1
|
St-Cyr G, Garneau D, Gévry N, Blouin R. Quantitative phosphoproteomics reveals that nestin is a downstream target of dual leucine zipper kinase during retinoic acid-induced neuronal differentiation of Neuro-2a cells. BMC Mol Cell Biol 2025; 26:10. [PMID: 40140778 PMCID: PMC11938613 DOI: 10.1186/s12860-025-00535-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Dual leucine zipper kinase (DLK) is critical for neurite outgrowth in the developing nervous system and during nerve regeneration, but the underlying mechanisms remain largely unknown. To address this issue, we generated stable shRNA-mediated DLK-depleted Neuro-2a cell lines and analyzed their phosphoproteome after induction of neuronal differentiation by retinoic acid (RA). RESULTS Here, we report the identification of 32 phosphopeptides that exhibited significant differences in relative abundance between control and DLK-depleted cells. Two of the most downregulated phosphopeptides identified after DLK depletion were derived from nestin, a type VI intermediate filament (IF) protein typically expressed in neural progenitor cells. The reduced abundance of these phosphopeptides in response to DLK knockdown was validated using parallel reaction monitoring (PRM)-based quantitative proteomics and paired with a concomitant reduction in nestin mRNA and protein expression, indicating that the decrease in nestin phosphorylation was due to a decrease in total nestin in DLK-depleted cells compared to control cells. This DLK-mediated regulation of nestin expression had no apparent effect on neurite formation because nestin knockdown alone was not sufficient to impair RA-induced neurite extension in parental Neuro-2a cells, and nestin overexpression failed to rescue the neurite outgrowth defect observed in DLK-depleted Neuro-2a cells. CONCLUSIONS Together, these results demonstrate that nestin is a novel downstream target of DLK signaling but not a mediator of its ability to promote neurite outgrowth during neuronal differentiation.
Collapse
Affiliation(s)
- Guillaume St-Cyr
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Daniel Garneau
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Nicolas Gévry
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Richard Blouin
- Département de biologie, Faculté des sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
2
|
Tong Z, Yin Z. Distribution, contribution and regulation of nestin + cells. J Adv Res 2024; 61:47-63. [PMID: 37648021 PMCID: PMC11258671 DOI: 10.1016/j.jare.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Nestin is an intermediate filament first reported in neuroepithelial stem cells. Nestin expression could be found in a variety of tissues throughout all systems of the body, especially during tissue development and tissue regeneration processes. AIM OF REVIEW This review aimed to summarize and discuss current studies on the distribution, contribution and regulation of nestin+ cells in different systems of the body, to discuss the feasibility ofusing nestin as a marker of multilineage stem/progenitor cells, and better understand the potential roles of nestin+ cells in tissue development, regeneration and pathological processes. KEY SCIENTIFIC CONCEPTS OF REVIEW This review highlights the potential of nestin as a marker of multilineage stem/progenitor cells, and as a key factor in tissue development and tissue regeneration. The article discussed the current findings, limitations, and potential clinical implications or applications of nestin+ cells. Additionally, it included the relationship of nestin+ cells to other cell populations. We propose potential future research directions to encourage further investigation in the field.
Collapse
Affiliation(s)
- Ziyang Tong
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zi Yin
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
3
|
Chen H, Cai J, Wang J, Qiu Y, Jiang C, Wang Y, Wang Y, Yi C, Guo Lv, Pan L, Guan Y, Zheng J, Qiu D, Du C, Liu Q, Chen G, Yang Y, Xu Y, Xiang AP, Zhang Q. Targeting Nestin + hepatic stellate cells ameliorates liver fibrosis by facilitating TβRI degradation. J Hepatol 2021; 74:1176-1187. [PMID: 33217494 DOI: 10.1016/j.jhep.2020.11.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Liver fibrosis is a wound healing response that arises from various aetiologies. The intermediate filament protein Nestin has been reported to participate in maintaining tissue homeostasis during wound healing responses. However, little is known about the role Nestin plays in liver fibrosis. This study investigated the function and precise regulatory network of Nestin during liver fibrosis. METHODS Nestin expression was assessed via immunostaining and quantitative real-time PCR (qPCR) in fibrotic/cirrhotic samples. The induction of Nestin expression by transforming growth factor beta (TGFβ)-Smad2/3 signalling was investigated through luciferase reporter assays. The functional role of Nestin in hepatic stellate cells (HSCs) was investigated by examining the pathway activity of profibrogenic TGFβ-Smad2/3 signalling and degradation of TGFβ receptor I (TβRI) after interfering with Nestin. The in vivo effects of knocking down Nestin were examined with an adeno-associated virus vector (serotype 6, AAV6) carrying short-hairpin RNA targeting Nestin in fibrotic mouse models. RESULTS Nestin was mainly expressed in activated HSCs and increased with the progression of liver fibrosis. The profibrogenic pathway TGFβ-Smad2/3 induced Nestin expression directly. Knocking down Nestin promoted caveolin 1-mediated TβRI degradation, resulting in TGFβ-Smad2/3 pathway impairment and reduced fibrosis marker expression in HSCs. In AAV6-treated murine fibrotic models, knocking down Nestin resulted in decreased levels of inflammatory infiltration, hepatocellular damage, and a reduced degree of fibrosis. CONCLUSION The expression of Nestin in HSCs was induced by TGFβ and positively correlated with the degree of liver fibrosis. Knockdown of Nestin decreased activation of the TGFβ pathway and alleviated liver fibrosis both in vitro and in vivo. Our data demonstrate a novel role of Nestin in controlling HSC activation in liver fibrosis. LAY SUMMARY Liver fibrosis has various aetiologies but represents a common process in chronic liver diseases that is associated with high morbidity and mortality. Herein, we demonstrate that the intermediate filament protein Nestin plays an essential profibrogenic role in liver fibrosis by forming a positive feedback loop with the TGFβ-Smad2/3 pathway, providing a potential therapeutic target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Huaxin Chen
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianye Cai
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China; Department of Hepatic Surgery and Liver Transplantation Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiancheng Wang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China; Scientific Research Centre, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuan Qiu
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Chenhao Jiang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Yi Wang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Yiqin Wang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Chenju Yi
- Scientific Research Centre, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Guo Lv
- Department of Hepatic Surgery and Liver Transplantation Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lijie Pan
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanjun Guan
- Core Facility Centre, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dongbo Qiu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cong Du
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiuli Liu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guihua Chen
- Department of Hepatic Surgery and Liver Transplantation Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yan Xu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Andy Peng Xiang
- Centre for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Qi Zhang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
4
|
Carter JL, Halmai JANM, Fink KD. The iNs and Outs of Direct Reprogramming to Induced Neurons. Front Genome Ed 2020; 2:7. [PMID: 34713216 PMCID: PMC8525349 DOI: 10.3389/fgeed.2020.00007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Understanding of cell-type specific transcription factors has promoted progress in methods for cellular reprogramming, such as directly reprogramming somatic cells to induced neurons (iN). Methods for direct reprogramming require neuronal-fate determining gene activation via neuron-specific microRNAs, chemical modulation of key neuronal signaling pathways or overexpression via viral vectors, with some reprogramming strategies requiring a combination of these methods to induce the neuronal-cell fate. These methods have been employed in a multitude of cell types, including fibroblasts, hepatocytes, peripheral blood mononuclear, and T cells. The ability to create iN from skin biopsies and blood samples coupled with recent advancements in artificially inducing age- and disease-associated phenotypes are accelerating the development of disease models for late-onset neurodegenerative disorders. Here, we review how activation of the neuronal transcriptome alters the epigenetic landscape of the donor cell to facilitate reprogramming to neurons. We also discuss the advantages of using DNA binding domains such as CRISPR/dCas9 to overcome epigenetic barriers to induce neuronal-cell fate by activating endogenous neuronal cell-fate determining genes.
Collapse
|
5
|
Velychko S, Kang K, Kim SM, Kwak TH, Kim KP, Park C, Hong K, Chung C, Hyun JK, MacCarthy CM, Wu G, Schöler HR, Han DW. Fusion of Reprogramming Factors Alters the Trajectory of Somatic Lineage Conversion. Cell Rep 2020; 27:30-39.e4. [PMID: 30943410 DOI: 10.1016/j.celrep.2019.03.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/23/2019] [Accepted: 03/07/2019] [Indexed: 01/28/2023] Open
Abstract
Simultaneous expression of Oct4, Klf4, Sox2, and cMyc induces pluripotency in somatic cells (iPSCs). Replacing Oct4 with the neuro-specific factor Brn4 leads to transdifferentiation of fibroblasts into induced neural stem cells (iNSCs). However, Brn4 was recently found to induce transient acquisition of pluripotency before establishing the neural fate. We employed genetic lineage tracing and found that induction of iNSCs with individual vectors leads to direct lineage conversion. In contrast, polycistronic expression produces a Brn4-Klf4 fusion protein that enables induction of pluripotency. Our study demonstrates that a combination of pluripotency and tissue-specific factors allows direct somatic cell transdifferentiation, bypassing the acquisition of a pluripotent state. This result has major implications for lineage conversion technologies, which hold potential for providing a safer alternative to iPSCs for clinical application both in vitro and in vivo.
Collapse
Affiliation(s)
- Sergiy Velychko
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Kyuree Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sung Min Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Tae Hwan Kwak
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Chanhyeok Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Jung Keun Hyun
- Department of Nanobiomedical Science, Dankook University, Cheonan 330714, Republic of Korea
| | - Caitlin M MacCarthy
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Guangming Wu
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Dong Wook Han
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; KU Open-Innovation Center, Institute of Biomedical Science and Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
6
|
Fukui M, Katayama S, Ikeya Y, Inazu T. Yokukansan, a Kampo medicine, enhances the level of neuronal lineage markers in differentiated P19 embryonic carcinoma cells. Heliyon 2019; 5:e02662. [PMID: 31692643 PMCID: PMC6806406 DOI: 10.1016/j.heliyon.2019.e02662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/11/2019] [Accepted: 10/11/2019] [Indexed: 02/01/2023] Open
Abstract
Yokukansan (YKS), a traditional Japanese Kampo medicine, affects neurological and psychiatric disorders. It ameliorates hippocampal neurogenesis in animals. However, its effect on neuronal cell differentiation remains unclear. Therefore, we investigated the effects of YKS on pluripotent P19 embryonic carcinoma cells as neuronal differentiation model cells. Western blotting and immunocytochemistry revealed that 10 μg/mL YKS treatment during embryoid body formation or neuronal differentiation increased the expression of the neuronal stem cell marker, Nestin, by 1.9-fold and 1.7-fold, respectively, and of the mature neuron marker, NeuN, by 1.5-fold and 1.4-fold, respectively. We examined the effect of YKS on intracellular signaling pathways in P19 cells and found significant elevation in phospho-PDK1 and phospho-mTOR expression (1.1-fold and 1.2-fold, respectively). Therefore, we investigated the effect of PDK1 and mTOR inhibitors on the level of neuronal lineage markers. We found that the mTOR inhibitor significantly abolished the YKS effect on the level of neuronal lineage markers. Moreover, to identify the target(s) of YKS, antibody array analysis that simultaneously detects 16 phosphorylated proteins was performed. YKS significantly upregulated 10 phosphorylated proteins including PDK1, Akt, AMPK, PRAS40, mTOR, p70 S6 kinase, GSK-3α, Bad and ERK1/2 under cell proliferation conditions. These results suggest that YKS simultaneously activates multiple signaling pathways. Thus, we concluded that YKS enhances the level of neuronal lineage markers in differentiated P19 cells, however it does not induce neuronal differentiation. Furthermore, mTOR is the predominant mediator of the YKS effect on these cells.
Collapse
Affiliation(s)
- Makoto Fukui
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Syouichi Katayama
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Yukinobu Ikeya
- Center for Supporting Pharmaceutical Education, Daiichi University of Pharmacy, Minami, Fukuoka, 815-8511, Japan
| | - Tetsuya Inazu
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
7
|
Direct reprogramming of fibroblasts into neural stem cells by single non-neural progenitor transcription factor Ptf1a. Nat Commun 2018; 9:2865. [PMID: 30030434 PMCID: PMC6054649 DOI: 10.1038/s41467-018-05209-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 06/23/2018] [Indexed: 01/29/2023] Open
Abstract
Induced neural stem cells (iNSCs) reprogrammed from somatic cells have great potentials in cell replacement therapies and in vitro modeling of neural diseases. Direct conversion of fibroblasts into iNSCs has been shown to depend on a couple of key neural progenitor transcription factors (TFs), raising the question of whether such direct reprogramming can be achieved by non-neural progenitor TFs. Here we report that the non-neural progenitor TF Ptf1a alone is sufficient to directly reprogram mouse and human fibroblasts into self-renewable iNSCs capable of differentiating into functional neurons, astrocytes and oligodendrocytes, and improving cognitive dysfunction of Alzheimer’s disease mouse models when transplanted. The reprogramming activity of Ptf1a depends on its Notch-independent interaction with Rbpj which leads to subsequent activation of expression of TF genes and Notch signaling required for NSC specification, self-renewal, and homeostasis. Together, our data identify a non-canonical and safer approach to establish iNSCs for research and therapeutic purposes. Fibroblasts can be reprogrammed into induced neural stem cells (iNSCs) using transcription factors expressed in neural progenitors. Here the authors show that Ptf1a, which is normally expressed in postmitotic neurons, can reprogram fibroblasts to iNSCs through Notch independent interaction with Rbpj.
Collapse
|
8
|
Kelly GM, Gatie MI. Mechanisms Regulating Stemness and Differentiation in Embryonal Carcinoma Cells. Stem Cells Int 2017; 2017:3684178. [PMID: 28373885 PMCID: PMC5360977 DOI: 10.1155/2017/3684178] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/10/2017] [Accepted: 02/08/2017] [Indexed: 02/06/2023] Open
Abstract
Just over ten years have passed since the seminal Takahashi-Yamanaka paper, and while most attention nowadays is on induced, embryonic, and cancer stem cells, much of the pioneering work arose from studies with embryonal carcinoma cells (ECCs) derived from teratocarcinomas. This original work was broad in scope, but eventually led the way for us to focus on the components involved in the gene regulation of stemness and differentiation. As the name implies, ECCs are malignant in nature, yet maintain the ability to differentiate into the 3 germ layers and extraembryonic tissues, as well as behave normally when reintroduced into a healthy blastocyst. Retinoic acid signaling has been thoroughly interrogated in ECCs, especially in the F9 and P19 murine cell models, and while we have touched on this aspect, this review purposely highlights how some key transcription factors regulate pluripotency and cell stemness prior to this signaling. Another major focus is on the epigenetic regulation of ECCs and stem cells, and, towards that end, this review closes on what we see as a new frontier in combating aging and human disease, namely, how cellular metabolism shapes the epigenetic landscape and hence the pluripotency of all stem cells.
Collapse
Affiliation(s)
- Gregory M. Kelly
- Department of Biology, Molecular Genetics Unit, Western University, London, ON, Canada
- Collaborative Program in Developmental Biology, Western University, London, ON, Canada
- Department of Paediatrics and Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Child Health Research Institute, London, ON, Canada
- Ontario Institute for Regenerative Medicine, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Mohamed I. Gatie
- Department of Biology, Molecular Genetics Unit, Western University, London, ON, Canada
- Collaborative Program in Developmental Biology, Western University, London, ON, Canada
| |
Collapse
|
9
|
Xiao J, Li X, Chen L, Han X, Zhao W, Li L, Chen JG. Apobec1 Promotes Neurotoxicity-Induced Dedifferentiation of Müller Glial Cells. Neurochem Res 2017; 42:1151-1164. [DOI: 10.1007/s11064-016-2151-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 11/10/2016] [Accepted: 12/19/2016] [Indexed: 01/16/2023]
|
10
|
Choi DH, Kim JH, Kim SM, Kang K, Han DW, Lee J. Therapeutic Potential of Induced Neural Stem Cells for Parkinson's Disease. Int J Mol Sci 2017; 18:E224. [PMID: 28117752 PMCID: PMC5297853 DOI: 10.3390/ijms18010224] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative disorder that results from the loss of cells in the substantia nigra (SN) which is located in the midbrain. However, no cure is available for PD. Recently, fibroblasts have been directly converted into induced neural stem cells (iNSCs) via the forced expression of specific transcription factors. Therapeutic potential of iNSC in PD has not been investigated yet. Here, we show that iNSCs directly converted from mouse fibroblasts enhanced functional recovery in an animal model of PD. The rotational behavior test was performed to assess recovery. Our results indicate that iNSC transplantation into the striatum of 6-hydroxydopamine (6-OHDA)-injected mice can significantly reduce apomorphine-induced rotational asymmetry. The engrafted iNSCs were able to survive in the striatum and migrated around the medial forebrain bundle and the SN pars compacta. Moreover, iNSCs differentiated into all neuronal lineages. In particular, the transplanted iNSCs that committed to the glial lineage were significantly increased in the striatum of 6-OHDA-injected mice. Engrafted iNSCs differentiated to dopaminergic (DA) neurons and migrated into the SN in the 6-OHDA lesion mice. Therefore, iNSC transplantation serves as a valuable tool to enhance the functional recovery in PD.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Department of Medical Science, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
| | - Ji-Hye Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
| | - Sung Min Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
| | - Kyuree Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
| | - Dong Wook Han
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
- Konkuk Univesity Open-Innovation Center, Institute of Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
- Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
| | - Jongmin Lee
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
- Department of Rehabilitation Medicine, Konkuk University School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.
| |
Collapse
|
11
|
Gomes CBF, Zechin KG, Xu S, Stelini RF, Nishimoto IN, Zhan Q, Xu T, Qin G, Treister NS, Murphy GF, Lian CG. TET2 Negatively Regulates Nestin Expression in Human Melanoma. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1427-34. [PMID: 27102770 PMCID: PMC4901139 DOI: 10.1016/j.ajpath.2016.01.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/09/2016] [Accepted: 01/27/2016] [Indexed: 01/31/2023]
Abstract
Although melanoma is an aggressive cancer, the understanding of the virulence-conferring pathways involved remains incomplete. We have demonstrated that loss of ten-eleven translocation methylcytosine dioxygenase (TET2)-mediated 5-hydroxymethylcytosine (5-hmC) is an epigenetic driver of melanoma growth and a biomarker of clinical virulence. We also have determined that the intermediate filament protein nestin correlates with tumorigenic and invasive melanoma growth. Here we examine the relationships between these two biomarkers. Immunohistochemistry staining of nestin and 5-hmC in 53 clinically annotated primary and metastatic patient melanomas revealed a significant negative correlation. Restoration of 5-hmC, as assessed in a human melanoma cell line by introducing full-length TET2 and TET2-mutated constructs, decreased nestin gene and protein expression in vitro. Genome-wide mapping using hydroxymethylated DNA immunoprecipitation sequencing disclosed significantly less 5-hmC binding in the 3' untranslated region of the nestin gene in melanoma compared to nevi, and 5-hmC binding in this region was significantly increased after TET2 overexpression in human melanoma cells in vitro. Our findings provide evidence suggesting that nestin regulation is negatively controlled epigenetically by TET2 via 5-hmC binding at the 3' untranslated region of the nestin gene, providing one potential pathway for understanding melanoma growth characteristics. Studies are now indicated to further define the interplay between 5-hmC, nestin expression, and melanoma virulence.
Collapse
Affiliation(s)
- Camilla B F Gomes
- Program in Oral Pathology, Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, Brazil; Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Karina G Zechin
- Program in Oral Pathology, Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, Brazil
| | - Shuyun Xu
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rafael F Stelini
- Department of Pathology, Medical Sciences School, University of Campinas, Piracicaba, Brazil
| | - Ines N Nishimoto
- Department of Head and Surgery and Otorhinolaryngology, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Qian Zhan
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ting Xu
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gungwei Qin
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nathaniel S Treister
- Division of Oral Medicine and Dentistry, Brigham and Women's Hospital, Harvard School of Dental Medicine, Boston, Massachusetts
| | - George F Murphy
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Christine G Lian
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
12
|
Kim SM, Kim JW, Kwak TH, Park SW, Kim KP, Park H, Lim KT, Kang K, Kim J, Yang JH, Han H, Lee I, Hyun JK, Bae YM, Schöler HR, Lee HT, Han DW. Generation of Integration-free Induced Neural Stem Cells from Mouse Fibroblasts. J Biol Chem 2016; 291:14199-14212. [PMID: 27189941 DOI: 10.1074/jbc.m115.713578] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Indexed: 01/10/2023] Open
Abstract
The viral vector-mediated overexpression of the defined transcription factors, Brn4/Pou3f4, Sox2, Klf4, and c-Myc (BSKM), could induce the direct conversion of somatic fibroblasts into induced neural stem cells (iNSCs). However, viral vectors may be randomly integrated into the host genome thereby increasing the risk for undesired genotoxicity, mutagenesis, and tumor formation. Here we describe the generation of integration-free iNSCs from mouse fibroblasts by non-viral episomal vectors containing BSKM. The episomal vector-derived iNSCs (e-iNSCs) closely resemble control NSCs, and iNSCs generated by retrovirus (r-iNSCs) in morphology, gene expression profile, epigenetic status, and self-renewal capacity. The e-iNSCs are functionally mature, as they could differentiate into all the neuronal cell types both in vitro and in vivo Our study provides a novel concept for generating functional iNSCs using a non-viral, non-integrating, plasmid-based system that could facilitate their biomedical applicability.
Collapse
Affiliation(s)
- Sung Min Kim
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; Department of Animal Biotechnology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jong-Wan Kim
- Department of Nanobiomedical Science, Dankook University Graduate School, Cheonan 31116, Republic of Korea
| | - Tae Hwan Kwak
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sang Woong Park
- Department of Physiology, School of Medicine, Konkuk University, Chungju, Chungbuk 27478, Republic of Korea
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hyunji Park
- Department of Physiology, School of Medicine, Konkuk University, Chungju, Chungbuk 27478, Republic of Korea
| | - Kyung Tae Lim
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kyuree Kang
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jonghun Kim
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ji Hun Yang
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Heonjong Han
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 04056, Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 04056, Korea
| | - Jung Keun Hyun
- Department of Nanobiomedical Science, Dankook University Graduate School, Cheonan 31116, Republic of Korea
| | - Young Min Bae
- Department of Physiology, School of Medicine, Konkuk University, Chungju, Chungbuk 27478, Republic of Korea
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany,; University of Münster, Medical Faculty, Domagkstraße 3, 48149 Münster, Germany
| | - Hoon Taek Lee
- Department of Animal Biotechnology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong Wook Han
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; KU Open-Innovation Center, Institute of Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
13
|
Matsuda Y, Ishiwata T, Yoshimura H, Yamashita S, Ushijima T, Arai T. Systemic Administration of Small Interfering RNA Targeting Human Nestin Inhibits Pancreatic Cancer Cell Proliferation and Metastasis. Pancreas 2016; 45:93-100. [PMID: 26335012 DOI: 10.1097/mpa.0000000000000427] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Nestin, a progenitor/stem cell marker, is expressed in human pancreatic cancer, where its expression correlates positively with invasiveness and metastasis. Here, we investigated the inhibition of nestin expression and the regulation of nestin expression. METHODS We analyzed the effects of small interfering RNA (siRNA) targeting nestin using pancreatic cancer cell lines. RESULTS Nestin siRNA inhibited the growth, migration, invasion, and sphere-forming ability of the pancreatic cancer cell lines. Pancreatic cancer cells cotreated with gemcitabine and nestin siRNA exhibited lower cell viability than cells treated with a control siRNA, gemcitabine alone, or nestin siRNA alone. Cells derived from the metastatic nodules of mice showed higher nestin expression than the parental cells, and nestin expression in pancreatic cancer cells was regulated by methylation of the nestin gene. In an orthotopic implantation model using mice, administration of nestin siRNA significantly decreased primary and metastatic tumor formation by human pancreatic cancer cells compared to tumor formation in control siRNA-treated mice. CONCLUSIONS Nestin plays a key role in pancreatic cancer cell metastasis and stemness and that administration of nestin siRNA may offer a novel therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Yoko Matsuda
- From the *Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Itabashi-ku; †Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, Bunkyo-ku; and ‡Division of Epigenomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
14
|
Pelch KE, Tokar EJ, Merrick BA, Waalkes MP. Differential DNA methylation profile of key genes in malignant prostate epithelial cells transformed by inorganic arsenic or cadmium. Toxicol Appl Pharmacol 2015; 286:159-67. [PMID: 25922126 PMCID: PMC4461502 DOI: 10.1016/j.taap.2015.04.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022]
Abstract
Previous work shows altered methylation patterns in inorganic arsenic (iAs)- or cadmium (Cd)-transformed epithelial cells. Here, the methylation status near the transcriptional start site was assessed in the normal human prostate epithelial cell line (RWPE-1) that was malignantly transformed by 10μM Cd for 11weeks (CTPE) or 5μM iAs for 29weeks (CAsE-PE), at which time cells showed multiple markers of acquired cancer phenotype. Next generation sequencing of the transcriptome of CAsE-PE cells identified multiple dysregulated genes. Of the most highly dysregulated genes, five genes that can be relevant to the carcinogenic process (S100P, HYAL1, NTM, NES, ALDH1A1) were chosen for an in-depth analysis of the DNA methylation profile. DNA was isolated, bisulfite converted, and combined bisulfite restriction analysis was used to identify differentially methylated CpG sites, which was confirmed with bisulfite sequencing. Four of the five genes showed differential methylation in transformants relative to control cells that was inversely related to altered gene expression. Increased expression of HYAL1 (>25-fold) and S100P (>40-fold) in transformants was correlated with hypomethylation near the transcriptional start site. Decreased expression of NES (>15-fold) and NTM (>1000-fold) in transformants was correlated with hypermethylation near the transcriptional start site. ALDH1A1 expression was differentially expressed in transformed cells but was not differentially methylated relative to control. In conclusion, altered gene expression observed in Cd and iAs transformed cells may result from altered DNA methylation status.
Collapse
Affiliation(s)
- Katherine E Pelch
- National Toxicology Program Laboratory, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Erik J Tokar
- National Toxicology Program Laboratory, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - B Alex Merrick
- Molecular Toxicology and Informatics Group, Biomolecular Screening Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Morrisville, NC 27560, USA
| | - Michael P Waalkes
- National Toxicology Program Laboratory, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
15
|
Neradil J, Veselska R. Nestin as a marker of cancer stem cells. Cancer Sci 2015; 106:803-11. [PMID: 25940879 PMCID: PMC4520630 DOI: 10.1111/cas.12691] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/14/2015] [Accepted: 04/27/2015] [Indexed: 12/14/2022] Open
Abstract
The crucial role of cancer stem cells (CSCs) in the pathology of malignant diseases has been extensively studied during the last decade. Nestin, a class VI intermediate filament protein, was originally detected in neural stem cells during development. Its expression has also been reported in different tissues under various pathological conditions. Specifically, nestin has been shown to be expressed in transformed cells of various human malignancies, and a correlation between its expression and the clinical course of some diseases has been proved. Furthermore, the coexpression of nestin with other stem cell markers was described as a CSC phenotype that was subsequently verified using tumorigenicity assays. The primary aim of this review is to summarize the recent findings regarding nestin expression in CSCs, its possible role in CSC phenotypes, particularly with respect to capacity for self-renewal, and its utility as a putative marker of CSCs.
Collapse
Affiliation(s)
- Jakub Neradil
- Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Brno, Czech Republic.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Brno, Czech Republic.,Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
16
|
Nestin depletion induces melanoma matrix metalloproteinases and invasion. J Transl Med 2014; 94:1382-95. [PMID: 25365206 PMCID: PMC4419570 DOI: 10.1038/labinvest.2014.130] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/25/2014] [Accepted: 09/08/2014] [Indexed: 12/20/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are key biological mediators of processes as diverse as wound healing, embryogenesis, and cancer progression. Although MMPs may be induced through multiple signaling pathways, the precise mechanisms for their regulation in cancer are incompletely understood. Because cytoskeletal changes are known to accompany MMP expression, we sought to examine the potential role of the poorly understood cytoskeletal protein, nestin, in modulating melanoma MMPs. Nestin knockdown (KD) upregulated the expression of specific MMPs and MMP-dependent invasion both through extracellular matrix barriers in vitro and in peritumoral connective tissue of xenografts in vivo. The development of three-dimensional melanospheres that in vitro partially recapitulate noninvasive tumorigenic melanoma growth was inhibited by nestin KD, although ECM invasion by aberrant melanospheres that did form was enhanced. Mechanistically, nestin KD-dependent melanoma invasion was associated with intracellular redistribution of phosphorylated focal adhesion kinase and increased melanoma cell responsiveness to transforming growth factor-beta, both implicated in pathways of melanoma invasion. The results suggest that the heretofore poorly understood intermediate filament, nestin, may serve as a novel mediator of MMPs critical to melanoma virulence.
Collapse
|
17
|
Hong JY, Lee SH, Lee SC, Kim JW, Kim KP, Kim SM, Tapia N, Lim KT, Kim J, Ahn HS, Ko K, Shin CY, Lee HT, Schöler HR, Hyun JK, Han DW. Therapeutic potential of induced neural stem cells for spinal cord injury. J Biol Chem 2014; 289:32512-25. [PMID: 25294882 DOI: 10.1074/jbc.m114.588871] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The spinal cord does not spontaneously regenerate, and treatment that ensures functional recovery after spinal cord injury (SCI) is still not available. Recently, fibroblasts have been directly converted into induced neural stem cells (iNSCs) by the forced expression defined transcription factors. Although directly converted iNSCs have been considered to be a cell source for clinical applications, their therapeutic potential has not yet been investigated. Here we show that iNSCs directly converted from mouse fibroblasts enhance the functional recovery of SCI animals. Engrafted iNSCs could differentiate into all neuronal lineages, including different subtypes of mature neurons. Furthermore, iNSC-derived neurons could form synapses with host neurons, thus enhancing the locomotor function recovery. A time course analysis of iNSC-treated SCI animals revealed that engrafted iNSCs effectively reduced the inflammatory response and apoptosis in the injured area. iNSC transplantation also promoted the active regeneration of the endogenous recipient environment in the absence of tumor formation. Therefore, our data suggest that directly converted iNSCs hold therapeutic potential for treatment of SCI and may thus represent a promising cell source for transplantation therapy in patients with SCI.
Collapse
Affiliation(s)
- Jin Young Hong
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea
| | | | | | - Jong-Wan Kim
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea
| | - Kee-Pyo Kim
- the Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | | | - Natalia Tapia
- the Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | | | | | - Hong-Sun Ahn
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea
| | - Kinarm Ko
- the Departments of Stem Cell Biology and
| | - Chan Young Shin
- Pharmacology, School of Medicine, and the Konkuk University Open-Innovation Center, Institute of Biomedical Science & Technology, Konkuk University, Gwangjin-gu, Seoul 143701, Republic of Korea
| | | | - Hans R Schöler
- the Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany, the University of Münster, Medical Faculty, 48149 Münster, Germany, and
| | - Jung Keun Hyun
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea, the Department of Rehabilitation Medicine, College of Medicine, Dankook University, Cheonan 330714, Republic of Korea
| | - Dong Wook Han
- the Departments of Stem Cell Biology and the Konkuk University Open-Innovation Center, Institute of Biomedical Science & Technology, Konkuk University, Gwangjin-gu, Seoul 143701, Republic of Korea,
| |
Collapse
|
18
|
Huang HS, Redmond TM, Kubish GM, Gupta S, Thompson RC, Turner DL, Uhler MD. Transcriptional regulatory events initiated by Ascl1 and Neurog2 during neuronal differentiation of P19 embryonic carcinoma cells. J Mol Neurosci 2014; 55:684-705. [PMID: 25189318 DOI: 10.1007/s12031-014-0408-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/20/2014] [Indexed: 11/25/2022]
Abstract
As members of the proneural basic-helix-loop-helix (bHLH) family of transcription factors, Ascl1 and Neurog2 direct the differentiation of specific populations of neurons at various times and locations within the developing nervous system. In order to characterize the mechanisms employed by these two bHLH factors, we generated stable, doxycycline-inducible lines of P19 embryonic carcinoma cells that express comparable levels of Ascl1 and Neurog2. Upon induction, both Ascl1 and Neurog2 directed morphological and immunocytochemical changes consistent with initiation of neuronal differentiation. Comparison of Ascl1- and Neurog2-regulated genes by microarray analyses showed both shared and distinct transcriptional changes for each bHLH protein. In both Ascl1- and Neurog2-differentiating cells, repression of Oct4 mRNA levels was accompanied by increased Oct4 promoter methylation. However, DNA demethylation was not detected for genes induced by either bHLH protein. Neurog2-induced genes included glutamatergic marker genes while Ascl1-induced genes included GABAergic marker genes. The Neurog2-specific induction of a gene encoding a protein phosphatase inhibitor, Ppp1r14a, was dependent on distinct, canonical E-box sequences within the Ppp1r14a promoter and the nucleotide sequences within these E-boxes were partially responsible for Neurog2-specific regulation. Our results illustrate multiple novel mechanisms by which Ascl1 and Neurog2 regulate gene repression during neuronal differentiation in P19 cells.
Collapse
Affiliation(s)
- Holly S Huang
- Molecular and Behavioral Neuroscience Institute, University of Michigan, 109 Zina Pitcher Pl, Ann Arbor, MI, 48109-2200, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Resendiz M, Mason S, Lo CL, Zhou FC. Epigenetic regulation of the neural transcriptome and alcohol interference during development. Front Genet 2014; 5:285. [PMID: 25206361 PMCID: PMC4144008 DOI: 10.3389/fgene.2014.00285] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/02/2014] [Indexed: 01/07/2023] Open
Abstract
Alcohol intoxicated cells broadly alter their metabolites – among them methyl and acetic acid can alter the DNA and histone epigenetic codes. Together with the promiscuous effect of alcohol on enzyme activities (including DNA methyltransferases) and the downstream effect on microRNA and transposable elements, alcohol is well placed to affect intrinsic transcriptional programs of developing cells. Considering that the developmental consequences of early alcohol exposure so profoundly affect neural systems, it is not unfounded to reason that alcohol exploits transcriptional regulators to challenge canonical gene expression and in effect, intrinsic developmental pathways to achieve widespread damage in the developing nervous system. To fully evaluate the role of epigenetic regulation in alcohol-related developmental disease, it is important to first gather the targets of epigenetic players in neurodevelopmental models. Here, we attempt to review the cellular and genomic windows of opportunity for alcohol to act on intrinsic neurodevelopmental programs. We also discuss some established targets of fetal alcohol exposure and propose pathways for future study. Overall, this review hopes to illustrate the known epigenetic program and its alterations in normal neural stem cell development and further, aims to depict how alcohol, through neuroepigenetics, may lead to neurodevelopmental deficits observed in fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Marisol Resendiz
- Stark Neuroscience Research Institute Indianapolis, IN, USA ; Indiana Alcohol Research Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Stephen Mason
- Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA
| | - Chiao-Ling Lo
- Indiana Alcohol Research Center, Indiana University School of Medicine Indianapolis, IN, USA ; Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA
| | - Feng C Zhou
- Stark Neuroscience Research Institute Indianapolis, IN, USA ; Indiana Alcohol Research Center, Indiana University School of Medicine Indianapolis, IN, USA ; Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA
| |
Collapse
|
20
|
Marthaler AG, Tiemann U, Araúzo-Bravo MJ, Wu G, Zaehres H, Hyun JK, Han DW, Schöler HR, Tapia N. Reprogramming to pluripotency through a somatic stem cell intermediate. PLoS One 2013; 8:e85138. [PMID: 24386457 PMCID: PMC3874029 DOI: 10.1371/journal.pone.0085138] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 11/24/2013] [Indexed: 12/28/2022] Open
Abstract
Transcription factor-based reprogramming can lead to the successful switching of cell fates. We have recently reported that mouse embryonic fibroblasts (MEFs) can be directly reprogrammed into induced neural stem cells (iNSCs) after the forced expression of Brn4, Sox2, Klf4, and Myc. Here, we tested whether iNSCs could be further reprogrammed into induced pluripotent stem cells (iPSCs). The two factors Oct4 and Klf4 were sufficient to induce pluripotency in iNSCs. Immunocytochemistry and gene expression analysis showed that iNSC-derived iPSCs (iNdiPSCs) are similar to embryonic stem cells at the molecular level. In addition, iNdiPSCs could differentiate into cells of all three germ layers, both in vitro and in vivo, proving that iNdiPSCs are bona fide pluripotent cells. Furthermore, analysis of the global gene expression profile showed that iNdiPSCs, in contrast to iNSCs, do not retain any MEF transcriptional memory even at early passages after reprogramming. Overall, our results demonstrate that iNSCs can be reprogrammed to pluripotency and suggest that cell fate can be redirected numerous times. Importantly, our findings indicate that the induced pluripotent cell state may erase the donor-cell type epigenetic memory more efficiently than other induced somatic cell fates.
Collapse
Affiliation(s)
- Adele G. Marthaler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Ulf Tiemann
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Marcos J. Araúzo-Bravo
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Guangming Wu
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Holm Zaehres
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Jung Keun Hyun
- Department of Nanobiomedical Science, Dankook University Graduate School, Cheonan, Republic of Korea
- Department of Rehabilitation Medicine, Dandook University, Cheonan, Republic of Korea
| | - Dong Wook Han
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hans R. Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Medical Faculty, University of Münster, Münster, Germany
| | - Natalia Tapia
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- * E-mail:
| |
Collapse
|
21
|
Svachova H, Kryukov F, Kryukova E, Sevcikova S, Nemec P, Greslikova H, Rihova L, Kubiczkova L, Hajek R. Nestin expression throughout multistep pathogenesis of multiple myeloma. Br J Haematol 2013; 164:701-9. [PMID: 24329895 DOI: 10.1111/bjh.12689] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/13/2013] [Indexed: 12/12/2022]
Abstract
The stem cell marker nestin (NES) is found in dividing cells of developing and regenerating tissues. Upon terminal differentiation, NES expression is diminished but may be re-expressed following injury or in cancer. Surprisingly, we recently confirmed NES as a tumour-specific marker for mature CD138(+) 38(+) plasma cells (PC) in multiple myeloma (MM). The present study analysed NES expression throughout the spectrum of MM developmental stages, starting with individuals with no haematological malignancy, through monoclonal gammopathy of undetermined significance (MGUS) and MM to plasma cell leukaemia (PCL) and MM cell lines. NES was analysed in bone marrow PC of 163 MM, four PCL and nine MGUS patients, 10 individuals with no haematological malignancy and 6 myeloma cell lines (OPM-2, RPMI-8226, MOLP-8, U-266, EJM, NCI-H929) by flow cytometry and/or real-time polymerase chain reaction or immunochemistry. We observed a tendency of increased NES expression in parallel with disease progression. NES was evaluated as a reliable marker for accurate discrimination between MM patients and the control group. High NES levels were strongly associated with the presence of 1q21 gain. For the first time, NES was demonstrated to predict worse response to conventional therapy/novel agents. These results suggest that NES might become a useful clinical parameter with an important role in MM pathogenesis.
Collapse
Affiliation(s)
- Hana Svachova
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Disclosing the crosstalk among DNA methylation, transcription factors, and histone marks in human pluripotent cells through discovery of DNA methylation motifs. Genome Res 2013; 23:2013-29. [PMID: 24149073 PMCID: PMC3847772 DOI: 10.1101/gr.155960.113] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gene expression regulation is gated by promoter methylation states modulating transcription factor binding. The known DNA methylation/unmethylation mechanisms are sequence unspecific, but different cells with the same genome have different methylomes. Thus, additional processes bringing specificity to the methylation/unmethylation mechanisms are required. Searching for such processes, we demonstrated that CpG methylation states are influenced by the sequence context surrounding the CpGs. We used such a property to develop a CpG methylation motif discovery algorithm. The newly discovered motifs reveal “methylation/unmethylation factors” that could recruit the “methylation/unmethylation machinery” to the loci specified by the motifs. Our methylation motif discovery algorithm provides a synergistic approach to the differently methylated region algorithms. Since our algorithm searches for commonly methylated regions inside the same sample, it requires only a single sample to operate. The motifs that were found discriminate between hypomethylated and hypermethylated regions. The hypomethylation-associated motifs have a high CG content, their targets appear in conserved regions near transcription start sites, they tend to co-occur within transcription factor binding sites, they are involved in breaking the H3K4me3/H3K27me3 bivalent balance, and they transit the enhancers from repressive H3K27me3 to active H3K27ac during ES cell differentiation. The new methylation motifs characterize the pluripotent state shared between ES and iPS cells. Additionally, we found a collection of motifs associated with the somatic memory inherited by the iPS from the initial fibroblast cells, thus revealing the existence of epigenetic somatic memory on a fine methylation scale.
Collapse
|
23
|
Han DW, Tapia N, Araúzo-Bravo MJ, Lim KT, Kim KP, Ko K, Lee HT, Schöler HR. Sox2 Level Is a Determinant of Cellular Reprogramming Potential. PLoS One 2013; 8:e67594. [PMID: 23825671 PMCID: PMC3688988 DOI: 10.1371/journal.pone.0067594] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/23/2013] [Indexed: 11/19/2022] Open
Abstract
Epiblast stem cells (EpiSCs) and embryonic stem cells (ESCs) differ in their in vivo differentiation potential. While ESCs form teratomas and efficiently contribute to the development of chimeras, EpiSCs form teratomas but very rarely chimeras. In contrast to their differentiation potential, the reprogramming potential of EpiSCs has not yet been investigated. Here we demonstrate that the epiblast-derived pluripotent stem cells EpiSCs and P19 embryonal carcinoma cells (ECCs) exhibit a lower reprogramming potential than ESCs and F9 ECCs. In addition, we show that the low reprogramming ability is due to the lower levels of Sox2 in epiblast-derived stem cells. Consistent with this observation, overexpression of Sox2 enhances reprogramming efficiency. In summary, these findings suggest that a low reprogramming potential is a general feature of epiblast-derived stem cells and that the Sox2 level is a determinant of the cellular reprogramming potential.
Collapse
Affiliation(s)
- Dong Wook Han
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
- Institute of Functional Genomics, Konkuk University, Seoul, Republic of Korea
- * E-mail:
| | - Natalia Tapia
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Marcos J. Araúzo-Bravo
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kyung Tae Lim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kee Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kinarm Ko
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
- Institute of Functional Genomics, Konkuk University, Seoul, Republic of Korea
| | - Hoon Taek Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hans R. Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- University of Münster, Medical Faculty, Münster, Germany
| |
Collapse
|
24
|
Abstract
The concept of cancer as a stem cell disease has slowly gained ground over the last decade. A 'stem-like' state essentially necessitates that some cells in the developing tumor express the properties of remaining quiescent, self-renewing and regenerating tumors through establishment of aberrant cellular hierarchies. Alternatively, such capacities may also be reacquired through a de-differentiation process. The abnormal cellular differentiation patterns involved during either process during carcinogenesis are likely to be driven through a combination of genetic events and epigenetic regulation. The role(s) of the latter is increasingly being appreciated in acquiring the requisite genomic specificity and flexibility required for phenotypic plasticity, specifically in a context wherein genome sequences are not altered for differentiation to ensue. In this chapter, the recent advances in elucidating epigenetic mechanisms that govern the self-renewal, differentiation and regenerative potentials of cancer stem cells will be presented.
Collapse
Affiliation(s)
- Sharmila A Bapat
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, 411 007, India,
| |
Collapse
|
25
|
Boulland JL, Mastrangelopoulou M, Boquest AC, Jakobsen R, Noer A, Glover JC, Collas P. Epigenetic regulation of nestin expression during neurogenic differentiation of adipose tissue stem cells. Stem Cells Dev 2012; 22:1042-52. [PMID: 23140086 DOI: 10.1089/scd.2012.0560] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Adipose-tissue-derived stem cells (ASCs) have received considerable attention due to their easy access, expansion potential, and differentiation capacity. ASCs are believed to have the potential to differentiate into neurons. However, the mechanisms by which this may occur remain largely unknown. Here, we show that culturing ASCs under active proliferation conditions greatly improves their propensity to differentiate toward osteogenic, adipogenic, and neurogenic lineages. Neurogenic-induced ASCs express early neurogenic genes as well as markers of mature neurons, including voltage-gated ion channels. Nestin, highly expressed in neural progenitors, is upregulated by mitogenic stimulation of ASCs, and as in neural progenitors, then repressed during neurogenic differentiation. Nestin gene (NES) expression under these conditions appears to be regulated by epigenetic mechanisms. The neural-specific, but not muscle-specific, enhancer regions of NES are DNA demethylated by mitogenic stimulation, and remethylated upon neurogenic differentiation. We observe dynamic changes in histone H3K4, H3K9, and H3K27 methylation on the NES locus before and during neurogenic differentiation that are consistent with epigenetic processes involved in the regulation of NES expression. We suggest that ASCs are epigenetically prepatterned to differentiate toward a neural lineage and that this prepatterning is enhanced by demethylation of critical NES enhancer elements upon mitogenic stimulation preceding neurogenic differentiation. Our findings provide molecular evidence that the differentiation repertoire of ASCs may extend beyond mesodermal lineages.
Collapse
Affiliation(s)
- Jean-Luc Boulland
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
26
|
Li P, Sun H, Du M, Fa Z, Qin K, Xu W, Zhang R, Chen L, Yao C, Xiao Z, Zhang S, Ke Y, Jiang X. Adult rat hippocampus soluble factors: a novel transplantation model mimicking intracranial microenvironment for tracing the induction and differentiation of adipose-derived stromal cells in vitro. Neurosci Lett 2012; 542:5-11. [PMID: 23103714 DOI: 10.1016/j.neulet.2012.10.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 08/30/2012] [Accepted: 10/18/2012] [Indexed: 12/16/2022]
Abstract
Intracranial transplantation of ADSCs induces recovery of CNS diseases, but how they develop in host is poorly understood. The aim of this study is to observe induction and differentiation of ADSCs in the presence of hippocampus soluble factors (HiSF) extracted from the hippocampus of adult Wistar rats to mimic an intracranial microenvironment. To determine the optimal microenvironment, five conditions were tested: 0μg/ml (as control), 50μg/ml, 100μg/ml, 200μg/ml, and 400μg/ml of HiSF. The number of neurospheres was significantly higher in 200μg/ml group than in other groups on the sixth day. Immunofluorescence demonstrated that the neurospheres induced from ADSCs in 200μg/ml group expressed both nestin and CD133, which are more highly expressed in neurospheres than in ADSCs. This result was confirmed by Western blot analysis. Quantitative PCR revealed that the mRNA levels of nestin and CD133 in the neurospheres were 145- and 220-fold higher, respectively, than those in ADSCs. In the presence of 200μg/ml HiSF and 1% FBS, the neurospheres can further differentiate into Schwann-like cells which expressing characteristic markers GFAP, S100 and P75 NGFR. These data indicated that HiSF, mimicking a destination of ADSCs transplanted model in vitro, could effectively induce and differentiate neurospheres, representing a new method to obtain NSCs and Schwann-like cells from ADSCs.
Collapse
Affiliation(s)
- Peng Li
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Xu W, Li P, Qin K, Wang X, Jiang X. miR-124 regulates neural stem cells in the treatment of spinal cord injury. Neurosci Lett 2012; 529:12-7. [PMID: 22999930 DOI: 10.1016/j.neulet.2012.09.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 09/05/2012] [Accepted: 09/12/2012] [Indexed: 01/17/2023]
Abstract
Several studies demonstrated that the overexpression of miR-124 in neural stem cells (NSCs) could lead the NSCs to differentiate into neurons and astrocytes, which may be important for functional recovery in spinal cord injury. The present study attempted to explore the potential repairing effect of the NSCs transfected with miR-124 for the rats with spinal cord injury (SCI). NSCs transfected with miR-124 were transplanted into rats by intravenous injection after SCI. The effects of miR-124 on the differentiation of NSCs and the treatment for the SCI-model rats were experimentally investigated. The reduction of cavity volume in focal lesions and Basso-Beattie-Bresnahan (BBB) scores were used as the criteria of functional recovery of the SCI-model rats. Up-regulation of miR-124 promoted the differentiation of NSCs. Transfection of miR-124 in NSCs dramatically increased the percentage of NeuN-positive cells, and reduced the percentage of GFAP-positive cells in vitro and in vivo respectively. All of the rats treated with NSCs transfected with miR-124 achieved the better functional recovery than the ones in NSCs and sham control groups. Furthermore, the systemic delivery of the NSCs transfected with miR-124 resulted in a reduction of lesion cavity volume of SCI-model rats. Thus, Overexpression of miR-124 can promote the differentiation of NSCs and play an important role in the repair of SCI. The utility of intravenous delivery of stem cells regulated with miR-124 to target lesion areas as a prospective therapeutic approach in acute spinal cord injury is very promising in the future.
Collapse
Affiliation(s)
- Weiwei Xu
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangdong Province 510630, China
| | | | | | | | | |
Collapse
|
28
|
Ring KL, Tong LM, Balestra ME, Javier R, Andrews-Zwilling Y, Li G, Walker D, Zhang WR, Kreitzer AC, Huang Y. Direct reprogramming of mouse and human fibroblasts into multipotent neural stem cells with a single factor. Cell Stem Cell 2012; 11:100-9. [PMID: 22683203 DOI: 10.1016/j.stem.2012.05.018] [Citation(s) in RCA: 419] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 04/25/2012] [Accepted: 05/15/2012] [Indexed: 01/01/2023]
Abstract
The generation of induced pluripotent stem cells (iPSCs) and induced neuronal cells (iNCs) from somatic cells provides new avenues for basic research and potential transplantation therapies for neurological diseases. However, clinical applications must consider the risk of tumor formation by iPSCs and the inability of iNCs to self-renew in culture. Here we report the generation of induced neural stem cells (iNSCs) from mouse and human fibroblasts by direct reprogramming with a single factor, Sox2. iNSCs express NSC markers and resemble wild-type NSCs in their morphology, self-renewal, ability to form neurospheres, and gene expression profiles. Cloned iNSCs differentiate into several types of mature neurons, as well as astrocytes and oligodendrocytes, indicating multipotency. Implanted iNSCs can survive and integrate in mouse brains and, unlike iPSC-derived NSCs, do not generate tumors. Thus, self-renewable and multipotent iNSCs without tumorigenic potential can be generated directly from fibroblasts by reprogramming.
Collapse
Affiliation(s)
- Karen L Ring
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Han DW, Tapia N, Hermann A, Hemmer K, Höing S, Araúzo-Bravo MJ, Zaehres H, Wu G, Frank S, Moritz S, Greber B, Yang JH, Lee HT, Schwamborn JC, Storch A, Schöler HR. Direct reprogramming of fibroblasts into neural stem cells by defined factors. Cell Stem Cell 2012; 10:465-72. [PMID: 22445517 DOI: 10.1016/j.stem.2012.02.021] [Citation(s) in RCA: 424] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/27/2012] [Accepted: 02/24/2012] [Indexed: 12/17/2022]
Abstract
Recent studies have shown that defined sets of transcription factors can directly reprogram differentiated somatic cells to a different differentiated cell type without passing through a pluripotent state, but the restricted proliferative and lineage potential of the resulting cells limits the scope of their potential applications. Here we show that a combination of transcription factors (Brn4/Pou3f4, Sox2, Klf4, c-Myc, plus E47/Tcf3) induces mouse fibroblasts to directly acquire a neural stem cell identity-which we term as induced neural stem cells (iNSCs). Direct reprogramming of fibroblasts into iNSCs is a gradual process in which the donor transcriptional program is silenced over time. iNSCs exhibit cell morphology, gene expression, epigenetic features, differentiation potential, and self-renewing capacity, as well as in vitro and in vivo functionality similar to those of wild-type NSCs. We conclude that differentiated cells can be reprogrammed directly into specific somatic stem cell types by defined sets of specific transcription factors.
Collapse
Affiliation(s)
- Dong Wook Han
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Reister S, Kordes C, Sawitza I, Häussinger D. The epigenetic regulation of stem cell factors in hepatic stellate cells. Stem Cells Dev 2011; 20:1687-99. [PMID: 21219128 DOI: 10.1089/scd.2010.0418] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The epigenetic regulation by DNA methylation is an important mechanism to control the expression of stem cell factors as demonstrated in tumor cells. It was recently shown that hepatic stellate cells (HSC) express stem/progenitor cell factors and have a differentiation potential. The aim of this work was to investigate if the expression of stem cell markers is regulated by DNA methylation during activation of rat HSC. It was found that CD133, Notch1, and Notch3 are regulated via DNA methylation in HSC, whereas Nestin shows no DNA methylation in HSC and other undifferentiated cells such as embryonic stem cells and umbilical cord blood stem cells from rats. In contrast to this, DNA methylation controls Nestin expression in differentiated cells like hepatocytes and the hepatoma cell line H4IIE. Demethylation by 5-Aza-2-deoxycytidine was sufficient to induce Nestin in H4IIE cells. In quiescent stellate cells and embryonic stem cells, the Nestin expression was suppressed by histone H3 methylation at lysine 9, which is another epigenetic mechanism. Apart from the known induction of Nestin in cultured HSC, this intermediate filament protein was also induced after partial hepatectomy, indicating activation of HSC during liver regeneration. Taken together, this study demonstrates for the first time that the expression of stem cell-associated factors such as CD133, Notch1, and Notch3 is controlled by DNA methylation in HSC. The regulation of Nestin by DNA methylation seems to be restricted to differentiated cells, whereas undifferentiated cells use different epigenetic mechanisms such as histone H3 methylation to control Nestin expression.
Collapse
Affiliation(s)
- Sven Reister
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Heinrich-Heine-Universität, Düsseldorf, Germany
| | | | | | | |
Collapse
|
31
|
Geminin promotes neural fate acquisition of embryonic stem cells by maintaining chromatin in an accessible and hyperacetylated state. Proc Natl Acad Sci U S A 2011; 108:3294-9. [PMID: 21300881 DOI: 10.1073/pnas.1012053108] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Formation of the complex vertebrate nervous system begins when pluripotent cells of the early embryo are directed to acquire a neural fate. Although cell intrinsic controls play an important role in this process, the molecular nature of this regulation is not well defined. Here we assessed the role for Geminin, a nuclear protein expressed in embryonic cells, during neural fate acquisition from mouse embryonic stem (ES) cells. Whereas Geminin knockdown does not affect the ability of ES cells to maintain or exit pluripotency, we found that it significantly impairs their ability to acquire a neural fate. Conversely, Geminin overexpression promotes neural gene expression, even in the presence of growth factor signaling that antagonizes neural transcriptional responses. These data demonstrate that Geminin's activity contributes to mammalian neural cell fate acquisition. We investigated the mechanistic basis of this phenomenon and found that Geminin maintains a hyperacetylated and open chromatin conformation at neural genes. Interestingly, recombinant Geminin protein also rapidly alters chromatin acetylation and accessibility even when Geminin is combined with nuclear extract and chromatin in vitro. Together, these data support a role for Geminin as a cell intrinsic regulator of neural fate acquisition that promotes expression of neural genes by regulating chromatin accessibility and histone acetylation.
Collapse
|
32
|
Ishiwata T, Matsuda Y, Naito Z. Nestin in gastrointestinal and other cancers: Effects on cells and tumor angiogenesis. World J Gastroenterol 2011; 17:409-18. [PMID: 21274370 PMCID: PMC3027007 DOI: 10.3748/wjg.v17.i4.409] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/01/2010] [Accepted: 12/08/2010] [Indexed: 02/06/2023] Open
Abstract
Nestin is a class VI intermediate filament protein that was originally described as a neuronal stem cell marker during central nervous system (CNS) development, and is currently widely used in that capacity. Nestin is also expressed in non-neuronal immature or progenitor cells in normal tissues. Under pathological conditions, nestin is expressed in repair processes in the CNS, muscle, liver, and infarcted myocardium. Furthermore, increased nestin expression has been reported in various tumor cells, including CNS tumors, gastrointestinal stromal tumors, pancreatic cancer, prostate cancer, breast cancer, malignant melanoma, dermatofibrosarcoma protuberances, and thyroid tumors. Nestin is reported to correlate with aggressive growth, metastasis, and poor prognosis in some tumors; however, the roles of nestin in cancer cells have not been well characterized. Furthermore, nestin is more specifically expressed in proliferating small-sized tumor vessels in glioblastoma and gastric, colorectal, and prostate cancers than are other tumor vessel markers. These findings indicate that nestin may be a marker for newly synthesized tumor vessels and a therapeutic target for tumor angiogenesis. It has received a lot of attention recently as a cancer stem cell marker in various cancer cells including brain tumors, malignant rhabdoid tumors, and uterine, cervical, prostate, bladder, head and neck, ovarian, testicular, and pancreatic cancers. The purpose of this review is to clarify the roles of nestin in cancer cells and in tumor angiogenesis, and to examine the association between nestin and cancer stem cells. Nestin has the potential to serve as a molecular target for cancers with nestin-positive cancer cells and nestin-positive tumor vasculature.
Collapse
|
33
|
Han DW, Tapia N, Joo JY, Greber B, Araúzo-Bravo MJ, Bernemann C, Ko K, Wu G, Stehling M, Do JT, Schöler HR. Epiblast stem cell subpopulations represent mouse embryos of distinct pregastrulation stages. Cell 2010; 143:617-27. [PMID: 21056461 DOI: 10.1016/j.cell.2010.10.015] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 08/17/2010] [Accepted: 10/12/2010] [Indexed: 01/01/2023]
Abstract
Embryonic stem cells (ESCs) comprise at least two populations of cells with divergent states of pluripotency. Here, we show that epiblast stem cells (EpiSCs) also comprise two distinct cell populations that can be distinguished by the expression of a specific Oct4-GFP marker. These two subpopulations, Oct4-GFP positive and negative EpiSCs, are capable of converting into each other in vitro. Oct4-GFP positive and negative EpiSCs are distinct from ESCs with respect to global gene expression pattern, epigenetic profile, and Oct4 enhancer utilization. Oct4-GFP negative cells share features with cells of the late mouse epiblast and cannot form chimeras. However, Oct4-GFP positive EpiSCs, which only represent a minor EpiSC fraction, resemble cells of the early epiblast and can readily contribute to chimeras. Our findings suggest that the rare ability of EpiSCs to contribute to chimeras is due to the presence of the minor EpiSC fraction representing the early epiblast.
Collapse
Affiliation(s)
- Dong Wook Han
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149 Münster, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Liu T, Cheng W, Liu T, Guo L, Huang Q, Jiang L, Du X, Xu F, Liu Z, Lai D. Human amniotic epithelial cell feeder layers maintain mouse embryonic stem cell pluripotency via epigenetic regulation of the c-Myc promoter. Acta Biochim Biophys Sin (Shanghai) 2010; 42:109-15. [PMID: 20119621 DOI: 10.1093/abbs/gmp115] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mouse embryonic stem cells (ESCs) are typically cultured on a feeder layer of mouse embryonic fibroblasts (MEFs), with leukemia inhibitory factor (LIF) added to maintain them in an undifferentiated state. We have previously shown that human amniotic epithelial cells (hAECs) can be used as feeder cells to maintain mouse ESC pluripotency, but the mechanism for this is unknown. In the present study, we found that CpG islands 5' of the c-Myc gene remain hypomethylated in mouse ESCs cultured on hAECs. In addition, levels of acetylation of histone H3 and trimethylation of histone H3K4 in the c-Myc gene promoter were higher in ES cells cultured on hAECs than those in ES cells cultured on MEFs. These data suggested that hAECs can alter mouse ESC gene expression via epigenetic modification of c-Myc, providing a possible mechanism for the hAEC-induced maintenance of ESCs in an undifferentiated state.
Collapse
Affiliation(s)
- Te Liu
- Tongji University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gombert WM, Krumm A. Targeted deletion of multiple CTCF-binding elements in the human C-MYC gene reveals a requirement for CTCF in C-MYC expression. PLoS One 2009; 4:e6109. [PMID: 19568426 PMCID: PMC2699473 DOI: 10.1371/journal.pone.0006109] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 06/01/2009] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Insulators and domain boundaries both shield genes from adjacent enhancers and inhibit intrusion of heterochromatin into transgenes. Previous studies examined the functional mechanism of the MYC insulator element MINE and its CTCF binding sites in the context of transgenes that were randomly inserted into the genome by transfection. However, the contribution of CTCF binding sites to both gene regulation and maintenance of chromatin has not been tested at the endogenous MYC gene. METHODOLOGY/PRINCIPAL FINDINGS To determine the impact of CTCF binding on MYC expression, a series of mutant human chromosomal alleles was prepared in homologous recombination-efficient DT40 cells and individually transferred by microcell fusion into murine cells. Functional tests reported here reveal that deletion of CTCF binding elements within the MINE does not impact the capacity of this locus to correctly organize an 'accessible' open chromatin domain, suggesting that these sites are not essential for the formation of a competent, transcriptionally active locus. Moreover, deletion of the CTCF site at the MYC P2 promoter reduces transcription but does not affect promoter acetylation or serum-inducible transcription. Importantly, removal of either CTCF site leads to DNA methylation of flanking sequences, thereby contributing to progressive loss of transcriptional activity. CONCLUSIONS These findings collectively demonstrate that CTCF-binding at the human MYC locus does not repress transcriptional activity but is required for protection from DNA methylation.
Collapse
Affiliation(s)
- Wendy M. Gombert
- Black Lowe & Graham PLLC, Seattle, Washington, United States of America
| | - Anton Krumm
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|