1
|
Kim SH, Kim CJ, Lee EY, Hwang YH, Joo ST. Chicken Embryo Fibroblast Viability and Trans-Differentiation Potential for Cultured Meat Production Across Passages. Cells 2024; 13:1734. [PMID: 39451252 PMCID: PMC11506350 DOI: 10.3390/cells13201734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
This study was conducted to analyze the viability of primary chicken embryo fibroblasts and the efficiency of adipogenic trans-differentiation for cultured meat production. In isolating chicken embryo fibroblasts (CEFs) from a heterogeneous cell pool containing chicken satellite cells (CSCs), over 90% of CEFs expressed CD29 and vimentin. The analysis of the proliferative capabilities of CEFs revealed no significant differences in EdU-positive cells (%), cumulative cell number, doubling time, and growth rate from passage 1 to passage 9 (p > 0.05). This indicates that CEFs can be isolated by 2 h of pre-plating and survive stably up to passage 9, and that primary fibroblasts can serve as a valuable cell source for the cultured meat industry. Adipogenic trans-differentiation was induced up to passage 9 of CEFs. As passages increased, lipid accumulation and adipocyte size significantly decreased (p < 0.05). The reduced differentiation rate of primary CEFs with increasing passages poses a major challenge to the cost and efficiency of cultured meat production. Thus, effective cell management and the maintenance of cellular characteristics for a long time are crucial for ensuring stable and efficient cultured fat production in the cultured meat industry.
Collapse
Affiliation(s)
- So-Hee Kim
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Republic of Korea; (S.-H.K.); (C.-J.K.); (E.-Y.L.)
| | - Chan-Jin Kim
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Republic of Korea; (S.-H.K.); (C.-J.K.); (E.-Y.L.)
| | - Eun-Yeong Lee
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Republic of Korea; (S.-H.K.); (C.-J.K.); (E.-Y.L.)
| | - Young-Hwa Hwang
- Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Seon-Tea Joo
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Republic of Korea; (S.-H.K.); (C.-J.K.); (E.-Y.L.)
- Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea;
| |
Collapse
|
2
|
Hou T, Du M, Gao X, An M. Human Vascular Endothelial Cells Promote the Secretion of Vascularization Factors and Migration of Human Skin Fibroblasts under Co-Culture and Its Preliminary Application. Int J Mol Sci 2022; 23:ijms232213995. [PMID: 36430476 PMCID: PMC9697737 DOI: 10.3390/ijms232213995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
The good treatment of skin defects has always been a challenge in the medical field, and the emergence of tissue engineering skin provides a new idea for the treatment of injured skin. However, due to the single seed cells, the tissue engineering skin has the problem of slow vascularization at the premonitory site after implantation into the human body. Cell co-culture technology can better simulate the survival and communication environment of cells in the human body. The study of multicellular co-culture hopes to bring a solution to the problem of tissue engineering. In this paper, human skin fibroblasts (HSFs) and human vascular endothelial cells (HVECs) were co-cultured in Transwell. The Cell Counting Kit 8 (CCK8), Transwell migration chamber, immunofluorescence, Western blot (WB), and real time quantitative PCR (RT-qPCR) were used to study the effects of HVECs on cell activity, migration factor (high mobility group protein 1, HMGB1) and vascularization factor (vascular endothelial growth factor A, VEGFA and fibroblast growth factor 2, FGF2) secretion of HSFs after co-cultured with HVECs in the Transwell. The biological behavior of HSFs co-cultured with HVECs was studied. The experimental results are as follows: (1) The results of cck8 showed that HVECS could promote the activity of HSFs. (2) HVECs could significantly promote the migration of HSFs and promote the secretion of HMGB1. (3) HVECs could promote the secretion of VEGFA and FGF2 of HSFs. (4) The HVECs and HSFs were inoculated on tissue engineering scaffolds at the ratio of 1:4 and were co-cultured and detected for 7 days. The results showed that from the third day, the number of HSFs was significantly higher than that of the control group without HVECs.
Collapse
|
3
|
The Role of Genetically Modified Human Feeder Cells in Maintaining the Integrity of Primary Cultured Human Deciduous Dental Pulp Cells. J Clin Med 2022; 11:jcm11206087. [PMID: 36294410 PMCID: PMC9605397 DOI: 10.3390/jcm11206087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/30/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
Tissue-specific stem cells exist in tissues and organs, such as skin and bone marrow. However, their pluripotency is limited compared to embryonic stem cells. Culturing primary cells on plastic tissue culture dishes can result in the loss of multipotency, because of the inability of tissue-specific stem cells to survive in feeder-less dishes. Recent findings suggest that culturing primary cells in medium containing feeder cells, particularly genetically modified feeder cells expressing growth factors, may be beneficial for their survival and proliferation. Therefore, the aim of this study was to elucidate the role of genetically modified human feeder cells expressing growth factors in maintaining the integrity of primary cultured human deciduous dental pulp cells. Feeder cells expressing leukemia inhibitory factor, bone morphogenetic protein 4, and basic fibroblast growth factor were successfully engineered, as evidenced by PCR. Co-culturing with mitomycin-C-treated feeder cells enhanced the proliferation of newly isolated human deciduous dental pulp cells, promoted their differentiation into adipocytes and neurons, and maintained their stemness properties. Our findings suggest that genetically modified human feeder cells may be used to maintain the integrity of primary cultured human deciduous dental pulp cells.
Collapse
|
4
|
Yde Ohki CM, McNeill RV, Nieberler M, Radtke F, Kittel-Schneider S, Grünblatt E. Promising Developments in the Use of Induced Pluripotent Stem Cells in Research of ADHD. Curr Top Behav Neurosci 2022; 57:483-501. [PMID: 35543866 DOI: 10.1007/7854_2022_346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Although research using animal models, peripheral and clinical biomarkers, multimodal neuroimaging techniques and (epi)genetic information has advanced our understanding of Attention-Deficit Hyperactivity Disorder (ADHD), the aetiopathology of this neurodevelopmental disorder has still not been elucidated. Moreover, as the primary affected tissue is the brain, access to samples is problematic. Alternative models are therefore required, facilitating cellular and molecular analysis. Recent developments in stem cell research have introduced the possibility to reprogram somatic cells from patients, in this case ADHD, and healthy controls back into their pluripotent state, meaning that they can then be differentiated into any cell or tissue type. The potential to translate patients' somatic cells into stem cells, and thereafter to use 2- and 3-dimensional (2D and 3D) neuronal cells to model neurodevelopmental disorders and/or test novel drug therapeutics, is discussed in this chapter.
Collapse
Affiliation(s)
- Cristine Marie Yde Ohki
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zürich, Switzerland
| | - Rhiannon V McNeill
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Matthias Nieberler
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Franziska Radtke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Würzburg, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zürich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Zürich, Switzerland.
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland.
| |
Collapse
|
5
|
Nakashima Y, Omasa T. What Kind of Signaling Maintains Pluripotency and Viability in Human-Induced Pluripotent Stem Cells Cultured on Laminin-511 with Serum-Free Medium? Biores Open Access 2016; 5:84-93. [PMID: 27096107 PMCID: PMC4834485 DOI: 10.1089/biores.2016.0001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Xeno-free medium contains no animal-derived components, but is composed of minimal growth factors and is serum free; the medium may be supplemented with insulin, transferrin, and selenium (ITS medium). Serum-free and xeno-free culture of human-induced pluripotent stem cells (hiPSCs) uses a variety of components based on ITS medium and Dulbecco's modified Eagle's medium/Ham's nutrient mixture F12 (DMEM/F12) that contain high levels of iron salt and glucose. Culture of hiPSCs also requires scaffolding materials, such as extracellular matrix, collagen, fibronectin, laminin, proteoglycan, and vitronectin. The scaffolding component laminin-511, which is composed of α5, β1, and γ1 chains, binds to α3β1, α6β1, and α6β4 integrins on the cell membrane to induce activation of the PI3K/AKT- and Ras/MAPK-dependent signaling pathways. In hiPSCs, the interaction of laminin-511/α6β1 integrin with the cell–cell adhesion molecule E-cadherin confers protection against apoptosis through the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathways for cell death) and the proto-oncogene tyrosine-protein kinase Fyn (Fyn)-RhoA-ROCK signaling pathway. The expression levels of α6β1 integrin and E-cadherin on cell membranes are controlled through the activation of insulin receptor/insulin, FGF receptor/FGF2, or activin-like kinase 5 (ALK5)-dependent TGF-β signaling. A combination of growth factors, medium constituents, cell membrane-located E-cadherin, and α6β1 integrin-induced signaling is required for pluripotent cell proliferation and for optimal cell survival on a laminin-511 scaffold. In this review, we discuss and explore the influence of growth factors on the cadherin and integrin signaling pathways in serum-free and xeno-free cultures of hiPSCs during the preparation of products for regenerative medicinal therapies. In addition, we suggest the optimum serum-free medium components for use with laminin-511, a new scaffold for hiPSC culture.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| | - Takeshi Omasa
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| |
Collapse
|
6
|
Yang H, Qiu Y, Zeng X, Ding Y, Zeng J, Lu K, Li D. Effect of a feeder layer composed of mouse embryonic and human foreskin fibroblasts on the proliferation of human embryonic stem cells. Exp Ther Med 2016; 11:2321-2328. [PMID: 27313670 DOI: 10.3892/etm.2016.3204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 10/29/2015] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to investigate the effects of feeder layers composed of various ratios of mouse embryonic fibroblasts (MEFs) and human foreskin fibroblasts (hFFs) on the growth of human embryonic stem cells (hESCs). In addition, the secretion levels of basic fibroblast growth factor (bFGF) by the feeder layers was detected. MEFs and hFFs were treated with mitomycin C and seeded onto gelatin-coated plates at a density of 1×108 cells/l. The hFFs and MEFs were combined and plated at the following ratios: 0:1, 1:2, 1:1, 2:1 and 1:0. The secretion of bFGF by the various hFF/MEF ratio feeder layers was detected using an enzyme-linked immunosorbent assay. Subsequently, hESCs were cultured on top of the various feeder layers. The differences in the cellular morphology of the hESCs were observed using microscopy, and the expression levels alkaline phosphatase (AKP) and octamer-binding transcription factor 4 (OCT-4) were detected using immunohistochemical analysis as indicators of differentiation status. The results showed that the hFFs secreted substantial quantities of bFGF, while no bFGF was secreted by the MEFs. The clones of hESC growing on the feeder layer containing MEF or hFF alone were flat. By contrast, hESC clones grown on a mixed feeder layer containing hFFs + MEFs at a ratio of 1:1 exhibited an accumulated growth with a clear edge, as compared with the other ratios. In addition, hESCs growing on the feeder layer were positive for the expression of AKP and OCT-4. In summary, feeder layer hFFs secreted bFGF, while MEFs did not, indicating that bFGF is not the only factor that supports the growth and differentiation of hESCs. The optimal growth of hESCs was achieved using a mixed feeder layer composed of hFFs + MEFs at a ratio of 1:1.
Collapse
Affiliation(s)
- Hua Yang
- Reproductive Medical Center of Nanning Second People's Hospital, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530031, P.R. China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi 530004, P.R. China
| | - Ying Qiu
- Reproductive Medical Center of Nanning Second People's Hospital, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530031, P.R. China
| | - Xianghui Zeng
- Reproductive Medical Center of Nanning Second People's Hospital, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530031, P.R. China
| | - Yan Ding
- Life Science Research Institute, Taihe Hospital, Shiyan, Hubei 442000, P.R. China
| | - Jianye Zeng
- Reproductive Medical Center of Nanning Second People's Hospital, Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530031, P.R. China
| | - Kehuan Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi 530004, P.R. China
| | - Dongsheng Li
- Life Science Research Institute, Taihe Hospital, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
7
|
Human Umbilical Cord Blood-Derived Serum for Culturing the Supportive Feeder Cells of Human Pluripotent Stem Cell Lines. Stem Cells Int 2015; 2016:4626048. [PMID: 26839561 PMCID: PMC4709772 DOI: 10.1155/2016/4626048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/02/2015] [Accepted: 09/13/2015] [Indexed: 12/24/2022] Open
Abstract
Although human pluripotent stem cells (hPSCs) can proliferate robustly on the feeder-free culture system, genetic instability of hPSCs has been reported in such environment. Alternatively, feeder cells enable hPSCs to maintain their pluripotency. The feeder cells are usually grown in a culture medium containing fetal bovine serum (FBS) prior to coculture with hPSCs. The use of FBS might limit the clinical application of hPSCs. Recently, human cord blood-derived serum (hUCS) showed a positive effect on culture of mesenchymal stem cells. It is interesting to test whether hUCS can be used for culture of feeder cells of hPSCs. This study was aimed to replace FBS with hUCS for culturing the human foreskin fibroblasts (HFFs) prior to feeder cell preparation. The results showed that HFFs cultured in hUCS-containing medium (HFF-hUCS) displayed fibroblastic features, high proliferation rates, short population doubling times, and normal karyotypes after prolonged culture. Inactivated HFF-hUCS expressed important genes, including Activin A, FGF2, and TGFβ1, which have been implicated in the maintenance of hPSC pluripotency. Moreover, hPSC lines maintained pluripotency, differentiation capacities, and karyotypic stability after being cocultured for extended period with inactivated HFF-hUCS. Therefore, the results demonstrated the benefit of hUCS for hPSCs culture system.
Collapse
|
8
|
Kim MH, Masuda E, Kino-oka M. Kinetic analysis of deviation from the undifferentiated state in colonies of human induced pluripotent stem cells on feeder layers. Biotechnol Bioeng 2014; 111:1128-38. [DOI: 10.1002/bit.25188] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 12/24/2013] [Accepted: 01/07/2014] [Indexed: 01/18/2023]
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering; Osaka University; Osaka 565-0871 Japan
| | - Eri Masuda
- Department of Biotechnology, Graduate School of Engineering; Osaka University; Osaka 565-0871 Japan
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering; Osaka University; Osaka 565-0871 Japan
| |
Collapse
|
9
|
Liu CX, Zhang RL, Gao J, Li T, Ren Z, Zhou CQ, Wen AM. Derivation of human embryonic stem cell lines without any exogenous growth factors. Mol Reprod Dev 2014; 81:470-9. [PMID: 24554631 DOI: 10.1002/mrd.22312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 02/11/2014] [Indexed: 01/16/2023]
Abstract
Human embryonic stem cell (hESC) lines are traditionally derived through immunosurgery. Their maintenance in culture requires the presence of mouse embryonic fibroblasts (MEFs) as feeder cells and media supplemented with basic fibroblast growth factor (bFGF) or other growth factors-both of which might introduce animal-derived culture components. The drawbacks associated with immunosurgery, MEF co-culture, and the cost of growth factors necessitate the exploration of a xeno-free method to maintain the self-renewal capacity of hESCs. Here, we describe an isolation method for the human inner cell mass (ICM), which was then cultured in the absence of exogenous growth factors and in the presence of human foreskin fibroblasts (HFFs) as feeder cells. Three hESC lines were obtained from poor-quality embryos by this near-xeno-free protocol. After culturing for more than 10 months, the hESCs retained normal morphology, expressed all expected cell surface markers, could differentiate to embryoid bodies upon culture in vitro, and formed teratomas in vivo. Furthermore, secretion of bFGF by HFFs was observed. In conclusion, this is the first study to describe an inexpensive, xeno-free culture system for the isolation and maintenance of hESCs that does not require bFGF supplementation.
Collapse
Affiliation(s)
- Cai Xia Liu
- Reproductive Medicine Center, Guangdong Academy of Medical Sciences/Guangdong General Hospital, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Wang X, Li S, Cao T, Fu X, Yu G. Evaluating biotoxicity with fibroblasts derived from human embryonic stem cells. Toxicol In Vitro 2012; 26:1056-63. [DOI: 10.1016/j.tiv.2012.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 02/19/2012] [Accepted: 04/04/2012] [Indexed: 12/28/2022]
|
11
|
HU JIABO, HU SANQIANG, MA QUANHUI, WANG XIAOHUI, ZHOU ZHONGWEI, ZHANG WEI, SUN XIAOCHUN, ZHU WEI, QIAN HUI, XU WENRONG. Immortalized mouse fetal liver stromal cells support growth and maintenance of human embryonic stem cells. Oncol Rep 2012; 28:1385-91. [DOI: 10.3892/or.2012.1909] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/19/2012] [Indexed: 11/06/2022] Open
|
12
|
Saitoh I, Sato M, Iwase Y, Inada E, Nomura T, Akasaka E, Yamasaki Y, Noguchi H. Generation of Mouse STO Feeder Cell Lines That Confer Resistance to Several Types of Selective Drugs. CELL MEDICINE 2012; 3:97-102. [PMID: 28058186 DOI: 10.3727/215517912x639414] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Feeder cells are generally required for establishment and maintenance of embryonic stem (ES)/induced pluripotent stem (iPS) cells. Increased demands for generation of those cells carrying various types of vectors (i.e., KO vectors and transgenes) also require feeder cells that confer resistance to any types of preexisting selective drugs. Unfortunately, the use of the feeders that are resistant to various drugs appears to be limited to a few laboratories. Here we generated a set of gene-engineered STO feeder cells that confer resistance to several commercially available drugs. The STO cells, which have long been used as a feeder for mouse ES and embryonal carcinoma (EC) cells, were transfected with pcBIH [carrying bleomycin resistance gene (ble) and hygromycin B phosphotransferase gene (Hyg)], pcBIP [carrying ble and puromycin resistance gene (puro)], or pcBSN [carrying ble and neomycin resistance gene (neo)]. The resulting stably transfectants (termed SHB for pcBIH, SPB for pcBIP, and SNB for pcBSN) exhibited bleomycin/hygromycin, bleomycin/puromycin, or bleomycin/neomycin, as expected. The morphology of these cells passaged over 18 generations was indistinguishable from that of parental STO cells. Of isolated clones, the SHB3, SPB3, and SNB2 clones successfully supported the growth of mouse ES cells in an undifferentiated state, when coculture was performed. PCR analysis revealed the presence of the selective markers in these clones, as expected. These SHB3, SPB3, and SNB2 cells will thus be useful for the acquisition and maintenance of genetically manipulated ES/iPS cells.
Collapse
Affiliation(s)
- Issei Saitoh
- Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshimashi, Kagoshima , Japan
| | - Masahiro Sato
- † Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University , Korimoto, Kagoshima, Kagoshima , Japan
| | - Yoko Iwase
- ‡ Department of Dental Anesthesia, Kagoshima University Medical and Dental Hospital , Sakuragaoka, Kagoshimashi, Kagoshima , Japan
| | - Emi Inada
- Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshimashi, Kagoshima , Japan
| | - Toshiki Nomura
- Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshimashi, Kagoshima , Japan
| | - Eri Akasaka
- Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshimashi, Kagoshima , Japan
| | - Youichi Yamasaki
- Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshimashi, Kagoshima , Japan
| | - Hirofumi Noguchi
- § Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Shikata-cho, Okayama , Japan
| |
Collapse
|
13
|
Lee EJ, Kang HJ, Lee HN, Kang SK, Kim KH, Lee SW, Lee G, Park YB, Kim HS. New culture system for human embryonic stem cells: autologous mesenchymal stem cell feeder without exogenous fibroblast growth factor 2. Differentiation 2011; 83:92-100. [PMID: 22099180 DOI: 10.1016/j.diff.2011.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 09/27/2011] [Accepted: 09/28/2011] [Indexed: 01/14/2023]
Abstract
Human embryonic stem (hES) cells have been successfully maintained using human-cell feeder systems or feeder-free systems. However, despite advances in culture techniques, hES cells require supplementation with fibroblast growth factor 2 (FGF-2), an exogenous stemness factor, which is needed to sustain the authentic undifferentiated status. We developed a new culture system for hES cells; this system does not require supplementation with FGF-2 to obtain hES cells that are suitable for tissue engineering and regenerative medicine. This culture system employed mesenchymal stem cells derived from hES cells (hESC-MSCs) as autologous human feeder cells in the absence of FGF-2. The hES cell line SNUhES3 cultured in this new autologous feeder culture system maintained the typical morphology of hES cells and expression of pluripotency-related proteins, SSEA-4, TRA-1-60, OCT4, and alkaline phosphatase, without development of abnormal karyotypes after more than 30 passages. RNA expression of the pluripotency-related genes OCT4 and NANOG was similar to the expression in SNUhES3 cells maintained on xenofeeder STO cells. To identify the mechanism that enables the cells to be maintained without exogenous FGF-2, we checked the secretion of FGF-2 from the mitomycin-C treated autofeeder hESC-MSCs versus xenofeeder STO cells, and confirmed that hESC-MSCs secreted FGF-2 whereas STO cells did not. The level of FGF-2 in the media from the autofeeder system without exogenous FGF-2 was comparable to that from the xenofeeder system with addition of FGF-2. In conclusion, our new culture system for hES cells, which employs a feeder layer of autologous hESC-MSCs, supplies sufficient amounts of secreted FGF-2 to eliminate the requirement for exogenous FGF-2.
Collapse
Affiliation(s)
- Eun Ju Lee
- National Research Laboratory for Stem Cell Niche and IRICT, Seoul National University Hospital, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Mamidi MK, Pal R, Mori NAB, Arumugam G, Thrichelvam ST, Noor PJ, Abdullah HMF, Gupta PK, Das AK, Zakaria Z, Bhonde R. Co-culture of mesenchymal-like stromal cells derived from human foreskin permits long term propagation and differentiation of human embryonic stem cells. J Cell Biochem 2011; 112:1353-63. [DOI: 10.1002/jcb.23052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
15
|
Xi J, Wang Y, Zhang P, He L, Nan X, Yue W, Pei X. Human fetal liver stromal cells that overexpress bFGF support growth and maintenance of human embryonic stem cells. PLoS One 2010; 5:e14457. [PMID: 21209880 PMCID: PMC3012692 DOI: 10.1371/journal.pone.0014457] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 12/07/2010] [Indexed: 12/23/2022] Open
Abstract
In guiding hES cell technology toward the clinic, one key issue to be addressed is to culture and maintain hES cells much more safely and economically in large scale. In order to avoid using mouse embryonic fibroblasts (MEFs) we isolated human fetal liver stromal cells (hFLSCs) from 14 weeks human fetal liver as new human feeder cells. hFLSCs feeders could maintain hES cells for 15 passages (about 100 days). Basic fibroblast growth factor (bFGF) is known to play an important role in promoting self-renewal of human embryonic stem (hES) cells. So, we established transgenic hFLSCs that stably express bFGF by lentiviral vectors. These transgenic human feeder cells — bFGF-hFLSCs maintained the properties of H9 hES cells without supplementing with any exogenous growth factors. H9 hES cells culturing under these conditions maintained all hES cell features after prolonged culture, including the developmental potential to differentiate into representative tissues of all three embryonic germ layers, unlimited and undifferentiated proliferative ability, and maintenance of normal karyotype. Our results demonstrated that bFGF-hFLSCs feeder cells were central to establishing the signaling network among bFGF, insulin-like growth factor 2 (IGF-2), and transforming growth factor β (TGF-β), thereby providing the framework in which hES cells were instructed to self-renew or to differentiate. We also found that the conditioned medium of bFGF-hFLSCs could maintain the H9 hES cells under feeder-free conditions without supplementing with bFGF. Taken together, bFGF-hFLSCs had great potential as feeders for maintaining pluripotent hES cell lines more safely and economically.
Collapse
Affiliation(s)
- Jiafei Xi
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Yunfang Wang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Peng Zhang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Lijuan He
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Xue Nan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, China
- * E-mail: (WY); (XP)
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, China
- * E-mail: (WY); (XP)
| |
Collapse
|
16
|
Pan C, Hicks A, Guan X, Chen H, Bishop CE. SNL fibroblast feeder layers support derivation and maintenance of human induced pluripotent stem cells. J Genet Genomics 2010; 37:241-8. [PMID: 20439100 DOI: 10.1016/s1673-8527(09)60042-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 01/22/2010] [Accepted: 01/26/2010] [Indexed: 01/09/2023]
Abstract
Induced pluripotent stem (iPS) cells can be derived from human somatic cells by cellular reprogramming. This technology provides a potential source of non-controversial therapeutic cells for tissue repair, drug discovery, and opportunities for studying the molecular basis of human disease. Normally, mouse embryonic fibroblasts (MEFs) are used as feeder layers in the initial derivation of iPS lines. The purpose of this study was to determine whether SNL fibroblasts can be used to support the growth of human iPS cells reprogrammed from somatic cells using lentiviral expressed reprogramming factors. In our study, iPS cells expressed common pluripotency markers, displayed human embryonic stem cells (hESCs) morphology and unmethylated promoters of NANOG and OCT4. These data demonstrate that SNL feeder cells can support the derivation and maintenance of human iPS cells.
Collapse
Affiliation(s)
- Chuanying Pan
- College of Animal Science and Technology, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Northwest A & F University, Yangling 712100, China
| | | | | | | | | |
Collapse
|
17
|
Fu X, Toh WS, Liu H, Lu K, Li M, Hande MP, Cao T. Autologous Feeder Cells from Embryoid Body Outgrowth Support the Long-Term Growth of Human Embryonic Stem Cells More Effectively than Those from Direct Differentiation. Tissue Eng Part C Methods 2010; 16:719-33. [DOI: 10.1089/ten.tec.2009.0360] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Xin Fu
- Stem Cell Laboratory, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Wei Seong Toh
- Stem Cell Laboratory, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Hua Liu
- Stem Cell Laboratory, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Kai Lu
- Stem Cell Laboratory, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Mingming Li
- Stem Cell Laboratory, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Manoor Prakash Hande
- Department of Physiology, Faculty of Medicine, National University of Singapore, Singapore, Singapore
| | - Tong Cao
- Stem Cell Laboratory, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| |
Collapse
|
18
|
Browning LM, Huang T, Xu XN. Electric pulses to prepare feeder cells for sustaining and culturing of undifferentiated embryonic stem cells. Biotechnol J 2010; 5:588-95. [PMID: 20518062 PMCID: PMC3014325 DOI: 10.1002/biot.201000040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Current challenges in embryonic-stem cell (ESC) research include the inability of sustaining and culturing of undifferentiated ESCs over time. Growth-arrested feeder cells are essential to the culture and sustaining of undifferentiated ESCs, and they are currently prepared using gamma-radiation and chemical inactivation. Both techniques have severe limitations. In this study, we developed a new, simple and effective technique (pulsed electric fields, PEFs) to produce viable growth-arrested cells (RTS34st) and used them as high-quality feeder cells to culture and sustain undifferentiated zebrafish ESCs over time. The cells were exposed to 25 sequential 10-ns electric pulses (10nsEPs) of 25, 40 and 150 kV/cm with 1-s pulse interval, or 2 sequential 50-mus electric pulses (50microsEPs) of 2.83, 1.78 and 0.78 kV/cm with 5-s pulse interval, respectively. We found that the cellular effects of PEFs depended directly upon the duration, number and electric field strength of the pulses, showing the feasibility of tuning them to produce various types of growth-arrested cells for culturing undifferentiated ESCs. Both 10nsEPs of 40 kV/cm produced by a 10nsEP generator and 50microsEPs of 1.78 kV/cm provided by inexpensive and widely available conventional electroporators, generated high-quality growth-arrested feeder cells for proliferation of undifferentiated ESCs over time. PEFs can therefore be used to replace radiation and chemical inactivation methods for preparation of growth-arrested feeder cells for advancing ESC research.
Collapse
Affiliation(s)
- Lauren M. Browning
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, Virginia 23529
| | - Tao Huang
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, Virginia 23529
| | - X. Nancy Xu
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, Virginia 23529
| |
Collapse
|
19
|
Camarasa M, Brison D, Kimber SJ, Handyside AH. Naturally immortalised mouse embryonic fibroblast lines support human embryonic stem cell growth. CLONING AND STEM CELLS 2009; 11:453-62. [PMID: 19594387 DOI: 10.1089/clo.2008.0082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human embryonic stem cell (hESC) growth is dependent on various factors released by feeder cells. Some of them have already been elucidated, although much research is still needed to understand the biology of stem cell maintenance in culture. Traditionally, primary mouse embryonic fibroblasts (PMEFs) have been used as feeder layers, and both murine and human fibroblast cell lines have been shown to support pluripotency and self-renewal of hESC. Here we report the derivation of three new mouse embryonic fibroblast cell lines, MEFLU-M, MEFLU-T, and MEFLU-TB, with different properties regarding growth and support for undifferentiated hESCs. MEFLU-TB is able to support continuous growth of the newly derived Man-1, as well as H1, HUES-1, HUES-7, HUES-8, and HUES-9 human embryonic stem cell lines. After more than 50 passages and doublings, MEFLU-TB feeders compare to early passage primary mouse embryonic fibroblasts in their ability to support undifferentiated hESC growth. Our results contradict a previous paradigm that PMEFs tend to lose their capacity to support proliferation of hESCs with increasing passages, and show that the MEFLU-TB mouse embryonic fibroblast cell line and its conditioned medium have the potential to support the maintenance of hESC lines. Also, our results clearly show that spontaneous immortalization of primary fibroblasts can be achieved in culture without any chemical addition or genetic modification.
Collapse
Affiliation(s)
- Mavi Camarasa
- North West Embryonic Stem Cell Centre, Faculty of life Sciences, University of Manchester, Manchester, United Kingdom.
| | | | | | | |
Collapse
|
20
|
Sivasubramaiyan K, Totey S, Bhat V, Totey SM, Deb K. Y-27632 enhances differentiation of blastocyst like cystic human embryoid bodies to endocrinologically active trophoblast cells on a biomimetic platform. J Biomed Sci 2009; 16:88. [PMID: 19772618 PMCID: PMC2754421 DOI: 10.1186/1423-0127-16-88] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 09/22/2009] [Indexed: 11/10/2022] Open
Abstract
Trophoblast differentiation and formation of the placenta are important events linked to post-implantation embryonic development. Models mimicking the biology of trophoblast differentiation in a post-implantation maternal microenvironment are needed for understanding disorders like placental-ischemia or for applications in drug-screening, and would help in overcoming the ethical impasse on using human embryos for such research. Here we attempt to create such a model by using embryoid bodies (EBs) and a biomimetic platform composed of a bilayer of fibronectin and gelatin on top of low-melting agarose. Using this model we test the hypothesis that cystic-EBs (day 30) that resemble blastocysts morphologically, are better sources as compared to noncytic EBs (day 10), for functional trophoblast differentiation; and that the Rho kinases inhibitor Y27632 can enhance this differentiation. Non/cytic EBs with/out Y27632 were grown on this platform for 28 days, and screened from secretion and expression of trophoblast and other lineage markers using ECLIA, RT-PCR, and Immunofluorescence. All EBs attached on this surface and rapidly proliferated into hCG and progesterone (P2) secreting functional trophoblast cells. However, the cells derived from cytic-EBs and cytic-EBs+ Y27632 showed the maximum secretion of these hormones and expressed IGF2, supporting our hypothesis. Also Y27632 reduced extraembryonic endoderm and trophoblast lineage differentiation from early noncystic-EBs, whereas, it specifically enhanced the induction of trophoblast and multinucleated syncitiotrophoblast differentiation from late cystic-EBs. In vivo trophoblast differentiation can be replicated in fibronectin based biomaterials, using cytic-EBs and by maneuvering the Rho-ROCK pathways. Response of EBs to a compound may vary temporally, and determination of their right stage is crucial for applications in directed-differentiation or drug-screening.
Collapse
|
21
|
Immortalized human skin fibroblast feeder cells support growth and maintenance of both human embryonic and induced pluripotent stem cells. Hum Reprod 2009; 24:2567-81. [DOI: 10.1093/humrep/dep232] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
22
|
Schneider A, Spitkovsky D, Riess P, Molcanyi M, Kamisetti N, Maegele M, Hescheler J, Schaefer U. "The good into the pot, the bad into the crop!"--a new technology to free stem cells from feeder cells. PLoS One 2008; 3:e3788. [PMID: 19023443 PMCID: PMC2582950 DOI: 10.1371/journal.pone.0003788] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 10/22/2008] [Indexed: 01/11/2023] Open
Abstract
A variety of embryonic and adult stem cell lines require an initial co-culturing with feeder cells for non-differentiated growth, self renewal and maintenance of pluripotency. However for many downstream ES cell applications the feeder cells have to be considered contaminations that might interfere not just with the analysis of experimental data but also with clinical application and tissue engineering approaches. Here we introduce a novel technique that allows for the selection of pure feeder-freed stem cells, following stem cell proliferation on feeder cell layers. Complete and reproducible separation of feeder and embryonic stem cells was accomplished by adaptation of an automated cell selection system that resulted in the aspiration of distinct cell colonies or fraction of colonies according to predefined physical parameters. Analyzing neuronal differentiation we demonstrated feeder-freed stem cells to exhibit differentiation potentials comparable to embryonic stem cells differentiated under standard conditions. However, embryoid body growth as well as differentiation of stem cells into cardiomyocytes was significantly enhanced in feeder-freed cells, indicating a feeder cell dependent modulation of lineage differentiation during early embryoid body development. These findings underline the necessity to separate stem and feeder cells before the initiation of in vitro differentiation. The complete separation of stem and feeder cells by this new technology results in pure stem cell populations for translational approaches. Furthermore, a more detailed analysis of the effect of feeder cells on stem cell differentiation is now possible, that might facilitate the identification and development of new optimized human or genetically modified feeder cell lines.
Collapse
Affiliation(s)
- Annette Schneider
- Institute for Research in Operative Medicine (IFOM), Faculty of Medicine, University of Witten/Herdecke, Witten, Germany
| | - Dimitry Spitkovsky
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Peter Riess
- Department of Trauma and Orthopedic Surgery, Faculty of Medicine, University of Witten-Herdecke at the Hospital Merheim, Cologne, Germany
| | - Marek Molcanyi
- Clinic of Neurosurgery and 2nd Department of Neurophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Naidu Kamisetti
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Marc Maegele
- Department of Trauma and Orthopedic Surgery, Faculty of Medicine, University of Witten-Herdecke at the Hospital Merheim, Cologne, Germany
| | - Jürgen Hescheler
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Ute Schaefer
- Department of Experimental Neurotraumatology, Medical University Graz, Graz, Austria
- * E-mail:
| |
Collapse
|