1
|
Grogan S, Kopcow J, D’Lima D. Challenges Facing the Translation of Embryonic Stem Cell Therapy for the Treatment of Cartilage Lesions. Stem Cells Transl Med 2022; 11:1186-1195. [PMID: 36493381 PMCID: PMC9801304 DOI: 10.1093/stcltm/szac078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 10/02/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis is a common disease resulting in significant disability without approved disease-modifying treatment (other than total joint replacement). Stem cell-based therapy is being actively explored for the repair of cartilage lesions in the treatment and prevention of osteoarthritis. Embryonic stem cells are a very attractive source as they address many of the limitations inherent in autologous stem cells, such as variability in function and limited expansion. Over the past 20 years, there has been widespread interest in differentiating ESC into mesenchymal stem cells and chondroprogenitors with successful in vitro, ex vivo, and early animal studies. However, to date, none have progressed to clinical trials. In this review, we compare and contrast the various approaches to differentiating ESC; and discuss the benefits and drawbacks of each approach. Approaches relying on spontaneous differentiation are simpler but not as efficient as more targeted approaches. Methods replicating developmental biology are more efficient and reproducible but involve many steps in a complicated process. The small-molecule approach, arguably, combines the advantages of the above two methods because of the relative efficiency, reproducibility, and simplicity. To better understand the reasons for lack of progression to clinical applications, we explore technical, scientific, clinical, and regulatory challenges that remain to be overcome to achieve success in clinical applications.
Collapse
Affiliation(s)
- Shawn Grogan
- Corresponding author: Darryl D’Lima, MD, PhD, Shiley Center for Orthopaedic Research and Education, Scripps Health, 10666 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Joel Kopcow
- Shiley Center for Orthopaedic Research and Education, Scripps Health, La Jolla, CA, USA
| | - Darryl D’Lima
- Shiley Center for Orthopaedic Research and Education, Scripps Health, La Jolla, CA, USA
| |
Collapse
|
2
|
The specialized mitotic behavior of human embryonic stem cells. Cell Tissue Res 2021; 387:85-93. [PMID: 34729647 DOI: 10.1007/s00441-021-03544-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 10/08/2021] [Indexed: 10/19/2022]
Abstract
Human embryonic stem cells (hESCs) are self-renewing and pluripotent cells that originate from the inner cell mass of the blastocyst. Mitosis is fundamental to organism survival and reproduction and is responsible for the equal distribution of duplicated chromosomes into daughter cells. Mitotic dysfunction is associated with a wide variety of human diseases, not least cancer. hESCs have a unique cell cycle distribution, but it is unclear exactly how the mitotic activity of hESCs is related to their proliferation and differentiation. Here, we established a cell line of hESCs stably expressing GFP-α-tubulin and mCherry-H2B by lentiviral infection to analyze and visualize mitosis in detail. During metaphase, the mitotic spindle was smaller and wider and contained a greater proportion of astral microtubules than normal cells. In addition, spindle microtubules were more stable, and chromosome alignment was faster in hESCs than in somatic cells. We also found that the spindle assembly checkpoint was functional in hESCs. These findings thus reveal a specialized mitotic behavior of hESCs.
Collapse
|
3
|
Baek SK, Jeon SB, Seo BG, Hwangbo C, Shin KC, Choi JW, An CS, Jeong MA, Kim TS, Lee JH. The Presence or Absence of Alkaline Phosphatase Activity to Discriminate Pluripotency Characteristics in Porcine Epiblast Stem Cell-Like Cells. Cell Reprogram 2021; 23:221-238. [PMID: 34227846 DOI: 10.1089/cell.2021.0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Porcine embryonic stem cells (pESCs) would provide potentials for agricultural- and biotechnological-related applications. However, authentic pESCs have not been established yet because standards for porcine stem cell-specific markers and culture conditions are not clear. Therefore, the present study reports attempts to derive pluripotent epiblast stem cells either from in vitro or in vivo derived porcine embryos. Nine epiblast cell lines (seven lines from Berkshire and two lines from Duroc) could only be isolated from day 9- to 9.5-old in vivo derived early conceptuses. Pluripotency features were analyzed in relation to the presence or absence of alkaline phosphatase (AP) activity. Interestingly, the mRNA expression of several marker genes for pluripotency or epiblast was different between putative epiblast stem cells of the two groups [AP-positive (+) pEpiSC-like cell 2 line and AP-negative (-) pEpiSC-like cell 8 line]. For example, expressions of OCT-3/4, NANOG, SOX2, c-MYC, FGF2, and NODAL in AP-negative (-) porcine epiblast stem cell (pEpiSC)-like cells were higher than those in AP-positive (+) pEpiSC-like cells. Expression of surface markers differed between the two groups to some extent. SSEA-1 was strongly expressed only in AP-negative (-) pEpiSC-like cells, whereas AP-positive (+) pEpiSC-like cells did not express. In addition, we report to have some differences in the in vitro differentiation capacity between AP-positive (+) and AP-negative (-) epiblast cell lines. Primary embryonic germ layer markers (cardiac actin, nestin, and GATA 6) and primordial germ cell markers (Dazl and Vasa) were strongly expressed in embryoid bodies (EBs) aggregated from AP-negative (-) pEpiSC-like cells, whereas EBs aggregated from AP-positive (+) pEpiSCs did not show expression of primary embryonic germ layers and primordial germ cell markers except GATA 6. These results indicate that pEpiSC-like cells display different pluripotency characteristics in relation to AP activity.
Collapse
Affiliation(s)
- Sang-Ki Baek
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Soo-Been Jeon
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Bo-Gyeong Seo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Cheol Hwangbo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Keum-Chul Shin
- Institute of Agriculture & Life Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Department of Forest Environmental Resources, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Jung-Woo Choi
- College of Animal Life Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Chang-Seop An
- Gyeongsangnamdo Livestock Experiment Station, Sancheong, Republic of Korea
| | - Mi-Ae Jeong
- Gyeongsangnamdo Livestock Experiment Station, Sancheong, Republic of Korea
| | - Tae-Suk Kim
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Joon-Hee Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea.,Institute of Agriculture & Life Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
4
|
Yi JK, Park S, Ha JJ, Kim DH, Huang H, Park SJ, Lee MH, Ryoo ZY, Kim SH, Kim MO. Effects of Dimethyl Sulfoxide on the Pluripotency and Differentiation Capacity of Mouse Embryonic Stem Cells. Cell Reprogram 2020; 22:244-253. [DOI: 10.1089/cell.2020.0006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- Jun-Koo Yi
- Department of Embryo Transfer Research, Gyeongbuk Livestock Research Institute, Yeongju, Korea
| | - Song Park
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea
| | - Jae-Jung Ha
- Department of Embryo Transfer Research, Gyeongbuk Livestock Research Institute, Yeongju, Korea
| | - Dae-Hyun Kim
- Department of Embryo Transfer Research, Gyeongbuk Livestock Research Institute, Yeongju, Korea
| | - Hai Huang
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Korea
| | - Si-Jun Park
- Department of Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, Korea
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju, Jeollanamdo, Korea
- China-US (Henan) Hormel Cancer Institute, No. 127 Dongming Road, Zhengzhou, Henan, China
| | - Zae-Young Ryoo
- Department of Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, Korea
| | - Sung-Hyun Kim
- Life Medicine Analysis Korea Polytechnics Institute, Nonsan, Korea
| | - Myoung-Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Korea
| |
Collapse
|
5
|
Kumar S, Curran JE, DeLeon E, Leandro AC, Howard TE, Lehman DM, Williams-Blangero S, Glahn DC, Blangero J. Role of miRNA-mRNA Interaction in Neural Stem Cell Differentiation of Induced Pluripotent Stem Cells. Int J Mol Sci 2020; 21:ijms21196980. [PMID: 32977388 PMCID: PMC7582477 DOI: 10.3390/ijms21196980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 11/16/2022] Open
Abstract
miRNA regulates the expression of protein coding genes and plays a regulatory role in human development and disease. The human iPSCs and their differentiated progenies provide a unique opportunity to identify these miRNA-mediated regulatory mechanisms. To identify miRNA-mRNA regulatory interactions in human nervous system development, well characterized NSCs were differentiated from six validated iPSC lines and analyzed for differentially expressed (DE) miRNome and transcriptome by RNA sequencing. Following the criteria, moderated t statistics, FDR-corrected p-value ≤ 0.05 and fold change-absolute (FC-abs) ≥2.0, 51 miRNAs and 4033 mRNAs were found to be significantly DE between iPSCs and NSCs. The miRNA target prediction analysis identified 513 interactions between 30 miRNA families (mapped to 51 DE miRNAs) and 456 DE mRNAs that were paradoxically oppositely expressed. These 513 interactions were highly enriched in nervous system development functions (154 mRNAs; FDR-adjusted p-value range: 8.06 × 10-15-1.44 × 10-4). Furthermore, we have shown that the upregulated miR-10a-5p, miR-30c-5p, miR23-3p, miR130a-3p and miR-17-5p miRNA families were predicted to down-regulate several genes associated with the differentiation of neurons, neurite outgrowth and synapse formation, suggesting their role in promoting the self-renewal of undifferentiated NSCs. This study also provides a comprehensive characterization of iPSC-generated NSCs as dorsal neuroepithelium, important for their potential use in in vitro modeling of human brain development and disease.
Collapse
Affiliation(s)
- Satish Kumar
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (E.D.); (S.W.-B.)
- Correspondence:
| | - Joanne E. Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.E.C.); (A.C.L.); (T.E.H.); (J.B.)
| | - Erica DeLeon
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (E.D.); (S.W.-B.)
| | - Ana C. Leandro
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.E.C.); (A.C.L.); (T.E.H.); (J.B.)
| | - Tom E. Howard
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.E.C.); (A.C.L.); (T.E.H.); (J.B.)
| | - Donna M. Lehman
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
| | - Sarah Williams-Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (E.D.); (S.W.-B.)
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.E.C.); (A.C.L.); (T.E.H.); (J.B.)
| | - David C. Glahn
- Department of Psychiatry, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
- Olin Neuropsychiatric Research Center, Institute of Living, Hartford, CT 06102, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.E.C.); (A.C.L.); (T.E.H.); (J.B.)
| |
Collapse
|
6
|
Sánchez-Maldonado B, Galicia MDL, Rojo C, González-Gil A, Flor-García M, Picazo RA. Spheroids Spontaneously Generated In Vitro from Sheep Ovarian Cortical Cells Contain Integrating Cells That Exhibit Hallmarks of Neural Stem/Progenitor Cells. Stem Cells Dev 2018; 27:1557-1576. [PMID: 30251912 DOI: 10.1089/scd.2017.0141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cell spheroids are inducible or spontaneously generated cell aggregates produced in vitro that can provide a valuable model for developmental biology, stem cell biology, and cancer therapy research. This investigation aimed to define the cellular identity of spheroids spontaneously generated in vitro from sheep ovarian cortical cells cultured under specific serum-free conditions. Spheroids were characterized during 21 days of culture by morphometric evaluation, detection of alkaline phosphatase (AP) activity, gene expression analyses of stemness transcription factors and several lineage markers, immunolocalization analyses, as well as assessment of self-renewal and differentiation potential. Cell aggregation, evidenced from day 3 of culture onward, resulted in efficient generation of 65-75 spheroids for every 500,000 cells seeded. The spheroids reached maximum diameter (187 ± 15.9 μm) during the second week of culture and exhibited AP activity. Sox2, Oct4, and Nanog were expressed throughout the culture period, with upregulation of Sox2. Neural lineage specification genes (eg, nestin, vimentin, Pax6, and p75NTR) were expressed from day 10 onward at levels above that of Oct4, Nanog and those for endoderm [alpha-fetoprotein (AFP)], and mesoderm (brachyury) specification. Neural stem cell (NSC)/neural progenitor cell (NPC) markers, nestin, Pax6, p75NTR, and vimentin, were extensively localized in cells on day 10, 15 (44.75% ± 5.84%; 93.54% ± 1.35%; 78.90% ± 4.80%; 73.82% ± 3.40%, respectively), and 21 (49.98% ± 5.30%; 91.84% ± 1.9%; 76.74% ± 11.0%; 95.80% ± 3.60%, respectively). Spheroid cell self-renewal was evidenced by cell proliferation and the generation of new spheroids during two consecutive expansion periods. Culture of spheroid cells under differentiation conditions gave rise to cells showing immunolocalization of the neuron-specific antigen NeuN and the astroglial antigen GFAP (glial fibrillary acidic protein). Our results indicate that spheroids spontaneously generated in this culture system were comprised of cells with molecular characteristics of NSC/NPC that can self-renew and differentiate into neurons and glia, supporting the identity of spheroids as neurospheres.
Collapse
Affiliation(s)
- Belén Sánchez-Maldonado
- 1 Departamento de Medicina y Cirugía, Facultad de Veterinaria, Universidad Complutense de Madrid , Madrid, España
| | - María de Lourdes Galicia
- 2 Sección Departamental de Fisiología, Facultad de Veterinaria, Universidad Complutense de Madrid , Madrid, España
| | - Concepción Rojo
- 3 Sección Departamental de Anatomía y Embriología, Facultad de Veterinaria, Universidad Complutense de Madrid , Madrid, España
| | - Alfredo González-Gil
- 2 Sección Departamental de Fisiología, Facultad de Veterinaria, Universidad Complutense de Madrid , Madrid, España
| | - Miguel Flor-García
- 4 Departamento de Neuropatología Molecular, Centro de Biología Molecular "Severo Ochoa" (CBMSO), CSIC-UAM , Madrid, España.,5 Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid , Madrid, España
| | - Rosa A Picazo
- 2 Sección Departamental de Fisiología, Facultad de Veterinaria, Universidad Complutense de Madrid , Madrid, España
| |
Collapse
|
7
|
Crompton LA, Cordero‐Llana O, Caldwell MA. Astrocytes in a dish: Using pluripotent stem cells to model neurodegenerative and neurodevelopmental disorders. Brain Pathol 2017; 27:530-544. [PMID: 28585380 PMCID: PMC8028895 DOI: 10.1111/bpa.12522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 02/06/2023] Open
Abstract
Neuroscience and Neurobiology have historically been neuron biased, yet up to 40% of the cells in the brain are astrocytes. These cells are heterogeneous and regionally diverse but universally essential for brain homeostasis. Astrocytes regulate synaptic transmission as part of the tripartite synapse, provide metabolic and neurotrophic support, recycle neurotransmitters, modulate blood flow and brain blood barrier permeability and are implicated in the mechanisms of neurodegeneration. Using pluripotent stem cells (PSC), it is now possible to study regionalised human astrocytes in a dish and to model their contribution to neurodevelopmental and neurodegenerative disorders. The evidence challenging the traditional neuron-centric view of degeneration within the CNS is reviewed here, with focus on recent findings and disease phenotypes from human PSC-derived astrocytes. In addition we compare current protocols for the generation of regionalised astrocytes and how these can be further refined by our growing knowledge of neurodevelopment. We conclude by proposing a functional and phenotypical characterisation of PSC-derived astrocytic cultures that is critical for reproducible and robust disease modelling.
Collapse
Affiliation(s)
- Lucy A. Crompton
- School of Biochemistry, Medical Sciences BldUniversity of BristolBristolBS8 1TDUK
| | - Oscar Cordero‐Llana
- Bristol Medical School, Medical Sciences BldUniversity of BristolBristolBS8 1TDUK
| | - Maeve A. Caldwell
- Trinity College Institute for NeuroscienceTrinity College Dublin 2Ireland
| |
Collapse
|
8
|
Mawad D, Figtree G, Gentile C. Current Technologies Based on the Knowledge of the Stem Cells Microenvironments. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1041:245-262. [DOI: 10.1007/978-3-319-69194-7_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
9
|
Thiesler CT, Cajic S, Hoffmann D, Thiel C, van Diepen L, Hennig R, Sgodda M, Weiβmann R, Reichl U, Steinemann D, Diekmann U, Huber NMB, Oberbeck A, Cantz T, Kuss AW, Körner C, Schambach A, Rapp E, Buettner FFR. Glycomic Characterization of Induced Pluripotent Stem Cells Derived from a Patient Suffering from Phosphomannomutase 2 Congenital Disorder of Glycosylation (PMM2-CDG). Mol Cell Proteomics 2016; 15:1435-52. [PMID: 26785728 PMCID: PMC4824866 DOI: 10.1074/mcp.m115.054122] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Indexed: 01/08/2023] Open
Abstract
PMM2-CDG, formerly known as congenital disorder of glycosylation-Ia (CDG-Ia), is caused by mutations in the gene encoding phosphomannomutase 2 (PMM2). This disease is the most frequent form of inherited CDG-diseases affecting protein N-glycosylation in human. PMM2-CDG is a multisystemic disease with severe psychomotor and mental retardation. In order to study the pathophysiology of PMM2-CDG in a human cell culture model, we generated induced pluripotent stem cells (iPSCs) from fibroblasts of a PMM2-CDG-patient (PMM2-iPSCs). Expression of pluripotency factors and in vitro differentiation into cell types of the three germ layers was unaffected in the analyzed clone PMM2-iPSC-C3 compared with nondiseased human pluripotent stem cells (hPSCs), revealing no broader influence of the PMM2 mutation on pluripotency in cell culture. Analysis of gene expression by deep-sequencing did not show obvious differences in the transcriptome between PMM2-iPSC-C3 and nondiseased hPSCs. By multiplexed capillary gel electrophoresis coupled to laser induced fluorescence detection (xCGE-LIF) we could show that PMM2-iPSC-C3 exhibit the common hPSC N-glycosylation pattern with high-mannose-type N-glycans as the predominant species. However, phosphomannomutase activity of PMM2-iPSC-C3 was 27% compared with control hPSCs and lectin staining revealed an overall reduced protein glycosylation. In addition, quantitative assessment of N-glycosylation by xCGE-LIF showed an up to 40% reduction of high-mannose-type N-glycans in PMM2-iPSC-C3, which was in concordance to the observed reduction of the Glc3Man9GlcNAc2 lipid-linked oligosaccharide compared with control hPSCs. Thus we could model the PMM2-CDG disease phenotype of hypoglycosylation with patient derived iPSCs in vitro. Knock-down of PMM2 by shRNA in PMM2-iPSC-C3 led to a residual activity of 5% and to a further reduction of the level of N-glycosylation. Taken together we have developed human stem cell-based cell culture models with stepwise reduced levels of N-glycosylation now enabling to study the role of N-glycosylation during early human development.
Collapse
Affiliation(s)
- Christina T Thiesler
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Samanta Cajic
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Dirk Hoffmann
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ‖Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Christian Thiel
- **Center for Child and Adolescent Medicine, Department Kinderheilkunde I, 69120 Heidelberg, Germany
| | - Laura van Diepen
- ‡‡Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt University, 17475 Greifswald, Germany
| | - René Hennig
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany; §§glyXera GmbH, 39120 Magdeburg, Germany
| | - Malte Sgodda
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ¶¶Translational Hepatology and Stem Cell Biology, Dept. of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Robert Weiβmann
- ‡‡Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt University, 17475 Greifswald, Germany
| | - Udo Reichl
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Doris Steinemann
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ‖‖Institute of Human Genetics, Hannover Medical School, 30625 Hannover, Germany
| | - Ulf Diekmann
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Nicolas M B Huber
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Astrid Oberbeck
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Cantz
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ¶¶Translational Hepatology and Stem Cell Biology, Dept. of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas W Kuss
- ‡‡Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt University, 17475 Greifswald, Germany
| | - Christian Körner
- **Center for Child and Adolescent Medicine, Department Kinderheilkunde I, 69120 Heidelberg, Germany
| | - Axel Schambach
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; ‖Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Erdmann Rapp
- ¶Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany; §§glyXera GmbH, 39120 Magdeburg, Germany
| | - Falk F R Buettner
- From the ‡REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; §Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
10
|
Li S, Nie EH, Yin Y, Benowitz LI, Tung S, Vinters HV, Bahjat FR, Stenzel-Poore MP, Kawaguchi R, Coppola G, Carmichael ST. GDF10 is a signal for axonal sprouting and functional recovery after stroke. Nat Neurosci 2015; 18:1737-45. [PMID: 26502261 PMCID: PMC4790086 DOI: 10.1038/nn.4146] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/24/2015] [Indexed: 02/08/2023]
Abstract
Stroke produces a limited process of neural repair. Axonal sprouting in cortex adjacent to the infarct is part of this recovery process, but the signal that initiates axonal sprouting is not known. Growth and differentiation factor 10 (GDF10) is induced in peri-infarct neurons in mice, non-human primates and humans. GDF10 promotes axonal outgrowth in vitro in mouse, rat and human neurons through TGFβRI and TGFβRII signaling. Using pharmacogenetic gain- and loss-of-function studies, we found that GDF10 produced axonal sprouting and enhanced functional recovery after stroke; knocking down GDF10 blocked axonal sprouting and reduced recovery. RNA sequencing from peri-infarct cortical neurons revealed that GDF10 downregulated PTEN, upregulated PI3 kinase signaling and induced specific axonal guidance molecules. Using unsupervised genome-wide association analysis of the GDF10 transcriptome, we found that it was not related to neurodevelopment, but may partially overlap with other CNS injury patterns. Thus, GDF10 is a stroke-induced signal for axonal sprouting and functional recovery.
Collapse
Affiliation(s)
- S Li
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - EH Nie
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Y Yin
- Laboratories for Neuroscience Research in Neurosurgery, Children’s Hospital, Boston, MA
| | - LI Benowitz
- Laboratories for Neuroscience Research in Neurosurgery, Children’s Hospital, Boston, MA
| | - S Tung
- Department of Pathology and Laboratory Medicine (Neuropathology), David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - HV Vinters
- Department of Pathology and Laboratory Medicine (Neuropathology), David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - FR Bahjat
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR
| | - MP Stenzel-Poore
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR
| | - R Kawaguchi
- Program in Neurogenetics, Department of Neurology and Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
| | - G Coppola
- Program in Neurogenetics, Department of Neurology and Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
| | - ST Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
11
|
Abstract
Compared with traditional 2D adherent cell culture, 3D spheroidal cell aggregates, or spheroids, are regarded as more physiological, and this technique has been exploited in the field of oncology, stem cell biology, and tissue engineering. Mesenchymal stem cells (MSCs) cultured in spheroids have enhanced anti-inflammatory, angiogenic, and tissue reparative/regenerative effects with improved cell survival after transplantation. Cytoskeletal reorganization and drastic changes in cell morphology in MSC spheroids indicate a major difference in mechanophysical properties compared with 2D culture. Enhanced multidifferentiation potential, upregulated expression of pluripotency marker genes, and delayed replicative senescence indicate enhanced stemness in MSC spheroids. Furthermore, spheroid formation causes drastic changes in the gene expression profile of MSC in microarray analyses. In spite of these significant changes, underlying molecular mechanisms and signaling pathways triggering and sustaining these changes are largely unknown.
Collapse
|
12
|
Pethe P, Pursani V, Bhartiya D. Lineage specific expression of Polycomb Group Proteins in human embryonic stem cells in vitro. Cell Biol Int 2015; 39:600-610. [PMID: 25572667 DOI: 10.1002/cbin.10431] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/26/2014] [Indexed: 02/05/2023]
Abstract
Human embryonic (hES) stem cells are an excellent model to study lineage specification and differentiation into various cell types. Differentiation necessitates repression of specific genes not required for a particular lineage. Polycomb Group (PcG) proteins are key histone modifiers, whose primary function is gene repression. PcG proteins form complexes called Polycomb Repressive Complexes (PRCs), which catalyze histone modifications such as H2AK119ub1, H3K27me3, and H3K9me3. PcG proteins play a crucial role during differentiation of stem cells. The expression of PcG transcripts during differentiation of hES cells into endoderm, mesoderm, and ectoderm lineage is yet to be shown. In-house derived hES cell line KIND1 was differentiated into endoderm, mesoderm, and ectoderm lineages; followed by characterization using RT-PCR for HNF4A, CDX2, MEF2C, TBX5, SOX1, and MAP2. qRT-PCR and western blotting was performed to compare expression of PcG transcripts and proteins across all the three lineages. We observed that cells differentiated into endoderm showed upregulation of RING1B, BMI1, EZH2, and EED transcripts. Mesoderm differentiation was characterized by significant downregulation of all PcG transcripts during later stages. BMI1 and RING1B were upregulated while EZH2, SUZ12, and EED remained low during ectoderm differentiation. Western blotting also showed distinct expression of BMI1 and EZH2 during differentiation into three germ layers. Our study shows that hES cells differentiating into endoderm, mesoderm, and ectoderm lineages show distinct PcG expression profile at transcript and protein level.
Collapse
Affiliation(s)
- Prasad Pethe
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (NIRRH), Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | | | | |
Collapse
|
13
|
Chen X, Xu B, Han X, Mao Z, Chen M, Du G, Talbot P, Wang X, Xia Y. The effects of triclosan on pluripotency factors and development of mouse embryonic stem cells and zebrafish. Arch Toxicol 2015; 89:635-646. [PMID: 24879426 DOI: 10.1007/s00204-014-1270-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 05/13/2014] [Indexed: 01/27/2023]
Abstract
Triclosan (TCS) poses potential risks to reproduction and development due to its endocrine-disrupting properties. However, the mechanism of TCS's effects on early embryonic development is little known. Embryonic stem cells (ESC) and zebrafish embryos provide valuable models for testing the toxic effects of environmental chemicals on early embryogenesis. In this study, mouse embryonic stem cells (mESC) were acutely exposed to TCS for 24 h, and general cytotoxicity and the effect of TCS on pluripotency were then evaluated. In addition, zebrafish embryos were exposed to TCS from 2- to 24-h post-fertilization (hpf), and their morphology was evaluated. In mESC, alkaline phosphatase staining was significantly decreased after treatment with the highest concentration of TCS (50 μM). Although the expression levels of Sox2 mRNA were not changed, the mRNA levels of Oct4 and Nanog in TCS-treated groups were significantly decreased compared to controls. In addition, the protein levels of Oct4, Sox2 and Nanog were significantly reduced in response to TCS treatment. MicroRNA (miR)-134, an expression inhibitor of pluripotency markers, was significantly increased in TCS-treated mESC. In zebrafish experiments, after 24 hpf of treatment, the controls had developed to the late stage of somitogenesis, while embryos exposed to 300 μg/L of TCS were still at the early stage of somitogenesis, and three genes (Oct4, Sox2 and Nanog) were upregulated in treated groups when compared with the controls. The two models demonstrated that TCS may affect early embryonic development by disturbing the expression of the pluripotency markers (Oct4, Sox2 and Nanog).
Collapse
Affiliation(s)
- Xiaojiao Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Maternity and Child Health Hospital, Nanjing Medical University, Nanjing, 210029, China
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Laperle A, Masters KS, Palecek SP. Influence of substrate composition on human embryonic stem cell differentiation and extracellular matrix production in embryoid bodies. Biotechnol Prog 2014; 31:212-9. [PMID: 25311359 DOI: 10.1002/btpr.2001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/28/2014] [Indexed: 12/14/2022]
Abstract
Stem cells reside in specialized niches in vivo. Specific factors, including the extracellular matrix (ECM), in these niches are directly responsible for maintaining the stem cell population. During development, components of the stem cell microenvironment also control differentiation with precise spatial and temporal organization. The stem cell microenvironment is dynamically regulated by the cellular component, including stem cells themselves. Thus, a mechanism exists whereby stem cells modify the ECM, which in turn affects the fate of the stem cell. In this study, we investigated whether the type of ECM initially adsorbed to the culture substrate can influence the composition of the ECM deposited by human embryonic stem cells (hESCs) differentiating in embryoid bodies, and whether different ECM composition and deposition profiles elicit distinct differentiation fates. We have shown that the initial ECM environment hESCs are exposed to affects the fate decisions of those cells and that this initial ECM environment is constantly modified during the differentiation process.
Collapse
Affiliation(s)
- Alex Laperle
- Dept. of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53706
| | | | | |
Collapse
|
15
|
De Paepe C, Krivega M, Cauffman G, Geens M, Van de Velde H. Totipotency and lineage segregation in the human embryo. ACTA ACUST UNITED AC 2014; 20:599-618. [DOI: 10.1093/molehr/gau027] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
16
|
Combined hydrogels that switch human pluripotent stem cells from self-renewal to differentiation. Proc Natl Acad Sci U S A 2014; 111:5580-5. [PMID: 24706900 DOI: 10.1073/pnas.1319685111] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The ability of materials to define the architecture and microenvironment experienced by cells provides new opportunities to direct the fate of human pluripotent stem cells (HPSCs) [Robinton DA, Daley GQ (2012) Nature 481(7381):295-305]. However, the conditions required for self-renewal vs. differentiation of HPSCs are different, and a single system that efficiently achieves both outcomes is not available [Giobbe GG, et al. (2012) Biotechnol Bioeng 109(12):3119-3132]. We have addressed this dual need by developing a hydrogel-based material that uses ionic de-cross-linking to remove a self-renewal permissive hydrogel (alginate) and switch to a differentiation-permissive microenvironment (collagen). Adjusting the timing of this switch can preferentially steer the HPSC differentiation to mimic lineage commitment during gastrulation to ectoderm (early switch) or mesoderm/endoderm (late switch). As an exemplar differentiated cell type, we showed that directing early lineage specification using this single system can promote cardiogenesis with increased gene expression in high-density cell populations. This work will facilitate regenerative medicine by allowing in situ HPSC expansion to be coupled with early lineage specification within defined tissue geometries.
Collapse
|
17
|
From pluripotency to forebrain patterning: an in vitro journey astride embryonic stem cells. Cell Mol Life Sci 2014; 71:2917-30. [PMID: 24643740 PMCID: PMC4098049 DOI: 10.1007/s00018-014-1596-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/17/2014] [Accepted: 02/26/2014] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) have been used extensively as in vitro models of neural development and disease, with special efforts towards their conversion into forebrain progenitors and neurons. The forebrain is the most complex brain region, giving rise to several fundamental structures, such as the cerebral cortex, the hypothalamus, and the retina. Due to the multiplicity of signaling pathways playing different roles at distinct times of embryonic development, the specification and patterning of forebrain has been difficult to study in vivo. Research performed on ESCs in vitro has provided a large body of evidence to complement work in model organisms, but these studies have often been focused more on cell type production than on cell fate regulation. In this review, we systematically reassess the current literature in the field of forebrain development in mouse and human ESCs with a focus on the molecular mechanisms of early cell fate decisions, taking into consideration the specific culture conditions, exogenous and endogenous molecular cues as described in the original studies. The resulting model of early forebrain induction and patterning provides a useful framework for further studies aimed at reconstructing forebrain development in vitro for basic research or therapy.
Collapse
|
18
|
Chen X, Xu B, Han X, Mao Z, Talbot P, Chen M, Du G, Chen A, Liu J, Wang X, Xia Y. Effect of bisphenol A on pluripotency of mouse embryonic stem cells and differentiation capacity in mouse embryoid bodies. Toxicol In Vitro 2013; 27:2249-2255. [PMID: 24090592 DOI: 10.1016/j.tiv.2013.09.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/13/2013] [Accepted: 09/23/2013] [Indexed: 12/01/2022]
Abstract
Bisphenol A (BPA) poses potential risks to reproduction and development. However, the mechanism of BPA's effects on early embryonic development is still unknown. Embryonic stem cells (ESC) and embryoid bodies (EB) provide valuable in vitro models for testing the toxic effects of environmental chemicals in early embryogenesis. In this study, mouse embryonic stem cells (mESC) were acutely exposed to BPA for 24h, and general cytotoxicity and the effect of BPA on pluripotency were then evaluated. Meanwhile, mouse embryoid bodies (mEB) were exposed to BPA up to 6 days and their differentiation capacity was evaluated. In mESC and mEB, we found that BPA up-regulated pluripotency markers (Oct4, Sox2 and Nanog) at mRNA and/or protein levels. Moreover, BPA increased the mRNA levels of endodermal markers (Gata4,Sox17) and mesodermal markers (Sma,Desmin), and reduced the mRNA levels of ectodermal markers (Nestin,Fgf5) in mEB. Furthermore, microRNA(miR)-134, an expression inhibitor of pluripotency markers including Oct4, Sox2 and Nanog, was decreased both in BPA-treated mESC and mEB. These results firstly indicate that BPA may disturb pluripotency in mESC and differentiation of mEB, and may inhibit ectodermal lineage differentiation of mEB while miR-134 may play a key role underlying this effect.
Collapse
Affiliation(s)
- Xiaojiao Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Maternity and Child Health Hospital, Nanjing Medical University, Nanjing 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Preda MB, Burlacu A, Simionescu M. Defined-size embryoid bodies formed in the presence of serum replacement increases the efficiency of the cardiac differentiation of mouse embryonic stem cells. Tissue Cell 2012; 45:54-60. [PMID: 23107982 DOI: 10.1016/j.tice.2012.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 09/11/2012] [Accepted: 09/23/2012] [Indexed: 11/26/2022]
Abstract
The pluripotent nature of embryonic stem (ES) cells makes them powerful tools in cell replacement therapy for severe degenerative diseases, such as heart failure. However, the development of strategies to increase the efficiency of cardiomyocyte (CMC) differentiation is still needed to produce a sufficient amount of cells for clinical applications. This paper evaluates the impact of the size and the aggregation of embryoid bodies (EBs) on the efficiency of ES cell differentiation into CMCs. ES cells were generated from RAP inbred mice. These cells expressed pluripotency markers and induced teratomas when injected into syngeneic mice, which made them suitable for differentiation into CMCs. We found that the EBs that were formed as a result of in vitro ES cell aggregation generated contractile tissue in direct correlation with the initial number of ES cells. Furthermore, the presence of knock-out serum replacement (KO-SR) during ES cell aggregation resulted in less compacted EBs and increased cell differentiation into CMCs compared to the presence of foetal bovine serum. In conclusion, cardiac differentiation of ES cells is dependent on the size and the degree of compaction of EBs, and the presence of KO-SR during initiation of EBs may lead to improved cardiogenic differentiation of ES cells.
Collapse
Affiliation(s)
- M B Preda
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, 8 BP Hasdeu 050568, Romania
| | | | | |
Collapse
|
20
|
Cooper A, Leung M, Zhang M. Polymeric Fibrous Matrices for Substrate-Mediated Human Embryonic Stem Cell Lineage Differentiation. Macromol Biosci 2012; 12:882-92. [DOI: 10.1002/mabi.201100269] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Revised: 09/16/2011] [Indexed: 12/30/2022]
|
21
|
Stachelscheid H, Wulf-Goldenberg A, Eckert K, Jensen J, Edsbagge J, Björquist P, Rivero M, Strehl R, Jozefczuk J, Prigione A, Adjaye J, Urbaniak T, Bussmann P, Zeilinger K, Gerlach JC. Teratoma formation of human embryonic stem cells in three-dimensional perfusion culture bioreactors. J Tissue Eng Regen Med 2012; 7:729-41. [PMID: 22438087 DOI: 10.1002/term.1467] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 10/14/2011] [Accepted: 01/05/2012] [Indexed: 11/10/2022]
Abstract
Teratoma formation in mice is today the most stringent test for pluripotency that is available for human pluripotent cells, as chimera formation and tetraploid complementation cannot be performed with human cells. The teratoma assay could also be applied for assessing the safety of human pluripotent cell-derived cell populations intended for therapeutic applications. In our study we examined the spontaneous differentiation behaviour of human embryonic stem cells (hESCs) in a perfused 3D multi-compartment bioreactor system and compared it with differentiation of hESCs and human induced pluripotent cells (hiPSCs) cultured in vitro as embryoid bodies and in vivo in an experimental mouse model of teratoma formation. Results from biochemical, histological/immunohistological and ultrastuctural analyses revealed that hESCs cultured in bioreactors formed tissue-like structures containing derivatives of all three germ layers. Comparison with embryoid bodies and the teratomas revealed a high degree of similarity of the tissues formed in the bioreactor to these in the teratomas at the histological as well as transcriptional level, as detected by comparative whole-genome RNA expression profiling. The 3D culture system represents a novel in vitro model that permits stable long-term cultivation, spontaneous multi-lineage differentiation and tissue formation of pluripotent cells that is comparable to in vivo differentiation. Such a model is of interest, e.g. for the development of novel cell differentiation strategies. In addition, the 3D in vitro model could be used for teratoma studies and pluripotency assays in a fully defined, controlled environment, alternatively to in vivo mouse models.
Collapse
Affiliation(s)
- H Stachelscheid
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Chen YS, Pelekanos RA, Ellis RL, Horne R, Wolvetang EJ, Fisk NM. Small molecule mesengenic induction of human induced pluripotent stem cells to generate mesenchymal stem/stromal cells. Stem Cells Transl Med 2012. [PMID: 23197756 DOI: 10.5966/sctm.2011-0022] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The translational potential of mesenchymal stem/stromal cells (MSCs) is limited by their rarity in somatic organs, heterogeneity, and need for harvest by invasive procedures. Induced pluripotent stem cells (iPSCs) could be an advantageous source of MSCs, but attempts to derive MSCs from pluripotent cells have required cumbersome or untranslatable techniques, such as coculture, physical manipulation, sorting, or viral transduction. We devised a single-step method to direct mesengenic differentiation of human embryonic stem cells (ESCs) and iPSCs using a small molecule inhibitor. First, epithelial-like monolayer cells were generated by culturing ESCs/iPSCs in serum-free medium containing the transforming growth factor-β pathway inhibitor SB431542. After 10 days, iPSCs showed upregulation of mesodermal genes (MSX2, NCAM, HOXA2) and downregulation of pluripotency genes (OCT4, LEFTY1/2). Differentiation was then completed by transferring cells into conventional MSC medium. The resultant development of MSC-like morphology was associated with increased expression of genes, reflecting epithelial-to-mesenchymal transition. Both ESC- and iPSC-derived MSCs exhibited a typical MSC immunophenotype, expressed high levels of vimentin and N-cadherin, and lacked expression of pluripotency markers at the protein level. Robust osteogenic and chondrogenic differentiation was induced in vitro in ES-MSCs and iPS-MSCs, whereas adipogenic differentiation was limited, as reported for primitive fetal MSCs and ES-MSCs derived by other methods. We conclude that treatment with SB431542 in two-dimensional cultures followed by culture-induced epithelial-to-mesenchymal transition leads to rapid and uniform MSC conversion of human pluripotent cells without the need for embryoid body formation or feeder cell coculture, providing a robust, clinically applicable, and efficient system for generating MSCs from human iPSCs.
Collapse
Affiliation(s)
- Yen Shun Chen
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Australia
| | | | | | | | | | | |
Collapse
|
23
|
Liu L, Liu C, Zhong Y, Apostolou A, Fang S. ER stress response during the differentiation of H9 cells induced by retinoic acid. Biochem Biophys Res Commun 2012; 417:738-743. [PMID: 22197812 DOI: 10.1016/j.bbrc.2011.12.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 12/08/2011] [Indexed: 02/05/2023]
Abstract
Endoplasmic reticulum (ER) stress occurs during early embryonic development. The aim of this study is to determine whether ER stress occurs during human embryonic stem cell differentiation induced by retinoic acid (RA). H9 human embryonic stem cells were subjected to RA treatment for up to 29days to induce differentiation. HEK293 cells were treated with RA as a control. The results demonstrate that several ER stress-responsive genes are differentially regulated in H9 and HEK293 cells in response to 5days of RA treatment. GRP78/Bip was upregulated in H9 cells but downregulated in HEK293 cells. eIF2α was downregulated in H9 cells but not in HEK293 cells. Phosphorylation of eIF2α was downregulated in H9 cells but upregulated in HEK293 cells. XBP-1 was downregulated immediately after RA treatment in H9 cells, but its downregulation was much slower in HEK293 cells. Additionally, two ER-resident E3 ubiquitin ligases, gp78 and Hrd1, were both upregulated in H9 cells following 5 days of exposure to RA. Moreover, the protein Bcl2 was undetectable in H9 cells and H9-derived cells but was expressed in HEK293 cells, and it expression in the two types of cells was unaltered by RA treatment. In H9 cells treated with RA for 29 days, GRP78/Bip, XBP-1 and Bcl2 were all upregulated. These results suggest that ER stress is involved in H9 cell differentiation induced by RA.
Collapse
Affiliation(s)
- Lihua Liu
- Clinical Pharmacology Institute, Anhui Medical University, Hefei, Anhui 230032, China
| | | | | | | | | |
Collapse
|
24
|
Campos PB, Sartore RC, Ramalho BL, Costa ES, Rehen SK. Cycle arrest and aneuploidy induced by zidovudine in murine embryonic stem cells. Mutagenesis 2012; 27:431-6. [DOI: 10.1093/mutage/ger093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
25
|
Kolle G, Shepherd JL, Gardiner B, Kassahn KS, Cloonan N, Wood DLA, Nourbakhsh E, Taylor DF, Wani S, Chy HS, Zhou Q, McKernan K, Kuersten S, Laslett AL, Grimmond SM. Deep-transcriptome and ribonome sequencing redefines the molecular networks of pluripotency and the extracellular space in human embryonic stem cells. Genome Res 2011; 21:2014-25. [PMID: 22042643 DOI: 10.1101/gr.119321.110] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent RNA-sequencing studies have shown remarkable complexity in the mammalian transcriptome. The ultimate impact of this complexity on the predicted proteomic output is less well defined. We have undertaken strand-specific RNA sequencing of multiple cellular RNA fractions (>20 Gb) to uncover the transcriptional complexity of human embryonic stem cells (hESCs). We have shown that human embryonic stem (ES) cells display a high degree of transcriptional diversity, with more than half of active genes generating RNAs that differ from conventional gene models. We found evidence that more than 1000 genes express long 5' and/or extended 3'UTRs, which was confirmed by "virtual Northern" analysis. Exhaustive sequencing of the membrane-polysome and cytosolic/untranslated fractions of hESCs was used to identify RNAs encoding peptides destined for secretion and the extracellular space and to demonstrate preferential selection of transcription complexity for translation in vitro. The impact of this newly defined complexity on known gene-centric network models such as the Plurinet and the cell surface signaling machinery in human ES cells revealed a significant expansion of known transcript isoforms at play, many predicting possible alternative functions based on sequence alterations within key functional domains.
Collapse
Affiliation(s)
- Gabriel Kolle
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Queensland 4072, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shweiki D, Ben-Yehudah A, Redinger C, Easley CA, Doron S, Schatten G. Sexually dimorphic gene expression in non-human primate ESCs analyzed stringently. Biochem Biophys Res Commun 2011; 414:631-4. [PMID: 22005468 DOI: 10.1016/j.bbrc.2011.09.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 09/28/2011] [Indexed: 10/17/2022]
Abstract
Human exhibit sexual dimorphism early in development and throughout life. Here we stringently analyzed gene expression in inbred non-human primate embryonic stem cells (nhpESCs) searching for sexually dimorphisms. We utilized location-specific probes solely, thus avoiding probe cross-reactivity between members of gene families and genomic gene duplications. Seventeen sexually dimorphic transcripts (15 genes, out of which 9 autosomals) were identified, of which five were verified using real-time q-PCR. We compared these results from pedigreed nhpESCs with available human ESCs datasets. Three human X-linked genes show sexual dimorphism. Thus, these results enhance our knowledge and deepen our understanding on early development processes for sexual dimorphism.
Collapse
Affiliation(s)
- Dorit Shweiki
- Bioinformatics Program, School of Computer Science, The Academic College of Tel Aviv-Yaffo, Tel Aviv, Israel.
| | | | | | | | | | | |
Collapse
|
27
|
Denis JA, Rochon-Beaucourt C, Champon B, Pietu G. Global Transcriptional Profiling of Neural and Mesenchymal Progenitors Derived from Human Embryonic Stem Cells Reveals Alternative Developmental Signaling Pathways. Stem Cells Dev 2011; 20:1395-409. [DOI: 10.1089/scd.2010.0331] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jérôme Alexandre Denis
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| | - Christelle Rochon-Beaucourt
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| | - Benoite Champon
- CECS/AFM, I-STEM, Centre d'Etude des Cellules Souches, Evry Cedex, France
| | - Geneviève Pietu
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| |
Collapse
|
28
|
Kim JM, Moon SH, Lee SG, Cho YJ, Hong KS, Lee JH, Lee HJ, Chung HM. Assessment of differentiation aspects by the morphological classification of embryoid bodies derived from human embryonic stem cells. Stem Cells Dev 2011; 20:1925-35. [PMID: 21388292 DOI: 10.1089/scd.2010.0476] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In general, the formation of embryoid bodies (EBs) is a commonly known method for initial induction of human embryonic stem cells (hESCs) into their derivatives in vitro. Despite the ability of EBs to mimic developmental processing, the specification and classifications of EBs are not yet well known. Because EBs show various differentiation potentials depending on the size and morphology of the aggregated cells, specification is difficult to attain. Here, we sought to classify the differentiation potentials of EBs by morphologies to enable one to control the differentiation of specific lineages from hESCs with high efficiency. To induce the differentiation of EB formation, we established floating cultures of undifferentiated hESCs in Petri dishes with hESC medium lacking basic fibroblast growth factor. Cells first aggregated into balls; ∼10 days after suspension culture, some different types of EB morphology were present, which we classified as cystic-, bright cavity-, and dark cavity-type EBs. Next, we analyzed the characteristics of each type of EB for its capacity to differentiate into the 3 germ layers via multiplex polymerase chain reaction (PCR), real-time PCR, and immunocytochemistry. Our results indicated that most cells within the cystic EBs were composed of endoderm lineage populations, and both of the cavity EB types were well organized with 3 germ-layer cells. However, the differentiation capacity of the bright cavity EBs was faster than that of the dark cavity EBs. Thus, the bright cavity EBs in this study, which showed equal differentiation tendencies compared with other types of EBs, may serve as the standard for in vitro engineering of EBs. These results indicate that the classification of EB morphologies allows the estimation of the differentiation status of the EBs and may allow the delineation of subsets of conditions necessary for EBs to differentiate into specific cell types.
Collapse
Affiliation(s)
- Jung Mo Kim
- Stem Cell Research Lab, CHA Stem Cell Institute, CHA University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
29
|
VandeVoort CA, Hill DL, Chaffin CL, Conley AJ. Ethanol, acetaldehyde, and estradiol affect growth and differentiation of rhesus monkey embryonic stem cells. Alcohol Clin Exp Res 2011; 35:1534-40. [PMID: 21438889 DOI: 10.1111/j.1530-0277.2011.01490.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND The timing of the origins of fetal alcohol syndrome has been difficult to determine, in part because of the challenge associated with in vivo studies of the peri-implantation stage of embryonic development. Because embryonic stem cells (ESCs) are derived from blastocyst stage embryos, they are used as a model for early embryo development. METHODS Rhesus monkey ESC lines (ORMES-6 and ORMES-7) were treated with 0, 0.01, 0.1, or 1.0% ethanol, 1.0% ethanol with estradiol, or 0.00025% acetaldehyde with or without estradiol for 4 weeks. RESULTS Although control ESCs remained unchanged, abnormal morphology of ESCs in the ethanol and acetaldehyde treatment groups was observed before 2 weeks of treatment. Immunofluorescence staining of key pluripotency markers (TRA-1-81 and alkaline phosphatase) indicated a loss of ESC pluripotency in the 1.0% ethanol group. ORMES-7 was more sensitive to effects of ethanol than ORMES-6. CONCLUSIONS Estradiol appeared to increase sensitivity to ethanol in the ORMES-6 and ORMES-7 cell line. The morphological changes and labeling for pluripotency, proliferation, and apoptosis demonstrated that how ethanol affects these early cells that develop in culture, their differentiation state in particular. The effects of ethanol may be mediated in part through metabolic pathways regulating acetaldehyde formation, and while potentially accentuated by estradiol in some individuals, how remains to be determined.
Collapse
|
30
|
Abstract
Maintenance of a pluripotent cell population during mammalian embryogenesis is crucial for the proper generation of extraembryonic and embryonic tissues to ensure intrauterine survival and fetal development. Pluripotent stem cells derived from early stage mammalian embryos are known as "embryonic stem cells." Such embryo-derived stem cells can proliferate indefinitely in vitro and give rise to derivatives of all three primary germ layers. Their potential for clinical and commercial applications has sparked great excitement within scientific and lay communities. Identification of the signaling pathways controlling stem cell pluripotency and differentiation provides knowledge-based approaches to manipulate stem cells for regenerative medicine. One of the signaling cascades that has been identified in the control of stem cell pluripotency and differentiation is the Activin/Nodal pathway. Here, we describe the differences among pluripotent cell types and discuss the latest findings on the molecular mechanisms involving Activin/Nodal signaling in controlling their pluripotency and differentiation.
Collapse
Affiliation(s)
- Zhenzhi Chng
- Institute of Medical Biology, Singapore, Singapore
| | | | | |
Collapse
|
31
|
Lukaszewicz AI, McMillan MK, Kahn M. Small molecules and stem cells. Potency and lineage commitment: the new quest for the fountain of youth. J Med Chem 2010; 53:3439-53. [PMID: 20047330 DOI: 10.1021/jm901361d] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Agnès I Lukaszewicz
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
32
|
Gerlach JC, Hout M, Edsbagge J, Björquist P, Lübberstedt M, Miki T, Stachelscheid H, Schmelzer E, Schatten G, Zeilinger K. Dynamic 3D culture promotes spontaneous embryonic stem cell differentiation in vitro. Tissue Eng Part C Methods 2010; 16:115-21. [PMID: 19382830 DOI: 10.1089/ten.tec.2008.0654] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spontaneous in vitro differentiation of mouse embryonic stem cells (mESC) is promoted by a dynamic, three-dimensional (3D), tissue-density perfusion technique with continuous medium perfusion and exchange in a novel four-compartment, interwoven capillary bioreactor. We compared ectodermal, endodermal, and mesodermal immunoreactive tissue structures formed by mESC at culture day 10 with mouse fetal tissue development at gestational day E9.5. The results show that the bioreactor cultures more closely resemble mouse fetal tissue development at gestational day E9.5 than control mESC cultured in Petri dishes.
Collapse
Affiliation(s)
- Jörg C Gerlach
- Department of Surgery and Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Collier TS, Sarkar P, Rao B, Muddiman DC. Quantitative top-down proteomics of SILAC labeled human embryonic stem cells. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2010; 21:879-889. [PMID: 20199872 DOI: 10.1016/j.jasms.2010.01.031] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Revised: 01/28/2010] [Accepted: 01/28/2010] [Indexed: 05/28/2023]
Abstract
Human embryonic stem cells (hESCs) are self-renewing pluripotent cells with relevance to treatment of numerous medical conditions. However, a global understanding of the role of the hESC proteome in maintaining pluripotency or triggering differentiation is still largely lacking. The emergence of top-down proteomics has facilitated the identification and characterization of intact protein forms that are not readily apparent in bottom-up studies. Combined with metabolic labeling techniques such as stable isotope labeling by amino acids in cell culture (SILAC), quantitative comparison of intact protein expression under differing experimental conditions is possible. Herein, quantitative top-down proteomics of hESCs is demonstrated using the SILAC method and nano-flow reverse phase chromatography directly coupled to a linear-ion-trap Fourier transform ion cyclotron resonance mass spectrometer (nLC-LTQ-FT-ICR-MS). In this study, which to the best of our knowledge represents the first top-down analysis of hESCs, we have confidently identified 11 proteins by accurate intact mass, MS/MS, and amino acid counting facilitated by SILAC labeling. Although quantification is challenging due to the incorporation of multiple labeled amino acids (i.e., lysine and arginine) and arginine to proline conversion, we are able to quantitatively account for these phenomena using a mathematical model.
Collapse
Affiliation(s)
- Timothy S Collier
- W. M. Keck FT-ICR Mass Spectrometry Laboratory, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, USA
| | | | | | | |
Collapse
|
34
|
Alternative splicing regulates mouse embryonic stem cell pluripotency and differentiation. Proc Natl Acad Sci U S A 2010; 107:10514-9. [PMID: 20498046 DOI: 10.1073/pnas.0912260107] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Two major goals of regenerative medicine are to reproducibly transform adult somatic cells into a pluripotent state and to control their differentiation into specific cell fates. Progress toward these goals would be greatly helped by obtaining a complete picture of the RNA isoforms produced by these cells due to alternative splicing (AS) and alternative promoter selection (APS). To investigate the roles of AS and APS, reciprocal exon-exon junctions were interrogated on a genome-wide scale in differentiating mouse embryonic stem (ES) cells with a prototype Affymetrix microarray. Using a recently released open-source software package named AltAnalyze, we identified 144 genes for 170 putative isoform variants, the majority (67%) of which were predicted to alter protein sequence and domain composition. Verified alternative exons were largely associated with pathways of Wnt signaling and cell-cycle control, and most were conserved between mouse and human. To examine the functional impact of AS, we characterized isoforms for two genes. As predicted by AltAnalyze, we found that alternative isoforms of the gene Serca2 were targeted by distinct microRNAs (miRNA-200b, miRNA-214), suggesting a critical role for AS in cardiac development. Analysis of the Wnt transcription factor Tcf3, using selective knockdown of an ES cell-enriched and characterized isoform, revealed several distinct targets for transcriptional repression (Stmn2, Ccnd2, Atf3, Klf4, Nodal, and Jun) as well as distinct differentiation outcomes in ES cells. The findings herein illustrate a critical role for AS in the specification of ES cells with differentiation, and highlight the utility of global functional analyses of AS.
Collapse
|
35
|
Integrated proteomic analysis of human cancer cells and plasma from tumor bearing mice for ovarian cancer biomarker discovery. PLoS One 2009; 4:e7916. [PMID: 19936259 PMCID: PMC2775948 DOI: 10.1371/journal.pone.0007916] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 10/26/2009] [Indexed: 01/09/2023] Open
Abstract
Background The complexity of the human plasma proteome represents a substantial challenge for biomarker discovery. Proteomic analysis of genetically engineered mouse models of cancer and isolated cancer cells and cell lines provide alternative methods for identification of potential cancer markers that would be detectable in human blood using sensitive assays. The goal of this work is to evaluate the utility of an integrative strategy using these two approaches for biomarker discovery. Methodology/Principal Findings We investigated a strategy that combined quantitative plasma proteomics of an ovarian cancer mouse model with analysis of proteins secreted or shed by human ovarian cancer cells. Of 106 plasma proteins identified with increased levels in tumor bearing mice, 58 were also secreted or shed from ovarian cancer cells. The remainder consisted primarily of host-response proteins. Of 25 proteins identified in the study that were assayed, 8 mostly secreted proteins common to mouse plasma and human cancer cells were significantly upregulated in a set of plasmas from ovarian cancer patients. Five of the eight proteins were confirmed to be upregulated in a second independent set of ovarian cancer plasmas, including in early stage disease. Conclusions/Significance Integrated proteomic analysis of cancer mouse models and human cancer cell populations provides an effective approach to identify potential circulating protein biomarkers.
Collapse
|
36
|
Tumour formation by single fibroblast growth factor receptor 3-positive rhabdomyosarcoma-initiating cells. Br J Cancer 2009; 101:2030-7. [PMID: 19888223 PMCID: PMC2795447 DOI: 10.1038/sj.bjc.6605407] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: The hypothesis that malignant tumours are generated by rare populations of cancer stem cells that are more tumourigenic than other cancer cells has gained increasing credence. The objective of this study was to identify and characterise a subpopulation of human sarcoma-initiating cells. Methods: We examined established rhabdomyosarcoma cell lines by flow cytometry. Tumourigenesis was examined by xenograft models. Real-time PCR and immunohistochemistry were performed to examine the gene expression using cell lines and biopsy specimens. Results: Rhabdomyosarcoma cell lines included small populations of fibroblast growth factor receptor 3 (FGFR3)-positive cells. FGFR3-positive KYM-1 and RD cells were more strongly tumourigenic than FGFR3-negative cells. In addition, xenoengraftment of 33% of single FGFR3-positive KYM-1 cells yielded tumour formation. Stem cell properties of FGFR3-positive cells were further established by real-time PCR, which demonstrated upregulation of undifferentiated cell markers and downregulation of differentiation markers. We showed that in the absence of serum, addition of basic fibroblast growth factor maintained and enriched FGFR3-positive cells. On the other hand, ciliary neurotrophic factor reduced the proportion of FGFR3-positive cells. Real-time PCR and immunohistochemical examination revealed that embryonal rhabdomyosarcoma patient biopsy specimens were found to over-express FGFR3. Conclusions: Our findings suggest that rhabdomyosarcoma cell lines include a minor subpopulation of FGFR3-positive sarcoma-initiating cells, which can be maintained indefinitely in culture and which is crucial for their malignancy.
Collapse
|
37
|
Nie Y, Bergendahl V, Hei DJ, Jones JM, Palecek SP. Scalable culture and cryopreservation of human embryonic stem cells on microcarriers. Biotechnol Prog 2009; 25:20-31. [PMID: 19197994 DOI: 10.1002/btpr.110] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As a result of their pluripotency and potential for unlimited self-renewal, human embryonic stem cells (hESCs) hold tremendous promise in regenerative medicine. An essential prerequisite for the widespread application of hESCs is the establishment of effective and efficient protocols for large-scale cell culture, storage, and distribution. At laboratory scales hESCs are cultured adherent to tissue culture plates; these culture techniques are labor-intensive and do not scale to high cell numbers. In an effort to facilitate larger scale hESC cultivation, we investigated the feasibility of culturing hESCs adherent to microcarriers. We modified the surface of Cytodex 3 microcarriers with either Matrigel or mouse embryonic fibroblasts (MEFs). hESC colonies were effectively expanded in a pluripotent, undifferentiated state on both Matrigel-coated microcarriers and microcarriers seeded with a MEF monolayer. While the hESC expansion rate on MEF-microcarriers was less than that on MEF-plates, the doubling time of hESCs on Matrigel-microcarriers was indistinguishable from that of hESCs expanded on Matrigel-coated tissue culture plates. Standard hESC cryopreservation methodologies are plagued by poor viability and high differentiation rates upon thawing. Here, we demonstrate that cryopreservation of hESCs adherent to microcarriers in cryovials provides a higher recovery of undifferentiated cells than cryopreservation of cells in suspension. Together, these results suggest that microcarrier-based stabilization and culture may facilitate hESC expansion and storage for research and therapeutic applications.
Collapse
Affiliation(s)
- Ying Nie
- Dept. of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
38
|
Xie Y, Yang Y, Kang X, Li R, Volakis LI, Zhang X, Lee LJ, Kniss DA. Bioassembly of three-dimensional embryonic stem cell-scaffold complexes using compressed gases. Biotechnol Prog 2009; 25:535-42. [DOI: 10.1002/btpr.151] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
39
|
Krtolica A, Genbacev O, Escobedo C, Zdravkovic T, Nordstrom A, Vabuena D, Nath A, Simon C, Mostov K, Fisher SJ. Disruption of Apical-Basal Polarity of Human Embryonic Stem Cells Enhances Hematoendothelial Differentiation. Stem Cells 2009; 25:2215-23. [PMID: 17569786 DOI: 10.1634/stemcells.2007-0230] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
During murine development, the formation of tight junctions and acquisition of polarity are associated with allocation of the blastomeres on the outer surface of the embryo to the trophoblast lineage, whereas the absence of polarization directs cells to the inner cell mass. Here, we report the results of ultrastructural analyses that suggest a similar link between polarization and cell fate in human embryos. In contrast, the five human embryonic stem cell (hESC) lines displayed apical-basal, epithelial-type polarity with electron-dense tight junctions, apical microvilli, and asymmetric distribution of organelles. Consistent with these findings, molecules that are components of tight junctions or play regulatory roles in polarization localized to the apical regions of the hESCs at sites of cell-cell contact. The tight junctions were functional, as shown by the ability of hESC colonies to exclude the pericellular passage of a biotin compound. Depolarization of hESCs produced multilayered aggregates of rapidly proliferating cells that continued to express transcription factors that are required for pluripotency at the same level as control cells. However, during embryoid body formation, depolarized cells differentiated predominantly along mesenchymal lineage and spontaneously produced hematoendothelial precursors more efficiently than control ESC. Our findings have numerous implications with regard to strategies for deriving, propagating, and differentiating hESC.
Collapse
Affiliation(s)
- Ana Krtolica
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Embryoid body morphology influences diffusive transport of inductive biochemicals: a strategy for stem cell differentiation. Biomaterials 2008; 29:4471-80. [PMID: 18793799 DOI: 10.1016/j.biomaterials.2008.08.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 08/20/2008] [Indexed: 02/08/2023]
Abstract
Differentiation of human embryonic stem (hES) cells into cells for regenerative medicine is often initiated by embryoid body (EB) formation. EBs may be treated with soluble biochemicals such as cytokines, growth factors and vitamins to induce differentiation. A scanning electron microscopy analysis, conducted over 14 days, revealed time-dependent changes in EB structure which led to the formation of a shell that significantly reduced the diffusive transport of a model molecule (374 Da) by >80%. We found that the shell consists of 1) an extracellular matrix (ECM) comprised of collagen type I; 2) a squamous cellular layer with tight cell-cell adhesions associated with E-cadherin; and 3) a collagen type IV lining indicative of a basement membrane. Disruption of the basement membrane, by either inhibiting its formation with noggin or permeabilizing it with collagenase, resulted in recovery of diffusive transport. Increasing the diffusive transport of retinoic acid (RA) and serum in EBs by a 15-min collagenase digestion on days 4, 5, 6 and 7 promoted neuronal differentiation. Flow cytometry and quantitative RT-PCR analysis of collagenase-treated EBs revealed 68% of cells expressing neural cell adhesion molecule (NCAM) relative to 28% for untreated EBs. Our results suggest that limitations in diffusive transport of biochemicals need to be considered when formulating EB differentiation strategies.
Collapse
|
41
|
Deconstructing human embryonic stem cell cultures: niche regulation of self-renewal and pluripotency. J Mol Med (Berl) 2008; 86:875-86. [PMID: 18521556 DOI: 10.1007/s00109-008-0356-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Revised: 03/17/2008] [Accepted: 03/31/2008] [Indexed: 02/08/2023]
Abstract
The factors and signaling pathways controlling pluripotent human cell properties, both embryonic and induced, have not been fully investigated. Failure to account for functional heterogeneity within human embryonic stem cell (hESC) cultures has led to inconclusive results in previous work examining extrinsic influences governing hESC fate (self renewal vs. differentiation vs. death). Here, we attempt to reconcile these inconsistencies with recent reports demonstrating that an autologously produced in vitro niche regulates hESCs. Moreover, we focus on the reciprocal paracrine signals within the in vitro hESC niche allowing for the maintenance and/or expansion of the hESC colony-initiating cell (CIC). Based on this, it is clear that separation of hESC-CICs, apart from their differentiated derivatives, will be essential in future studies involving their molecular regulation. Understanding how extrinsic factors control hESC self-renewal and differentiation will allow us to culture and differentiate these pluripotent cells with higher efficiency. This knowledge will be essential for clinical applications using human pluripotent cells in regenerative medicine.
Collapse
|
42
|
Vallier L, Alexander M, Pedersen RA. Activin/Nodal and FGF pathways cooperate to maintain pluripotency of human embryonic stem cells. J Cell Sci 2006; 118:4495-509. [PMID: 16179608 DOI: 10.1242/jcs.02553] [Citation(s) in RCA: 668] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Maintenance of pluripotency is crucial to the mammalian embryo's ability to generate the extra-embryonic and embryonic tissues that are needed for intrauterine survival and foetal development. The recent establishment of embryonic stem cells from human blastocysts (hESCs) provides an opportunity to identify the factors supporting pluripotency at early stages of human development. Using this in vitro model, we have recently shown that Nodal can block neuronal differentiation, suggesting that TGFbeta family members are involved in cell fate decisions of hESCs, including preservation of their pluripotency. Here, we report that Activin/Nodal signalling through Smad2/3 activation is necessary to maintain the pluripotent status of hESCs. Inhibition of Activin/Nodal signalling by follistatin and by overexpression of Lefty or Cerberus-Short, or by the Activin receptor inhibitor SB431542, precipitates hESC differentiation. Nevertheless, neither Nodal nor Activin is sufficient to sustain long-term hESC growth in a chemically defined medium without serum. Recent studies have shown that FGF2 can also maintain long-term expression of pluripotency markers, and we find that inhibition of the FGF signalling pathway by the tyrosine kinase inhibitor SU5402 causes hESC differentiation. However, this effect of FGF on hESC pluripotency depends on Activin/Nodal signalling, because it is blocked by SB431542. Finally, long-term maintenance of in-vitro pluripotency can be achieved with a combination of Activin or Nodal plus FGF2 in the absence of feeder-cell layers, conditioned medium or Serum Replacer. These findings suggest that the Activin/Nodal pathway maintains pluripotency through mechanism(s) in which FGF acts as a competence factor and therefore provide further evidence of distinct mechanisms for preservation of pluripotency in mouse and human ESCs.
Collapse
Affiliation(s)
- Ludovic Vallier
- Department of Surgery and Cambridge Institute for Medical Research, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge CB2 2XY, UK.
| | | | | |
Collapse
|