1
|
Gene Profiles in the Early Stage of Neuronal Differentiation of Mouse Bone Marrow Stromal Cells Induced by Basic Fibroblast Growth Factor. Stem Cells Int 2021; 2020:8857057. [PMID: 33424980 PMCID: PMC7775150 DOI: 10.1155/2020/8857057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 11/20/2022] Open
Abstract
A stably established population of mouse bone marrow stromal cells (BMSCs) with self-renewal and multilineage differentiation potential was expanded in vitro for more than 50 passages. These cells express high levels of mesenchymal stem cell markers and can be differentiated into adipogenic, chondrogenic, and osteogenic lineages in vitro. Subjected to basic fibroblast growth factor (bFGF) treatment, a typical neuronal phenotype was induced in these cells, as supported by neuronal morphology, induction of neuronal markers, and relevant electrophysiological excitability. To identify the genes regulating neuronal differentiation, cDNA microarray analysis was conducted using mRNAs isolated from cells differentiated for different time periods (0, 4, 24, and 72 h) after bFGF treatment. Various expression patterns of neuronal genes were stimulated by bFGF. These gene profiles were shown to be involved in developmental, functional, and structural integration of the nervous system. The expression of representative genes stimulated by bFGF in each group was verified by RT-PCR. Amongst proneural genes, the mammalian achate-schute homolog 1 (Mash-1), a basic helix-loop-helix transcriptional factor, was further demonstrated to be significantly upregulated. Overexpression of Mash-1 in mouse BMSCs was shown to induce the expression of neuronal specific enolase (NSE) and terminal neuronal morphology, suggesting that Mash-1 plays an important role in the induction of neuronal differentiation of mouse BMSCs.
Collapse
|
2
|
Terrigno M, Bertacchi M, Pandolfini L, Baumgart M, Calvello M, Cellerino A, Studer M, Cremisi F. The microRNA miR-21 Is a Mediator of FGF8 Action on Cortical COUP-TFI Translation. Stem Cell Reports 2018; 11:756-769. [PMID: 30174317 PMCID: PMC6135738 DOI: 10.1016/j.stemcr.2018.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
The morphogen FGF8 plays a pivotal role in neocortical area patterning through its inhibitory effect on COUP-TFI/Nr2f1 anterior expression, but its mechanism of action is poorly understood. We established an in vitro model of mouse embryonic stem cell corticogenesis in which COUP-TFI protein expression is inhibited by the activation of FGF8 in a time window corresponding to cortical area patterning. Interestingly, overexpression of the COUP-TFI 3'UTR reduces the inhibitory effect of FGF8 on COUP-TFI translation. FGF8 induces the expression of few miRNAs targeting COUP-TFI 3'UTR in silico. We found that the functional inhibition of miR-21 can effectively counteract the inhibitory effect of FGF8 in vitro and regulate COUP-TFI protein levels in vivo. Accordingly, miR-21 expression is complementary to COUP-TFI expression during corticogenesis. These data support a translational control of COUP-TFI gradient expression by FGF8 via miR-21 and contribute to our understanding of how regionalized expression is established during neocortical area mapping.
Collapse
Affiliation(s)
- Marco Terrigno
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy
| | | | - Luca Pandolfini
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy
| | - Mario Baumgart
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | | | - Alessandro Cellerino
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy; Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Michèle Studer
- Université Côte d'Azur (UCA), CNRS, Inserm, iBV, 06108 Nice, France
| | - Federico Cremisi
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy; Istituto di Biofisica CNR, Via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
3
|
Dung TTM, Yi YS, Heo J, Yang WS, Kim JH, Kim HG, Park JG, Yoo BC, Cho JY, Hong S. Critical role of protein L-isoaspartyl methyltransferase in basic fibroblast growth factor-mediated neuronal cell differentiation. BMB Rep 2017; 49:437-42. [PMID: 26973341 PMCID: PMC5070731 DOI: 10.5483/bmbrep.2016.49.8.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Indexed: 11/29/2022] Open
Abstract
We aimed to study the role of protein L-isoaspartyl methyltransferase (PIMT) in neuronal differentiation using basic fibroblast growth factor (bFGF)-induced neuronal differentiation, characterized by cell-body shrinkage, long neurite outgrowth, and expression of neuronal differentiation markers light and medium neurofilaments (NF). The bFGF-mediated neuronal differentiation of PC12 cells was induced through activation of mitogen-activated protein kinase (MAPK) signaling molecules [MAPK kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2 (ERK1/2), and p90RSK], and phosphatidylinositide 3-kinase (PI3K)/Akt signaling molecules PI3Kp110β, PI3Kp110γ, Akt, and mTOR. Inhibitors (adenosine dialdehyde and S-adenosylhomocysteine) of protein methylation suppressed bFGF-mediated neuronal differentiation of PC12 cells. PIMT-eficiency caused by PIMT-specific siRNA inhibited neuronal differentiation of PC12 cells by suppressing phosphorylation of MEK1/2 and ERK1/2 in the MAPK signaling pathway and Akt and mTOR in the PI3K/Akt signaling pathway. Therefore, these results suggested that PIMT was critical for bFGF-mediated neuronal differentiation of PC12 cells and regulated the MAPK and Akt signaling pathways. [BMB Reports 2016; 49(8): 437-442]
Collapse
Affiliation(s)
- To Thi Mai Dung
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Young-Su Yi
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419; Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28503, Korea
| | - Jieun Heo
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Han Gyung Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Jae Gwang Park
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Sungyoul Hong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
4
|
Iinuma M, Umehara T, Arimitsu N, Shimizu J, Misawa H, Takai K, Fujiwara N, Fujii A, Ueda Y, Wakisaka S, Suzuki T, Hirotsu C, Beppu M, Suzuki N. Induction of neural cells with spinal motoneuron phenotype from human iPS cells and the transplantation to totally transected spinal cords in mice. Inflamm Regen 2015. [DOI: 10.2492/inflammregen.35.154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Masahiro Iinuma
- Department of Orthopedics, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Tasuku Umehara
- Department of Orthopedics, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Nagisa Arimitsu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Jun Shimizu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Hiroko Misawa
- Department of Orthopedics, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Kenji Takai
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Naruyoshi Fujiwara
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Atsushi Fujii
- Department of Orthopedics, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Yuji Ueda
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Sueshige Wakisaka
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Tomoko Suzuki
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Chieko Hirotsu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Moroe Beppu
- Department of Orthopedics, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
| | - Noboru Suzuki
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawaksaki, Kanagawa, Japan
- Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawaksaki, Kanagawa, Japan
| |
Collapse
|
5
|
Nakano R, Edamura K, Nakayama T, Teshima K, Asano K, Narita T, Okabayashi K, Sugiya H. Differentiation of canine bone marrow stromal cells into voltage- and glutamate-responsive neuron-like cells by basic fibroblast growth factor. J Vet Med Sci 2014; 77:27-35. [PMID: 25284120 PMCID: PMC4349535 DOI: 10.1292/jvms.14-0284] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We investigated the in vitro differentiation of canine bone marrow stromal cells (BMSCs) into voltage- and glutamate-responsive neuron-like cells. BMSCs were obtained from the bone marrow of healthy beagle dogs. Canine BMSCs were incubated with the basal medium for neurons containing recombinant human basic fibroblast growth factor (bFGF; 100 ng/ml). The viability of the bFGF-treated cells was assessed by a trypan blue exclusion assay, and the morphology was monitored. Real-time RT-PCR was performed to evaluate mRNA expression of neuronal, neural stem cell and glial markers. Western blotting and immunocytochemical analysis for the neuronal markers were performed to evaluate the protein expression and localization. The Ca(2+) mobilization of the cells was evaluated using the Ca(2+) indicator Fluo3 to monitor Ca(2+) influx. To investigate the mechanism of bFGF-induced neuronal differentiation, the fibroblast growth factor receptor inhibitor, the phosphoinositide 3-kinase inhibitor or the Akt inhibitor was tested. The bFGF treatment resulted in the maintenance of the viability of canine BMSCs for 10 days, in the expression of neuronal marker mRNAs and proteins and in the manifestation of neuron-like morphology. Furthermore, in the bFGF-treated BMSCs, a high concentration of KCl and L-glutamate induced an increase in intracellular Ca(2+) levels. Each inhibitor significantly attenuated the bFGF-induced increase in neuronal marker mRNA expression. These results suggest that bFGF contributes to the differentiation of canine BMSCs into voltage- and glutamate-responsive neuron-like cells and may lead to the development of new cell-based treatments for neuronal diseases.
Collapse
Affiliation(s)
- Rei Nakano
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | | | | | | | | | | | | | | |
Collapse
|
6
|
From pluripotency to forebrain patterning: an in vitro journey astride embryonic stem cells. Cell Mol Life Sci 2014; 71:2917-30. [PMID: 24643740 PMCID: PMC4098049 DOI: 10.1007/s00018-014-1596-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/17/2014] [Accepted: 02/26/2014] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) have been used extensively as in vitro models of neural development and disease, with special efforts towards their conversion into forebrain progenitors and neurons. The forebrain is the most complex brain region, giving rise to several fundamental structures, such as the cerebral cortex, the hypothalamus, and the retina. Due to the multiplicity of signaling pathways playing different roles at distinct times of embryonic development, the specification and patterning of forebrain has been difficult to study in vivo. Research performed on ESCs in vitro has provided a large body of evidence to complement work in model organisms, but these studies have often been focused more on cell type production than on cell fate regulation. In this review, we systematically reassess the current literature in the field of forebrain development in mouse and human ESCs with a focus on the molecular mechanisms of early cell fate decisions, taking into consideration the specific culture conditions, exogenous and endogenous molecular cues as described in the original studies. The resulting model of early forebrain induction and patterning provides a useful framework for further studies aimed at reconstructing forebrain development in vitro for basic research or therapy.
Collapse
|
7
|
Feng N, Han Q, Li J, Wang S, Li H, Yao X, Zhao RC. Generation of highly purified neural stem cells from human adipose-derived mesenchymal stem cells by Sox1 activation. Stem Cells Dev 2014; 23:515-29. [PMID: 24138016 DOI: 10.1089/scd.2013.0263] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells (NSCs) are ideal candidates in stem cell-based therapy for neurodegenerative diseases. However, it is unfeasible to get enough quantity of NSCs for clinical application. Generation of NSCs from human adipose-derived mesenchymal stem cells (hAD-MSCs) will provide a solution to this problem. Currently, the differentiation of hAD-MSCs into highly purified NSCs with biological functions is rarely reported. In our study, we established a three-step NSC-inducing protocol, in which hAD-MSCs were induced to generate NSCs with high purity after sequentially cultured in the pre-inducing medium (Step1), the N2B27 medium (Step2), and the N2B27 medium supplement with basic fibroblast growth factor and epidermal growth factor (Step3). These hAD-MSC-derived NSCs (adNSCs) can form neurospheres and highly express Sox1, Pax6, Nestin, and Vimentin; the proportion was 96.1% ± 1.3%, 96.8% ± 1.7%, 96.2% ± 1.3%, and 97.2% ± 2.5%, respectively, as detected by flow cytometry. These adNSCs can further differentiate into astrocytes, oligodendrocytes, and functional neurons, which were able to generate tetrodotoxin-sensitive sodium current. Additionally, we found that the neural differentiation of hAD-MSCs were significantly suppressed by Sox1 interference, and what's more, Step1 was a key step for the following induction, probably because it was associated with the initiation and nuclear translocation of Sox1, an important transcriptional factor for neural development. Finally, we observed that bone morphogenetic protein signal was inhibited, and Wnt/β-catenin signal was activated during inducing process, and both signals were related with Sox1 expression. In conclusion, we successfully established a three-step inducing protocol to derive NSCs from hAD-MSCs with high purity by Sox1 activation. These findings might enable to acquire enough autologous transplantable NSCs for the therapy of neurodegenerative diseases in clinic.
Collapse
Affiliation(s)
- Nianhua Feng
- 1 Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences and School of Basic Medicine , Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
8
|
Fujiwara N, Shimizu J, Takai K, Arimitsu N, Saito A, Kono T, Umehara T, Ueda Y, Wakisaka S, Suzuki T, Suzuki N. Restoration of spatial memory dysfunction of human APP transgenic mice by transplantation of neuronal precursors derived from human iPS cells. Neurosci Lett 2013. [DOI: 10.1016/j.neulet.2013.10.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
9
|
Abstract
The potential of cell therapy is promising in nerve regeneration, but is limited by ethical considerations about the proper and technically safe source of stem cells. We report the successful differentiation of human EnSCs (endometrial stem cells) as a rich source of renewable and safe progenitors into high-efficiency cholinergic neurons. The extracellular signals of NGF (nerve growth factor) and bFGF (basic fibroblast growth factor) could induce cholinergic neuron differentiation. ChAT (choline acetyltransferase), MAP2 (microtubule associated protein 2) and NF-l (neurofilament L) increased after administration of bFGF and NGF to the EnSC cultures. trkC and FGFR2 (fibroblast growth factor receptor 2), which belong to the NGF and bFGF receptors respectively, were determined in populations of EnSCs. NGF, bFGF and their combination differentially influenced human EnSCs high efficiency differentiation. By inducing cholinergic neurons from EnSCs in a chemically defined medium, we could produce human neural cells without resorting to primary culture of neurons. This in vitro method provides an unlimited source of human neural cells and facilitates clinical applications of EnSCs for neurological diseases.
Collapse
|
10
|
The positional identity of mouse ES cell-generated neurons is affected by BMP signaling. Cell Mol Life Sci 2012; 70:1095-111. [PMID: 23069989 PMCID: PMC3578729 DOI: 10.1007/s00018-012-1182-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/24/2012] [Accepted: 09/25/2012] [Indexed: 01/10/2023]
Abstract
We investigated the effects of bone morphogenetic proteins (BMPs) in determining the positional identity of neurons generated in vitro from mouse embryonic stem cells (ESCs), an aspect that has been neglected thus far. Classical embryological studies in lower vertebrates indicate that BMPs inhibit the default fate of pluripotent embryonic cells, which is both neural and anterior. Moreover, mammalian ESCs generate neurons more efficiently when cultured in a minimal medium containing BMP inhibitors. In this paper, we show that mouse ESCs produce, secrete, and respond to BMPs during in vitro neural differentiation. After neuralization in a minimal medium, differentiated ESCs show a gene expression profile consistent with a midbrain identity, as evaluated by the analysis of a number of markers of anterior-posterior and dorsoventral identity. We found that BMPs endogenously produced during neural differentiation mainly act by inhibiting the expression of a telencephalic gene profile, which was revealed by the treatment with Noggin or with other BMP inhibitors. To better characterize the effect of BMPs on positional fate, we compared the global gene expression profiles of differentiated ESCs with those of embryonic forebrain, midbrain, and hindbrain. Both Noggin and retinoic acid (RA) support neuronal differentiation of ESCs, but they show different effects on their positional identity: whereas RA supports the typical gene expression profile of hindbrain neurons, Noggin induces a profile characteristic of dorsal telencephalic neurons. Our findings show that endogenously produced BMPs affect the positional identity of the neurons that ESCs spontaneously generate when differentiating in vitro in a minimal medium. The data also support the existence of an intrinsic program of neuronal differentiation with dorsal telencephalic identity. Our method of ESC neuralization allows for fast differentiation of neural cells via the same signals found during in vivo embryonic development and for the acquisition of cortical identity by the inhibition of BMP alone.
Collapse
|
11
|
Jung GA, Shin BS, Jang YS, Sohn JB, Woo SR, Kim JE, Choi G, Lee KM, Min BH, Lee KH, Park GH. Methylation of eukaryotic elongation factor 2 induced by basic fibroblast growth factor via mitogen-activated protein kinase. Exp Mol Med 2012; 43:550-60. [PMID: 21778808 DOI: 10.3858/emm.2011.43.10.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Protein arginine methylation is important for a variety of cellular processes including transcriptional regulation, mRNA splicing, DNA repair, nuclear/cytoplasmic shuttling and various signal transduction pathways. However, the role of arginine methylation in protein biosynthesis and the extracellular signals that control arginine methylation are not fully understood. Basic fibroblast growth factor (bFGF) has been identified as a potent stimulator of myofibroblast dedifferentiation into fibroblasts. We demonstrated that symmetric arginine dimethylation of eukaryotic elongation factor 2 (eEF2) is induced by bFGF without the change in the expression level of eEF2 in mouse embryo fibroblast NIH3T3 cells. The eEF2 methylation is preceded by ras-raf-mitogen-activated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK1/2)- p21Cip/WAF1 activation, and suppressed by the mitogenactivated protein kinase (MAPK) inhibitor PD98059 and p21Cip/WAF1 short interfering RNA (siRNA). We determined that protein arginine methyltransferase 7 (PRMT7) is responsible for the methylation, and that PRMT5 acts as a coordinator. Collectively, we demonstrated that eEF2, a key factor involved in protein translational elongation is symmetrically arginine-methylated in a reversible manner, being regulated by bFGF through MAPK signaling pathway.
Collapse
Affiliation(s)
- Gyung Ah Jung
- Department of Biochemistry, BK 21 Program, College of Medicine, Korea University, Seoul
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Bonilla AQS, Oliveira LJ, Ozawa M, Newsom EM, Lucy MC, Hansen PJ. Developmental changes in thermoprotective actions of insulin-like growth factor-1 on the preimplantation bovine embryo. Mol Cell Endocrinol 2011; 332:170-9. [PMID: 20965229 DOI: 10.1016/j.mce.2010.10.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/06/2010] [Accepted: 10/12/2010] [Indexed: 11/15/2022]
Abstract
Insulin-like growth factor 1 (IGF1) is an important endocrine signal for regulation of early embryonic development. It increases the proportion of preimplantation embryos becoming blastocysts, alters blastocyst gene expression, improves resistance of embryos to various stresses and can enhance survival of embryos after transfer to recipients. The present study had two objectives. The first was to determine whether the thermoprotective actions of IGF1 on the preimplantation bovine embryo was developmentally regulated, with the two-cell embryo being refractory to IGF1. The second was to determine the molecular basis for the improved competence of embryos treated with IGF1 to establish pregnancy after transfer to heat-stressed recipients. Treatment of embryos with 100 ng/ml IGF1 reduced the effects of heat shock on embryos ≥16 cells at day 5 after insemination but did not provide thermoprotection to two-cell embryos. Failure of IGF1 to alter embryo survival after heat shock was not associated with reduced expression of genes involved in IGF1 signaling (IGF1R, RAF1, PI3K, and MAPK) or immunoreactive IGF1R protein. Treatment with IGF1 had little effect on the transcriptome at the blastocyst stage of development, with a total of 102 differentially expressed genes identified. Among the differentially expressed genes were several involved in apoptosis, protection against free radicals and development. Changes in gene expression were consistent with IGF1 acting to induce an anti-apoptotic state and inhibit neurulation. In conclusion, thermoprotective actions of IGF1 are developmentally regulated. Failure of IGF1 to protect the two-cell embryo from heat shock could reflect the fact that these embryos are maximally sensitive to damage caused by heat shock or reflect the quiescence of the embryonic genome at this stage of development. Changes in gene expression at the blastocyst stage induced by IGF1 could contribute to the increased survival of IGF1-treated embryos when transferred during periods of heat stress.
Collapse
Affiliation(s)
- A Q S Bonilla
- Department of Animal Sciences and DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL 32611-0910, USA
| | | | | | | | | | | |
Collapse
|
13
|
Young A, Assey KS, Sturkie CD, West FD, Machacek DW, Stice SL. Glial cell line-derived neurotrophic factor enhances in vitro differentiation of mid-/hindbrain neural progenitor cells to dopaminergic-like neurons. J Neurosci Res 2010; 88:3222-32. [DOI: 10.1002/jnr.22499] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
14
|
Wada T, Honda M, Minami I, Tooi N, Amagai Y, Nakatsuji N, Aiba K. Highly efficient differentiation and enrichment of spinal motor neurons derived from human and monkey embryonic stem cells. PLoS One 2009; 4:e6722. [PMID: 19701462 PMCID: PMC2726947 DOI: 10.1371/journal.pone.0006722] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 07/25/2009] [Indexed: 01/21/2023] Open
Abstract
Background There are no cures or efficacious treatments for severe motor neuron diseases. It is extremely difficult to obtain naïve spinal motor neurons (sMNs) from human tissues for research due to both technical and ethical reasons. Human embryonic stem cells (hESCs) are alternative sources. Several methods for MN differentiation have been reported. However, efficient production of naïve sMNs and culture cost were not taken into consideration in most of the methods. Methods/Principal Findings We aimed to establish protocols for efficient production and enrichment of sMNs derived from pluripotent stem cells. Nestin+ neural stem cell (NSC) clusters were induced by Noggin or a small molecule inhibitor of BMP signaling. After dissociation of NSC clusters, neurospheres were formed in a floating culture containing FGF2. The number of NSCs in neurospheres could be expanded more than 30-fold via several passages. More than 33% of HB9+ sMN progenitor cells were observed after differentiation of dissociated neurospheres by all-trans retinoic acid (ATRA) and a Shh agonist for another week on monolayer culture. HB9+ sMN progenitor cells were enriched by gradient centrifugation up to 80% purity. These HB9+ cells differentiated into electrophysiologically functional cells and formed synapses with myotubes during a few weeks after ATRA/SAG treatment. Conclusions and Significance The series of procedures we established here, namely neural induction, NSC expansion, sMN differentiation and sMN purification, can provide large quantities of naïve sMNs derived from human and monkey pluripotent stem cells. Using small molecule reagents, reduction of culture cost could be achieved.
Collapse
Affiliation(s)
- Tamaki Wada
- Stem Cell and Drug Discovery Institute (SCDI), Shimogyo-ku, Kyoto, Japan
- * E-mail: (TW); (KA)
| | - Makoto Honda
- Stem Cell and Drug Discovery Institute (SCDI), Shimogyo-ku, Kyoto, Japan
| | - Itsunari Minami
- Stem Cell and Drug Discovery Institute (SCDI), Shimogyo-ku, Kyoto, Japan
| | - Norie Tooi
- Stem Cell and Drug Discovery Institute (SCDI), Shimogyo-ku, Kyoto, Japan
| | - Yuji Amagai
- Stem Cell and Drug Discovery Institute (SCDI), Shimogyo-ku, Kyoto, Japan
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo-ku, Kyoto, Japan
- Institute for Frontier Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Kazuhiro Aiba
- Stem Cell and Drug Discovery Institute (SCDI), Shimogyo-ku, Kyoto, Japan
- * E-mail: (TW); (KA)
| |
Collapse
|
15
|
Smith J, Wardle F, Loose M, Stanley E, Patient R. Germ layer induction in ESC--following the vertebrate roadmap. ACTA ACUST UNITED AC 2008; Chapter 1:Unit 1D.1. [PMID: 18785165 DOI: 10.1002/9780470151808.sc01d01s1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Controlled differentiation of pluripotential cells takes place routinely and with great success in developing vertebrate embryos. It therefore makes sense to take note of how this is achieved and use this knowledge to control the differentiation of embryonic stem cells (ESCs). An added advantage is that the differentiated cells resulting from this process in embryos have proven functionality and longevity. This unit reviews what is known about the embryonic signals that drive differentiation in one of the most informative of the vertebrate animal models of development, the amphibian Xenopus laevis. It summarizes their identities and the extent to which their activities are dose-dependent. The unit details what is known about the transcription factor responses to these signals, describing the networks of interactions that they generate. It then discusses the target genes of these transcription factors, the effectors of the differentiated state. Finally, how these same developmental programs operate during germ layer formation in the context of ESC differentiation is summarized.
Collapse
Affiliation(s)
- Jim Smith
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
16
|
Chiba S, Lee YM, Zhou W, Freed CR. Noggin enhances dopamine neuron production from human embryonic stem cells and improves behavioral outcome after transplantation into Parkinsonian rats. Stem Cells 2008; 26:2810-20. [PMID: 18772316 DOI: 10.1634/stemcells.2008-0085] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Symptoms of Parkinson's disease have been improved by transplantation of fetal dopamine neurons recovered from aborted fetal tissue, but tissue recovery is difficult. Human embryonic stem cells may provide unlimited cells for transplantation if they can be converted to dopamine neurons and survive transplantation into brain. We have found that the bone morphogenic protein antagonist Noggin increased the number of dopamine neurons generated in vitro from human and mouse embryonic stem cells differentiated on mouse PA6 stromal cells. Noggin effects were seen with either early (for mouse, days 0-7, and for human, days 0-9) or continuous treatment. After transplant into cyclosporin-immunosuppressed rats, human dopamine neurons improved apomorphine circling in direct relation to the number of surviving dopamine neurons, which was fivefold greater after Noggin treatment than with control human embryonic stem cell transplants differentiated only on PA6 cells. We conclude that Noggin promotes dopamine neuron differentiation and survival from human and mouse embryonic stem cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Shunmei Chiba
- Department of Medicine, Division of Clinical Pharmacology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | |
Collapse
|
17
|
Blomberg LA, Schreier LL, Talbot NC. Expression analysis of pluripotency factors in the undifferentiated porcine inner cell mass and epiblast during in vitro culture. Mol Reprod Dev 2008; 75:450-63. [PMID: 17680630 DOI: 10.1002/mrd.20780] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Limited understanding of the importance of known pluripotency factors in pig embryonic stem cells (ESC) impedes the establishment and validation of porcine ESC lines. This study evaluated the expression of known mouse ESC and human ESC (hESC) pluripotency markers in in vivo inner cell mass (ICM) and in vitro-cultured undifferentiated porcine epiblast cells isolated from 8-day porcine blastocysts, primary cultures of epiblast-derived neuroprogenitor cells, and endoderm cells. The expression profile of common pluripotency markers (POU domain 5 transcript factor 1, SRY-box containing gene 2, and Nanog homeobox), species-specific markers, ESC-associated factors, and differentiation markers was evaluated. The mRNA of uncultured ICMs, cultured epiblast cells, epiblast-derived neuroprogenitor cells, and endoderm cells was amplified prior to expression analysis of candidate genes by real-time RT-PCR. ESC factors whose expression correlated best with the undifferentiated epiblast state were identified by comparative mRNA expression analysis between porcine epiblast-derived somatic cell lines, fetal fibroblasts, and adult tissues. Across tissue types Nanog homeobox exhibited ubiquitous expression, whereas POU domain 5 transcript factor 1, teratocarcinoma-derived growth factor 1, and RNA exonuclease homolog 1 transcript expression was restricted primarily to undifferentiated epiblasts. Our results suggested that expression of pluripotency markers in undifferentiated pig epiblast cells more closely resembled that observed in hESC. Expression alterations of ESC-associated factors in epiblast cells were also observed during in vitro culture. Our data demonstrate the potential use of some pluripotency factors as markers of porcine epiblast stem cells and indicate that the in vitro environment may influence the cultured epiblast's developmental state.
Collapse
Affiliation(s)
- Le Ann Blomberg
- Biotechnology and Germplasm Laboratory, USDA Agricultural Research Service, Beltsville, Maryland 20705, USA.
| | | | | |
Collapse
|
18
|
Villanueva S, Cespedes C, Gonzalez AA, Roessler E, Vio CP. Inhibition of bFGF-receptor type 2 increases kidney damage and suppresses nephrogenic protein expression after ischemic acute renal failure. Am J Physiol Regul Integr Comp Physiol 2008; 294:R819-28. [PMID: 18184769 DOI: 10.1152/ajpregu.00273.2007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recovery from acute renal failure (ARF) requires the replacement of injured cells by new cells that are able to restore tubule epithelial integrity. We have recently described the expression of nephrogenic proteins [Vimentin, neural cell adhesion molecule, basic fibroblast growth factor (bFGF), Pax-2, bone morphogen protein-7, Noggin, Smad 1-5-8, p-Smad, hypoxia-inducible factor-1alpha, vascular endothelial growth factor], in a time frame similar to that observed in kidney development, after ischemic ARF induced in an ischemia-reperfusion (I/R) model. Furthermore, we show that bFGF, a morphogen involved in mesenchyme/epithelial transition in kidney development, induces a reexpression of morphogenic proteins in an earlier time frame and accelerates the recovery process after renal damage. Herein, we confirm that renal morphogenes are modulated by bFGF and hypothesized that a decrease in bFGF receptor 2 (bFGFR2) levels by the use of antisense oligonucleotides diminishes the expression of morphogenes. Male Sprague-Dawley rats submitted to ischemic injury were injected with 112 microg/kg bFGFR2 antisense oligonucleotide (bFGFR2-ASO) followed by reperfusion. Rats were killed, and the expression of nephrogenic proteins and renal marker damage was analyzed by immunohistochemistry and immunoblot. Animals subjected to I/R treated with bFGFR2-ASO showed a significant reduction in morphogen levels (P < 0.05). In addition, we observed an increase in markers of renal damage: macrophages (ED-1) and interstitial alpha-smooth muscle actin. These results confirm that bFGF participates in the recovery process and that treatment with bFGFR2-ASO induces an altered expression of morphogen proteins.
Collapse
Affiliation(s)
- Sandra Villanueva
- Laboratorio de Fisiologia Integrativa y Molecular, Universidad de Los Andes, San Carlos Apoquindo 2200, Santiago, Chile.
| | | | | | | | | |
Collapse
|
19
|
Yang H, Xia Y, Lu SQ, Soong TW, Feng ZW. Basic fibroblast growth factor-induced neuronal differentiation of mouse bone marrow stromal cells requires FGFR-1, MAPK/ERK, and transcription factor AP-1. J Biol Chem 2008; 283:5287-95. [PMID: 18171671 DOI: 10.1074/jbc.m706917200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It has been reported recently that bone marrow stromal cells (BMSCs) are able to differentiate into various neural cells both in vivo and in vitro (Egusa, H., Schweizer, F. E., Wang, C. C., Matsuka, Y., and Nishimura, I. (2005) J. Biol. Chem. 280, 23691-23697). However, the underlying mechanisms remain largely unknown. In this report, we have demonstrated that basic fibroblast growth factor (bFGF) alone effectively induces mouse BMSC neuronal differentiation. These differentiated neuronal cells exhibit characteristic electrophysiological properties and elevated levels of the neuronal differentiation marker, growth-associated protein-43 (GAP-43). To explore possible signaling pathways, we first analyzed the expression of various FGF receptors in mouse BMSCs. FGF receptor-1, -2, and -3 were detected, but only FGFR-1 was shown to be activated by bFGF. Small interfering RNA knock down of FGFR-1 in BMSCs significantly inhibited neuronal differentiation. Moreover, we have shown that the mitogen-activated protein kinase (ERK1/2) is persistently activated and blockage of ERK activity with the ERK-specific inhibitor U0126 prevents neuronal differentiation. It appears that activation of ERK cascade and neuronal differentiation of BMSCs induced by bFGF are independent of Ras activity but require functions of phospholipase C-gamma pathway. Lastly, we examined the role of the immediate-early transcription factors AP-1 and NF-kappaB and have found that phospholipase C-gamma-dependent c-Jun and ERK-dependent c-fos, but not the NF-kappaB, are strongly activated by bFGF, which in turn regulates the neuronal differentiation of BMSCs.
Collapse
Affiliation(s)
- Haijie Yang
- Research Laboratories, National Neuroscience Institute, Singapore 308433
| | | | | | | | | |
Collapse
|
20
|
Atoh KI, Kurokawa MS, Yoshikawa H, Masuda C, Takada E, Kumagai N, Suzuki N. Induction of melanocyte precursors from neural crest cells surrounding the neural tube-like structures developed in vitro using mouse ES cell culture. Inflamm Regen 2007. [DOI: 10.2492/inflammregen.27.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
21
|
Villanueva S, Cespedes C, Gonzalez A, Vio CP. bFGF induces an earlier expression of nephrogenic proteins after ischemic acute renal failure. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1677-87. [PMID: 16873559 DOI: 10.1152/ajpregu.00023.2006] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recovery from acute renal failure (ARF) requires the replacement of injured cells with new cells that restore tubule epithelial integrity. We described recently the expression of a wide range of nephrogenic proteins in tubular cells after ARF induced by ischemia-reperfusion (I/R) (Villanueva S, Cespedes C, and Vio CP. Am J Physiol Regul Integr Comp Physiol 290: R861-R870, 2006). These markers, namely, Vimentin, neural cell adhesion molecules (Ncam), basic fibroblast growth factor (bFGF), paired homeobox-2 (Pax-2), bone morphogene protein-7 (BMP-7), Noggin, Lim-1, Engrailed, Smad, phospho-Smad, hypoxia-induced factor-1alpha (HIF-1alpha), VEGF, and Tie-2, are expressed in a time frame similar to that observed in normal kidney development. bFGF participates in early kidney development as a morphogen involved in mesenchyme/epithelial transition, and it is reexpressed in the recovery phase of ARF. To test the hypothesis that bFGF can accelerate the regeneration after renal damage, we used recombinant bFGF and studied the expression pattern of the above described morphogens in ARF. Male Sprague-Dawley rats were subjected to 30 min of renal ischemic injury and were injected with bFGF 30 microg/kg followed by reperfusion. Rats were killed and the expression of nephrogenic proteins were analyzed by immunohistochemistry and Western blot analysis. In the animals subjected to I/R treated with bFGF, we observed a 12- to 24-h earlier and more abundant reexpression of the proteins Ncam, bFGF, Pax-2, BMP-7, Noggin, Lim-1, Engrailed, VEGF, and Tie-2 than the I/R untreated rats. In addition, we observed a reduction in renal damage markers ED-1 and alpha-smooth muscle actin. These results indicate that bFGF can participate in the regeneration process and suggest that the treatment with bFGF can induce an earlier regeneration process after ischemic acute renal failure.
Collapse
Affiliation(s)
- Sandra Villanueva
- Dept. de Fisiologia, Pontificia Universidad Catolica de Chile, Casilla 114-D, Santiago, Chile.
| | | | | | | |
Collapse
|