Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 263
Filter
1.
PLoS One ; 19(5): e0298274, 2024.
Article in English | MEDLINE | ID: mdl-38753762

ABSTRACT

The membrane peroxisomal proteins PEX11, play a crucial role in peroxisome proliferation by regulating elongation, membrane constriction, and fission of pre-existing peroxisomes. In this study, we evaluated the function of PEX11B gene in neural differentiation of human embryonic stem cell (hESC) by inducing shRNAi-mediated knockdown of PEX11B expression. Our results demonstrate that loss of PEX11B expression led to a significant decrease in the expression of peroxisomal-related genes including ACOX1, PMP70, PEX1, and PEX7, as well as neural tube-like structures and neuronal markers. Inhibition of SIRT1 using pharmacological agents counteracted the effects of PEX11B knockdown, resulting in a relative increase in PEX11B expression and an increase in differentiated neural tube-like structures. However, the neuroprotective effects of SIRT1 were eliminated by PPAR inhibition, indicating that PPARÉ£ may mediate the interaction between PEX11B and SIRT1. Our findings suggest that both SIRT1 and PPARÉ£ have neuroprotective effects, and also this study provides the first indication for a potential interaction between PEX11B, SIRT1, and PPARÉ£ during hESC neural differentiation.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells , Membrane Proteins , PPAR gamma , Sirtuin 1 , Humans , Sirtuin 1/metabolism , Sirtuin 1/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Cell Differentiation/drug effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Neurons/metabolism , Neurons/cytology , Neurons/drug effects , Cell Line , Peroxisomes/metabolism
2.
Environ Sci Technol ; 58(19): 8215-8227, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38687897

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) are extensively utilized in varieties of products and tend to accumulate in the human body including umbilical cord blood and embryos/fetuses. In this study, we conducted an assessment and comparison of the potential early developmental toxicity of perfluorooctanoic acid (PFOA), undecafluorohexanoic acid (PFHxA), heptafluorobutyric acid, perfluorooctanesulfonate (PFOS), perfluorohexanesulfonate, and perfluorobutyric acid at noncytotoxic concentrations relevant to human exposure using models based on human embryonic stem cells in both three-dimensional embryoid body (EB) and monolayer differentiation configurations. All six compounds influenced the determination of cell fate by disrupting the expression of associated markers in both models and, in some instances, even led to alterations in the formation of cystic EBs. The expression of cilia-related gene IFT122 was significantly inhibited. Additionally, PFOS and PFOA inhibited ciliogenesis, while PFOA specifically reduced the cilia length. Transcriptome analysis revealed that PFOS altered 1054 genes and disrupted crucial signaling pathways such as WNT and TGF-ß, which play integral roles in cilia transduction and are critical for early embryonic development. These results provide precise and comprehensive insights into the potential adverse health effects of these six PFAS compounds directly concerning early human embryonic development.


Subject(s)
Fluorocarbons , Human Embryonic Stem Cells , Humans , Human Embryonic Stem Cells/drug effects , Fluorocarbons/toxicity , Cell Differentiation/drug effects
3.
Reprod Sci ; 31(5): 1363-1372, 2024 May.
Article in English | MEDLINE | ID: mdl-38151652

ABSTRACT

Knowledge of action of progesterone (P4) on the human preimplantation embryo is lacking. The objective of this study was to determine expression of a mitochondrial P4 receptor (PR-M) in the trophectoderm (TE) and the inner cell mass (ICM) of the human blastocyst and to determine P4-induced gene expression during growth from the cleavage to the blastocyst stage. Previously cryopreserved cleavage stage embryos were treated with P4 (10-6 M) or vehicle until blastocyst development. Cells from the TE and the ICM of dissected euploid embryos underwent RNA-seq analysis, while other embryos were used for analysis of nuclear PR (nPR) and PR-M expression.PR-M expression was confirmed in the TE, the ICM, and a human embryonic stem cell line (HESC). Conversely, nPR expression was absent in the TE and the ICM with low expression in the HESC line. RNA-seq analysis revealed P4 effects greater in the TE with 183 significant pathway changes compared to 27 in the ICM. The TE response included significant upregulation of genes associated with DNA replication, cell cycle phase transition and others, exemplified by a 7.6-fold increase in the cell proliferation gene, F-Box Associated Domain Containing. The majority of ICM pathways were downregulated including chromosome separation, centromere complex assembly and chromatin remodeling at centromere. This study confirms that human blastocysts express PR-M in both the TE and the ICM, but lack expression of nPR. P4-induced gene regulation differs greatly in the two cell fractions with the predominant effect of cell proliferation in the TE and not the ICM.


Subject(s)
Blastocyst Inner Cell Mass , Blastocyst , Gene Expression Regulation, Developmental , Progesterone , Humans , Progesterone/pharmacology , Blastocyst/metabolism , Blastocyst/drug effects , Blastocyst Inner Cell Mass/metabolism , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Female , Embryonic Development/drug effects , Embryonic Development/physiology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/drug effects
4.
Stem Cell Rev Rep ; 19(8): 2980-2990, 2023 11.
Article in English | MEDLINE | ID: mdl-37702917

ABSTRACT

Embryonic development is a continuum in vivo. Transcriptional analysis can separate established human embryonic stem cells (hESC) into at least four distinct developmental pluripotent stages, two naïve and two primed, early and late relative to the intact epiblast. In this study we primarily show that exposure of frozen human blastocysts to an inhibitor of checkpoint kinase 1 (CHK1) upon thaw greatly enhances establishment of karyotypically normal late naïve hESC cultures. These late naïve cells are plastic and can be toggled back to early naïve and forward to early primed pluripotent stages. The early primed cells are transcriptionally equivalent to the post inner cell mass intermediate (PICMI) stage seen one day following transfer of human blastocysts into in vitro culture and are stable at an earlier stage than conventional primed hESC.


Subject(s)
Cell Culture Techniques , Checkpoint Kinase 1 , Human Embryonic Stem Cells , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Checkpoint Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Blastocyst/cytology , Pluripotent Stem Cells/cytology
5.
Cell Biochem Funct ; 40(4): 359-368, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35445405

ABSTRACT

Porcupine (Porcn) enzyme plays an essential role in Wnt signaling activation. Stearoyl-CoA desaturase-1 (SCD1) is required to provide Porcn substrates. The aim of this study was to determine the effect of a novel Porcn inhibitor on the fate of human embryonic stem cells (hESCs) and the reliance of Porcn on SCD1 activity. hESCs were cultured on a feeder layer or Matrigel-coated plates. Small molecules WNT974 (LGK-974) and CAY10566 were used to inhibit Porcn and SCD1 activity, respectively. We assessed the effect of Porcn inhibition on viability, expression of Wnt signaling targets, pluripotency markers, proliferation, differentiation, and protein fatty acylation. hESCs' conditioned medium (CM) containing secreted Wnt proteins were applied in rescue experiments. To examine the catalytic dependency of Porcn on SCD1, the results of combined inhibitor treatment were compared with the SCD1 inhibitor alone. LGK-974 at the selected concentrations showed mild effects on hESCs viability, but significantly reduced messenger RNA and protein expression of Wnt signaling targets (Axin-2 and c-Myc) and pluripotency markers (OCT-4 and SOX-2) (p < .05). Adding 1 µM of Porcn inhibitor reduced proliferation (p = .03) and enhanced differentiation capacity into ectodermal progenitors (p = .02), which were reverted by CM. Click chemistry reaction did not show significant alteration in protein fatty acylation upon LGK-974 treatment. Moreover, combined inhibitor treatment caused no further substantial reduction in Wnt signaling targets, pluripotency markers, and protein fatty acylation relative to CAY10566-treated cultures. The substrate availability for Porcn activity is regulated by SCD1 and targeting Porcn by LGK-974 prompts the transition of hESCs from self-renewal state to ectodermal lineage.


Subject(s)
Human Embryonic Stem Cells , Wnt Signaling Pathway , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Pyrazines/pharmacology , Pyridines/pharmacology , Stearoyl-CoA Desaturase
6.
Sci Rep ; 12(1): 2516, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169157

ABSTRACT

Clinical use of human pluripotent stem cells (hPSCs) is hampered by the technical limitations of their expansion. Here, we developed a chemically synthetic culture substrate for human pluripotent stem cell attachment and maintenance. The substrate comprises a hydrophobic polyvinyl butyral-based polymer (PVB) and a short peptide that enables easy and uniform coating of various types of cell culture ware. The coated ware exhibited thermotolerance, underwater stability and could be stored at room temperature. The substrate supported hPSC expansion in combination with most commercial culture media with an efficiency similar to that of commercial substrates. It supported not only the long-term expansion of examined iPS and ES cell lines with normal karyotypes during their undifferentiated state but also directed differentiation of three germ layers. This substrate resolves major concerns associated with currently used recombinant protein substrates and could be applied in large-scale automated manufacturing; it is suitable for affordable and stable production of clinical-grade hPSCs and hPSC-derived products.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Human Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Peptides/pharmacology , Polyvinyls/pharmacology , Tissue Scaffolds/chemistry , Cell Adhesion/drug effects , Cell Line , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Peptides/metabolism , Polyvinyls/metabolism
7.
Toxicology ; 465: 153065, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34896440

ABSTRACT

Trichloroethylene (TCE) is a major environmental contaminant. Maternal exposure of TCE is linked to developmental defects, but the mechanisms remain to be elucidated. Along with a strategy of 3Rs principle, human embryonic stem cells (hESCs) are regarded as most promising in vitro models for developmental toxicity studies. TCE interfered with hESCs differentiation, but no report was available for TCE effects on hESCs proliferation. Here, we aimed to explore the toxic effects and mechanisms of TCE on hESCs proliferation. Treatment with TCE, did not affect the pluripotency genes expression. However, TCE enhanced hESCs proliferation, manifested by increased cell number, PCNA expression and EdU incorporation. Moreover, TCE exposure upregulated the protein expression levels of Cx43 and cyclin-dependent kinases. Knockdown of Cx43 attenuated the TCE-induced cell hyper-proliferation and CDK2 upregulation. Furthermore, TCE increased Akt phosphorylation, and the inhibition of Akt blocked the TCE-induced Cx43 overexpression and cell proliferation. In conclusion, TCE exposure resulted in upregulation of Cx43 via Akt phosphorylation, consequently stimulated CDK2 expression, contributing to hyper-proliferation in hESCs. Our study brings to light that TCE stimulated the proliferation of hESCs via Cx43, providing a new research avenue for the causes of TCE-induced developmental toxicity.


Subject(s)
Cell Proliferation/drug effects , Connexin 43/metabolism , Environmental Pollutants/toxicity , Human Embryonic Stem Cells/drug effects , Trichloroethylene/toxicity , Cell Line , Connexin 43/genetics , Cyclin-Dependent Kinase 2/metabolism , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Up-Regulation
8.
Biochem Biophys Res Commun ; 586: 163-170, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34852960

ABSTRACT

SOX2, a well-established pluripotency factor supporting the self-renewal of pluripotent stem cells (PSCs), is also a crucial factor for maintaining the properties and functionalities of neural progenitor cells (NPCs). It regulates the transcription of target genes by forming complexes with its partner factors, but systematic comparison of SOX2 binding partners in human PSCs versus NPCs is lacking. Here, by deciphering and comparing the SOX2-protein interactomes in human embryonic stem cells (hESCs) versus the NPCs derived from them, we identified 23 proteins with high reproducibility that are most differentially associated with SOX2, of which 9 are DNA repair proteins (PARP1, PARP2, PRKDC, XRCC1, XRCC5, XRCC6, RPA1, LIG3, DDB1). Genetic knocking-down or pharmacological inhibiting two of the DNA repair proteins (PARP1 and PRKDC) significantly up-regulated certain NPC or ectodermal biomarkers that are transcriptionally-suppressed by the SOX2/DNA repair protein complexes. These findings point to a crucial role of DNA repair proteins in pluripotent state transition and neural induction.


Subject(s)
DNA Repair , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , Human Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , SOXB1 Transcription Factors/genetics , Animals , Cell Differentiation/drug effects , Cell Line , Computational Biology/methods , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Phthalazines/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Pyrans/pharmacology , SOXB1 Transcription Factors/metabolism , Signal Transduction , Triazoles/pharmacology
9.
EBioMedicine ; 74: 103713, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34837851

ABSTRACT

BACKGROUND: Exercise can protect myocardial infarction (MI) and downregulate cardiac Homeodomain-Interacting Protein Kinase 2 (HIPK2). However, the role of HIPK2 in MI is unclear. METHODS: HIPK2-/- mice and miR-222-/- rats, HIPK2 inhibitor (PKI1H) and adeno-associated virus serotype 9 (AAV9) carrying miR-222 were applied in the study. Animals were subjected to running, swimming, acute MI or post-MI remodeling. HIPK2 inhibition and P53 activator were used in neonatal rat cardiomyocytes (NRCMs) and human embryonic stem cell-derived cardiomyocytes (hESC-CMs) subjected to oxygen glucose deprivation/reperfusion (OGD/R). Serum miR-222 levels were analyzed in healthy people and MI patients that were survival or readmitted to the hospital and/or died. FINDINGS: Cardiac HIPK2 protein levels were reduced by exercise while increased in MI. In vitro, HIPK2 suppression by lentiviral vectors or inhibitor prevented apoptosis induced by OGD/R in NRCMs and hESC-CMs. HIPK2 inhibitor-treated mice and HIPK2-/- mice reduced infarct size after acute MI, and preserved cardiac function in MI remodeling. Mechanistically, protective effect against apoptosis by HIPK2 suppression was reversed by P53 activators. Furthermore, increasing levels of miR-222, targeting HIPK2, protected post-MI cardiac dysfunction, whereas cardiac dysfunction post-MI was aggravated in miR-222-/- rats. Moreover, serum miR-222 levels were significantly reduced in MI patients, as well as in MI patients that were readmitted to the hospital and/or died compared to those not. INTERPRETATION: Exercise-induced HIPK2 suppression attenuates cardiomyocytes apoptosis and protects MI by decreasing P-P53. Inhibition of HIPK2 represents a potential novel therapeutic intervention for MI. FUNDING: This work was supported by the grants from National Key Research and Development Project (2018YFE0113500 to JJ Xiao), National Natural Science Foundation of China (82020108002, 81722008, and 81911540486 to JJ Xiao, 81400647 to MJ Xu, 81800265 to YJ Liang), Innovation Program of Shanghai Municipal Education Commission (2017-01-07-00-09-E00042 to JJ Xiao), the grant from Science and Technology Commission of Shanghai Municipality (18410722200 and 17010500100 to JJ Xiao), the "Dawn" Program of Shanghai Education Commission (19SG34 to JJ Xiao), Shanghai Sailing Program (21YF1413200 to QL Zhou). JS is supported by Horizon2020 ERC-2016-COG EVICARE (725229).


Subject(s)
Carrier Proteins/genetics , Down-Regulation , Exercise/physiology , MicroRNAs/blood , MicroRNAs/genetics , Myocardial Infarction/genetics , Protein Serine-Threonine Kinases/genetics , Adult , Animals , Animals, Newborn , Carrier Proteins/metabolism , Case-Control Studies , Cells, Cultured , Dependovirus/genetics , Disease Models, Animal , Gene Knockout Techniques , Human Embryonic Stem Cells/chemistry , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Mice , Middle Aged , Myocardial Infarction/chemically induced , Myocardial Infarction/therapy , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats , Running/physiology , Swimming/physiology
10.
Cells ; 10(11)2021 10 26.
Article in English | MEDLINE | ID: mdl-34831120

ABSTRACT

Medullary thyroid carcinoma contributes to about 3-4% of thyroid cancers and affects C cells rather than follicular cells. Thyroid C cell differentiation from human pluripotent stem cells has not been reported. We report the stepwise differentiation of human embryonic stem cells into thyroid C cell-like cells through definitive endoderm and anterior foregut endoderm and ultimobranchial body-like intermediates in monolayer and 3D Matrigel culture conditions. The protocol involved sequential treatment with interferon/transferrin/selenium/pyruvate, foetal bovine serum, and activin A, then IGF-1 (Insulin-like growth factor 1), on the basis of embryonic thyroid developmental sequence. As well as expressing C cell lineage relative to follicular-lineage markers by qPCR (quantitative polymerase chain reaction) and immunolabelling, these cells by ELISA (enzyme-linked immunoassay) exhibited functional properties in vitro of calcitonin storage and release of calcitonin on calcium challenge. This method will contribute to developmental studies of the human thyroid gland and facilitate in vitro modelling of medullary thyroid carcinoma and provide a valuable platform for drug screening.


Subject(s)
Pluripotent Stem Cells/cytology , Thyroid Gland/cytology , Tissue Scaffolds/chemistry , Biomarkers/metabolism , Calcitonin/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Collagen/pharmacology , Drug Combinations , Endoderm/cytology , Gastrointestinal Tract/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Laminin/pharmacology , Neurosecretory Systems/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proteoglycans/pharmacology
11.
Stem Cell Reports ; 16(12): 2928-2941, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34767749

ABSTRACT

The immature characteristics and metabolic phenotypes of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) restrict their applications for disease modeling, drug discovery, and cell-based therapy. Leveraging on the metabolic shifts from glycolysis to fatty acid oxidation as CMs mature, a human hexokinase1-GFP metabolic reporter cell line (H7 HK1-GFP) was generated to facilitate the isolation of fetal or more matured hPSC-CMs. RNA sequencing of fetal versus more matured CMs uncovered a potential role of interferon-signaling pathway in regulating CM maturation. Indeed, IFN-γ-treated CMs resulted in an upregulation of the JAK-STAT pathway, which was found to be associated with increased expression of CM maturation genes, shift from MYH6 to MYH7 expression, and improved sarcomeric structure. Functionally, IFN-γ-treated CMs exhibited a more matured electrophysiological profile, such as increased calcium dynamics and action potential upstroke velocity, demonstrated through calcium imaging and MEA. Expectedly, the functional improvements were nullified with a JAK-STAT inhibitor, ruxolitinib.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Janus Kinases/metabolism , Myocytes, Cardiac/cytology , STAT Transcription Factors/metabolism , Signal Transduction , Up-Regulation , CRISPR-Cas Systems/genetics , Cell Differentiation/drug effects , Cell Line , Electrophysiological Phenomena/drug effects , Genes, Reporter , Green Fluorescent Proteins/metabolism , Human Embryonic Stem Cells/drug effects , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects
12.
Cells ; 10(10)2021 09 30.
Article in English | MEDLINE | ID: mdl-34685588

ABSTRACT

The heart tissue is a potential target of various noxae contributing to the onset of cardiovascular diseases. However, underlying pathophysiological mechanisms are largely unknown. Human stem cell-derived models are promising, but a major concern is cell immaturity when estimating risks for adults. In this study, 3D aggregates of human embryonic stem cell-derived cardiomyocytes were cultivated for 300 days and characterized regarding degree of maturity, structure, and cell composition. Furthermore, effects of ionizing radiation (X-rays, 0.1-2 Gy) on matured aggregates were investigated, representing one of the noxae that are challenging to assess. Video-based functional analyses were correlated to changes in the proteome after irradiation. Cardiomyocytes reached maximum maturity after 100 days in cultivation, judged by α-actinin lengths, and displayed typical multinucleation and branching. At this time, aggregates contained all major cardiac cell types, proven by the patch-clamp technique. Matured and X-ray-irradiated aggregates revealed a subtle increase in beat rates and a more arrhythmic sequence of cellular depolarisation and repolarisation compared to non-irradiated sham controls. The proteome analysis provides first insights into signaling mechanisms contributing to cardiotoxicity. Here, we propose an in vitro model suitable to screen various noxae to target adult cardiotoxicity by preserving all the benefits of a 3D tissue culture.


Subject(s)
Cell Differentiation/drug effects , Human Embryonic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Noxae/pharmacology , X-Rays , Adult , Cardiotoxicity/drug therapy , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Noxae/metabolism
13.
Cells ; 10(10)2021 10 14.
Article in English | MEDLINE | ID: mdl-34685725

ABSTRACT

Mature cardiomyocytes (CMs) obtained from human pluripotent stem cells (hPSCs) have been required for more accurate in vitro modeling of adult-onset cardiac disease and drug discovery. Here, we found that FGF4 and ascorbic acid (AA) induce differentiation of BG01 human embryonic stem cell-cardiogenic mesoderm cells (hESC-CMCs) into mature and ventricular CMs. Co-treatment of BG01 hESC-CMCs with FGF4+AA synergistically induced differentiation into mature and ventricular CMs. FGF4+AA-treated BG01 hESC-CMs robustly released acute myocardial infarction (AMI) biomarkers (cTnI, CK-MB, and myoglobin) into culture medium in response to hypoxic injury. Hypoxia-responsive genes and potential cardiac biomarkers proved in the diagnosis and prognosis of coronary artery diseases were induced in FGF4+AA-treated BG01 hESC-CMs in response to hypoxia based on transcriptome analyses. This study demonstrates that it is feasible to model hypoxic stress in vitro using hESC-CMs matured by soluble factors.


Subject(s)
Ascorbic Acid/pharmacology , Cell Differentiation , Fibroblast Growth Factor 4/pharmacology , Human Embryonic Stem Cells/pathology , Models, Biological , Myocytes, Cardiac/pathology , Stress, Physiological , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Line , Culture Media/pharmacology , Gene Expression Regulation/drug effects , Heart Ventricles/pathology , Human Embryonic Stem Cells/drug effects , Humans , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Stress, Physiological/drug effects , Transcriptome/genetics
14.
Toxicology ; 461: 152898, 2021 09.
Article in English | MEDLINE | ID: mdl-34403730

ABSTRACT

Silver nanoparticles (AgNPs) are widely used in medical and commercial products for their unique antibacterial functions. However, the impact of AgNPs on human neural development is not well understood. To investigate the effect of AgNPs on human neural development, various doses of 20 nm citrate-coated AgNP (AgSC) were administered to human embryonic stem cell derived neural progenitors during the neuronal differentiation. Immunofluorescence staining with neuronal progenitor markers SOX2 (sex determining region Y-box 2) and Nestin (VI intermediate filament protein) showed that AgSC inhibited rosette formation, neuronal progenitor proliferation, and neurite outgrowth. Furthermore, AgSC promoted astrocyte activation and neuronal apoptosis. These adverse effects can be partially recovered with ascorbic acid. A genome-wide transcriptome analysis of both AgSC treated and untreated samples indicated that the most up-graduated genes were a group of Metallothionein (1F, 1E, 2A) proteins, a metal-binding protein that plays an essential role in metal homeostasis, heavy metal detoxification, and cellular anti-oxidative defence. The most significantly down-regulated genes were neuronal differentiation 6 (NEUROD6) and fork head box G1 (FOXG1). GO analyse indicated that the regulation of cholesterol biosynthetic process, neuron differentiation, synapse organization and pattern specification, oliogenesis, and neuronal apoptosis were the most impacted biological processes. KEGG pathway analyse showed that the most significantly impacted pathways were C5 isoprenoid, axon guidance, Notch, WNT, RAS-MAPK signalling pathways, lysosome, and apoptosis. Our data suggests that AgSCs interfered with metal homeostasis and cholesterol biosynthesis which induced oxidative stress, inhibited neurogenesis, axon guidance, and promoted apoptosis. Supplementation with ascorbic acid could act as an antioxidant to prevent AgSC-mediated neurotoxicity.


Subject(s)
Citric Acid/chemistry , Human Embryonic Stem Cells/drug effects , Metal Nanoparticles/toxicity , Silver , Apoptosis/drug effects , Cell Differentiation/genetics , Cell Line , Cholesterol/biosynthesis , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Human Embryonic Stem Cells/cytology , Humans , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/genetics , Oxidative Stress/drug effects , Signal Transduction/drug effects
15.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209900

ABSTRACT

Adult human cardiomyocytes have an extremely limited proliferative capacity, which poses a great barrier to regenerative medicine and research. Human embryonic stem cells (hESCs) have been proposed as an alternative source to generate large numbers of clinical grade cardiomyocytes (CMs) that can have potential therapeutic applications to treat cardiac diseases. Previous studies have shown that bioactive lipids are involved in diverse cellular responses including cardiogenesis. In this study, we explored the novel function of the chemically synthesized bioactive lipid O-cyclic phytosphingosine-1-phosphate (cP1P) as an inducer of cardiac differentiation. Here, we identified cP1P as a novel factor that significantly enhances the differentiation potential of hESCs into cardiomyocytes. Treatment with cP1P augments the beating colony number and contracting area of CMs. Furthermore, we elucidated the molecular mechanism of cP1P regulating SMAD1/5/8 signaling via the ALK3/BMP receptor cascade during cardiac differentiation. Our result provides a new insight for cP1P usage to improve the quality of CM differentiation for regenerative therapies.


Subject(s)
Cell Differentiation/drug effects , Human Embryonic Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Sphingosine/analogs & derivatives , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Regulation/drug effects , Human Embryonic Stem Cells/physiology , Humans , Lipids/chemistry , Lipids/pharmacology , Myocytes, Cardiac/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/chemistry , Sphingosine/pharmacology
16.
Stem Cell Reports ; 16(7): 1686-1696, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34214484

ABSTRACT

Human extended pluripotent stem cells (EPSCs), with bidirectional chimeric ability to contribute to both embryonic and extraembryonic lineages, can be obtained and maintained by converting conventional pluripotent stem cells using chemicals. However, the transition system is based on inactivated mouse fibroblasts, and the underlying mechanism is not clear. Here we report a Matrigel-based feeder-free method to convert human embryonic stem cells and induced pluripotent stem cells into EPSCs and demonstrate the extended pluripotency in terms of molecular features, chimeric ability, and transcriptome. We further identify chemicals targeting glycolysis and histone methyltransferase to facilitate the conversion to and maintenance of feeder-free EPSCs. Altogether, our data not only establish a feeder-free system to generate human EPSCs, which should facilitate the mechanistic studies of extended pluripotency and further applications, but also provide additional insights into the transitions among different pluripotent states.


Subject(s)
Feeder Cells/cytology , Pluripotent Stem Cells/cytology , Cell Line , Chimera/physiology , Feeder Cells/drug effects , Glycolysis/drug effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Indoles/pharmacology , Pluripotent Stem Cells/drug effects , Pyridones/pharmacology
17.
STAR Protoc ; 2(3): 100659, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34286291

ABSTRACT

Training and experiences are usually required to successfully culture and differentiate human embryonic stem cells (hESCs). Here, we describe a simple but highly efficient protocol to induce endoderm differentiation of hESCs with crotonate, a precursor of crotonyl-CoA for histone crotonylation deposition on endodermal genes. In this protocol, adding crotonate in different endoderm differentiation media significantly increases the differentiation efficiency and substantially reduces the amount of required reagents. For complete details on the use and execution of this protocol, please refer to Fang et al. (2021).


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/drug effects , Crotonates/pharmacology , Endoderm/cytology , Human Embryonic Stem Cells/drug effects , Cells, Cultured , Humans
18.
Endocrinology ; 162(9)2021 09 01.
Article in English | MEDLINE | ID: mdl-34125902

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus play a key role in the regulation of reproductive function. In this study, we sought an efficient method for generating GnRH neurons from human embryonic and induced pluripotent stem cells (hESC and hiPSC, respectively). First, we found that exposure of primitive neuroepithelial cells, rather than neuroprogenitor cells, to fibroblast growth factor 8 (FGF8), was more effective in generating GnRH neurons. Second, addition of kisspeptin to FGF8 further increased the efficiency rates of GnRH neurogeneration. Third, we generated a fluorescent marker mCherry labeled human embryonic GnRH cell line (mCh-hESC) using a CRISPR-Cas9 targeting approach. Fourth, we examined physiological characteristics of GnRH (mCh-hESC) neurons: similar to GnRH neurons in vivo, they released the GnRH peptide in a pulsatile manner at ~60 min intervals; GnRH release increased in response to high potassium, kisspeptin, estradiol, and neurokinin B challenges; and injection of depolarizing current induced action potentials. Finally, we characterized developmental changes in transcriptomes of GnRH neurons using hESC, hiPSC, and mCh-hESC. The developmental pattern of transcriptomes was remarkably similar among the 3 cell lines. Collectively, human stem cell-derived GnRH neurons will be an important tool for establishing disease models to understand diseases, such as idiopathic hypothalamic hypogonadism, and testing contraceptive drugs.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Human Embryonic Stem Cells/physiology , Neurogenesis/genetics , Neurons/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Fibroblast Growth Factor 8/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Human Embryonic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Transcriptome/drug effects
19.
Biomed Res Int ; 2021: 6695663, 2021.
Article in English | MEDLINE | ID: mdl-33937411

ABSTRACT

BACKGROUND: When vascular endothelial cells are subjected to external stimuli, paracrine hormones and cytokines act on adjacent cells. The regulation of the biological behaviour of cells is closely related to the maintenance of organ function and the occurrence and development of disease. However, it is unclear whether vascular endothelial cells affect the biological behaviour of cells involved in wound repair through autocrine and paracrine mechanisms and ultimately play a role in wound healing. We aimed to verify the effect of the autocrine and paracrine functions of vascular endothelial cells on wound healing. MATERIALS AND METHODS: ELISA was used to detect platelet-derived growth factor, basic fibroblast growth factor, epidermal growth factor, and vascular endothelial growth factor in human umbilical vascular endothelial cell-conditioned medium (HUVEC-CM). Different concentrations of HUVEC-CM were used to treat different stem cells. CCK-8 and scratch assays were used to detect the proliferation and migration ability of each cell. A full-thickness dorsal skin defect model was established in mice, and skin wound healing was observed after the local injection of HUVEC-CM, endothelial cell medium (ECM), or normal saline. H&E staining and immunofluorescence were used to observe the gross morphology of the wound tissue, the epithelial cell migration distance, and the expression of CD3 and CD31. RESULTS: HUVEC-CM promotes the proliferation and migration of epidermal stem cells, skin fibroblasts, bone marrow mesenchymal stem cells, and HUVECs themselves. Furthermore, HUVEC-CM can promote angiogenesis in mouse skin wounds and granulation tissue formation and can accelerate wound surface epithelialization and collagen synthesis, thereby promoting wound healing. CONCLUSION: Our results clearly suggest that it is practicable and effective to promote wound healing with cytokines secreted by vascular endothelial cells in a mouse model.


Subject(s)
Autocrine Communication , Human Umbilical Vein Endothelial Cells/metabolism , Paracrine Communication , Skin/pathology , Wound Healing , Antigens, CD/metabolism , Autocrine Communication/drug effects , Biomarkers/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Paracrine Communication/drug effects , Wound Healing/drug effects
20.
Diabetes ; 70(8): 1689-1702, 2021 08.
Article in English | MEDLINE | ID: mdl-33958328

ABSTRACT

Metformin is becoming a popular treatment before and during pregnancy, but current literature on in utero exposure to metformin lacks long-term clinical trials and mechanistic studies. Current literature on the effects of metformin on mature pancreatic ß-cells highlights its dual, opposing, protective, or inhibitory effects, depending on metabolic environment. However, the impact of metformin on developing human pancreatic ß-cells remains unknown. In this study, we investigated the potential effects of metformin exposure on human pancreatic ß-cell development and function in vitro. In the absence of metabolic challenges such as high levels of glucose and fatty acids, metformin exposure impaired the development and function of pancreatic ß-cells, with downregulation of pancreatic genes and dysfunctional mitochondrial respiration. It also affected the insulin secretion function of pancreatic ß-cells. These findings call for further in-depth evaluation of the exposure of human embryonic and fetal tissue during pregnancy to metformin and its implications for long-term offspring health.


Subject(s)
Cell Differentiation/drug effects , Human Embryonic Stem Cells/drug effects , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Metformin/pharmacology , Pancreas/drug effects , Cell Survival/drug effects , Human Embryonic Stem Cells/cytology , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pancreas/cytology , Pancreas/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...