using isogenic human induced pluripotent stem cells, to evaluate the effects of schizophrenia (SZ)-associated common variants predicted to function as SZ expression quantitative trait loci (eQTLs). By integrating CRISPR-mediated gene editing, activation and repression technologies to study one putative SZ eQTL (FURIN rs4702) and four top-ranked SZ eQTL genes (FURIN, SNAP91, TSNARE1 and CLCN3), our platform resolves pre- and postsynaptic neuronal deficits, recapitulates genotype-dependent gene expression differences and identifies convergence downstream of SZ eQTL gene perturbations. Our observations highlight the cell-type-specific effects of common variants and demonstrate a synergistic effect between SZ eQTL genes that converges on synaptic function. We propose that the links between rare and common variants implicated in psychiatric disease risk constitute a potentially generalizable phenomenon occurring more widely in complex genetic disorders.


Sujet(s)
Régulation de l'expression des gènes , Prédisposition génétique à une maladie , Cellules souches pluripotentes induites/anatomopathologie , Polymorphisme de nucléotide simple , Locus de caractère quantitatif , Schizophrénie/génétique , Schizophrénie/anatomopathologie , Systèmes CRISPR-Cas , Canaux chlorure/antagonistes et inhibiteurs , Canaux chlorure/génétique , Canaux chlorure/métabolisme , Femelle , Furine/antagonistes et inhibiteurs , Furine/génétique , Furine/métabolisme , Édition de gène , Étude d'association pangénomique , Humains , Cellules souches pluripotentes induites/métabolisme , Mâle , Protéines d'assemblage monomériques de la clathrine/antagonistes et inhibiteurs , Protéines d'assemblage monomériques de la clathrine/génétique , Protéines d'assemblage monomériques de la clathrine/métabolisme , Protéines SNARE/antagonistes et inhibiteurs , Protéines SNARE/génétique , Protéines SNARE/métabolisme
9.
Science ; 362(6420)2018 12 14.
Article de Anglais | MEDLINE | ID: mdl-30545851

RÉSUMÉ

To explore the developmental reorganization of the three-dimensional genome of the brain in the context of neuropsychiatric disease, we monitored chromosomal conformations in differentiating neural progenitor cells. Neuronal and glial differentiation was associated with widespread developmental remodeling of the chromosomal contact map and included interactions anchored in common variant sequences that confer heritable risk for schizophrenia. We describe cell type-specific chromosomal connectomes composed of schizophrenia risk variants and their distal targets, which altogether show enrichment for genes that regulate neuronal connectivity and chromatin remodeling, and evidence for coordinated transcriptional regulation and proteomic interaction of the participating genes. Developmentally regulated chromosomal conformation changes at schizophrenia-relevant sequences disproportionally occurred in neurons, highlighting the existence of cell type-specific disease risk vulnerabilities in spatial genome organization.


Sujet(s)
Chromosomes humains/composition chimique , Connectome , Épigenèse génétique , Régulation de l'expression des gènes au cours du développement , Prédisposition génétique à une maladie , Cellules souches neurales/cytologie , Neurogenèse/génétique , Schizophrénie/génétique , Encéphale/croissance et développement , Encéphale/métabolisme , Cellules cultivées , Chromatine/composition chimique , Assemblage et désassemblage de la chromatine , Génome humain , Étude d'association pangénomique , Humains , Mâle , Cellules souches neurales/métabolisme , Névroglie/cytologie , Neurones/cytologie , Neurones/métabolisme , Conformation d'acide nucléique , Cartes d'interactions protéiques/génétique , Protéomique , Risque , Transcription génétique , Transcriptome
10.
Stem Cell Reports ; 9(2): 615-628, 2017 08 08.
Article de Anglais | MEDLINE | ID: mdl-28757163

RÉSUMÉ

Modulation of transcription, either synthetic activation or repression, via dCas9-fusion proteins is a relatively new methodology with the potential to facilitate high-throughput up- or downregulation studies of gene function. Genetic studies of neurodevelopmental disorders have identified a growing list of risk variants, including both common single-nucleotide variants and rare copy-number variations, many of which are associated with genes having limited functional annotations. By applying a CRISPR-mediated gene-activation/repression platform to populations of human-induced pluripotent stem cell-derived neural progenitor cells, neurons, and astrocytes, we demonstrate that it is possible to manipulate endogenous expression levels of candidate neuropsychiatric risk genes across these three cell types. Although proof-of-concept studies using catalytically inactive Cas9-fusion proteins to modulate transcription have been reported, here we present a detailed survey of the reproducibility of gRNA positional effects across a variety of neurodevelopmental disorder-relevant risk genes, donors, neural cell types, and dCas9 effectors.


Sujet(s)
Astrocytes/cytologie , Astrocytes/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Neurones/cytologie , Neurones/métabolisme , Calcium/métabolisme , Différenciation cellulaire , Cellules cultivées , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes au cours du développement , Humains , Cellules souches pluripotentes induites/métabolisme , Imagerie moléculaire , Transcriptome
11.
Nat Genet ; 49(8): 1239-1250, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28671686

RÉSUMÉ

We report locus-specific disintegration of megabase-scale chromosomal conformations in brain after neuronal ablation of Setdb1 (also known as Kmt1e; encodes a histone H3 lysine 9 methyltransferase), including a large topologically associated 1.2-Mb domain conserved in humans and mice that encompasses >70 genes at the clustered protocadherin locus (hereafter referred to as cPcdh). The cPcdh topologically associated domain (TADcPcdh) in neurons from mutant mice showed abnormal accumulation of the transcriptional regulator and three-dimensional (3D) genome organizer CTCF at cryptic binding sites, in conjunction with DNA cytosine hypomethylation, histone hyperacetylation and upregulated expression. Genes encoding stochastically expressed protocadherins were transcribed by increased numbers of cortical neurons, indicating relaxation of single-cell constraint. SETDB1-dependent loop formations bypassed 0.2-1 Mb of linear genome and radiated from the TADcPcdh fringes toward cis-regulatory sequences within the cPcdh locus, counterbalanced shorter-range facilitative promoter-enhancer contacts and carried loop-bound polymorphisms that were associated with genetic risk for schizophrenia. We show that the SETDB1 repressor complex, which involves multiple KRAB zinc finger proteins, shields neuronal genomes from excess CTCF binding and is critically required for structural maintenance of TADcPcdh.


Sujet(s)
Chromatine/métabolisme , Histone-lysine N-methyltransferase/métabolisme , Neurones/métabolisme , Animaux , Facteur de liaison à la séquence CCCTC , Cadhérines/génétique , Lignée cellulaire , Méthylation de l'ADN , Épigenèse génétique , Femelle , Régulation de l'expression des gènes , Histone-lysine N-methyltransferase/génétique , Humains , Mâle , Souris , Mutation , Conformation d'acide nucléique , Liaison aux protéines , Domaines protéiques , Protéines de répression/métabolisme
12.
Methods ; 101: 113-24, 2016 05 15.
Article de Anglais | MEDLINE | ID: mdl-26626326

RÉSUMÉ

Since the discovery of somatic reprogramming, human induced pluripotent stem cells (hiPSCs) have been exploited to model a variety of neurological and psychiatric disorders. Because hiPSCs represent an almost limitless source of patient-derived neurons that retain the genetic variations thought to contribute to disease etiology, they have been heralded as a patient-specific platform for high throughput drug screening. However, the utility of current protocols for generating neurons from hiPSCs remains limited by protracted differentiation timelines and heterogeneity of the neuronal phenotypes produced. Neuronal induction via the forced expression of exogenous transcription factors rapidly induces defined populations of functional neurons from fibroblasts and hiPSCs. Here, we describe an adapted protocol that accelerates maturation of functional excitatory neurons from hiPSC-derived neural progenitor cells (NPCs) via lentiviral transduction of Neurogenin 2 (using both mNgn2 and hNGN2). This methodology, relying upon a robust and scalable starting population of hiPSC NPCs, should be readily amenable to scaling for hiPSC-based high-throughput drug screening.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/physiologie , Cellules souches pluripotentes induites/physiologie , Protéines de tissu nerveux/physiologie , Techniques de culture cellulaire , Cellules cultivées , Reprogrammation cellulaire , Expression des gènes , Humains , Lentivirus , Cellules souches neurales/physiologie , Activation de la transcription
13.
Biomark Insights ; 10(Suppl 1): 31-41, 2015.
Article de Anglais | MEDLINE | ID: mdl-26045654

RÉSUMÉ

Aberrant behavior and function of neurons are believed to be the primary causes of most neurological diseases and psychiatric disorders. Human postmortem samples have limited availability and, while they provide clues to the state of the brain after a prolonged illness, they offer limited insight into the factors contributing to disease onset. Conversely, animal models cannot recapitulate the polygenic origins of neuropsychiatric disease. Novel methods, such as somatic cell reprogramming, deliver nearly limitless numbers of pathogenic human neurons for the study of the mechanism of neuropsychiatric disease initiation and progression. First, this article reviews the advent of human induced pluripotent stem cell (hiPSC) technology and introduces two major methods, "directed differentiation" and "neuronal induction," by which it is now possible to generate neurons for modeling neuropsychiatric disease. Second, it discusses the recent applications, and the limitations, of these technologies to in vitro studies of psychiatric disorders.

14.
NPJ Schizophr ; 12015 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-26985448

RÉSUMÉ

Neurodevelopmental disorders, such as autism spectrum disorders (ASD) and schizophrenia (SZ), are complex disorders with a high degree of heritability. Genetic studies have identified several candidate genes associated with these disorders, including contactin-associated protein-like 2 (CNTNAP2). Traditionally, in animal models or in vitro, the function of CNTNAP2 has been studied by genetic deletion or transcriptional knockdown, which reduce the expression of the entire gene; however, it remains unclear whether the mutations identified in clinical settings are sufficient to alter CNTNAP2 expression in human neurons. Here, using human induced pluripotent stem cells (hiPSCs) derived from two individuals with a large (289kb) and heterozygous deletion in CNTNAP2 (affecting exons 14-15) and discordant clinical outcomes, we have characterized CNTNAP2 expression patterns in hiPSC neural progenitor cells (NPCs), two independent populations of hiPSC-derived neurons and hiPSC-derived oligodendrocyte precursor cells (OPCs). First, we observed exon-specific changes in CNTNAP2 expression in both carriers; although the expression of exons 14-15 is significantly decreased, the expression of other exons is upregulated. Second, we observed significant differences in patterns of allele-specific expression in CNTNAP2 carriers that were consistent with clinical outcome. Third, we observed a robust neural migration phenotype that correlated with diagnosis and exon- and allele-specific CNTNAP2 expression patterns, but not with genotype. In all, our data highlight the importance of considering the nature, location and regulation of mutated alleles when attempting to connect GWAS studies to gene function.

15.
Nat Commun ; 5: 5455, 2014 Nov 19.
Article de Anglais | MEDLINE | ID: mdl-25406935

RÉSUMÉ

Canonical Wnt signalling regulates expansion of neural progenitors and functions as a dorsalizing signal in the developing forebrain. In contrast, the multifunctional co-receptor Cdo promotes neuronal differentiation and is important for the function of the ventralizing signal, Shh. Here we show that Cdo negatively regulates Wnt signalling during neurogenesis. Wnt signalling is enhanced in Cdo-deficient cells, leading to impaired neuronal differentiation. The ectodomains of Cdo and Lrp6 interact via the Ig2 repeat of Cdo and the LDLR repeats of Lrp6, and the Cdo Ig2 repeat is necessary for Cdo-dependent Wnt inhibition. Furthermore, the Cdo-deficient dorsal forebrain displays stronger Wnt signalling activity, increased cell proliferation and enhanced expression of the dorsal markers and Wnt targets, Pax6, Gli3, Axin2. Therefore, in addition to promoting ventral central nervous system cell fates with Shh, Cdo promotes neuronal differentiation by suppression of Wnt signalling and provides a direct link between two major dorsoventral morphogenetic signalling pathways.


Sujet(s)
Cysteine dioxygenase/métabolisme , Régulation de l'expression des gènes au cours du développement , Protéine-6 apparentée au récepteur des LDL/métabolisme , Neurogenèse , Neurones/métabolisme , Voie de signalisation Wnt , Animaux , Axine/métabolisme , Différenciation cellulaire , Embryon de mammifère , Protéines de l'oeil/métabolisme , Protéines Hedgehog/métabolisme , Protéines à homéodomaine/métabolisme , Facteurs de transcription Krüppel-like/métabolisme , Souris , Protéines de tissu nerveux/métabolisme , Neurones/cytologie , Facteur de transcription PAX6 , Facteurs de transcription PAX/métabolisme , Protéines de répression/métabolisme , Protéines de type Wingless/métabolisme , Protéine à doigts de zinc Gli3 , bêta-Caténine/métabolisme
16.
ACS Nano ; 6(8): 7151-61, 2012 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-22845739

RÉSUMÉ

Dual-function poly(L-lysine) (PLL) composites that function as antibacterial agents and promote the growth of human cell culture have been sought by researchers for a long period. In this paper, we report the preparation of new graphene derivative-PLL composites via electrostatic interactions and covalent bonding between graphene derivatives and PLL. The resulting composites were characterized by infrared spectroscopy, scanning electron microscopy, and X-ray photoelectron spectroscopy. The novel dual function of PLL composites, specifically antibacterial activity and biocompatibility with human cells [human adipose-derived stem cells and non-small-cell lung carcinoma cells (A549)], was carefully investigated. Graphene-DS-PLL composites composed of 4-carboxylic acid benzene diazonium salt (DS) generated more anionic carboxylic acid groups to bind to cationic PLLs, forming the most potent antibacterial agent among PLL and PLL composites with high biocompatibility with human cell culture. This dual functionality can be used to inhibit bacterial growth while enhancing human cell growth.


Sujet(s)
Antibactériens/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Graphite/pharmacologie , Nanostructures/administration et posologie , Nanostructures/composition chimique , Polylysine/pharmacologie , Antibactériens/composition chimique , Lignée cellulaire , Escherichia coli/effets des médicaments et des substances chimiques , Graphite/composition chimique , Humains , Test de matériaux , Cellules souches/effets des médicaments et des substances chimiques
17.
J Biol Chem ; 287(15): 11602-15, 2012 Apr 06.
Article de Anglais | MEDLINE | ID: mdl-22337877

RÉSUMÉ

p38MAPK plays an essential role in the transition of myoblasts to differentiated myotubes through the activation of MyoD family transcription factors. A promyogenic cell surface molecule, Cdo, promotes myogenic differentiation mainly through activation of the p38MAPK pathway. Two MAP3Ks, TAK1 and ASK1, can activate p38MAPK via MKK6 in various cell systems. Moreover TAK1 has been shown to promote myogenic differentiation via p38MAPK activation. In this study, we hypothesized that TAK1 and ASK1 might function as MAP3Ks in Cdo-mediated p38MAPK activation during myoblast differentiation. Both ASK1 and TAK1 were expressed in myoblasts and interacted with the cytoplasmic tail of Cdo and a scaffold protein, JLP. The depletion of TAK1 or ASK1 in C2C12 cells decreased myoblast differentiation, whereas overexpression of TAK1 or ASK1 in C2C12 cells enhanced myotube formation. In agreement with this, overexpression of ASK1 or TAK1 resulted in enhanced p38MAPK activation, and their knockdown inhibited p38MAPK in C2C12 cells. Overexpression of TAK1 or ASK1 in Cdo(-/-) myoblasts and Cdo-depleted C2C12 cells restored p38MAPK activation as well as myotube formation. Furthermore, ASK1 and TAK1 compensated for each other in p38MAPK activation and myoblast differentiation. Taken together, these findings suggest that ASK1 and TAK1 function as MAP3Ks in Cdo-mediated p38MAPK activation to promote myogenic differentiation.


Sujet(s)
Molécules d'adhérence cellulaire/métabolisme , MAP Kinase Kinase Kinase 5/métabolisme , MAP Kinase Kinase Kinases/métabolisme , Système de signalisation des MAP kinases , Animaux , Molécules d'adhérence cellulaire/génétique , Différenciation cellulaire , Lignée cellulaire , Prolifération cellulaire , Activation enzymatique , Techniques de knock-out de gènes , Humains , MAP Kinase Kinase Kinase 5/génétique , MAP Kinase Kinase Kinases/génétique , Souris , Complexes multiprotéiques/génétique , Complexes multiprotéiques/métabolisme , Développement musculaire , Fibres musculaires squelettiques/cytologie , Fibres musculaires squelettiques/métabolisme , Myoblastes/métabolisme , Myoblastes/physiologie , Liaison aux protéines , p38 Mitogen-Activated Protein Kinases/métabolisme
18.
Cell Signal ; 23(12): 2021-9, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21820049

RÉSUMÉ

Skeletal myogenesis is a multistep process that involves cell cycle exit, expression of muscle-specific genes and formation of multinucleated myotubes. Growth arrest specific gene 1 (Gas1) is a GPI-linked membrane protein and originally identified as a growth arrest-linked gene in fibroblasts. Promyogenic cell surface protein, Cdo functions as a component of multiprotein complexes that include other cell adhesion molecules, like Cadherins to mediate cell contact signaling. Here we report that Gas1 and Cdo are coexpressed in muscle cells and form a complex in differentiating myoblasts. Interestingly, Cdo(-/-) myoblasts display defects in Gas1 induction during differentiation. Overexpression or depletion of Gas1 enhances or decreases myogenic differentiation, respectively. During myoblast differentiation, Gas1 depletion causes defects in downregulation of Cdk2 and Cyclin D1 and up-regulation of miR-322, a negative regulator of Cdk2 activities. Furthermore overexpression or knockdown of Gas1 either enhances or decreases activation of p38MAPK that functions downstream of Cdo. Additionally, Gas1 overexpression in Cdo-depleted C2C12 cells restores p38MAPK activities and differentiation abilities. These data suggest that Gas1 promotes myogenic differentiation through regulation of cell cycle arrest and is critical to activate p38MAPK, most likely via association with Cdo/Cadherin multiprotein complexes.


Sujet(s)
Molécules d'adhérence cellulaire/métabolisme , Protéines du cycle cellulaire/métabolisme , Différenciation cellulaire , Activation enzymatique , Développement musculaire , p38 Mitogen-Activated Protein Kinases/métabolisme , Animaux , Molécules d'adhérence cellulaire/génétique , Points de contrôle du cycle cellulaire , Protéines du cycle cellulaire/génétique , Lignée cellulaire , Cycline D1/métabolisme , Kinase-2 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/métabolisme , Protéines liées au GPI/génétique , Protéines liées au GPI/métabolisme , Techniques de knock-down de gènes , Humains , Souris , Protéine MyoD/métabolisme , Myoblastes/métabolisme , Myoblastes/physiologie , Myogénine/métabolisme , Chaînes lourdes de myosine/métabolisme , Liaison aux protéines , Interférence par ARN , cdc25 Phosphatases/génétique , cdc25 Phosphatases/métabolisme
19.
Mol Biol Cell ; 21(14): 2399-411, 2010 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-20484574

RÉSUMÉ

Cell-cell interactions between muscle precursors are required for myogenic differentiation; however, underlying mechanisms are largely unknown. Promyogenic cell surface protein Cdo functions as a component of multiprotein complexes containing other cell adhesion molecules, Boc, Neogenin and N-cadherin, and mediates some of signals triggered by cell-cell interactions between muscle precursors. Cdo activates p38MAPK via interaction with two scaffold proteins JLP and Bnip-2 to promote myogenesis. p38MAPK and Akt signaling are required for myogenic differentiation and activation of both signaling pathways is crucial for efficient myogenic differentiation. We report here that APPL1, an interacting partner of Akt, forms complexes with Cdo and Boc in differentiating myoblasts. Both Cdo and APPL1 are required for efficient Akt activation during myoblast differentiation. The defective differentiation of Cdo-depleted cells is fully rescued by overexpression of a constitutively active form of Akt, whereas overexpression of APPL1 fails to do so. Taken together, Cdo activates Akt through association with APPL1 during myoblast differentiation, and this complex likely mediates some of the promyogenic effect of cell-cell interaction. The promyogenic function of Cdo involves a coordinated activation of p38MAPK and Akt via association with scaffold proteins, JLP and Bnip-2 for p38MAPK and APPL1 for Akt.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Molécules d'adhérence cellulaire/métabolisme , Différenciation cellulaire , Myoblastes/cytologie , Myoblastes/enzymologie , Protéines proto-oncogènes c-akt/métabolisme , Acides aminés/métabolisme , Animaux , Cadhérines/métabolisme , Molécules d'adhérence cellulaire/déficit , Lignée cellulaire , Activation enzymatique , Humains , Protéines membranaires/métabolisme , Souris , Développement musculaire , Liaison aux protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...