• Title/Summary/Keyword: mESCs

Search Result 39, Processing Time 0.031 seconds

Transcriptional Properties of the BMP, $TGF-\beta$, RTK, Wnt, Hh, Notch, and JAK/STAT Signaling Molecules in Mouse Embryonic Stem Cells

  • Rho Jeung-Yon;Bae Gab-Yong;Chae Jung-Il;Yu Kweon;Koo Deog-Bon;Lee Kyung-Kwang;Han Yong-Mahn
    • Reproductive and Developmental Biology
    • /
    • v.30 no.2
    • /
    • pp.143-156
    • /
    • 2006
  • Major characteristics of embryonic stem cells (ESCs) are sustaining of sternness and pluripotency by self-renewal. In this report, transcriptional profiles of the molecules in the developmentally important signaling pathways including Wnt, BMP4, $TGF-\beta$, RTK, Hh, Notch, and JAK/STAT signaling pathways were investigated to understand the self-renewal of mouse ESCs (mESCs), J1 line, and compared with the NIH3T3 cell line and mouse embryonic fibroblast (MEF) cells as controls. In the Wnt signaling pathway, the expression of Wnt3 was seen widely in mESCs, suggesting that the ligand may be an important regulator for self-renewal in mESCs. In the Hh signaling pathway, the expression of Gli and N-myc were observed extensively in mESCs, whereas the expression levels of in a Shh was low, suggesting that intracellular molecules may be essential for the self-renewal of mESCs. IGF-I, IGF-II, IGF-IR and IGF-IIR of RTK signaling showed a lower expression in mESCs, these molecules related to embryo development may be restrained in mESCs. The expression levels of the Delta and HESS in Notch signaling were enriched in mESCs. The expression of the molecules related to BMP and JAK-STAT signaling pathways were similar or at a slightly lower level in mESCs compared to those in MEF and NIH3T3 cells. It is suggested that the observed differences in gene expression profiles among the signaling pathways may contribute to the self-renewal and differentiation of mESCs in a signaling-specific manner.

miRNA-222 Modulates Differentiation of Mouse Embryonic Stem Cells

  • Ahn, Hee-Jin;Jung, Jee-Eun;Park, Kyung-Soon
    • Development and Reproduction
    • /
    • v.15 no.4
    • /
    • pp.331-338
    • /
    • 2011
  • MicroRNAs (miRNAs) function as a key regulator of diverse cellular functions. To find out novel miRNAs that promote the differentiation of mouse embryonic stem cells (mESCs), we compared the miRNAs expression profiles of mESCs under self-renewal vs. differentiation states. We noticed that miR-222 was highly expressed during the differentiation of mESCs. Quantitative RT-PCR analysis revealed that expression of miR-222 was up-regulated during the embryonic bodies formation and retinoic acid -dependent differentiation. When miR-222 was suppressed by antogomiR-222, the differentiation of mESCs was delayed compared to control. Self-renewal marker expression or cell proliferation was not affected but the expression of lineage specific marker was suppressed by the treatment of miR-222 inhibitor during the differentiation of mESCs. Taken together, these results suggest that miR-222 functions to promote the differentiation of mESCs by regulating expression of differentiation related genes.

Ganglioside GT1b Mediates Neuronal Differentiation of Mouse Embryonic Stem Cells

  • Lee, So-Dam;Jin, Jung-Woo;Choi, Jin;Choo, Young-Kug
    • Development and Reproduction
    • /
    • v.13 no.3
    • /
    • pp.155-161
    • /
    • 2009
  • It has been reported that ganglioside GT1b is expressed during neuronal cell differentiation from undifferentiated mouse embryonic stem cells (mESCs), which suggests that ganglioside GT1b has a direct effect on neuronal cell differentiation. Therefore, this study was conducted to evaluate the effect of exogenous addition of ganglioside GT1b to an in vitro model of neuronal cell differentiation from undifferentiated mESCs. The results revealed that a significant increase in the expression of ganglioside GT1b occurred during neuronal differentiation of undifferentiated mESCs. Next, we evaluated the effect of retinoic acid (RA) on GT1b-treated undifferentiated mESCs, which was found to lead to increased neuronal differentiation. Taken together, the results of this study suggest that ganglioside GT1b plays a crucial role in neuronal differentiation of mESCs.

  • PDF

Regulation of Pluripotency-related Genes and Differentiation in Mouse Embryonic Stem Cells by Direct Delivery of Cell-penetrating Peptide-conjugated CARM1 Recombinant Protein

  • Choi, Sara;Jo, Junghyun;Seol, Dong-Won;Cha, Soo Kyung;Lee, Jeoung Eun;Lee, Dong Ryul
    • Development and Reproduction
    • /
    • v.17 no.1
    • /
    • pp.9-16
    • /
    • 2013
  • Coactivator-associated arginine methyltransferase 1 (CARM1) is included in the protein arginine methyltransferase (PRMT) family, which methylates histone arginine residues through posttranslational modification. It has been proposed that CARM1 may up-regulate the expression of pluripotency-related genes through the alteration of the chromatin structure. Mouse embryonic stem cells (mESCs) are pluripotent and have the ability to self-renew. The cells are mainly used to study the genetic function of novel genes, because the cells facilitate the transmission of the manipulated genes into target mice. Since the up-regulated methylation levels of histone arginine residue lead to the maintenance of pluripotency in embryos and stem cells, it may be suggested that CARM1 overexpressing mESCs elevate the expression of pluripotency-related genes in reconstituted embryos for transgenic mice and may resist the differentiation into trophectoderm (TE). We constructed a fusion protein by connecting CARM1 and 7X-arginine (R7). As a cell-penetrating peptide (CPP), can translocate CARM1 protein into mESCs. CPP-CARM1 protein was detected in the nuclei of the mESCs after a treatment of 24 hours. Accordingly, the expression of pluripotency-related genes was up-regulated in CPP-CARM1-treated mESCs. In addition, CPP-CARM1-treated mESC-derived embryoid bodies (EBs) showed an elevated expression of pluripotency-related genes and delayed spontaneous differentiation. This result suggests that the treatment of recombinant CPP-CARM1 protein elevates the expression of pluripotency-related genes of mESCs by epigenetic modification, and this protein-delivery system could be used to modify embryonic fate in reconstituted embryos with mESCs.

Effects of Extracellular Signaling on the Endogenous Expression of Self-Renewal-Stimulating Factor Genes in Mouse Embryonic Stem Cells

  • Gong, Seung-Pyo;Lee, Seung-Tae
    • Reproductive and Developmental Biology
    • /
    • v.36 no.1
    • /
    • pp.13-19
    • /
    • 2012
  • In order to provide the basis for developing practical mouse embryonic stem cells (mESCs) culture method, how the endogenous level of self-renewal-stimulating factor genes was altered in the mESCs by different extracellular signaling was investigated in this study. For different extracellular signaling, mESCs were cultured in 2 dimension (D), 3D and integrin-stimulating 3D culture system in the presence or absence of leukemia inhibitory factor (LIF) and transcriptional level of $Lif$, $Bmp4$ and $Wnt3a$ was evaluated in the mESCs cultured in each system. The expression of three genes was significantly increased in 3D system relative to 2D system under LIF-containing condition, while only $Wnt3a$ expression was increased by 3D culture under LIF-free condition. Stimulation of integrin signaling in mESCs within 3D system with exogenous LIF significantly up-regulated transcriptional level of $Bmp4$, but did not induce transcriptional regulation of $Lif$ and $Wnt3a$. In the absence of LIF inside 3D system, the expression of $Lif$ and $Bmp4$ was significantly increased by integrin signaling, while it significantly decreased $Wnt3a$ expression. Finally, the signal from exogenous LIF significantly caused increased expression of $Lif$ in 2D system, decreased expression of $Bmp4$ in both 2D and 3D system, and decreased expression of $Wnt3a$ in integrin-stimulating 3D system. From these results, we identified that endogenous expression level of self-renewal-stimulating factor genes in mESCs could be effectively regulated through artificial and proper manipulation of extracellular signaling. Moreover, synthetic 3D niche stimulating endogenous secretion of self-renewal-stimulating factors will be able to help develop growth factor-free maintenance system of mESCs.

Effects of Extracellular Stimulation of Different Niche Condition on the Transcriptional Regulation of Matrix Metalloproteinase Genes in the Mouse Embryonic Stem Cells

  • Yun, Jung Im;Kim, Min Seong;Lee, Seung Tae
    • Reproductive and Developmental Biology
    • /
    • v.37 no.2
    • /
    • pp.79-83
    • /
    • 2013
  • Matrix metalloproteinases (MMPs) have been known to affect to cell migration, proliferation, morphogenesis and apoptosis by degrading the extracellular matrix. In the previous studies, undifferentiated mouse embryonic stem cells (ESCs) were successfully proliferated inside the extracellular matrix (ECM) analog-conjugated three-dimensional (3D) poly ethylene glycol (PEG)-based hydrogel. However, there is no report about MMP secretion in ESCs, which makes it difficult to understand and explain how ESCs enlarge space and proliferate inside 3D PEG-based hydrogel constructed by crosslinkers containing MMP-specific cleavage peptide sequence. Therefore, we investigated what types of MMPs are released from undifferentiated ESCs and how extracellular signals derived from various niche conditions affect MMP expression of ESCs at the transcriptional level. Results showed that undifferentiated ESCs expressed specifically MMP2 and MMP3 mRNAs. Transcriptional up-regulation of MMP2 was caused by the 3D scaffold, and activation of integrin inside the 3D scaffold upregulated MMP2 mRNAs synergistically. Moreover, mouse embryonic fibroblasts (MEFs) on 2D matrix and 3D scaffold induced upregulation of MMP3 mRNAs, and activation of integrins through conjugation of extracellular matrix (ECM) analogs with 3D scaffold upregulated MMP3 mRNAs synergistically. These results suggest that successful proliferation of ESCs inside the 3D PEG-based hydrogel may be caused by increase of MMP2 and MMP3 expression resulting from 3D scaffold itself as well as activation of integrins inside the 3D PEG-based scaffold.

Suppression of HIF-1α by Valproic Acid Sustains Self-Renewal of Mouse Embryonic Stem Cells under Hypoxia In Vitro

  • Lee, Hyo-Jong;Kim, Kyu-Won
    • Biomolecules & Therapeutics
    • /
    • v.20 no.3
    • /
    • pp.280-285
    • /
    • 2012
  • The developing embryo naturally experiences relatively low oxygen conditions in vivo. Under in vitro hypoxia, mouse embryonic stem cells (mESCs) lose their self-renewal activity and display an early differentiated morphology mediated by the hypoxia-inducible factor-$1{\alpha}$ (HIF-$1{\alpha}$). Previously, we demonstrated that histone deacetylase (HDAC) is activated by hypoxia and increases the protein stability and transcriptional activity of HIF-$1{\alpha}$ in many human cancer cells. Furthermore HDAC1 and 3 mediate the differentiation of mECSs and hematopoietic stem cells. However, the role of HDACs and their inhibitors in hypoxia-induced early differentiation of mESCs remains largely unknown. Here, we examined the effects of several histone deacetylase inhibitors (HDACIs) on the self-renewal properties of mESCs under hypoxia. Inhibition of HDAC under hypoxia effectively decreased the HIF-$1{\alpha}$ protein levels and substantially improved the expression of the LIF-specific receptor (LIFR) and phosphorylated-STAT3 in mESCs. In particular, valproic acid (VPA), a pan HDACI, showed dramatic changes in HIF-$1{\alpha}$ protein levels and LIFR protein expression levels compared to other HDACIs, including sodium butyrate (SB), trichostatin A (TSA), and apicidin (AP). Importantly, our RT-PCR data and alkaline phosphatase assays indicate that VPA helps to maintain the self-renewal activity of mESCs under hypoxia. Taken together, these results suggest that VPA may block the early differentiation of mESCs under hypoxia via the destabilization of HIF-$1{\alpha}$.

Adequate concentration of B cell leukemia/lymphoma 3 (Bcl3) is required for pluripotency and self-renewal of mouse embryonic stem cells via downregulation of Nanog transcription

  • Kang, Songhwa;Yun, Jisoo;Kim, Da Yeon;Jung, Seok Yun;Kim, Yeon Ju;Park, Ji Hye;Ji, Seung Taek;Jang, Woong Bi;Ha, Jongseong;Kim, Jae Ho;Baek, Sang Hong;Kwon, Sang-Mo
    • BMB Reports
    • /
    • v.51 no.2
    • /
    • pp.92-97
    • /
    • 2018
  • B cell leukemia/lymphoma 3 (Bcl3) plays a pivotal role in immune homeostasis, cellular proliferation, and cell survival, as a co-activator or co-repressor of transcription of the $NF-{\kappa}B$ family. Recently, it was reported that Bcl3 positively regulates pluripotency genes, including Oct4, in mouse embryonic stem cells (mESCs). However, the role of Bcl3 in the maintenance of pluripotency and self-renewal activity is not fully established. Here, we report the dynamic regulation of the proliferation, pluripotency, and self-renewal of mESCs by Bcl3 via an influence on Nanog transcriptional activity. Bcl3 expression is predominantly observed in immature mESCs, but significantly decreased during cell differentiation by LIF depletion and in mESC-derived EBs. Importantly, the knockdown of Bcl3 resulted in the loss of self-renewal ability and decreased cell proliferation. Similarly, the ectopic expression of Bcl3 also resulted in a significant reduction of proliferation, and the self-renewal of mESCs was demonstrated by alkaline phosphatase staining and clonogenic single cell-derived colony assay. We further examined that Bcl3-mediated regulation of Nanog transcriptional activity in mESCs, which indicated that Bcl3 acts as a transcriptional repressor of Nanog expression in mESCs. In conclusion, we demonstrated that a sufficient concentration of Bcl3 in mESCs plays a critical role in the maintenance of pluripotency and the self-renewal of mESCs via the regulation of Nanog transcriptional activity.

Clinical Effect through Histological Characteristics of Focal Ischemia Region (뇌허혈성 부위의 조직학적 특성을 통한 임상적 영향)

  • Lee, Tae-Hoon
    • Journal of Industrial Convergence
    • /
    • v.17 no.4
    • /
    • pp.39-43
    • /
    • 2019
  • Mouse embryonic stem cell could show an substitutional materials of cells of neuron differentiation, positively increasing their effectiveness in the treatment of nervous symptom. We examined that mouse embryonic stem cells (mESCs) can be induced to undergo neuronal differentiation. After neuronal induction, the phenotype of mESCs changed towards neuronal morphology and mESCs were injected into the lateral ventricle of the experimental animal brain. Transplanted cells migrated to various parts of the brain and ischemic brain injury by middle cerebral artery occlusion (MCAO) increased their migration to the injured cortex. Intracerebral grafting of mESCs mostly improve sensory and motor nervous system of neurological injury in focal cerebral rats.

Identification and Characterization of Secreted Phosphoprotein 2 as a Novel Bioactive Protein for Myocardial Differentiation (심근세포로의 분화에 관여하는 새로운 생리활성 단백질 SPP2의 발굴)

  • Sejin Jeon
    • Journal of Life Science
    • /
    • v.33 no.1
    • /
    • pp.64-72
    • /
    • 2023
  • Despite several advances in identification of cardiac transcription factors, there are still needs to find new bioactive molecules that promote cardiomyogenesis from stem cells to highly efficient myocardial differentiation. We analyzed Illumina expression microarray data of mouse embryonic stem cells (mESCs)-derived cardiomyocytes. 276 genes were upregulated (≥ 4fold) in mESCs-derived cardiomyocytes compared undifferentiated ESCs. Secreted phosphoprotein 2 (Spp2) is one of candidates and is known to inhibit bone morphogenetic protein 2 (BMP2) signal transduction as a pseudoreceptor for BMP2. However, its function in cardiomyogenesis is unknown. We confirmed that Spp2 expression increased during the differentiation into functional cardiomyocytes using mESCs, TC-1/Kh2 and E14. Interestingly, Spp2 secretion transiently increased 3 days after formation of embryoid bodies (EBs), indicating that the extracellular secretion of Spp2 is involved in the differentiation of ESCs into cardiomyocytes. To characterize Spp2, we performed experiments using the C2C12 mouse myoblast cell line, which has the property of shifting the differentiation pathway from myoblastic to osteoblastic by treatment with BMP2. Similar to the differentiation of ESCs, transcription of Spp2 increased as C2C12 myoblasts differentiated into myotubes. In particular, Spp2 secretion increased dramatically in the early stage of differentiation. Furthermore, treatment with Spp2-Flag recombinant protein promoted the differentiation of C2C12 myoblasts into myotubes. Taken together, we suggest a novel bioactive protein Spp2 that differentiates ESCs into cardiomyocytes. This may be useful for understanding the molecular pathways of cardiomyogenesis and for experimental or clinical promotion of stem cell therapy for ischemic heart diseases.