Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Commun Biol ; 6(1): 452, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37095219

ABSTRACT

Familial hypercholesterolemia (FH) patients suffer from excessively high levels of Low Density Lipoprotein Cholesterol (LDL-C), which can cause severe cardiovascular disease. Statins, bile acid sequestrants, PCSK9 inhibitors, and cholesterol absorption inhibitors are all inefficient at treating FH patients with homozygous LDLR gene mutations (hoFH). Drugs approved for hoFH treatment control lipoprotein production by regulating steady-state Apolipoprotein B (apoB) levels. Unfortunately, these drugs have side effects including accumulation of liver triglycerides, hepatic steatosis, and elevated liver enzyme levels. To identify safer compounds, we used an iPSC-derived hepatocyte platform to screen a structurally representative set of 10,000 small molecules from a proprietary library of 130,000 compounds. The screen revealed molecules that could reduce the secretion of apoB from cultured hepatocytes and from humanized livers in mice. These small molecules are highly effective, do not cause abnormal lipid accumulation, and share a chemical structure that is distinct from any known cholesterol lowering drug.


Subject(s)
Anticholesteremic Agents , Homozygous Familial Hypercholesterolemia , Hyperlipoproteinemia Type II , Induced Pluripotent Stem Cells , Humans , Animals , Mice , Proprotein Convertase 9/genetics , Proprotein Convertase 9/pharmacology , Proprotein Convertase 9/therapeutic use , Cholesterol, LDL , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/genetics , Anticholesteremic Agents/pharmacology , Apolipoproteins B/genetics , Apolipoproteins B/pharmacology , Apolipoproteins B/therapeutic use , Hepatocytes
2.
Cell Rep ; 35(7): 109145, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34010638

ABSTRACT

In addition to driving specific gene expression profiles, transcriptional regulators are becoming increasingly recognized for their capacity to modulate chromatin structure. GATA6 is essential for the formation of definitive endoderm; however, the molecular basis defining the importance of GATA6 to endoderm commitment is poorly understood. The members of the GATA family of transcription factors have the capacity to bind and alter the accessibility of chromatin. Using pluripotent stem cells as a model of human development, we reveal that GATA6 is integral to the establishment of the endoderm enhancer network via the induction of chromatin accessibility and histone modifications. We additionally identify the chromatin-modifying complexes that interact with GATA6, defining the putative mechanisms by which GATA6 modulates chromatin architecture. The identified GATA6-dependent processes further our knowledge of the molecular mechanisms that underpin cell-fate decisions during formative development.


Subject(s)
Chromatin/metabolism , GATA6 Transcription Factor/metabolism , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Humans
3.
PeerJ ; 8: e9060, 2020.
Article in English | MEDLINE | ID: mdl-32391204

ABSTRACT

Genome editing in human induced pluripotent stem cells (iPSCs) provides the potential for disease modeling and cell therapy. By generating iPSCs with specific mutations, researchers can differentiate the modified cells to their lineage of interest for further investigation. However, the low efficiency of targeting in iPSCs has hampered the application of genome editing. In this study we used a CRISPR-Cas9 system that introduces a specific point substitution into the sequence of the Na+/K+-ATPase subunit ATP1A1. The introduced mutation confers resistance to cardiac glycosides, which can then be used to select successfully targeted cells. Using this system, we introduced different formats of donor DNA for homology-directed repair (HDR), including single-strand DNAs, double-strand DNAs, and plasmid donors. We achieved a 35-fold increase in HDR when using plasmid donor with a 400 bp repair template. We further co-targeted ATP1A1 and a second locus of interest to determine the enrichment of mutagenesis after cardiac glycoside selection. Through this approach, INDEL rate was increased after cardiac glycoside treatment, while HDR enrichment was only observed at certain loci. Collectively, these results suggest that a plasmid donor with a 400 bp repair template is an optimal donor DNA for targeted substitution and co-targeting ATP1A1 with the second locus enriches for mutagenesis events through cardiac glycoside selection in human iPSCs.

4.
J Vis Exp ; (135)2018 05 19.
Article in English | MEDLINE | ID: mdl-29863663

ABSTRACT

The ability to differentiate human induced pluripotent stem cells (iPSCs) into hepatocyte-like cells (HLCs) provides new opportunities to study inborn errors in hepatic metabolism. However, to provide a platform that supports the identification of small molecules that can potentially be used to treat liver disease, the procedure requires a culture format that is compatible with screening thousands of compounds. Here, we describe a protocol using completely defined culture conditions, which allow the reproducible differentiation of human iPSCs to hepatocyte-like cells in 96-well tissue culture plates. We also provide an example of using the platform to screen compounds for their ability to lower Apolipoprotein B (APOB) produced from iPSC-derived hepatocytes generated from a familial hypercholesterolemia patient. The availability of a platform that is compatible with drug discovery should allow researchers to identify novel therapeutics for diseases that affect the liver.


Subject(s)
Drug Discovery/methods , Animals , Disease Models, Animal , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice
5.
Toxicol Sci ; 162(1): 64-78, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29121352

ABSTRACT

Chronic arsenic exposure can result in adverse development effects including decreased intellectual function, reduced birth weight, and altered locomotor activity. Previous in vitro studies have shown that arsenic inhibits stem cell differentiation. MicroRNAs (miRNAs) are small noncoding RNAs that regulate multiple cellular processes including embryonic development and cell differentiation. The purpose of this study was to examine whether altered miRNA expression was a mechanism by which arsenic inhibited cellular differentiation. The pluripotent P19 mouse embryonal carcinoma cells were exposed to 0 or 0.5 µM sodium arsenite for 9 days during cell differentiation, and changes in miRNA expression was analyzed using microarrays. We found that the expression of several miRNAs important in cellular differentiation, such as miR-9 and miR-199 were decreased by 1.9- and 1.6-fold, respectively, following arsenic exposure, while miR-92a, miR-291a, and miR-709 were increased by 3-, 3.7-, and 1.6-fold, respectively. The members of the miR-466-669 cluster and its host gene, Scm-like with 4 Mbt domains 2 (Sfmbt2), were significantly induced by arsenic from 1.5- to 4-fold in a time-dependent manner. Multiple miRNA target prediction programs revealed that several neurogenic transcription factors appear to be targets of the cluster. When consensus anti-miRNAs targeting the miR-466-669 cluster were transfected into P19 cells, arsenic-exposed cells were able to more effectively differentiate. The consensus anti-miRNAs appeared to rescue the inhibitory effects of arsenic on cell differentiation due to an increased expression of NeuroD1. Taken together, we conclude that arsenic induces the miR-466-669 cluster, and that this induction acts to inhibit cellular differentiation in part due to a repression of NeuroD1.


Subject(s)
Arsenic/toxicity , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/drug effects , Environmental Pollutants/toxicity , Gene Expression/drug effects , MicroRNAs/genetics , Animals , Cell Differentiation/genetics , Embryonal Carcinoma Stem Cells , Mice , Multigene Family
6.
Toxicol Rep ; 3: 405-413, 2016.
Article in English | MEDLINE | ID: mdl-27158593

ABSTRACT

Millions of people are exposed to arsenic through their drinking water and food, but the mechanisms by which it impacts embryonic development are not well understood. Arsenic exposure during embryogenesis is associated with neurodevelopmental effects, reduced weight gain, and altered locomotor activity, and in vitro data indicates that arsenic exposure inhibits stem cell differentiation. This study investigated whether arsenic disrupted the Wnt3a signaling pathway, critical in the formation of myotubes and neurons, during the differentiation in P19 mouse embryonic stem cells. Cells were exposed to 0, 0.1, or 0.5 µM arsenite, with or without exogenous Wnt3a, for up to 9 days of differentiation. Arsenic exposure alone inhibits the differentiation of stem cells into neurons and skeletal myotubes, and reduces the expression of both ß-catenin and GSK3ß mRNA to ~55% of control levels. Co-culture of the arsenic-exposed cells with exogenous Wnt3a rescues the morphological phenotype, but does not alter transcript, protein, or phosphorylation status of GSK3ß or ß-catenin. However, arsenic exposure maintains high levels of Hes5 and decreases the expression of MASH1 by 2.2-fold, which are anti- and pro-myogenic and neurogenic genes, respectively, in the Notch signaling pathway. While rescue with exogenous Wnt3a reduced Hes5 levels, MASH1 levels stay repressed. Thus, while Wnt3a can partially rescue the inhibition of differentiation from arsenic, it does so by also modulating Notch target genes rather than only working through the canonical Wnt signaling pathway. These results indicate that arsenic alters the interplay between multiple signaling pathways, leading to reduced stem cell differentiation.

7.
Chem Res Toxicol ; 28(7): 1409-21, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26024302

ABSTRACT

Exposure to arsenic in food and drinking water has been correlated with adverse developmental outcomes, such as reductions in birth weight and neurological deficits. Additionally, studies have shown that arsenic suppresses sensory neuron formation and skeletal muscle myogenesis, although the reason why arsenic targets both of these cell types in unclear. Thus, P19 mouse embryonic stem cells were used to investigate the mechanisms by which arsenic could inhibit cellular differentiation. P19 cells were exposed to 0, 0.1, or 0.5 µM sodium arsenite and induced to form embryoid bodies over a period of 5 days. The expression of transcription factors necessary to form neural plate border specifier (NPBS) cells, neural crest cells and their progenitors, and myocytes and their progenitors were examined. Early during differentiation, arsenic significantly reduced the transcript and protein expression of Msx1 and Pax3, both needed for NPBS cell formation. Arsenic also significantly reduced the protein expression of Sox 10, needed for neural crest progenitor cell production, by 31-50%, and downregulated the protein and mRNA levels of NeuroD1, needed for neural crest cell differentiation, in a time- and dose-dependent manner. While the overall protein expression of transcription factors in the skeletal muscle lineage was not changed, arsenic did alter their nuclear localization. MyoD nuclear translocation was significantly reduced on days 2-5 between 15 and 70%. At a 10-fold lower concentration, monomethylarsonous acid (MMA III) appeared to be just as potent as inorganic arsenic at reducing the mRNA levels Pax3 (79% vs84%), Sox10 (49% vs 65%), and Msx1 (56% vs 56%). Dimethylarsinous acid (DMA III) also reduced protein and transcript expression, but the changes were less dramatic than those with MMA or arsenite. All three arsenic species reduced the nuclear localization of MyoD and NeuroD1 in a similar manner. The early changes in the differentiation of neural plate border specifier cells may provide a mechanism for arsenic to suppress both neurogenesis and myogenesis.


Subject(s)
Arsenites/toxicity , Cell Differentiation/drug effects , Sodium Compounds/toxicity , Animals , Arsenites/chemistry , Arsenites/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cacodylic Acid/analogs & derivatives , Cacodylic Acid/chemical synthesis , Cacodylic Acid/chemistry , Cacodylic Acid/toxicity , Cell Line , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Neural Plate/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Organometallic Compounds/toxicity , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Sodium Compounds/chemistry , Sodium Compounds/metabolism , Transcription Factors/metabolism
8.
Toxicol Appl Pharmacol ; 281(3): 243-53, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25448440

ABSTRACT

Arsenic is a toxicant found in ground water around the world, and human exposure mainly comes from drinking water or from crops grown in areas containing arsenic in soils or water. Epidemiological studies have shown that arsenic exposure during development decreased intellectual function, reduced birth weight, and altered locomotor activity, while in vitro studies have shown that arsenite decreased muscle and neuronal cell differentiation. The sonic hedgehog (Shh) signaling pathway plays an important role during the differentiation of both neurons and skeletal muscle. The purpose of this study was to investigate whether arsenic can disrupt Shh signaling in P19 mouse embryonic stem cells, leading to changes muscle and neuronal cell differentiation. P19 embryonic stem cells were exposed to 0, 0.25, or 0.5 µM of sodium arsenite for up to 9 days during cell differentiation. We found that arsenite exposure significantly reduced transcript levels of genes in the Shh pathway in both a time and dose-dependent manner. This included the Shh ligand, which was decreased 2- to 3-fold, the Gli2 transcription factor, which was decreased 2- to 3-fold, and its downstream target gene Ascl1, which was decreased 5-fold. GLI2 protein levels and transcriptional activity were also reduced. However, arsenic did not alter GLI2 primary cilium accumulation or nuclear translocation. Moreover, additional extracellular SHH rescued the inhibitory effects of arsenic on cellular differentiation due to an increase in GLI binding activity. Taken together, we conclude that arsenic exposure affected Shh signaling, ultimately decreasing the expression of the Gli2 transcription factor. These results suggest a mechanism by which arsenic disrupts cell differentiation.


Subject(s)
Arsenic/pharmacology , Down-Regulation/drug effects , Embryonic Stem Cells/drug effects , Hedgehog Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Water Pollutants, Chemical/pharmacology , Animals , Arsenic/chemistry , Arsenites/antagonists & inhibitors , Arsenites/pharmacology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Line , Cilia/drug effects , Cilia/metabolism , Embryonic Stem Cells/cytology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Muscle Development/drug effects , Neurogenesis/drug effects , Protein Transport/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium Compounds/antagonists & inhibitors , Sodium Compounds/pharmacology , Water Pollutants, Chemical/antagonists & inhibitors , Zinc Finger Protein Gli2
9.
J Agric Food Chem ; 60(12): 3092-7, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22380926

ABSTRACT

Cinnamomum osmophloeum Kaneh is an indigenous tree species in Taiwan. In this study, phytochemical characteristics and antioxidant activities of the essential oils and key constituents from the leaves of two C. osmophloeum clones were investigated. The two trees possess two chemotypes, which were classified as the cinnamaldehyde type and camphor type. We demonstrated that the essential oils from C. osmophloeum leaves exerted in vivo antioxidant activities in Caenorhabditis elegans. In addition, trans-cinnamaldehyde and D-(+)-camphor, which respectively represent the major compounds in the cinnamaldehyde-type and camphor-type trees, exerted significant in vivo antioxidant activities against juglone-induced oxidative stress in C. elegans. Moreover, expressions of antioxidative-related genes, including superoxide dismutase (SOD) and glutathione S-transferase (GST), were significantly induced by trans-cinnamaldehyde and D-(+)-camphor from C. osmophloeum leaves. Our results showed that the essential oils from C. osmophloeum leaves and their major compounds might have good potential for further development as nutraceuticals or antioxidant remedies.


Subject(s)
Antioxidants/pharmacology , Cinnamomum/chemistry , Oils, Volatile/pharmacology , Plant Leaves/chemistry , Acrolein/analogs & derivatives , Acrolein/pharmacology , Animals , Caenorhabditis elegans/drug effects , Camphor/pharmacology , Gene Expression/drug effects , Glutathione Transferase/genetics , Naphthoquinones/pharmacology , Oils, Volatile/chemistry , Oxidation-Reduction , Oxidative Stress/drug effects , Superoxide Dismutase/genetics , Taiwan
10.
Food Chem Toxicol ; 49(4): 812-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21145367

ABSTRACT

Selenium is an essential trace nutrient that has a narrow exposure window between its beneficial and detrimental effects. We investigated how selenium affected the development, fertility, and cholinergic signaling of the nematode, Caenorhabditis elegans. Our results showed that selenite supplementation at 0.01 and 0.05 µM accelerated development and increased the brood size, while the addition of 20 µM selenite retarded the developmental rate and decreased the brood size. We also showed that the 0.01 µM selenite-pretreated nematodes were more resistant to paralysis induced by an acetylcholinesterase inhibitor, aldicarb, and a nicotinic acetylcholine receptor agonist, levamisole, compared to untreated worms. In contrast, 20 µM selenite-pretreated animals were more sensitive to aldicarb- and levamisole-induced paralysis compared to untreated worms. We measured the internal selenium in supplemented worms using inductively coupled plasma atomic emission spectroscopy, and the data obtained suggested that selenite added to growth medium was taken up by the worms. Taken together, these results suggest that selenite exerts both ameliorative and toxic effects on C.elegans, depending on the amount. Our investigations here thus reinforce our understanding of the ameliorative and toxic effects of selenium on development, reproduction, and cholinergic signaling.


Subject(s)
Caenorhabditis elegans/drug effects , Sodium Selenite/toxicity , Animals , Caenorhabditis elegans/physiology , Signal Transduction
11.
Chem Res Toxicol ; 23(5): 926-32, 2010 May 17.
Article in English | MEDLINE | ID: mdl-20423156

ABSTRACT

Arsenic poisoning affects millions of people worldwide. Although there is accumulating evidence to suggest that the nervous system is a target of arsenic, relatively little information is known regarding its effects on the nervous system. The effects of arsenite on the nervous system in Caenorhabditis elegans were investigated in the present study. We found that abts-1, which encodes a Na(+)-dependent Cl(-)/HCO(3)(-) transporter, is required to protect C. elegans from arsenite toxicity. The abts-1::GFP transgene is primarily expressed in neurons and the hypodermis, but stronger expression was also observed in the pharynx and body wall muscle cells after exposure to arsenite. The steady-state level of ABTS-1 mRNA increased in response to arsenite exposure. We showed that worms lacking abts-1 are hypersensitive to the paralytic effects of the cholinesterase inhibitor, aldicarb, and the nicotinic acetylcholine receptor agonist, levamisole. We also showed that arsenite enhanced sensitivity to aldicarb and levamisole in abts-1 mutant worms. Our results indicate neuronal effects of arsenite and the ABTS-1 bicarbonate transporter.


Subject(s)
Anion Transport Proteins/metabolism , Arsenites/toxicity , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Cholinergic Agents/toxicity , Aldicarb/pharmacology , Animals , Animals, Genetically Modified , Anion Transport Proteins/genetics , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/genetics , Chloride-Bicarbonate Antiporters/genetics , Cholinesterase Inhibitors/pharmacology , Levamisole/pharmacology , Nicotinic Agonists/pharmacology , RNA Interference , RNA, Messenger/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL