Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(10): 5529-5548, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38512058

ABSTRACT

The process of induced pluripotent stem cells (iPSCs) reprogramming involves several crucial events, including the mesenchymal-epithelial transition (MET), activation of pluripotent genes, metabolic reprogramming, and epigenetic rewiring. Although these events intricately interact and influence each other, the specific element that regulates the reprogramming network remains unclear. Dux, a factor known to promote totipotency during the transition from embryonic stem cells (ESC) to 2C-like ESC (2CLC), has not been extensively studied in the context of iPSC reprogramming. In this study, we demonstrate that the modification of H3K18la induced by Dux overexpression controls the metabolism-H3K18la-MET network, enhancing the efficiency of iPSC reprogramming through a metabolic switch and the recruitment of p300 via its C-terminal domain. Furthermore, our proteomic analysis of H3K18la immunoprecipitation experiment uncovers the specific recruitment of Brg1 during reprogramming, with both H3K18la and Brg1 being enriched on the promoters of genes associated with pluripotency and epithelial junction. In summary, our study has demonstrated the significant role of Dux-induced H3K18la in the early reprogramming process, highlighting its function as a potent trigger. Additionally, our research has revealed, for the first time, the binding of Brg1 to H3K18la, indicating its role as a reader of histone lactylation.


Subject(s)
Cellular Reprogramming , Epithelial-Mesenchymal Transition , Histones , Homeodomain Proteins , Induced Pluripotent Stem Cells , Transcription Factors , Animals , Humans , Mice , Cellular Reprogramming/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Epithelial-Mesenchymal Transition/genetics , Histones/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
2.
Basic Res Cardiol ; 119(1): 151-168, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38145999

ABSTRACT

A deficiency of striated preferentially expressed gene (Speg), a member of the myosin light chain kinase family, results in abnormal myofibril structure and function of immature cardiomyocytes (CMs), corresponding with a dilated cardiomyopathy, heart failure and perinatal death. Mitochondrial development plays a role in cardiomyocyte maturation. Therefore, this study investigated whether Speg deficiency ( - / - ) in CMs would result in mitochondrial abnormalities. Speg wild-type and Speg-/- C57BL/6 littermate mice were utilized for assessment of mitochondrial structure by transmission electron and confocal microscopies. Speg was expressed in the first and second heart fields at embryonic (E) day 7.5, prior to the expression of mitochondrial Na+/Ca2+/Li+ exchanger (NCLX) at E8.5. Decreases in NCLX expression (E11.5) and the mitochondrial-to-nuclear DNA ratio (E13.5) were observed in Speg-/- hearts. Imaging of E18.5 Speg-/- hearts revealed abnormal mitochondrial cristae, corresponding with decreased ATP production in cells fed glucose or palmitate, increased levels of mitochondrial superoxide and depolarization of mitochondrial membrane potential. Interestingly, phosphorylated (p) PGC-1α, a key mediator of mitochondrial development, was significantly reduced in Speg-/- hearts during screening for targeted genes. Besides Z-line expression, Speg partially co-localized with PGC-1α in the sarcomeric region and was found in the same complex by co-immunoprecipitation. Overexpression of a Speg internal serine/threonine kinase domain in Speg-/- CMs promoted translocation of pPGC-1α into the nucleus, and restored ATP production that was abolished by siRNA-mediated silencing of PGC-1α. Our results demonstrate a critical role of Speg in mitochondrial development and energy metabolism in CMs, mediated in part by phosphorylation of PGC-1α.


Subject(s)
Cardiomyopathy, Dilated , Mitochondrial Diseases , Mice , Animals , Pregnancy , Female , Myocytes, Cardiac/metabolism , Mice, Inbred C57BL , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , DNA, Mitochondrial/metabolism , Adenosine Triphosphate/metabolism , Mitochondrial Diseases/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Muscle Proteins/genetics , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism
3.
JHEP Rep ; 5(7): 100746, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37274776

ABSTRACT

Background & Aims: Non-alcoholic fatty liver disease (NAFLD) affects nearly a quarter of the population with no approved pharmacological therapy. Liver steatosis is a primary characteristic of NAFLD. Recent studies suggest that human umbilical cord mesenchymal stem cell-derived exosomes (MSC-ex) may provide a promising strategy for treating liver injury; however, the role and underlying mechanisms of MSC-ex in steatosis are not fully understood. Methods: Oleic-palmitic acid-treated hepatic cells and high-fat diet (HFD)-induced NAFLD mice were established to observe the effect of MSC-ex. Using non-targeted lipidomics and transcriptome analyses, we analysed the gene pathways positively correlated with MSC-ex. Mass spectrometry and gene knockdown/overexpression analyses were performed to evaluate the effect of calcium/calmodulin-dependent protein kinase 1 (CAMKK1) transferred by MSC-ex on lipid homoeostasis regulation. Results: Here, we demonstrate that MSC-ex promote fatty acid oxidation and reduce lipogenesis in oleic-palmitic acid-treated hepatic cells and HFD-induced NAFLD mice. Non-targeted lipidomics and transcriptome analyses suggested that the effect of MSC-ex on lipid accumulation positively correlated with the phosphorylation of AMP-activated protein kinase. Furthermore, mass spectrometry and gene knockdown/overexpression analyses revealed that MSC-ex-transferred CAMKK1 is responsible for ameliorating lipid accumulation in an AMP-activated protein kinase-dependent manner, which subsequently inhibits SREBP-1C-mediated fatty acid synthesis and enhances peroxisome proliferator-activated receptor alpha (PPARα)-mediated fatty acid oxidation. Conclusions: MSC-ex may prevent HFD-induced NAFLD via CAMKK1-mediated lipid homoeostasis regulation. Impact and Implications: NAFLD includes many conditions, from simple steatosis to non-alcoholic steatohepatitis, which can lead to fibrosis, cirrhosis, and even hepatocellular carcinoma. So far, there is no approved drug for treating liver steatosis of NAFLD. Thus, better therapies are needed to regulate lipid metabolism and prevent the progression from liver steatosis to chronic liver disease. By using a combination of non-targeted lipidomic and transcriptome analyses, we revealed that human umbilical cord mesenchymal stem cell-derived exosomes (MSC-ex) effectively reduced lipid deposition and improved liver function from HFD-induced liver steatosis. Our study highlights the importance of exosomal CAMKK1 from MSC-ex in mediating lipid metabolism regulation via AMPK-mediated PPARα/CPT-1A and SREBP-1C/fatty acid synthase signalling in hepatocytes. These findings are significant in elucidating novel mechanisms related to MSC-ex-based therapies for preventing NAFLD.

4.
Sensors (Basel) ; 22(13)2022 Jun 26.
Article in English | MEDLINE | ID: mdl-35808333

ABSTRACT

The use of fiber Bragg grating (FBG) sensors is proposed to solve the technical problem of poor sensor stability in the long-term safety monitoring of shaft lining structures. The auxiliary shaft of the Zhuxianzhuang coal mine was considered as the engineering background, and a test system implementing FBG sensors was established to monitor the long-term safety of the shaft lining structure. Indoor simulation testing revealed that the coefficient of determination (r2) between the test curves of the FBG sensor and the resistance strain gauge is greater than 0.99 in both the transverse and vertical strains. Therefore, the FBG sensor and resistance strain gauge test values are similar, and the error is small. The early warning value was obtained by calculation, according to the specific engineering geological conditions and shaft lining structure. The monitoring data obtained for the shaft lining at three test levels over more than three years reveal that the measured vertical strain value is less than the warning value, indicating that the shaft lining structure is currently in a safe state. The analysis of the monitoring data reveals that the vertical strain increment caused by the vertical additional force is approximately 0.0752 µÎµ/d. As the mine drainage progresses, the increasing vertical additional force acting on the shaft lining will compromise the safety of the shaft lining structure. Therefore, the monitoring must be enhanced to facilitate decision-making for safe shaft operation.


Subject(s)
Fiber Optic Technology , Optical Fibers , Monitoring, Physiologic
5.
Stem Cell Res Ther ; 12(1): 430, 2021 07 31.
Article in English | MEDLINE | ID: mdl-34332630

ABSTRACT

The study of human brain physiology, including cellular interactions in normal and disease conditions, has been a challenge due to its complexity and unavailability. Induced pluripotent stem cell (iPSC) study is indispensable in the study of the pathophysiology of neurological disorders. Nevertheless, monolayer systems lack the cytoarchitecture necessary for cellular interactions and neurological disease modeling. Brain organoids generated from human pluripotent stem cells supply an ideal environment to model both cellular interactions and pathophysiology of the human brain. This review article discusses the composition and interactions among neural lineage and non-central nervous system cell types in brain organoids, current studies, and future perspectives in brain organoid research. Ultimately, the promise of brain organoids is to unveil previously inaccessible features of neurobiology that emerge from complex cellular interactions and to improve our mechanistic understanding of neural development and diseases.


Subject(s)
Induced Pluripotent Stem Cells , Nervous System Diseases , Brain , Humans , Organoids , Technology
6.
Biol Reprod ; 105(5): 1089-1103, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34296246

ABSTRACT

Two-cell-like (2C-like) embryonic stem cells (ESCs) are a small group of ESCs that spontaneously express zygotic genome activation (ZGA) genes and repeats, such as Zscan4 and murine endogenous retrovirus with leucine (MERVL), and are specifically expressed in 2-cell-stage mouse embryos. Although numerous types of treatment and agents elevate the transition of ESCs to 2C-like ESCs, Dux serves as a critical factor in this transition by increasing the expression of Zscan4 and MERVL directly. However, the loss of Dux did not impair the birth of mice, suggesting that Dux may not be the primary transitioning factor in fertilized embryos. It has been reported that for 2-cell embryos derived from somatic cell nuclear transfer (SCNT) and whose expression of ZGA genes and repeats was aberrant, Dux improved the reprogramming efficiency by correcting aberrant H3K9ac modification via its C-terminal domain. We confirmed that the overexpression of full-length Dux mRNA in SCNT embryos improved the efficiency of preimplantation development (62.16% vs. 41.26% with respect to controls) and also increased the expression of Zscan4 and MERVL. Furthermore, we found that the N-terminal double homeodomains of Dux were indispensable for Dux localization and function. The intermediate region was essential for MERVL and Zscan4 activation, and the C-terminal domain was important for elevating level of H3K27ac. Mutant Dux mRNA containing N-terminal double homeodomains with the intermediate region or the C-terminal domain also improved the preimplantation development of SCNT embryos. This is the first report focusing on distinguishing functional domains of Dux in embryos derived from SCNT.


Subject(s)
Embryo, Mammalian/embryology , Embryonic Development/genetics , Homeodomain Proteins/genetics , Mice/embryology , Nuclear Transfer Techniques , Animals , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice/genetics , Protein Domains/genetics
7.
Stem Cell Rev Rep ; 16(6): 1049-1061, 2020 12.
Article in English | MEDLINE | ID: mdl-32939647

ABSTRACT

Induced pluripotent stem cells (iPSCs) are mainly characterized by their unlimited proliferation abilities and potential to develop into almost any cell type. The creation of this technology has been of great interest to many scientific fields, especially regenerative biology. However, concerns about the safety of iPSC application in transplantation have arisen due to the tumorigenic and immunogenic properties of iPSCs. This review will briefly introduce the developing history of somatic reprogramming and applications of iPSC technology in regenerative medicine. In addition, the review will highlight two challenges to the efficient usage of iPSCs and the underlying mechanisms of these challenges. Finally, the review will discuss the expanding application of iPSC technology in cancer immunotherapy as a potential cancer vaccine and its advantages in auxiliary treatment compared with oncofetal antigen-based and embryonic stem cell (ESC)-based vaccines.


Subject(s)
Cancer Vaccines/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/pathology , Animals , Carcinogenesis/genetics , Epigenesis, Genetic , Humans , Regeneration
8.
Sci Rep ; 10(1): 7315, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355273

ABSTRACT

SLC16A family members play crucial roles in tumorigenesis and tumor progression. However, the exact role of distinct members in the SLC16A family in human pancreatic cancer remains unclear. Integrated bioinformatics analysis for the identification of therapeutic targets for certain cancers based on transcriptomics, proteomics and high-throughput sequencing could help us obtain novel information and understand potential underlying molecular mechanisms. In the present study, we investigated SLC16A family members in pancreatic cancer through accumulated data from GEO (Gene Expression Omnibus), TCGA (The Cancer Genome Atlas) and other available databases. The expression profile, clinical application significance and prognostic value of the SLC16A family for patients with pancreatic cancer were explored. SLC16A1, SLC16A3 and SLC16A13 exhibited biomarker potential for prognosis, and we further identified their related genes and regulatory networks, revealing core molecular pathways that require further investigation for pancreatic cancer.


Subject(s)
Computational Biology/methods , Gene Expression Regulation, Neoplastic , Monocarboxylic Acid Transporters/genetics , Pancreatic Neoplasms/genetics , Symporters/genetics , Biomarkers , Biomarkers, Tumor , Cell Transformation, Neoplastic/genetics , Disease Progression , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Multigene Family , Oligonucleotide Array Sequence Analysis , Prognosis , Protein Interaction Mapping , Protein Interaction Maps , Treatment Outcome
9.
Stem Cells ; 38(8): 960-972, 2020 08.
Article in English | MEDLINE | ID: mdl-32346926

ABSTRACT

Aberrant epigenetic reprogramming is one of the major barriers for somatic cell reprogramming. Although our previous study has indicated that H3K27me3 demethylase KDM6A can improve the nuclear reprogramming efficiency, the mechanism remains unclear. In this study, we demonstrate that the overexpression of Kdm6a may improve induced pluripotent stem cell (iPSC) reprogramming efficiency in a demethylase enzymatic activity-dependent manner. KDM6A erased H3K27me3 on pluripotency- and metabolism-related genes, and consequently facilitated changing the gene expression profile and metabolic pattern to an intermediate state. Furthermore, KDM6A may promote IL-6 expression, and the secreted IL-6 may further improve iPSC reprogramming efficiency. In addition, KDM6A may promote PTEN expression to decrease p-AKT and p-mTOR levels, which in turn facilitates reprogramming. Overall, our results reveal that KDM6A may promote iPSC reprogramming efficiency by accelerating changes in the gene expression profile and the metabolic pattern in a demethylation-activity-dependent manner. These results may provide an insight into the relationship between epigenomics, transcriptomics, metabolomics, and reprogramming.


Subject(s)
Histone Demethylases/metabolism , Induced Pluripotent Stem Cells/cytology , Interleukin-6/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Cellular Reprogramming/physiology , Epigenesis, Genetic , Female , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Signal Transduction
10.
Cytokine Growth Factor Rev ; 52: 1-14, 2020 04.
Article in English | MEDLINE | ID: mdl-32057701

ABSTRACT

Immune checkpoint inhibitors (ICIs) and immunotherapy have proven to be a transformative therapy for many forms of cancer treatment. While many antibodies targeting the PD-1, PD-L1, and CTLA-4 pathways have been approved for clinical use by the FDA, it is clear that a single ICI is not sufficient to eradicate disease. ICI combination strategies are being extensively investigated to advance cancer treatment to next curative stage. Among the immune checkpoint inhibitors being actively investigated, the potential of VISTA (V-domain Ig suppressor of T cell activation), a unique B7 family member that functions as both ligand and receptor, is being actively pursued. This article summarizes the expression and immunomodulatory effects of VISTA in autoimmune diseases and cancer, and assesses its potential as an additional component of immune checkpoint cancer therapy.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , B7 Antigens/genetics , Neoplasms/genetics , Neoplasms/immunology , Animals , B7 Antigens/immunology , Gene Expression Regulation , Humans , Immunomodulation , Lymphocyte Activation , Mice
11.
Sci Rep ; 8(1): 10955, 2018 Jul 19.
Article in English | MEDLINE | ID: mdl-30026469

ABSTRACT

Patient-specific induced pluripotent stem cells (iPSCs) have the potential to be useful in the treatment of human diseases. While prior studies have reported multiple methods to generate iPSCs, DNA methylation continues to limit the totipotency and reprogramming efficiency of iPSCs. Here, we first show the competency of embryonic germ cells (EGCs) as a reprogramming catalyst capable of effectively promoting reprogramming induced by four defined factors, including Oct4, Sox2, Klf4 and c-Myc. Combining EGC extracts with these four factors resulted in formation of more embryonic stem cell-like colonies than did factors alone. Notably, expression of imprinted genes was higher with combined induction than with factors alone. Moreover, iPSCs derived from the combined inductors tended to have more global hypomethylation. Our research not only provides evidence that EGC extracts could activate DNA demethylation and reprogram imprinted genes, but also establishes a new way to enhance reprogramming of iPSCs, which remains a critical safety concern for potential use of iPSCs in regenerative medicine.


Subject(s)
Embryonic Germ Cells/cytology , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cells, Cultured , Cellular Reprogramming , DNA Methylation , Embryonic Germ Cells/metabolism , Female , Genomic Imprinting , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Male , Mice , Proto-Oncogene Proteins c-myc , Regenerative Medicine
12.
Stem Cell Res Ther ; 7(1): 112, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27515169

ABSTRACT

BACKGROUND: rDNA, the genes encoding ribosomal RNA (rRNA), is highly demanded for ribosome production and protein synthesis in growing cells such as pluripotent stem cells. rDNA transcription activity varies between cell types, metabolism conditions, and specific environmental challenges. Embryonic stem cells (ESCs), partially reprogrammed cells, and somatic cells reveal different epigenetic signatures, including rDNA epigenetic marks. rDNA epigenetic characteristic resetting is not quite clear during induced pluripotent stem cell (iPSC) generation. Little is known that whether the different rDNA epigenetic status in donor cells will result in different rDNA transcription activities, and furthermore affect reprogramming efficiency. METHODS: We utilized serum starvation-synchronized mouse embryonic fibroblasts (MEFs) to generate S-iPSCs. Both MEFs and serum-refeeding MEFs (S-MEFs) were reprogrammed to a pluripotent state. rDNA-related genes, UBF proteins, and rDNA methylation levels were detected during the MEF and S-MEF cell reprogramming process. RESULTS: We demonstrated that, after transient inhibition, retroviral induced rRNA transcriptional activity was reprogrammed towards a pluripotent state. Serum starvation would stimulate rDNA transcription reactivation during somatic cell reprogramming. Serum starvation improved the methylation status of donor cells at rRNA gene promoter regions. CONCLUSIONS: Our results provide insight into regulation of rDNA transcriptional activity during somatic cell reprogramming and allow for comparison of rDNA regulation patterns between iPSCs and S-iPSCs. Eventually, regulation of rDNA transcriptional activity will benefit partially reprogrammed cells to overcome the epigenetic barrier to pluripotency.


Subject(s)
Cell Cycle/physiology , Cellular Reprogramming/genetics , DNA, Ribosomal/genetics , Induced Pluripotent Stem Cells/physiology , Transcriptional Activation/genetics , Animals , Cellular Reprogramming/physiology , Embryonic Stem Cells , Epigenesis, Genetic/genetics , Epigenomics/methods , Fibroblasts/physiology , Methylation , Mice , Promoter Regions, Genetic/genetics
13.
Dev Growth Differ ; 58(3): 270-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26991405

ABSTRACT

Mouse parthenogenetic embryonic stem cells (PgESCs) could be applied to study imprinting genes and are used in cell therapy. Our previous study found that stem cells established by aggregation of two parthenogenetic embryos at 8-cell stage (named as a2 PgESCs) had a higher efficiency than that of PgESCs, and the paternal expressed imprinting genes were observably upregulated. Therefore, we propose that increasing the number of parthenogenetic embryos in aggregation may improve the development of parthenogenetic mouse and imprinting gene expression of PgESCs. To verify this hypothesis, we aggregated four embryos together at the 4-cell stage and cultured to the blastocyst stage (named as 4aPgB). qPCR detection showed that the expression of imprinting genes Igf2, Mest, Snrpn, Igf2r, H19, Gtl2 in 4aPgB were more similar to that of fertilized blastocyst (named as fB) compared to 2aPgB (derived from two 4-cell stage parthenogenetic embryos aggregation) or PgB (single parthenogenetic blastocyst). Post-implantation development of 4aPgB extended to 11 days of gestation. The establishment efficiency of GFP-a4 PgESCs which derived from GFP-4aPgB is 62.5%. Moreover, expression of imprinting genes Igf2, Mest, Snrpn, notably downregulated and approached the level of that in fertilized embryonic stem cells (fESCs). In addition, we acquired a 13.5-day fetus totally derived from GFP-a4 PgESCs with germline contribution by 8-cell under zona pellucida (ZP) injection. In conclusion, four embryos aggregation improves parthenogenetic development, and compensates imprinting genes expression in PgESCs. It implied that a4 PgESCs could serve as a better scientific model applied in translational medicine and imprinting gene study.


Subject(s)
Blastomeres/metabolism , Gene Expression Regulation, Developmental , Genomic Imprinting/genetics , Mouse Embryonic Stem Cells/metabolism , Parthenogenesis/genetics , Animals , Blastocyst/cytology , Blastocyst/metabolism , Blastomeres/cytology , Cell Aggregation/genetics , Cell Differentiation/genetics , Embryonic Development/genetics , Female , Fluorescent Antibody Technique , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
14.
Arch Med Sci ; 12(1): 199-207, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26925137

ABSTRACT

INTRODUCTION: Stem cells involved cell replacement therapies for type 1 diabetes mellitus is promising, yet time-consuming and inefficient. Exendin-4 is a glucagon-like peptide-1 (GLP-1) receptor agonist which has been reported to possess anti-apoptotic effects, thereby increasing ß-cell mass and improving ß-cell function. The present study aimed to investigate whether exendin-4 would enhance the differentiation of embryonic stem cells into insulin-secreting cells and improve the pancreatic differentiation strategy. MATERIAL AND METHODS: R1 embryonic stem cells were treated with different concentrations of exendin-4 and divided into three groups. In the high dosage group (group H), exendin-4 was added at the dosage of 10 nmol/l. In the low dosage group (group L), exendin-4 was added at the dosage of 0.1 nmol/l. Group C was a control. Expression of genes related to the ß-cell phenotype and immunofluorescence staining of insulin and C-peptide were detected. RESULTS: Compared with groups L and C, group H had the highest mRNA expression levels of Isl1, Pdx1, Ngn3, and Insulin1 (p < 0.05). Neurod1 and Glut2 only emerged at the final stage of differentiation in group H. Immunofluorescence analysis revealed that exendin-4 upregulated the protein expression of insulin and C-peptide. CONCLUSIONS: Exendin-4 remarkably facilitated Neurod1 and Glut2 gene transcription, and was able to induce differentiation of embryonic stem cells into endocrine and insulin-producing cells.

15.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 33(2): 382-7, 2016 Apr.
Article in Chinese | MEDLINE | ID: mdl-29708710

ABSTRACT

A critical shortage of donor organs raises a question of needs for alternative organ sources for regenerative medicine.Over the last decade,three-dimensional(3D)culture has become a new approach for organ regeneration.The 3Dculture takes significant advantages of cells spatial relationships between multiple cellular types and surrounding matrices of dynamic cellular interactions,which plays a key role in structural self-formation of complex organ buds.Here we present major classic cases of 3Dculture organ regeneration to show how it works,and then we try to find the way of future organ regeneration.


Subject(s)
Organogenesis , Regenerative Medicine/methods , Tissue Engineering/methods , Animals , Humans , Regeneration , Stem Cells/cytology
16.
Sci Rep ; 5: 17829, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26643778

ABSTRACT

Autophagy is an essential cellular mechanism that degrades cytoplasmic proteins and organelles to recycle their components. Moreover, autophagy is essential for preimplantation development in mammals. Here we show that autophagy is also important for reprogramming in somatic cell nuclear transfer (SCNT). Our data indicate that unlike fertilized oocytes, autophagy is not triggered in SCNT embryos during 6 hours of activation. Mechanistically, the inhibited autophagic induction during SCNT activation is due to the cytochalasin B (CB) caused depolymerization of actin filaments. In this study, we induced autophagy during SCNT activation by rapamycin and pp242, which could restore the expected level of autophagy and significantly enhance the development of SCNT embryos to the blastocyst stage when compared with the control (68.5% and 68.7% vs. 41.5%, P < 0.05). Furthermore, the treatment of rapamycin and pp242 accelerates active DNA demethylation indicated by the conversion of 5 mC to 5 hmC, and treatment of rapamycin improves degradation of maternal mRNA as well. Thus, our findings reveal that autophagy is important for development of SCNT embryos and inhibited autophagic induction during SCNT activation might be one of the serious causes of low efficiency of SCNT.


Subject(s)
Autophagy , Cloning, Organism , Embryo, Mammalian , Actin Cytoskeleton/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , DNA Methylation , Female , Fertilization in Vitro , Gene Expression , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/antagonists & inhibitors , Nuclear Transfer Techniques , Oocytes/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
17.
ACS Appl Mater Interfaces ; 7(48): 26792-801, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26558750

ABSTRACT

Herein we describe a novel AC electrothermal (ACET) fluidic circulatory pumping chip to overcome the challenge of fluid-to-tissue ratio for "human-on-a-chip" cell culture systems. To avoid the deleterious effects of Joule heating and electric current on sample cells, a rectangular microchannel was designed with distantly separated regions for pumping and cell culture. Temperature variations were examined using a commercial thermocouple sensor to detect temperature values in both pumping and culture regions. To generate a sufficient ACET circulatory pumping rate, 30 pairs of asymmetrical electrodes were employed in the pumping region; generated ACET velocity was measured by fluorescent microparticle image velocimetry. The benefits of our pumping chip were demonstrated by culturing human embryonic kidney cells (HEK293T) and human colon carcinoma cells (SW620) for 72 h with an energized voltage of 3 V and 10 MHz. Cells grew and proliferated well, implying our ACET circulatory pumping chip has great potential for cell culture and tissue engineering applications.


Subject(s)
Cell Culture Techniques/methods , Electric Conductivity , Electrochemistry/methods , Cell Line, Tumor , Cell Survival , Computer Simulation , Culture Media , HEK293 Cells , Humans , Numerical Analysis, Computer-Assisted , Rheology , Temperature
18.
J Vet Sci ; 15(3): 417-22, 2014.
Article in English | MEDLINE | ID: mdl-24962406

ABSTRACT

A purely laparoscopic four-port approach was created for left hepatectomy in pigs. A polyethylene loop was placed on the left two hepatic lobes for traction and lift. Next, penetrating ligation of the lobes using of a double row of silk sutures was performed to control bleeding. A direct hepatic transection was completed using a monopolar hook electrode without meticulous dissection of the left hepatic vein. The raw surface of the liver was coagulated and sealed with fibrin glue. Lobes were retrieved through an enlarged portal. Laparoscopic hepatic lobectomy was completed in all pigs without the use of specialized instruments and with a mean operative time of 179 ± 9 min. No significant perioperative complications were observed. The average weight of each resected lobe was 180 ± 51 g. Complete blood count as well as serum organics and enzyme levels normalized after about 2 weeks. During necropsy, adhesion of the hepatic raw surface to the gastric wall and omentum were observed. No other abnormalities were identified. This minimally invasive left hepatectomy technique in swine could serve as a useful model for investigating liver diseases and regeneration, and offer preclinical information to improve hepatobiliary surgical procedures.


Subject(s)
Hepatectomy/veterinary , Swine/surgery , Animals , Female , Hepatectomy/methods , Laparoscopy/methods , Laparoscopy/veterinary , Liver/surgery , Male , Postoperative Care/methods , Postoperative Care/veterinary , Swine, Miniature/surgery
19.
Stem Cell Rev Rep ; 10(4): 548-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24828831

ABSTRACT

The most effective reprogramming methods, somatic cell nuclear transfer (SCNT) and induced pluripotent stem cells (iPSCs), are widely used in biological research and regenerative medicine, yet the mechanism that reprograms somatic cells to totipotency remains unclear and thus reprogramming efficiency is still low. Microarray technology has been employed in analyzing the transcriptomes changes during iPS reprogramming. Unfortunately, it is difficult to obtain enough DNA from SCNT reconstructed embryos to take advantage of this technology. In this study, we aimed to identify critical genes from the transcriptional profile for iPS reprogramming and compared expression levels of these genes in SCNT reprogramming. By integrating gene expression information from microarray databases and published studies comparing somatic cells with either miPSCs or mouse embryonic stem cells (ESCs), we obtained two lists of co-upregulated genes. The gene ontology (GO) enriched analysis of these two lists demonstrated that the reprogramming process is associated with numerous biological processes. Specifically, we selected 32 genes related to heterochromatin, embryonic development, and cell cycle from our co-upregulated gene datasets and examined the gene expression level in iPSCs and SCNT embryos by qPCR. The results revealed that some reprogramming related genes in iPSCs were also expressed in SCNT reprogramming. We established the network of gene interactions that occur with genes differentially expressed in iPS and SCNT reprogramming and then performed GO analysis on the genes in the network. The network genes function in chromatin organization, heterochromatin, transcriptional regulation, and cell cycle. Further researches to improve reprogramming efficiency, especially in SCNT, will focus on functional studies of these selected genes.


Subject(s)
Cellular Reprogramming , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Animals , Cells, Cultured , Clone Cells , Embryonic Stem Cells/cytology , Female , Gene Regulatory Networks , Induced Pluripotent Stem Cells/cytology , Male , Mice , Mice, Inbred C57BL , Nuclear Transfer Techniques , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Regenerative Medicine , Reverse Transcriptase Polymerase Chain Reaction
20.
Cell Reprogram ; 16(1): 77-83, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24387163

ABSTRACT

Induced pluripotent stem cells (iPSCs) are usually generated by reprogramming somatic cells through transduction with a transcription factor cocktail. However, the low efficiency of this procedure has kept iPSCs away from the study of the clinical application of stem cell biology. Our research shows that continuous passage increases the efficiency of reprogramming. Compared with conventional method of establishment of iPSCs, more embryonic stem cell (ESC)-like clones are generated by continuous passage during early reprogramming. These inchoate clones, indistinguishable from genuine ESC clones, are closer to fully reprogrammed cells compared with those derived from classical iPSC induction, which increased the expression of pluripotent gene markers and the levels of demethylation of Oct4 and Nanog. These results suggested that full reprogramming is a gradual process that does not merely end at the point of the activation of endogenous pluripotency-associated genes. Continuous passage could increase the pluripotency of induced cells and accelerate the process of reprogramming by epigenetic modification. In brief, we have provided an advanced strategy to accelerate the reprogramming and generate more nearly fully reprogrammed iPSCs efficiently and rapidly.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Dedifferentiation , Induced Pluripotent Stem Cells/metabolism , Transcription Factors/biosynthesis , Animals , Antigens, Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Mice , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...