Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
Add more filters

Complementary Medicines
Therapeutic Methods and Therapies TCIM
Publication year range
1.
Chem Biol Interact ; 352: 109783, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-34932953

ABSTRACT

This paper provides an identification and detailed assessment of hormetic dose responses of embryonic stem cells (ESCs) with particular emphasis on cell renewal (proliferation) and differentiation, underlying mechanistic foundations and potential therapeutic implications. Hormetic dose responses were commonly reported, being induced by a broad range of chemicals, including pharmaceuticals (e.g., atorvastatin, isoproterenol, lithium, nicotine, ouabain), dietary supplements (e.g., curcumin, multiple ginsenosides, resveratrol), endogenous agents (e.g., estrogen, hydrogen peroxide, melatonin), and physical stressor agents (e.g., hypoxia, ionizing radiation). ESC-hormetic dose responses are similar for other stem cell types (e.g., adipose-derived stem cells, apical papilla, bone marrow stem cells, dental pulp stem cells, endothelial stem cells, muscle stem cells, periodontal ligament stem cells, neural stem cells), indicating a high degree of generality for the hormetic-stem cells response. The widespread occurrence of hormetic dose responses shown by ESCs and other stem cells suggests that the hormetic dose response may represent a fundamental and highly conserved evolutionary strategy.


Subject(s)
Embryonic Stem Cells/drug effects , Hormesis , Animals , Biological Evolution , Cell Differentiation/drug effects , Cell Hypoxia/physiology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dietary Supplements , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Fatty Acids/administration & dosage , Hormesis/physiology , Humans
2.
Article in English | MEDLINE | ID: mdl-34044158

ABSTRACT

An important bottleneck in fish aquaculture research is the supply and maintenance of embryos, larvae, juvenile and adult specimens. In this context, cell lines represent alternative experimental models for in vitro studies that complement in vivo assays. This allows us to perform easier experimental design and sampling and avoid the sacrifice of animals. Embryonic stem (ES) cell lines have attracted increasing attention because they have the capability to proliferate indefinitely and could be differentiated into any cell type of the organism. To minimise cell heterogeneity and increase uniformity of in vitro studies results, in this manuscript we report the development and characterisation of two single cell-derived ES cell lines (monoclonal) from the morula stage embryos of the gilthead seabream, Sparus aurata, named as SAEC-A3 and SAEC-H7. Both cell lines have been passaged for over 100 times, indicating the establishment of long-term, immortalised ES cell cultures. Sequence analyses confirmed the seabream origin of the cell lines, and growth analyses evidenced their high viability and proliferating activity, particularly in culture medium supplemented with 10-15% fetal bovine serum and 22 °C. Both cell lines showed the ability to generate embryoid bodies and show different sensitivity and response to all-trans retinoic acid. The analysis of epithelial (col1α1) and neuronal (sox3) markers in differentiated cultures revealed that SAEC-A3 tended to differentiate towards epithelial-like cells whereas SAEC-H7 tended to differentiate towards neuronal-like cells. Both cell lines were efficiently transfected with pDsRed2-ER and/or pEGFP-N1 plasmids, indicating that they could represent useful biotechnological tools. Daily expression of pcna showed significant expression rhythms, with maximum levels of cell proliferation during the day-night transition. Currently, these cell lines are being successfully used as experimental models for the study of cellular metabolism, physiology and rhythms as well as for toxicological, pharmacological and gene expression analyses.


Subject(s)
Cell Differentiation , Circadian Rhythm , Embryonic Stem Cells/cytology , Fish Proteins/metabolism , Sea Bream/physiology , Animals , Cell Proliferation , Embryonic Stem Cells/metabolism , Fish Proteins/genetics
4.
Sci Rep ; 10(1): 15469, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32963285

ABSTRACT

Mechanical forces are important in the regulation of physiological homeostasis and the development of disease. The application of mechanical forces to cultured cells is often performed using specialized systems that lack the flexibility and throughput of other biological techniques. In this study, we developed a high throughput platform for applying complex dynamic mechanical forces to cultured cells. We validated the system for its ability to accurately apply parallel mechanical stretch in a 96 well plate format in 576 well simultaneously. Using this system, we screened for optimized conditions to stimulate increases in Oct-4 and other transcription factor expression in mouse fibroblasts. Using high throughput mechanobiological screening assays, we identified small molecules that can synergistically enhance the increase in reprograming-related gene expression in mouse fibroblasts when combined with mechanical loading. Taken together, our findings demonstrate a new powerful tool for investigating the mechanobiological mechanisms of disease and performing drug screening in the presence of applied mechanical load.


Subject(s)
Cellular Reprogramming/drug effects , Drug Evaluation, Preclinical/methods , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Small Molecule Libraries/pharmacology , Stress, Mechanical , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Female , Fibroblasts/cytology , Fibroblasts/drug effects , High-Throughput Screening Assays , Male , Mice
5.
J Cell Mol Med ; 24(22): 12886-12899, 2020 11.
Article in English | MEDLINE | ID: mdl-32969187

ABSTRACT

Yin yang 2 (YY2) is a multifunctional zinc finger protein that belongs to the yin yang (YY) family. YY2 has dual function in regulating gene expression, as it could act either as a transcriptional activator or as a repressor of its target genes. YY2 could regulate genes that have been previously identified as targets of yin yang 1 (YY1), another member of the YY family, by binding to their common binding sequences. However, recent studies revealed that YY2 also has its own specific binding sequences, leading to its particular biological functions distinct from those of YY1. Furthermore, they have different levels or even opposite regulatory effects on common target genes, suggesting the importance of balanced YY1 and YY2 regulations in maintaining proper cellular homeostasis and biological functions. Recent studies revealed that YY2 plays crucial roles in maintaining stemness and regulating differentiation potential of embryonic stem cells, as well as in the development of the brain, nervous and cardiovascular systems. YY2 expression is also closely related to diseases, as it could act as a tumour suppressor gene that regulates tumour cell proliferation and metastasis. Moreover, YY2 is also involved in immune regulation and immune surveillance. Herein, we summarize recent perspectives regarding the regulatory functions of YY2, as well as its biological functions and relation with diseases.


Subject(s)
Embryonic Stem Cells/cytology , Transcription Factors/metabolism , YY1 Transcription Factor/metabolism , Animals , Binding Sites , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/metabolism , Homeostasis , Humans , Immune System , Mice , Neoplasms/immunology , Neoplasms/metabolism , Prognosis , Protein Processing, Post-Translational , Stem Cells/cytology , Transcriptional Activation
6.
Antioxid Redox Signal ; 32(1): 35-59, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31656084

ABSTRACT

Aims: The naive or primitive states of stem cells (SCs) residing in specific niches are unstable and difficult to preserve in vitro. Vitamin C (VitC), in addition to suppressing oxygen radicals, exerts pleiotropic effects to preserve the core functions of SCs. However, this compound is labile and readily oxidized, resulting in cellular toxicity and preventing its reliable application in this context. We found that a VitC derivative, ascorbic acid 2-glucoside (AA2G), stably maintains the naive pluripotency of murine embryonic SCs (mESCs) and the primitiveness of human mesenchymal SCs (hMSCs) without cellular toxicity. Results: The beneficial effects of AA2G and related molecular mechanisms were evaluated in mESCs, induced pluripotent-SCs (iPSCs), and hMSCs. AA2G was stable in aqueous solution and barely induced cellular toxicity in cultured SCs, unlike VitC. AA2G supplementation recapitulated the well-known effects of VitC, including induction of ten-eleven translocation-dependent DNA demethylation in mESCs and suppression of p53 during generation of murine iPSCs. Furthermore, supplementation of hMSCs with AA2G improved therapeutic outcomes in an asthma mouse model by promoting their self-renewal, engraftment, and anti-inflammatory properties. Particularly, activation of the cAMP-responsive element-binding protein-1 (CREB1) pathway contributed to the ability of AA2G to maintain naive pluripotency of mESCs and functionality of hMSCs. Innovation and Conclusion: Given its long-lasting effects and low cellular toxicity, AA2G supplementation is useful to support the naive pluripotency of mESCs and the primitiveness of hMSCs, affecting their developmental potency and therapeutic efficacy. Furthermore, we demonstrate the significance of the CREB1 pathway in the mechanism of action of AA2G.


Subject(s)
Ascorbic Acid/analogs & derivatives , Asthma/therapy , Cyclic AMP Response Element-Binding Protein/metabolism , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Animals , Ascorbic Acid/pharmacology , Asthma/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Stem Cell Niche
7.
Epigenetics Chromatin ; 12(1): 69, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31722724

ABSTRACT

BACKGROUND: Neural tube defects (NTDs) are common congenital malformations resulting in failure of the neural tube closure during early embryonic development. Although it is known that maternal folate deficiency increases the risk of NTDs, the mechanism remains elusive. RESULTS: Herein, we report that histone H2A monoubiquitination (H2AK119ub1) plays a role in neural tube closure. We found that the folate antagonist methotrexate induced H2AK119ub1 in mouse embryonic stem cells. We demonstrated that an increase in H2AK119ub1 downregulated expression of the neural tube closure-related genes Cdx2, Nes, Pax6, and Gata4 in mouse embryonic stem cells under folate deficiency conditions. We also determined that the E3 ligase Mdm2 was responsible for the methotrexate-induced increase in H2AK119ub1 and downregulation of neural tube closure-related genes. Surprisingly, we found that Mdm2 is required for MTX-induced H2A ubiquitination and is recruited to the sites of DSB, which is dependent on DNA damage signaling kinase ATM. Furthermore, folic acid supplementation restored H2AK119ub1 binding to neural tube closure-related genes. Downregulation of these genes was also observed in both brain tissue of mouse and human NTD cases, and high levels of H2AK119ub1 were found in the corresponding NTDs samples with their maternal serum folate under low levels. Pearson correlation analysis showed a significant negative correlation between expression of the neural precursor genes and H2AK119ub1. CONCLUSION: Our results indicate that folate deficiency contributes to the onset of NTDs by altering H2AK119ub1 and subsequently affecting expression of neural tube closure-related genes. This may be a potential risk factor for NTDs in response to folate deficiency.


Subject(s)
Down-Regulation , Histones/metabolism , Neural Tube Defects/pathology , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , DNA Damage , Down-Regulation/drug effects , Embryonic Development/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Folic Acid/metabolism , Folic Acid/pharmacology , Folic Acid/therapeutic use , Methotrexate/pharmacology , Mice , Mice, Inbred C57BL , Neural Tube Defects/metabolism , Neural Tube Defects/prevention & control , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Protein Binding , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Ubiquitination
8.
Endocrinology ; 160(7): 1701-1718, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31135891

ABSTRACT

Tanycytes have recently been accepted as neural stem/progenitor cells in the postnatal hypothalamus. Persistent retina and anterior neural fold homeobox (Rax) expression is characteristic of tanycytes in contrast to its transient expression of whole hypothalamic precursors. In this study, we found that Rax+ residual cells in the maturation phase of hypothalamic differentiation in mouse embryonic stem cell (mESC) cultures had similar characteristics to ventral tanycytes. They expressed typical neural stem/progenitor cell markers, including Sox2, vimentin, and nestin, and differentiated into mature neurons and glial cells. Quantitative RT-PCR analysis showed that Rax+ residual cells expressed Fgf-10, Fgf-18, and Lhx2, which are expressed by ventral tanycytes. They highly expressed tanycyte-specific genes Dio2 and Gpr50 compared with Rax+ early hypothalamic progenitor cells. Therefore, Rax+ residual cells in the maturation phase of hypothalamic differentiation were considered to be more differentiated and similar to late progenitor cells and tanycytes. They self-renewed and formed neurospheres when cultured with exogenous FGF-2. Additionally, these Rax+ neurospheres differentiated into three neuronal lineages (neurons, astrocytes, and oligodendrocytes), including neuropeptide Y+ neuron, that are reported to be differentiated from ventral tanycytes toward the arcuate nuclei. Thus, Rax+ residual cells were multipotent neural stem/progenitor cells. Rax+ neurospheres were stably passaged and retained high Sox2 expression even after multiple passages. These results suggest the successful induction of Rax+ tanycyte-like cells from mESCs [induced tanycyte-like (iTan) cells]. These hypothalamic neural stem/progenitor cells may have potential in regenerative medicine and as a research tool.


Subject(s)
Cell Lineage/physiology , Embryonic Stem Cells/metabolism , Ependymoglial Cells/metabolism , Hypothalamus/metabolism , Neural Stem Cells/metabolism , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Ependymoglial Cells/cytology , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factors/metabolism , Hypothalamus/cytology , LIM-Homeodomain Proteins/metabolism , Mice , Neural Stem Cells/cytology , Transcription Factors/metabolism
9.
Methods Mol Biol ; 1953: 23-31, 2019.
Article in English | MEDLINE | ID: mdl-30912013

ABSTRACT

The steadily increasing throughput in next-generation sequencing technologies is revolutionizing a number of fields in biology. One application requiring massive parallel sequencing is forward genetic screening based on chemical mutagenesis. Such screens interrogate the entire genome in an entirely unbiased fashion and can be applied to a number of research questions. CRISPR/Cas9-based screens, which are largely limited to a gene's loss of function, have already been very successful in identifying drug targets and pathways related to the drug's mode of action. By inducing single nucleotide changes using an alkylating reagent, it is possible to generate amino acid changes that perturb the interaction between a drug and its direct target, resulting in drug resistance. This chemogenomic approach combined with latest sequencing technologies allows deconvolution of drug targets and characterization of drug-target binding interfaces at amino acid resolution, therefore nicely complementing existing biochemical approaches. Here we describe a general protocol for a chemical mutagenesis-based forward genetic screen applicable for drug-target deconvolution.


Subject(s)
CRISPR-Cas Systems , Drug Evaluation, Preclinical/methods , Gene Editing/methods , Mutagenesis , Animals , Cell Culture Techniques/methods , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Discovery/methods , Drug Resistance , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Mice , Mutation , Sequence Analysis, DNA/methods
10.
Differentiation ; 106: 15-22, 2019.
Article in English | MEDLINE | ID: mdl-30844688

ABSTRACT

Drug-induced liver injury is a major cause of drug discovery failure in clinical trials and a leading cause of liver disease. Current preclinical drug testing does not predict hepatotoxicity which highlights the importance of developing highly predictive cell-based models. The use of stem cell technology and differentiation into hepatocyte-like cells (HLCs) could provide a stable source of hepatocytes for multiple applications, including drug screening. HLCs derived from both embryonic and induced pluripotent stem cells have been used to accurately predict hepatotoxicity as well as to test individual-specific toxicity. Although there are still many limitations, mainly related to the lack of fully maturity of the HLCs derived from pluripotent stem cells, they could provide a relative unlimited and consistent supply of cells with stable phenotype, that could be obtained from different donors, enabling the generation of a library of HLCs representative of the variability of human population.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Drug Evaluation, Preclinical/methods , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Embryonic Stem Cells/drug effects , Hepatocytes/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Phenotype
11.
Int J Mol Sci ; 20(5)2019 Mar 11.
Article in English | MEDLINE | ID: mdl-30862029

ABSTRACT

Peanut sprouts (PS), which are germinated peanut seeds, have recently been reported to have anti-oxidant, anti-inflammatory, and anti-obesity effects. However, the underlying mechanisms by which PS modulates lipid metabolism are largely unknown. To address this question, serial doses of PS extract (PSE) were added to 3T3-L1 cells during adipocyte differentiation. PSE (25 µg/mL) significantly attenuated adipogenesis by inhibiting lipid accumulation in addition to reducing the level of adipogenic protein and gene expression with the activation of AMP-activated protein kinase (AMPK). Other adipocyte cell models such as mouse embryonic fibroblasts C3H10T1/2 and primary adipocytes also confirmed the anti-adipogenic properties of PSE. Next, we investigated whether PSE attenuated lipid accumulation in mature adipocytes. We found that PSE significantly suppressed lipogenic gene expression, while fatty acid (FA) oxidation genes were upregulated. Augmentation of FA oxidation by PSE in mature 3T3-L1 adipocytes was confirmed via a radiolabeled-FA oxidation rate experiment by measuring the conversion of [³H]-oleic acid (OA) to [³H]-H2O. Furthermore, PSE enhanced the mitochondrial oxygen consumption rate (OCR), especially maximal respiration, and beige adipocyte formation in adipocytes. In summary, PSE was effective in reducing lipid accumulation in 3T3-L1 adipocytes through mitochondrial fatty acid oxidation involved in AMPK and mitochondrial activation.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Arachis/chemistry , Fatty Acids/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Plant Extracts/pharmacology , Triglycerides/metabolism , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipogenesis/drug effects , Animals , Cell Respiration/drug effects , Cell Survival/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Flavonoids/chemistry , Lipid Metabolism/drug effects , Mice , Oxygen Consumption , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/chemistry , Polyphenols/chemistry , Resveratrol/chemistry
12.
Biofactors ; 45(3): 427-438, 2019 May.
Article in English | MEDLINE | ID: mdl-30907984

ABSTRACT

One of the major issues in cell therapy of myocardial infarction (MI) is early death of engrafted cells in a harsh oxidative stress environment, which limits the potential therapeutic utility of this strategy in the clinical setting. Increasing evidence implicates beneficial effects of omega-3 fatty acids including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and ascorbic acid (AA) in cardiovascular diseases, in particular their role in ameliorating fibrosis. In the current study, we aim to assess the cytoprotective role of EPA + DHA and AA in protecting embryonic stem cell (ESC)-derived cardiac lineage cells and amelioration of fibrosis. Herein, we have shown that preincubation of the cells with EPA + DHA + AA prior to H2 O2 treatment attenuated generation of reactive oxygen species (ROS) and enhanced cell viability. Gene expression analysis revealed that preincubation with EPA + DHA + AA followed by H2 O2 treatment, upregulated heme oxygenase-1 (HO-1) along with cardiac markers (GATA4, myosin heavy chain, α isoform [MYH6]), connexin 43 [CX43]) and attenuated oxidative stress-induced upregulation of fibroblast markers (vimentin and collagen type 1 [Col1]). Alterations in gene expression patterns were followed by marked elevation of cardiac troponin (TNNT2) positive cells and reduced numbers of vimentin positive cells. An injection of EPA + DHA + AA-pretreated ESC-derived cardiac lineage cells into the ischemic myocardium of a rat model of MI significantly reduced fibrosis compared to the vehicle group. This study provided evidence that EPA + DHA + AA may be an appropriate preincubation regimen for regenerative purposes. © 2019 BioFactors, 45(3):427-438, 2019.


Subject(s)
Ascorbic Acid/therapeutic use , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Fatty Acids, Omega-3/therapeutic use , Animals , Biomarkers/metabolism , Blotting, Western , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Docosahexaenoic Acids/therapeutic use , Echocardiography , Eicosapentaenoic Acid/therapeutic use , Heme Oxygenase-1/metabolism , Humans , Hydrogen Peroxide/metabolism , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
13.
Cell Reprogram ; 20(5): 289-300, 2018 10.
Article in English | MEDLINE | ID: mdl-30277824

ABSTRACT

Establishing pig embryonic stem cells (pESCs) remains a challenge due to differences in the genetic backgrounds of mouse, human, and pig. Therefore, pig-specific pluripotency markers and cellular signaling must be identified. In this study, doxycycline (DOX)-inducible vectors carrying Oct4, sex-determining region Y-box 2 (Sox2), Nanog, Kruppel-like family 4 (Klf4), or Myc, which are known reprogramming factors, were transduced into pESCs. And pluripotency genes were analyzed in one or two reprogramming factor-expressed pESCs. When cultured without DOX, pESCs were stably maintained in basic fibroblast growth factor-supplemented media. However, when treated with DOX, the cells lost their alkaline phosphatase (AP) activity and differentiated within 2 weeks. Subsequently, we investigated the expression of genes related to pluripotency in DOX-treated pESCs using quantitative reverse transcription-polymerase chain reaction (PCR). Expression levels of Oct4, E-cadherin, and Fut4 were significantly increased by Oct4 overexpression, and Oct4 and Fut4 were upregulated in the Sox2-infected group. When a combination of two reprogramming factors, including Oct4 or Sox2, was introduced, weak AP activity remained. In addition, several of the two reprogramming factor transduction groups could be maintained after subculturing with transgene activation. Although long-term culture failed, pESCs transduced with Oct4 and Nanog, Oct4 and Klf4, or Sox2 and Nanog combinations could be subcultured even under transgene activation conditions. Analysis of the cause of long-term culture failure by quantitative PCR confirmed that the expression of intermediate reprogramming markers was not maintained. Given these results, additional methods are needed to support the completion of each reprogramming phase to succeed in the conversion of the pluripotent state of pESCs. This study improves our understanding of pluripotent networks and can be used to aid in the establishment of bona fide pig pluripotent stem cells.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Embryoid Bodies/cytology , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Animals , Cells, Cultured , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lentivirus/genetics , Pluripotent Stem Cells/metabolism , Swine , Transcription Factors/genetics
14.
Development ; 145(11)2018 06 11.
Article in English | MEDLINE | ID: mdl-29784672

ABSTRACT

Haploid embryonic stem cells (haESCs) have been extensively applied in forward and reverse genetic screening. However, a mammalian haploid somatic cell line is difficult to achieve because of spontaneous diploidization in differentiation. As a non-human primate species, monkeys are widely used in basic and pre-clinical research in which haploid cells are restricted to ESCs. Here, we report that rhesus monkey haESCs in an optimized culture medium show naïve-state pluripotency and stable haploidy. This model facilitated the derivation of haploid neural progenitor cells (haNPCs), which maintained haploidy and differentiation potential into neurons and glia for a long period in vitro High-throughput trapping mutations can be efficiently introduced into haNPCs via piggyBac transposons. This system proves useful when identifying gene targets of neural toxicants via a proof-of-concept experiment. Using CRISPR/Cas9 editing, we confirmed that B4GALT6, from the candidate gene list, is a resistance gene of A803467 (a tetrodotoxin-like toxicant). This model is the first non-human primate haploid somatic cell line with proliferative ability, multipotency and an intact genome, thus providing a cellular resource for recessive genetic and potential drug screening.


Subject(s)
Drug Evaluation, Preclinical/methods , Embryonic Stem Cells/cytology , Galactosyltransferases/genetics , Gene Editing/methods , Genetic Testing/veterinary , Macaca mulatta/embryology , Neural Stem Cells/cytology , Aniline Compounds/pharmacology , Animals , CRISPR-Cas Systems , DNA Transposable Elements/genetics , Furans/pharmacology , Genetic Testing/methods , Haploidy , Poisons/pharmacology
15.
Stem Cell Res ; 25: 98-106, 2017 12.
Article in English | MEDLINE | ID: mdl-29125995

ABSTRACT

Muscular dystrophy is a disease characterized by progressive muscle weakness and degeneration. There are currently no available treatments for most muscular diseases, such as muscular dystrophy. Moreover, current therapeutics are focused on improving the quality of life of patients by relieving the symptoms or stress caused by the disease. Although the causative genes for many muscular diseases have been identified, the mechanisms underlying their pathogenesis remain unclear. Patient-derived induced pluripotent stem cells (iPSCs) have become a powerful tool for understanding the pathogenesis of intractable diseases, as well as for phenotype screening, which can serve as the basis for developing new drugs. However, it is necessary to develop an efficient and reproducible myogenic differentiation system. Previously, we reported a tetracycline-inducible MyoD overexpression model of myogenic differentiation using human iPSCs (hiPSCs). However, this model has certain disadvantages that limit its use in various applications, such as a drug screening. In this study, we developed an efficient and reproducible myogenic differentiation system by further modifying our previous protocol. The new protocol achieves efficient differentiation of feeder-free hiPSCs to myogenic cells via small-scale culture in six-well microplates to large-scale culture in 384-well microplates for high-throughput applications.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Drug Evaluation, Preclinical , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Muscle Development/physiology , Quality of Life
16.
Cell Rep ; 21(2): 517-532, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29020636

ABSTRACT

The human cerebral cortex possesses distinct structural and functional features that are not found in the lower species traditionally used to model brain development and disease. Accordingly, considerable attention has been placed on the development of methods to direct pluripotent stem cells to form human brain-like structures termed organoids. However, many organoid differentiation protocols are inefficient and display marked variability in their ability to recapitulate the three-dimensional architecture and course of neurogenesis in the developing human brain. Here, we describe optimized organoid culture methods that efficiently and reliably produce cortical and basal ganglia structures similar to those in the human fetal brain in vivo. Neurons within the organoids are functional and exhibit network-like activities. We further demonstrate the utility of this organoid system for modeling the teratogenic effects of Zika virus on the developing brain and identifying more susceptibility receptors and therapeutic compounds that can mitigate its destructive actions.


Subject(s)
Anti-Retroviral Agents/pharmacology , Cerebral Cortex/cytology , Drug Evaluation, Preclinical/methods , Organoids/virology , Primary Cell Culture/methods , Zika Virus/drug effects , Cell Line , Cerebral Cortex/virology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Humans , Neurons/cytology , Neurons/metabolism , Neurons/virology , Organoids/cytology , Organoids/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , c-Mer Tyrosine Kinase/metabolism
17.
PLoS One ; 12(8): e0183358, 2017.
Article in English | MEDLINE | ID: mdl-28817722

ABSTRACT

INTRODUCTION: Vitamin B3 has been shown to play an important role during embryogenesis. Specifically, there is growing evidence that nicotinamide, the biologically active form of vitamin B3, plays a critical role as a morphogen in the differentiation of stem cells to mature cell phenotypes, including those of the central nervous system (CNS). Detailed knowledge of the action of small molecules during neuronal differentiation is not only critical for uncovering mechanisms underlying lineage-specification, but also to establish more effective differentiation protocols to obtain clinically relevant cells for regenerative therapies for neurodegenerative conditions such as Huntington's disease (HD). Thus, this study aimed to investigate the potential of nicotinamide to promote the conversion of stem cells to mature CNS neurons. METHODS: Nicotinamide was applied to differentiating mouse embryonic stem cells (mESC; Sox1GFP knock-in 46C cell line) during their conversion towards a neural fate. Cells were assessed for changes in their proliferation, differentiation and maturation; using immunocytochemistry and morphometric analysis methods. RESULTS: Results presented indicate that 10 mM nicotinamide, when added at the initial stages of differentiation, promoted accelerated progression of ESCs to a neural lineage in adherent monolayer cultures. By 14 days in vitro (DIV), early exposure to nicotinamide was shown to increase the numbers of differentiated ßIII-tubulin-positive neurons. Nicotinamide decreased the proportion of pluripotent stem cells, concomitantly increasing numbers of neural progenitors at 4 DIV. These progenitors then underwent rapid conversion to neurons, observed by a reduction in Sox 1 expression and decreased numbers of neural progenitors in the cultures at 14 DIV. Furthermore, GABAergic neurons generated in the presence of nicotinamide showed increased maturity and complexity of neurites at 14 DIV. Therefore, addition of nicotinamide alone caused an accelerated passage of pluripotent cells through lineage specification and further to non-dividing mature neurons. CONCLUSIONS: Our results show that, within an optimal dose range, nicotinamide is able to singly and selectively direct the conversion of embryonic stem cells to mature neurons, and therefore may be a critical factor for normal brain development, thus supporting previous evidence of the fundamental role of vitamins and their metabolites during early CNS development. In addition, nicotinamide may offer a simple effective supplement to enhance the conversion of stem cells to clinically relevant neurons.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Niacinamide/pharmacology , Animals , Cell Lineage , Cell Proliferation/drug effects , Embryonic Stem Cells/cytology , Green Fluorescent Proteins/genetics , Mice
18.
Mamm Genome ; 28(7-8): 302-314, 2017 08.
Article in English | MEDLINE | ID: mdl-28752194

ABSTRACT

Since its domestication over 100 years ago, the laboratory rat has been the preferred experimental animal in many areas of biomedical research (Lindsey and Baker The laboratory rat. Academic, New York, pp 1-52, 2006). Its physiology, size, genetics, reproductive cycle, cognitive and behavioural characteristics have made it a particularly useful animal model for studying many human disorders and diseases. Indeed, through selective breeding programmes numerous strains have been derived that are now the mainstay of research on hypertension, obesity and neurobiology (Okamoto and Aoki Jpn Circ J 27:282-293, 1963; Zucker and Zucker J Hered 52(6):275-278, 1961). Despite this wealth of genetic and phenotypic diversity, the ability to manipulate and interrogate the genetic basis of existing phenotypes in rat strains and the methodology to generate new rat models has lagged significantly behind the advances made with its close cousin, the laboratory mouse. However, recent technical developments in stem cell biology and genetic engineering have again brought the rat to the forefront of biomedical studies and enabled researchers to exploit the increasingly accessible wealth of genome sequence information. In this review, we will describe how a breakthrough in understanding the molecular basis of self-renewal of the pluripotent founder cells of the mammalian embryo, embryonic stem (ES) cells, enabled the derivation of rat ES cells and their application in transgenesis. We will also describe the remarkable progress that has been made in the development of gene editing enzymes that enable the generation of transgenic rats directly through targeted genetic modifications in the genomes of zygotes. The simplicity, efficiency and cost-effectiveness of the CRISPR/Cas gene editing system, in particular, mean that the ability to engineer the rat genome is no longer a limiting factor. The selection of suitable targets and gene modifications will now become a priority: a challenge where ES culture and gene editing technologies can play complementary roles in generating accurate bespoke rat models for studying biological processes and modelling human disease.


Subject(s)
Gene Editing , Genetic Engineering , Genome , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Cell Differentiation , Embryo, Mammalian , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Rearrangement , Gene Targeting/methods , Mice , Oligodeoxyribonucleotides , Rats , Transcription Activator-Like Effector Nucleases/metabolism , Zinc Finger Nucleases/metabolism
19.
Stem Cells ; 35(8): 1913-1923, 2017 08.
Article in English | MEDLINE | ID: mdl-28580685

ABSTRACT

During cardiac development, DNA binding transcription factors and epigenetic modifiers regulate gene expression in cardiac progenitor cells (CPCs). We have previously shown that Yin Yang 1 (YY1) is essential for the commitment of mesodermal precursors into CPCs. However, the role of YY1 in the maintenance of CPC phenotype and their differentiation into cardiomyocytes is unknown. In this study, we found, by genome-wide transcriptional profiling and phenotypic assays, that YY1 overexpression prevents cardiomyogenic differentiation and maintains the proliferative capacity of CPCs. We show further that the ability of YY1 to regulate CPC phenotype is associated with its ability to modulate histone modifications specifically at a developmentally critical enhancer of Nkx2-5 and other key cardiac transcription factor such as Tbx5. Specifically, YY1 overexpression helps to maintain markers of gene activation such as the acetylation of histone H3 at lysine 9 (H3K9Ac) and lysine 27 (H3K27Ac) as well as trimethylation at lysine 4 (H3K4Me3) at the Nkx2-5 cardiac enhancer. Furthermore, transcription factors associated proteins such as PoIII, p300, and Brg1 are also enriched at the Nkx2-5 enhancer with YY1 overexpression. The biological activities of YY1 in CPCs appear to be cell autonomous, based coculture assays in differentiating embryonic stem cells. Altogether, these results demonstrate that YY1 overexpression is sufficient to maintain a CPC phenotype through its ability to sustain the presence of activating epigenetic/chromatin marks at key cardiac enhancers. Stem Cells 2017;35:1913-1923.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Myocardium/cytology , YY1 Transcription Factor/metabolism , Animals , Binding Sites , Cell Differentiation/genetics , Cell Line , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gain of Function Mutation , Gene Expression Regulation , Homeobox Protein Nkx-2.5/genetics , Mice
20.
Tissue Eng Part C Methods ; 23(5): 286-297, 2017 05.
Article in English | MEDLINE | ID: mdl-28401793

ABSTRACT

Skeletal development is a multistep process that involves the complex interplay of multiple cell types at different stages of development. Besides biochemical and physical cues, oxygen tension also plays a pivotal role in influencing cell fate during skeletal development. At physiological conditions, bone cells generally reside in a relatively oxygenated environment whereas chondrocytes reside in a hypoxic environment. However, it is technically challenging to achieve such defined, yet diverse oxygen distribution on traditional in vitro cultivation platforms. Instead, engineered osteochondral constructs are commonly cultivated in a homogeneous, stable environment. In this study, we describe a customized perfusion bioreactor having stable positional variability in oxygen tension at defined regions. Further, engineered collagen constructs were coaxed into adopting the shape and dimensions of defined cultivation platforms that were precasted in 1.5% agarose bedding. After cultivating murine embryonic stem cells that were embedded in collagen constructs for 50 days, mineralized constructs of specific dimensions and a stable structural integrity were achieved. The end-products, specifically constructs cultivated without chondroitin sulfate A (CSA), showed a significant increase in mechanical stiffness compared with their initial gel-like constructs. More importantly, the localization of osteochondral cell types was specific and corresponded to the oxygen tension gradient generated in the bioreactor. In addition, CSA in complementary with low oxygen tension was also found to be a potent inducer of chondrogenesis in this system. In summary, we have demonstrated a customized perfusion bioreactor prototype that is capable of generating a more dynamic, yet specific cultivation environment that could support propagation of multiple osteochondral lineages within a single engineered construct in vitro. Our system opens up new possibilities for in vitro research on human skeletal development.


Subject(s)
Bioreactors , Bone and Bones/cytology , Cartilage, Articular/cytology , Chondrocytes/cytology , Embryonic Stem Cells/cytology , Oxygen/metabolism , Tissue Engineering/methods , Animals , Bone and Bones/physiology , Cartilage, Articular/physiology , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Chondrocytes/physiology , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Embryonic Stem Cells/physiology , Mice , Partial Pressure , Perfusion
SELECTION OF CITATIONS
SEARCH DETAIL