Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
1.
ESC Heart Fail ; 10(6): 3559-3572, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37752740

RÉSUMÉ

AIMS: Mechanochemical signalling drives organogenesis and is highly conserved in mammal evolution. Regaining recovery in myocardial jeopardy by inducing principles linking cardiovascular therapy and clinical outcome has been the dream of scientists for decades. Concepts involving embryonic pathways to regenerate adult failing hearts became popular in the early millennium. Since then, abundant data on stem cell research have been published, never reaching widespread application in heart failure therapy. Another conceptual access, using mechanotransduction in cardiac veins to limit myocardial decay, is pressure-controlled intermittent coronary sinus occlusion (PICSO). Recently, we reported acute molecular signs and signals of PICSO activating regulatory miRNA and inducing cell proliferation mimicking cardiac development in adult failing hearts. According to a previously formulated hypothesis, 'embryonic recall', this study aimed to define molecular signals involved in endogenous heart repair during PICSO and study their relation to patient survival. METHODS AND RESULTS: We previously reported a study on the acute molecular effects of PICSO in an observational non-randomized study. Eight out of the thirty-two patients with advanced heart failure undergoing cardiac resynchronization therapy (CRT) were treated with PICSO. Survival was monitored over 10 years, and coronary sinus blood samples were collected during intervention before and after 20 min and tested for miRNA signalling and proliferation when co-cultured with cardiomyocytes. A numerically lower death rate post-CRT and PICSO as compared with control CRT only, and a non-significant reduction in all-cause mortality risk of 42% was observed (37.5% vs. 54.0%, relative risk = 0.58, 95% confidence interval: 0.17-2.05; P = 0.402). Four miRNAs involved in cell cycle, proliferation, morphogenesis, embryonic development, and apoptosis significantly increased concomitantly in survivors and PICSO compared with a decrease in non-survivors (hsa-miR Let7b, P < 0.01; hsa-miR- 421, P < 0.006; hsa-miR 363-3p, P < 0.03 and hsa-miR 19b-3p P < 0.01). In contrast, three miRNAs involved in proliferation and survival, determining cell fate, and recycling endosomes decreased in survivors and PICSO (hsa miR 101-3p, P < 0.03; hsa-miR 25-3p, P < 002; hsa-miR 30d-5p P < 0.04). In vitro cellular proliferation increased in survivors and lowered in non-survivors showing a pattern distinction, discriminating longevity according to up to 10-year survival in heart failure patients. CONCLUSIONS: This study proposes that generating regenerative signals observed during PICSO intervention relate to patient outcomes. Morphogenetic pathways induced by periods of flow reversal in cardiac veins in a domino-like pattern transform embryonic into regenerative signals. Studies supporting the conversion of mechanochemical signals into regenerative molecules during PICSO are warranted to substantiate predictive power on patient longevity, opening new therapeutic avenues in otherwise untreatable heart failure.


Sujet(s)
MicroARN circulant , Défaillance cardiaque , microARN , Adulte , Animaux , Humains , Myocytes cardiaques/métabolisme , Mécanotransduction cellulaire , microARN/génétique , microARN/métabolisme , Défaillance cardiaque/thérapie , Prolifération cellulaire , Mammifères/métabolisme
2.
Stem Cell Reports ; 15(5): 1067-1079, 2020 11 10.
Article de Anglais | MEDLINE | ID: mdl-33125875

RÉSUMÉ

The role of leptin receptor (OB-R) signaling in linking pluripotency with growth and development and the consequences of dysfunctional leptin signaling on progression of metabolic disease is poorly understood. Using a global unbiased proteomics approach we report that embryonic fibroblasts (MEFs) carrying the db/db mutation exhibit metabolic abnormalities, while their reprogrammed induced pluripotent stem cells (iPSCs) show altered expression of proteins involved in embryonic development. An upregulation in expression of eukaryotic translation initiation factor 4e (Eif4e) and Stat3 binding to the Eif4e promoter was supported by enhanced protein synthesis in mutant iPSCs. Directed differentiation of db/db iPSCs toward the neuronal lineage showed defects. Gene editing to correct the point mutation in db/db iPSCs using CRISPR-Cas9, restored expression of neuronal markers and protein synthesis while reversing the metabolic defects. These data imply a direct role for OB-R in regulating metabolism in embryonic fibroblasts and key developmental pathways in iPSCs.


Sujet(s)
Facteur-4E d'initiation eucaryote/métabolisme , Cellules souches pluripotentes induites/métabolisme , Biosynthèse des protéines , Récepteurs à la leptine/métabolisme , Facteur de transcription STAT-3/métabolisme , Transduction du signal , Animaux , Systèmes CRISPR-Cas , Différenciation cellulaire , Lignage cellulaire , Facteur-4E d'initiation eucaryote/génétique , Fibroblastes/métabolisme , Édition de gène , Régulation de l'expression des gènes au cours du développement , Métabolome , Souris , Souris knockout , Neurogenèse , Protéines , Protéomique , Récepteurs à la leptine/génétique
3.
Mol Autism ; 11(1): 2, 2020.
Article de Anglais | MEDLINE | ID: mdl-31921404

RÉSUMÉ

Background: Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder with frequent occurrence of epilepsy, autism spectrum disorder (ASD), intellectual disability (ID), and tumors in multiple organs. The aberrant activation of mTORC1 in TSC has led to treatment with mTORC1 inhibitor rapamycin as a lifelong therapy for tumors, but TSC-associated neurocognitive manifestations remain unaffected by rapamycin. Methods: Here, we generated patient-specific, induced pluripotent stem cells (iPSCs) from a TSC patient with a heterozygous, germline, nonsense mutation in exon 15 of TSC1 and established an isogenic set of heterozygous (Het), null and corrected wildtype (Corr-WT) iPSCs using CRISPR/Cas9-mediated gene editing. We differentiated these iPSCs into neural progenitor cells (NPCs) and examined neurodevelopmental phenotypes, signaling and changes in gene expression by RNA-seq. Results: Differentiated NPCs revealed enlarged cell size in TSC1-Het and Null NPCs, consistent with mTORC1 activation. TSC1-Het and Null NPCs also revealed enhanced proliferation and altered neurite outgrowth in a genotype-dependent manner, which was not reversed by rapamycin. Transcriptome analyses of TSC1-NPCs revealed differentially expressed genes that display a genotype-dependent linear response, i.e., genes upregulated/downregulated in Het were further increased/decreased in Null. In particular, genes linked to ASD, epilepsy, and ID were significantly upregulated or downregulated warranting further investigation. In TSC1-Het and Null NPCs, we also observed basal activation of ERK1/2, which was further activated upon rapamycin treatment. Rapamycin also increased MNK1/2-eIF4E signaling in TSC1-deficient NPCs. Conclusion: MEK-ERK and MNK-eIF4E pathways regulate protein translation, and our results suggest that aberrant translation distinct in TSC1/2-deficient NPCs could play a role in neurodevelopmental defects. Our data showing upregulation of these signaling pathways by rapamycin support a strategy to combine a MEK or a MNK inhibitor with rapamycin that may be superior for TSC-associated CNS defects. Importantly, our generation of isogenic sets of NPCs from TSC patients provides a valuable platform for translatome and large-scale drug screening studies. Overall, our studies further support the notion that early developmental events such as NPC proliferation and initial process formation, such as neurite number and length that occur prior to neuronal differentiation, represent primary events in neurogenesis critical to disease pathogenesis of neurodevelopmental disorders such as ASD.


Sujet(s)
Facteur-4E d'initiation eucaryote/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Cellules souches neurales/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Complexe de la sclérose tubéreuse , Systèmes CRISPR-Cas , Codon non-sens , Édition de gène , Mutation germinale , Humains , Cellules souches pluripotentes induites/cytologie , Complexe-1 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Neurogenèse , Phénotype , RNA-Seq , Transduction du signal , Sirolimus , Protéine-1 du complexe de la sclérose tubéreuse/génétique
4.
Mol Metab ; 24: 108-119, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30940487

RÉSUMÉ

OBJECTIVE: Impaired expansion of peripheral fat contributes to the pathogenesis of insulin resistance and Type 2 Diabetes (T2D). We aimed to identify novel disease-gene interactions during adipocyte differentiation. METHODS: Genes in disease-associated loci for T2D, adiposity and insulin resistance were ranked according to expression in human adipocytes. The top 125 genes were ablated in human pre-adipocytes via CRISPR/CAS9 and the resulting cellular phenotypes quantified during adipocyte differentiation with high-content microscopy and automated image analysis. Morphometric measurements were extracted from all images and used to construct morphologic profiles for each gene. RESULTS: Over 107 morphometric measurements were obtained. Clustering of the morphologic profiles accross all genes revealed a group of 14 genes characterized by decreased lipid accumulation, and enriched for known lipodystrophy genes. For two lipodystrophy genes, BSCL2 and AGPAT2, sub-clusters with PLIN1 and CEBPA identifed by morphological similarity were validated by independent experiments as novel protein-protein and gene regulatory interactions. CONCLUSIONS: A morphometric approach in adipocytes can resolve multiple cellular mechanisms for metabolic disease loci; this approach enables mechanistic interrogation of the hundreds of metabolic disease loci whose function still remains unknown.


Sujet(s)
Adipocytes/cytologie , Adipogenèse , Diabète/génétique , Réseaux de régulation génique , Cartes d'interactions protéiques , Acyltransferases/génétique , Acyltransferases/métabolisme , Adipocytes/métabolisme , Adipocytes/anatomopathologie , Protéines liant les séquences stimulatrices de type CCAAT/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Cellules cultivées , Diabète/anatomopathologie , Sous-unités gamma des protéines G/génétique , Sous-unités gamma des protéines G/métabolisme , Cellules HEK293 , Humains , Insulinorésistance , Périlipine-1/génétique , Périlipine-1/métabolisme , Phénotype , Transcriptome
5.
ESC Heart Fail ; 5(6): 1176-1183, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30230713

RÉSUMÉ

AIMS: Cardiac repair has steered clinical attention and remains an unmet need, because available regenerative therapies lack robust mechanistic evidence. Pressure-controlled intermittent coronary sinus occlusion (PICSO), known to induce angiogenetic and vasoactive molecules as well as to reduce regional ischemia, may activate endogenous regenerative processes in failing myocardium. We aimed to investigate the effects of PICSO in patients with advanced heart failure undergoing cardiac resynchronization therapy. METHODS AND RESULTS: Eight out of 32 patients were treated with PICSO, and the remainder served as controls. After electrode testing including left ventricular leads, PICSO was performed for 20 min. To test immediate molecular responses, in both patient groups, coronary venous blood samples were taken at baseline and after 20 min, the time required for the intervention. Sera were tested for microRNAs and growth factors. To test the ability of up-regulated soluble factors on cell proliferation and expression of transcription factors [e.g. Krüppel-like factor 4 (KLF-4)], sera were co-cultured with human cardiomyocytes and fibroblasts. As compared with controls, significant differential expression (differences between pre-values and post-values in relation to both patient cohorts) of microRNA patterns associated with cardiac development was observed with PICSO. Importantly, miR-143 (P < 0.048) and miR-145 (P < 0,047) increased, both targeting a network of transcription factors (including KLF-4) that promote differentiation and repress proliferation of vascular smooth muscle cells. Additionally, an increase of miR-19b (P < 0.019) known to alleviate endothelial cell apoptosis was found, whereas disadvantageous miR-320b (P < 0.023) suspect to impair expression of c-myc, normally provoking cell cycle re-entry in post-mitotic myocytes and miR-25 (P < 0.023), decreased, a target of anti-miR application to improve contractility in the failing heart. Co-cultured post-PICSO sera significantly increased cellular proliferation both in fibroblasts (P < 0.001) and adult cardiomycytes (P < 0.004) sampled from a transplant recipient as compared with controls. Adult cardiomyocytes showed a seven-fold increase of the transcription factor KLF-4 protein when co-cultured with treated sera as compared with controls. CONCLUSIONS: Here, we show for the first time that PICSO, a trans-coronary sinus catheter intervention, is associated with an increase in morphogens secreted into cardiac veins, normally present during cardiac development, and a significant induction of cell proliferation. Present findings support the notion that epigenetic modifications, that is, haemodynamic stimuli on venous vascular cells, may reverse myocardial deterioration. Further investigations are needed to decipher the maze of complex interacting molecular pathways in failing myocardium and the potential role of PICSO to reinitiate developmental processes to prevent further myocardial decay eventually reaching clinical significance.


Sujet(s)
Occlusion par ballonnet/méthodes , Cathétérisme cardiaque/méthodes , Circulation coronarienne/physiologie , Sinus coronaire/physiopathologie , Vaisseaux coronaires/physiopathologie , Défaillance cardiaque/thérapie , Sujet âgé , Marqueurs biologiques/sang , Femelle , Défaillance cardiaque/sang , Défaillance cardiaque/physiopathologie , Humains , Facteur-4 de type Kruppel , Mâle , Adulte d'âge moyen , Pression
6.
Mol Ther Methods Clin Dev ; 5: 116-129, 2017 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-28480311

RÉSUMÉ

The role of striatin interacting protein 2 (Strip2) in differentiation of embryonic stem cells (ESCs) is still under debate. Strip2-silenced murine (KD) ESCs were differentiated for 4, 8, 12, and 16 days. We show that Strip2 is distributed in the perinucleus or nuclei of wild-type (WT) undifferentiated ESCs, but is localized in high-density nuclear bodies in differentiated cells. CellNet analysis of microarray gene expression data for the KD and scrambled control (SCR) embryoid bodies (EBs), as well as immunostainings of key pluripotent factors, demonstrated that differentiation of KD ESCs is repressed. This occurs even in 16-day-old EBs, which possessed a high tumorigenic potential. Correlated with very high expression levels of epigenetic regulator genes, Hat1 and Dnmt3, enzymatic activities of the histone acetyltransferase type B (Hat1) and DNA (cytosine-5)-methyltransferase 3 beta (Dnmt3b) were higher in differentiated 16-day-old KD EBs than in SCR or WT EBs. The expression levels of let-7, 290, and 302 microRNA families were opposed in KD ESCs, while KD EBs had levels comparable to WT and SCR ESCs during differentiation. Strip2 is critical for the regular differentiation of ESCs. Moreover, Strip2 deficient ESCs showed a dysregulation of epigenetic regulators and microRNAs regulating pluripotency.

7.
Amino Acids ; 49(12): 1955-1963, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-28421296

RÉSUMÉ

Drug-induced off-target cardiotoxicity, particularly following anti-cancer therapy, is a major concern in new drug discovery and development. To ensure patient safety and efficient pharmaceutical drug development, there is an urgent need to develop more predictive cell model systems and distinct toxicity signatures. In this study, we applied our previously proposed repeated exposure toxicity methodology and performed 1H NMR spectroscopy-based extracellular metabolic profiling in culture medium of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) exposed to doxorubicin (DOX), an anti-cancer agent. Single exposure to DOX did not show alteration in the basal level of extracellular metabolites while repeated exposure to DOX caused reduction in the utilization of pyruvate and acetate, and accumulation of formate compared to control culture medium. During drug washout, only pyruvate showed reversible effect and restored its utilization by hiPSC-CMs. On the other hand, formate and acetate showed irreversible effect in response to DOX exposure. DOX repeated exposure increased release of lactate dehydrogenase (LDH) in culture medium suggesting cytotoxicity events, while declined ATP levels in hiPSC-CMs. Our data suggests DOX perturbed mitochondrial metabolism in hiPSC-CMs. Pyruvate, acetate and formate can be used as metabolite signatures of DOX induced cardiotoxicity. Moreover, the hiPSC-CMs model system coupled with metabolomics technology offers a novel and powerful approach to strengthen cardiac safety assessment during new drug discovery and development.


Sujet(s)
Doxorubicine/toxicité , Cellules souches pluripotentes induites/cytologie , Métabolome/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Acide acétique/analyse , Adénosine triphosphate/analyse , Adénosine triphosphate/métabolisme , Biomarqueurs pharmacologiques/analyse , Biomarqueurs pharmacologiques/métabolisme , Cardiotoxines/toxicité , Différenciation cellulaire , Cellules cultivées , Formiates/analyse , Humains , L-Lactate dehydrogenase/analyse , L-Lactate dehydrogenase/métabolisme , Métabolomique , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Spectroscopie par résonance magnétique du proton , Acide pyruvique/analyse , Facteurs temps
8.
Arch Toxicol ; 90(11): 2763-2777, 2016 Nov.
Article de Anglais | MEDLINE | ID: mdl-26537877

RÉSUMÉ

The currently available techniques for the safety evaluation of candidate drugs are usually cost-intensive and time-consuming and are often insufficient to predict human relevant cardiotoxicity. The purpose of this study was to develop an in vitro repeated exposure toxicity methodology allowing the identification of predictive genomics biomarkers of functional relevance for drug-induced cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The hiPSC-CMs were incubated with 156 nM doxorubicin, which is a well-characterized cardiotoxicant, for 2 or 6 days followed by washout of the test compound and further incubation in compound-free culture medium until day 14 after the onset of exposure. An xCELLigence Real-Time Cell Analyser was used to monitor doxorubicin-induced cytotoxicity while also monitoring functional alterations of cardiomyocytes by counting of the beating frequency of cardiomyocytes. Unlike single exposure, repeated doxorubicin exposure resulted in long-term arrhythmic beating in hiPSC-CMs accompanied by significant cytotoxicity. Global gene expression changes were studied using microarrays and bioinformatics tools. Analysis of the transcriptomic data revealed early expression signatures of genes involved in formation of sarcomeric structures, regulation of ion homeostasis and induction of apoptosis. Eighty-four significantly deregulated genes related to cardiac functions, stress and apoptosis were validated using real-time PCR. The expression of the 84 genes was further studied by real-time PCR in hiPSC-CMs incubated with daunorubicin and mitoxantrone, further anthracycline family members that are also known to induce cardiotoxicity. A panel of 35 genes was deregulated by all three anthracycline family members and can therefore be expected to predict the cardiotoxicity of compounds acting by similar mechanisms as doxorubicin, daunorubicin or mitoxantrone. The identified gene panel can be applied in the safety assessment of novel drug candidates as well as available therapeutics to identify compounds that may cause cardiotoxicity.


Sujet(s)
Anthracyclines/effets indésirables , Cardiotoxines/effets indésirables , Médicaments en essais cliniques/effets indésirables , Myocytes cardiaques/effets des médicaments et des substances chimiques , Antibiotiques antinéoplasiques/effets indésirables , Biomarqueurs pharmacologiques/métabolisme , Cellules cultivées , Biologie informatique , Daunorubicine/effets indésirables , Doxorubicine/effets indésirables , Évaluation préclinique de médicament , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Cellules souches pluripotentes induites/cytologie , Mitoxantrone/effets indésirables , Annotation de séquence moléculaire , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Séquençage par oligonucléotides en batterie , Réaction de polymérisation en chaine en temps réel , RT-PCR , Inhibiteurs de la topoisomérase-II/effets indésirables , Tests de toxicité chronique
9.
PLoS One ; 10(10): e0140192, 2015.
Article de Anglais | MEDLINE | ID: mdl-26445504

RÉSUMÉ

The multi-subunit mammalian Mediator complex acts as an integrator of transcriptional regulation by RNA Polymerase II, and has emerged as a master coordinator of development and cell fate determination. We previously identified the Mediator subunit, MED28, as a cytosolic binding partner of merlin, the Neurofibromatosis 2 (NF2) tumor suppressor, and thus MED28 is distinct in having a cytosolic role as an NF2 interacting protein as well as a nuclear role as a Mediator complex subunit. Although limited in vitro studies have been performed on MED28, its in vivo function remains unknown. Employing a knockout mouse model, we describe for the first time the requirement for Med28 in the developing mouse embryo. Med28-deficiency causes peri-implantation lethality resulting from the loss of pluripotency of the inner cell mass accompanied by reduced expression of key pluripotency transcription factors Oct4 and Nanog. Further, overexpression of Med28 in mouse embryonic fibroblasts enhances the efficiency of their reprogramming to pluripotency. Cre-mediated inactivation of Med28 in induced pluripotent stem cells shows that Med28 is required for their survival. Intriguingly, heterozygous loss of Med28 results in differentiation of induced pluripotent stem cells into extraembryonic trophectoderm and primitive endoderm lineages. Our findings document the essential role of Med28 in the developing embryo as well as in acquisition and maintenance of pluripotency during reprogramming.


Sujet(s)
Implantation embryonnaire , Régulation de l'expression des gènes au cours du développement , Cellules souches pluripotentes induites/métabolisme , Complexe médiateur/métabolisme , Animaux , Différenciation cellulaire , Reprogrammation cellulaire , Perte de l'embryon/génétique , Perte de l'embryon/métabolisme , Délétion de gène , Cellules souches pluripotentes induites/cytologie , Complexe médiateur/génétique , Souris , Souris de lignée C57BL , Souris knockout
10.
Oncotarget ; 6(19): 16981-97, 2015 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-26219339

RÉSUMÉ

Meningiomas are the most common primary intracranial adult tumor. All Neurofibromatosis 2 (NF2)-associated meningiomas and ~60% of sporadic meningiomas show loss of NF2 tumor suppressor protein. There are no effective medical therapies for progressive and recurrent meningiomas. Our previous work demonstrated aberrant activation of mTORC1 signaling that led to ongoing clinical trials with rapamycin analogs for NF2 and sporadic meningioma patients. Here we performed a high-throughput kinome screen to identify kinases responsible for mTORC1 pathway activation in NF2-deficient meningioma cells. Among the emerging top candidates were the mTORC2-specific target serum/glucocorticoid-regulated kinase 1 (SGK1) and p21-activated kinase 1 (PAK1). In NF2-deficient meningioma cells, inhibition of SGK1 rescues mTORC1 activation, and SGK1 activation is sensitive to dual mTORC1/2 inhibitor AZD2014, but not to rapamycin. PAK1 inhibition also leads to attenuated mTORC1 but not mTORC2 signaling, suggesting that mTORC2/SGK1 and Rac1/PAK1 pathways are independently responsible for mTORC1 activation in NF2-deficient meningiomas. Using CRISPR-Cas9 genome editing, we generated isogenic human arachnoidal cell lines (ACs), the origin cell type for meningiomas, expressing or lacking NF2. NF2-null CRISPR ACs recapitulate the signaling of NF2-deficient meningioma cells. Interestingly, we observe increased SGK1 transcription and protein expression in NF2-CRISPR ACs and in primary NF2-negative meningioma lines. Moreover, we demonstrate that the dual mTORC1/mTORC2 inhibitor, AZD2014 is superior to rapamycin and PAK inhibitor FRAX597 in blocking proliferation of meningioma cells. Importantly, AZD2014 is currently in use in several clinical trials of cancer. Therefore, we believe that AZD2014 may provide therapeutic advantage over rapalogs for recurrent and progressive meningiomas.


Sujet(s)
Protéines précoces immédiates/métabolisme , Tumeurs des méninges/enzymologie , Méningiome/enzymologie , Protein-Serine-Threonine Kinases/métabolisme , Antinéoplasiques/pharmacologie , Benzamides , Lignée cellulaire tumorale , Techniques de knock-down de gènes , Séquençage nucléotidique à haut débit , Humains , Immunotransfert , Tumeurs des méninges/génétique , Méningiome/génétique , Morpholines/pharmacologie , Neurofibromatose de type 2/génétique , Réaction de polymérisation en chaîne , Pyrimidines , Transduction du signal/effets des médicaments et des substances chimiques
11.
Stem Cell Res Ther ; 5(3): 75, 2014 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-24906886

RÉSUMÉ

INTRODUCTION: Posttranscriptional control of mRNA by microRNA (miRNA) has been implicated in the regulation of diverse biologic processes from directed differentiation of stem cells through organism development. We describe a unique pathway by which miRNA regulates the specialized differentiation of cardiomyocyte (CM) subtypes. METHODS: We differentiated human embryonic stem cells (hESCs) to cardiac progenitor cells and functional CMs, and characterized the regulated expression of specific miRNAs that target transcriptional regulators of left/right ventricular-subtype specification. RESULTS: From >900 known human miRNAs in hESC-derived cardiac progenitor cells and functional CMs, a subset of differentially expressed cardiac miRNAs was identified, and in silico analysis predicted highly conserved binding sites in the 3'-untranslated regions (3'UTRs) of Hand-and-neural-crest-derivative-expressed (HAND) genes 1 and 2 that are involved in left and right ventricular development. We studied the temporal and spatial expression patterns of four miRNAs in differentiating hESCs, and found that expression of miRNA (miR)-363, miR-367, miR-181a, and miR-181c was specific for stage and site. Further analysis showed that miR-363 overexpression resulted in downregulation of HAND1 mRNA and protein levels. A dual luciferase reporter assay demonstrated functional interaction of miR-363 with the full-length 3'UTR of HAND1. Expression of anti-miR-363 in-vitro resulted in enrichment for HAND1-expressing CM subtype populations. We also showed that BMP4 treatment induced the expression of HAND2 with less effect on HAND1, whereas miR-363 overexpression selectively inhibited HAND1. CONCLUSIONS: These data show that miR-363 negatively regulates the expression of HAND1 and suggest that suppression of miR-363 could provide a novel strategy for generating functional left-ventricular CMs.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/biosynthèse , Cellules souches embryonnaires/cytologie , Régulation de l'expression des gènes au cours du développement/génétique , Ventricules cardiaques/cytologie , microARN/génétique , Myocytes cardiaques/cytologie , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Différenciation cellulaire/génétique , Lignée cellulaire , Humains , Immunotransfert , Hybridation fluorescente in situ , Cellules souches pluripotentes induites/cytologie , Réaction de polymérisation en chaine en temps réel
12.
PLoS One ; 8(8): e70913, 2013.
Article de Anglais | MEDLINE | ID: mdl-23936474

RÉSUMÉ

Catecholamine release is known to modulate cardiac output by increasing heart rate. Although much is known about catecholamine function and regulation in adults, little is known about the presence and role of catecholamines during heart development. The present study aimed therefore to evaluate the effects of different catecholamines on early heart development in an in vitro setting using embryonic stem (ES) cell-derived cardiomyocytes. Effects of catecholamine depletion induced by reserpine were examined in murine ES cells (line D3, αPIG44) during differentiation. Cardiac differentiation was assessed by immunocytochemistry, qRT-PCR, quantification of beating clusters, flow cytometry and pharmacological approaches. Proliferation was analyzed by EB cross-section measurements, while functionality of cardiomyocytes was studied by extracellular field potential (FP) measurements using microelectrode arrays (MEAs). To further differentiate between substance-specific effects of reserpine and catecholamine action via α- and ß-receptors we proved the involvement of adrenergic receptors by application of unspecific α- and ß-receptor antagonists. Reserpine treatment led to remarkable down-regulation of cardiac-specific genes, proteins and mesodermal marker genes. In more detail, the average ratio of ∼40% spontaneously beating control clusters was significantly reduced by 100%, 91.1% and 20.0% on days 10, 12, and 14, respectively. Flow cytometry revealed a significant reduction (by 71.6%, n = 11) of eGFP positive CMs after reserpine treatment. By contrast, reserpine did not reduce EB growth while number of neuronal cells in reserpine-treated EBs was significantly increased. MEA measurements of reserpine-treated EBs showed lower FP frequencies and weak responsiveness to adrenergic and muscarinic stimulation. Interestingly we found that developmental inhibition after α- and ß-adrenergic blocker application mimicked developmental changes with reserpine. Using several methodological approaches our data suggest that reserpine inhibits cardiac differentiation. Thus catecholamines play a critical role during development.


Sujet(s)
Catécholamines/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignage cellulaire/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/cytologie , Myocytes cardiaques/cytologie , Inhibiteurs de la capture adrénergique/pharmacologie , Animaux , Techniques de biocapteur , Cellules cultivées , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/métabolisme , Cytométrie en flux , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Immunotransfert , Techniques immunoenzymatiques , Souris , Analyse sur microréseau , Microélectrodes , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Neurones/cytologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , Récepteurs adrénergiques/composition chimique , Récepteurs adrénergiques/métabolisme , Réserpine/pharmacologie , RT-PCR
13.
Stem Cell Res ; 10(2): 228-40, 2013 Mar.
Article de Anglais | MEDLINE | ID: mdl-23314291

RÉSUMÉ

Human embryonic stem cells (hESCs) can be used to model the cellular and molecular mechanisms that underlie embryonic development. Understanding the cellular mechanisms and pathways involved in extraembryonic (ExE) differentiation is of great interest because of the important role of this process in maternal health and fertility. Fibroblast growth factor 2 (FGF-2) is widely used to maintain the self-renewal of hESCs and induced pluripotent stem cells, while all trans retinoic acid (RA) is used to facilitate the directed differentiation of hESCs. Here, we monitored the RA induced differentiation of hESCs to the ExE lineage with and without FGF-2 over a 7-day period via global transcriptional profiling. The stemness markers POU5F1, NANOG and TDGF1 were markedly downregulated, whereas an upregulation of the ExE markers KRT7, CGA, DDAH2 and IGFBP3 was observed. Many of the differentially expressed genes were involved in WNT and TGF-ß signaling. RA inactivated WNT signaling even in the presence of exogenous FGF-2, which that promotes the maintenance of the pluripotent state. We also show that BMP4 was upregulated and that RA was able to modulate the TGF-ß signaling pathway and direct hESCs toward the ExE lineage. In addition, an epigenetic study revealed hypermethylation of the DDAH2, TDGF1 and GATA3 gene promoters, suggesting a role for epigenetic regulation during ExE differentiation. These data reveals that the effect of RA prevails in the presence of exogenous FGF-2 thus resulting in the direction of hESCs toward the ExE lineage.


Sujet(s)
Lignage cellulaire/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/cytologie , Membranes extraembryonnaires/cytologie , Facteur de croissance fibroblastique de type 2/pharmacologie , Cellules souches pluripotentes/cytologie , Trétinoïne/pharmacologie , Animaux , Marqueurs biologiques/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , Lignée cellulaire , Lignage cellulaire/génétique , Forme de la cellule/effets des médicaments et des substances chimiques , Forme de la cellule/génétique , Analyse de regroupements , Synergie des médicaments , Ectoderme/cytologie , Ectoderme/effets des médicaments et des substances chimiques , Ectoderme/métabolisme , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/métabolisme , Membranes extraembryonnaires/effets des médicaments et des substances chimiques , Membranes extraembryonnaires/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Souris , Cellules souches pluripotentes/effets des médicaments et des substances chimiques , Cellules souches pluripotentes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Facteurs temps , Facteurs de transcription/métabolisme , Transcriptome/génétique , Trophoblastes/cytologie , Trophoblastes/effets des médicaments et des substances chimiques , Trophoblastes/métabolisme
14.
PLoS One ; 7(8): e44228, 2012.
Article de Anglais | MEDLINE | ID: mdl-22952932

RÉSUMÉ

Embryonic development can be partially recapitulated in vitro by differentiating human embryonic stem cells (hESCs). Thalidomide is a developmental toxicant in vivo and acts in a species-dependent manner. Besides its therapeutic value, thalidomide also serves as a prototypical model to study teratogenecity. Although many in vivo and in vitro platforms have demonstrated its toxicity, only a few test systems accurately reflect human physiology. We used global gene expression and proteomics profiling (two dimensional electrophoresis (2DE) coupled with Tandem Mass spectrometry) to demonstrate hESC differentiation and thalidomide embryotoxicity/teratogenecity with clinically relevant dose(s). Proteome analysis showed loss of POU5F1 regulatory proteins PKM2 and RBM14 and an over expression of proteins involved in neuronal development (such as PAK2, PAFAH1B2 and PAFAH1B3) after 14 days of differentiation. The genomic and proteomic expression pattern demonstrated differential expression of limb, heart and embryonic development related transcription factors and biological processes. Moreover, this study uncovered novel possible mechanisms, such as the inhibition of RANBP1, that participate in the nucleocytoplasmic trafficking of proteins and inhibition of glutathione transferases (GSTA1, GSTA2), that protect the cell from secondary oxidative stress. As a proof of principle, we demonstrated that a combination of transcriptomics and proteomics, along with consistent differentiation of hESCs, enabled the detection of canonical and novel teratogenic intracellular mechanisms of thalidomide.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/métabolisme , Analyse de profil d'expression de gènes , Protéomique , Thalidomide/pharmacologie , Relation dose-effet des médicaments , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Développement embryonnaire/effets des médicaments et des substances chimiques , Développement embryonnaire/génétique , Cellules souches embryonnaires/cytologie , Membres/embryologie , Régulation de l'expression des gènes au cours du développement/effets des médicaments et des substances chimiques , Glutathione transferase/génétique , Glutathione transferase/métabolisme , Coeur/effets des médicaments et des substances chimiques , Coeur/embryologie , Humains , Cinétique , Transport des protéines/effets des médicaments et des substances chimiques , ARN messager/génétique , ARN messager/métabolisme , Facteurs temps , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines de type Wingless/génétique , Protéines de type Wingless/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Voie de signalisation Wnt/génétique
15.
Stem Cells Dev ; 21(13): 2471-84, 2012 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-22420508

RÉSUMÉ

Investigating the molecular mechanisms controlling the in vivo developmental program postembryogenesis is challenging and time consuming. However, the developmental program can be partly recapitulated in vitro by the use of cultured embryonic stem cells (ESCs). Similar to the totipotent cells of the inner cell mass, gene expression and morphological changes in cultured ESCs occur hierarchically during their differentiation, with epiblast cells developing first, followed by germ layers and finally somatic cells. Combination of high throughput -omics technologies with murine ESCs offers an alternative approach for studying developmental processes toward organ-specific cell phenotypes. We have made an attempt to understand differentiation networks controlling embryogenesis in vivo using a time kinetic, by identifying molecules defining fundamental biological processes in the pluripotent state as well as in early and the late differentiation stages of ESCs. Our microarray data of the differentiation of the ESCs clearly demonstrate that the most critical early differentiation processes occur at days 2 and 3 of differentiation. Besides monitoring well-annotated markers pertinent to both self-renewal and potency (capacity to differentiate to different cell lineage), we have identified candidate molecules for relevant signaling pathways. These molecules can be further investigated in gain and loss-of-function studies to elucidate their role for pluripotency and differentiation. As an example, siRNA knockdown of MageB16, a gene highly expressed in the pluripotent state, has proven its influence in inducing differentiation when its function is repressed.


Sujet(s)
Différenciation cellulaire , Cellules souches embryonnaires/cytologie , Régulation de l'expression des gènes au cours du développement , Cellules souches pluripotentes/cytologie , Transcriptome , Animaux , Marqueurs biologiques/métabolisme , Techniques de culture cellulaire , Développement embryonnaire , Cellules souches embryonnaires/métabolisme , Feuillets embryonnaires/cytologie , Feuillets embryonnaires/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Souris , Protéine homéotique Nanog , Facteur de transcription Oct-3/génétique , Facteur de transcription Oct-3/métabolisme , Séquençage par oligonucléotides en batterie , Cellules souches pluripotentes/métabolisme , Analyse en composantes principales , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal , Facteurs temps
16.
Front Biosci (Elite Ed) ; 4(1): 156-68, 2012 01 01.
Article de Anglais | MEDLINE | ID: mdl-22201861

RÉSUMÉ

Chemopreventive agents are derived from edible plants and from ancient time is a part of daily intake for many humans and animals. There are several lines of compelling evidence from epidemiological, clinical and laboratory studies that these dietary constituents are associated in reducing cancer risks. However, developmental toxicity of these natural compounds cannot be excluded. In the present study, we examined the effect of chemopreventive agents on the differentiation of mouse embryonic stem cells (ESCs) as an in vitro embryotoxicity model. We assumed that inhibition of developmentally regulated genes in vitro might predict developmental toxicity also under in vivo conditions. We found that epigallocatechin gallate (EGCG) (20 microM) induced the expression of mesodermal and cardiomyocyte genes and a significant increase in the number and the percentage of cardiomyocytes. The increase of the subpopulation correlated with higher numbers of beating foci and beating frequencies. Curcumin on the other hand at 0.4 mM was seen to enhance expression of ectodermal transcripts. Quercetin (2.5 microM) was found to inhibit several developmentally regulated genes.


Sujet(s)
Anticarcinogènes/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Animaux , Séquence nucléotidique , Technique de Western , Lignée cellulaire , Amorces ADN , Cellules souches embryonnaires/cytologie , Immunohistochimie , Souris , Réaction de polymérisation en chaine en temps réel
17.
Front Biosci (Elite Ed) ; 3(4): 1273-88, 2011 06 01.
Article de Anglais | MEDLINE | ID: mdl-21622134

RÉSUMÉ

The deregulated activity of the Bcr-Abl tyrosine kinase provides a rational basis for the development therapeutics in all phases of Chronic Myelogenous Leukemia (CML). Although a well studied imatinib therapy has clinical success against CML, resistance to imatinib due to mutations in the kinase domain, especially T315I poses a major problem for the ultimate success of CML therapy by this agent. Herein we describe an NPB001-05, derived from extract of Piper betle leafs, which is highly active in specifically inhibiting Bcr-Abl expressing cells. NPB001-05 inhibited the proliferation of BaF3 cells ectopically expressing wild type Bcr-Abl phenotype and 12 different imatinib-resistant mutations of clinical relevance (average IC50 5.7 microg/ml). Moreover, NPB001-05 was highly inhibitory to wild type P210(Bcr-Abl) and P210(Bcr-Abl-T315I) kinase activity and abrogated the autophosphorylating enzyme in time- and dose- dependent manner. NPB001-05 was non-toxic on normal cells, but was inhibitory to CML patient derived peripheral blood mononuclear cells. Treatment with NPB001-05 caused apoptosis induction and G0G1 cell cycle arrest in both Bcr-Abl wild type and T315I mutant cell lines.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Protéines de fusion bcr-abl/antagonistes et inhibiteurs , Pipérazines/pharmacologie , Extraits de plantes/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Protein-tyrosine kinases/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Benzamides , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Protéines de fusion bcr-abl/génétique , Humains , Mésilate d'imatinib , Mutation , Tumeurs/anatomopathologie , Phosphorylation , Protein-tyrosine kinases/génétique
18.
Front Biosci (Elite Ed) ; 3(4): 1349-64, 2011 06 01.
Article de Anglais | MEDLINE | ID: mdl-21622141

RÉSUMÉ

Scientists are constantly searching for phytochemical compounds with anti-cancer activity. In this study, activity of plant extract NPB001-05 from Piper betle was tested on human chronic myelogenous leukemia (CML) xenograft models. NPB001-05 was active when dosed orally (500 mg/kg) once or twice a day in xenograft tumor models. NPB001-05 showed activity to T315I tumor xenograft, where imatinib failed to show antitumor activity. NPB001-05 showed no relevant toxicity in animal models during 2 weeks exposure to drug. Responsive tumor showed inhibition of tyrosine kinase activity with lowered Bcr-Abl protein levels and increased apoptosis. Microarray based transcription profiling studies demonstrated that both imatinib and NPB001-05 dysregulated imatinib- responsive genes. NPB001-05 showed additional genes selectively dysregulated from ER stress, PI3K/AKT, MAPK pathways. Additionally, we tested gene expression of PI3K, AKT1, JUN, CASP3 and DDIT3 in K562, BaF3P210(BCR-ABL) and BaF3 P210(BCR-ABLT315I) cell line treated for 6- and 12- hours with NPB001-05 and imatinib. The data indicates that NPB001-05 mediated cell death in K562 affects the function of ER stress. NPB001-05 shows antitumor activity with favorable toxicity profile.


Sujet(s)
Antinéoplasiques/pharmacologie , Antienzymes/pharmacologie , Protéines de fusion bcr-abl/antagonistes et inhibiteurs , Extraits de plantes/pharmacologie , Protein-tyrosine kinases/antagonistes et inhibiteurs , Administration par voie orale , Animaux , Antinéoplasiques/administration et posologie , Séquence nucléotidique , Amorces ADN , Antienzymes/administration et posologie , Humains , Mâle , Souris , Souris SCID , Extraits de plantes/administration et posologie , RT-PCR
19.
Stem Cell Rev Rep ; 7(3): 506-17, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-21279480

RÉSUMÉ

Human embryonic stem cells (hESCs) can be propagated indefinitely in vitro in an undifferentiated pluripotent state, can differentiate into derivatives of all three germ layers and are of considerable interest for applications in regenerative medicine. Clinical application of hESCs, however, requires reliable protocols for cryopreservation. Current protocols for cryopreservation of hESCs suffer from low recovery rates of hESCs and loss of pluripotency after thawing. We therefore studied the effects of cryopreservation on the viability, proliferation potential, and the pluripotency status of hESCs by combining cellular readouts and transcriptomics. We identified biological processes and pathways affected by cryopreservation in order to understand the limited survival rate of hESCs by comparing transcriptomes of hESCs at different time points after thawing with cells that did not undergo cryopreservation. While the transcriptomes of cells post thawing were very similar to those of control non-frozen hESCs for the early time points, we observed increased expression of genes involved in apoptosis, embryonic morphogenesis, ossification, tissue morphogenesis, regeneration, vasculature development and cell death at later time points. Our data suggest that inhibition of anoikis apoptosis and the stress-induced differentiation pathways are promising targets for improving the survival rate and maintaining pluripotency of hESCs after cryopreservation.


Sujet(s)
Techniques de culture cellulaire/méthodes , Cryoconservation/méthodes , Cellules souches embryonnaires/physiologie , Analyse de profil d'expression de gènes , Marqueurs biologiques/métabolisme , Mort cellulaire , Différenciation cellulaire/physiologie , Prolifération cellulaire , Survie cellulaire , Cellules souches embryonnaires/cytologie , Humains , Analyse sur microréseau , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/physiologie
20.
Cell Physiol Biochem ; 25(6): 595-604, 2010.
Article de Anglais | MEDLINE | ID: mdl-20511704

RÉSUMÉ

Early mammalian heart development is characterized by transient expression of alpha-smooth muscle actin (Acta2). To date, cardiomyocytes expressing Acta2 in the early stages of in vivo development have not been characterized. To functionally characterize Acta2-expressing cardiomyocytes, we used a transgenic ES cell line expressing both the puromycin acetyl transferase (Pac) and enhanced green fluorescent protein (EGFP) cassettes under the control of the Acta2 promoter. The onset of Acta2 expression occurred in parallel with the appearance of beating areas, indicating the formation of cardiomyocytes. Antibiotic selection resulted in a high yield of cardiomyocytes and smooth muscle cells. The green fluorescent beating areas stained positively for multiple cardiomyocyte markers. Comparative electrophysiological analysis including fetal and alpha-MHC-expressing ES cell-derived cardiomyocyte controls showed that Acta2-positive cardiomyocytes contained pacemaker-, atrial- and ventricular-like phenotypes. Interestingly, the proportion of ventricular-like cells was much higher in the Acta2-positive cardiomyocytes population than in control alpha-MHC-expressing cardiomyocytes (75 % and 12 %, respectively). The findings of the present study provide a novel approach for the identification and enrichment of Acta2-positive cardiomyocytes, especially of the ventricular phenotype under in vitro conditions.


Sujet(s)
Actines/isolement et purification , Actines/métabolisme , Cellules souches embryonnaires/cytologie , Muscles lisses/métabolisme , Myocytes cardiaques/métabolisme , Acetyltransferases/génétique , Actines/génétique , Animaux , Lignée cellulaire , Séparation cellulaire , Expression des gènes , Protéines à fluorescence verte/génétique , Souris , Myocytes cardiaques/cytologie , Techniques de patch-clamp , RT-PCR , Transgènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...