51
|
Sustained intrinsic WNT and BMP4 activation impairs hESC differentiation to definitive endoderm and drives the cells towards extra-embryonic mesoderm. Sci Rep 2021; 11:8242. [PMID: 33859268 PMCID: PMC8050086 DOI: 10.1038/s41598-021-87547-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
We identified a human embryonic stem cell subline that fails to respond to the differentiation cues needed to obtain endoderm derivatives, differentiating instead into extra-embryonic mesoderm. RNA-sequencing analysis showed that the subline has hyperactivation of the WNT and BMP4 signalling. Modulation of these pathways with small molecules confirmed them as the cause of the differentiation impairment. While activation of WNT and BMP4 in control cells resulted in a loss of endoderm differentiation and induction of extra-embryonic mesoderm markers, inhibition of these pathways in the subline restored its ability to differentiate. Karyotyping and exome sequencing analysis did not identify any changes in the genome that could account for the pathway deregulation. These findings add to the increasing evidence that different responses of stem cell lines to differentiation protocols are based on genetic and epigenetic factors, inherent to the line or acquired during cell culture.
Collapse
|
52
|
Mus LM, Van Haver S, Popovic M, Trypsteen W, Lefever S, Zeltner N, Ogando Y, Jacobs EZ, Denecker G, Sanders E, Van Neste C, Vanhauwaert S, Decaesteker B, Deforce D, Van Nieuwerburgh F, Mestdagh P, Vandesompele J, Menten B, De Preter K, Studer L, Heindryckx B, Durinck K, Roberts S, Speleman F. Recurrent chromosomal imbalances provide selective advantage to human embryonic stem cells under enhanced replicative stress conditions. Genes Chromosomes Cancer 2021; 60:272-281. [PMID: 33336840 DOI: 10.1002/gcc.22931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/07/2022] Open
Abstract
Human embryonic stem cells (hESCs) and embryonal tumors share a number of common features, including a compromised G1/S checkpoint. Consequently, these rapidly dividing hESCs and cancer cells undergo elevated levels of replicative stress, inducing genomic instability that drives chromosomal imbalances. In this context, it is of interest that long-term in vitro cultured hESCs exhibit a remarkable high incidence of segmental DNA copy number gains, some of which are also highly recurrent in certain malignancies such as 17q gain (17q+). The selective advantage of DNA copy number changes in these cells has been attributed to several underlying processes including enhanced proliferation. We hypothesized that these recurrent chromosomal imbalances become rapidly embedded in the cultured hESCs through a replicative stress driven Darwinian selection process. To this end, we compared the effect of hydroxyurea-induced replicative stress vs normal growth conditions in an equally mixed cell population of isogenic euploid and 17q + hESCs. We could show that 17q + hESCs rapidly overtook normal hESCs. Our data suggest that recurrent chromosomal segmental gains provide a proliferative advantage to hESCs under increased replicative stress, a process that may also explain the highly recurrent nature of certain imbalances in cancer.
Collapse
Affiliation(s)
- Liselot M Mus
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Stéphane Van Haver
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Mina Popovic
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Wim Trypsteen
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Steve Lefever
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Nadja Zeltner
- Center for Molecular Medicine, Department of Biochemistry & Molecular Biology and Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Yudelca Ogando
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Eva Z Jacobs
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Geertrui Denecker
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Ellen Sanders
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Christophe Van Neste
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Suzanne Vanhauwaert
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Bieke Decaesteker
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Dieter Deforce
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Filip Van Nieuwerburgh
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Pieter Mestdagh
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jo Vandesompele
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Björn Menten
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Katleen De Preter
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, USA
- Developmental Biology Program, Sloan Kettering Institute, New York, USA
| | - Björn Heindryckx
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kaat Durinck
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Stephen Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Frank Speleman
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Ghent University, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
53
|
Between a Rock and a Hard Place: An Epigenetic-Centric View of Testicular Germ Cell Tumors. Cancers (Basel) 2021; 13:cancers13071506. [PMID: 33805941 PMCID: PMC8036638 DOI: 10.3390/cancers13071506] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary This minireview focuses on the role of epigenetics in testicular cancer. A working model is developed that postulates that epigenetic features that drive testicular cancer malignancy also enable these tumors to be cured at a high rate with chemotherapy. Chemoresistance may occur by epigenetic uncoupling of malignancy and chemosensitivity, a scenario that may be amenable to epigenetic-based therapies. Abstract Compared to many common solid tumors, the main genetic drivers of most testicular germ cell tumors (TGCTs) are unknown. Decades of focus on genomic alterations in TGCTs including awareness of a near universal increase in copies of chromosome 12p have failed to uncover exceptional driver genes, especially in genes that can be targeted therapeutically. Thus far, TGCT patients have missed out on the benefits of targeted therapies available to treat most other malignancies. In the past decade there has been a greater appreciation that epigenetics may play an especially prominent role in TGCT etiology, progression, and hypersensitivity to conventional chemotherapy. While genetics undoubtedly plays a role in TGCT biology, this mini-review will focus on the epigenetic “states” or features of testicular cancer, with an emphasis on DNA methylation, histone modifications, and miRNAs associated with TGCT susceptibility, initiation, progression, and response to chemotherapy. In addition, we comment on the current status of epigenetic-based therapy and epigenetic biomarker development for TGCTs. Finally, we suggest a unifying “rock and a hard place” or “differentiate or die” model where the tumorigenicity and curability of TGCTs are both dependent on common but still ill-defined epigenetic states.
Collapse
|
54
|
Vasudevan A, Schukken KM, Sausville EL, Girish V, Adebambo OA, Sheltzer JM. Aneuploidy as a promoter and suppressor of malignant growth. Nat Rev Cancer 2021; 21:89-103. [PMID: 33432169 DOI: 10.1038/s41568-020-00321-1] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Aneuploidy has been recognized as a hallmark of tumorigenesis for more than 100 years, but the connection between chromosomal errors and malignant growth has remained obscure. New evidence emerging from both basic and clinical research has illuminated a complicated relationship: despite its frequency in human tumours, aneuploidy is not a universal driver of cancer development and instead can exert substantial tumour-suppressive effects. The specific consequences of aneuploidy are highly context dependent and are influenced by a cell's genetic and environmental milieu. In this Review, we discuss the diverse facets of cancer biology that are shaped by aneuploidy, including metastasis, drug resistance and immune recognition, and we highlight aneuploidy's distinct roles as both a tumour promoter and an anticancer vulnerability.
Collapse
|
55
|
Abstract
Acquired chromosomal abnormalities may occur during the reprogramming and culture of human pluripotent stem cells (hPSCs). Therefore, it is required that regular testing of genetic integrity be conducted. G-banded karyotyping is a widely used genetic assay that is capable of detecting chromosomal abnormalities. Karyotyping of hPSC cultures can be a challenging undertaking for inexperienced investigators; here, we provide detailed procedures for karyotyping, including sample preparation and analysis, as well as the interpretation of hPSC karyotype results.
Collapse
|
56
|
Avior Y, Lezmi E, Eggan K, Benvenisty N. Cancer-Related Mutations Identified in Primed Human Pluripotent Stem Cells. Cell Stem Cell 2020; 28:10-11. [PMID: 33321073 DOI: 10.1016/j.stem.2020.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yishai Avior
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 9190401, Israel
| | - Elyad Lezmi
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 9190401, Israel
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, and The Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 9190401, Israel.
| |
Collapse
|
57
|
Functional in vivo and in vitro effects of 20q11.21 genetic aberrations on hPSC differentiation. Sci Rep 2020; 10:18582. [PMID: 33122739 PMCID: PMC7596514 DOI: 10.1038/s41598-020-75657-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 10/15/2020] [Indexed: 01/01/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) have promising therapeutic applications due to their infinite capacity for self-renewal and pluripotency. Genomic stability is imperative for the clinical use of hPSCs; however, copy number variation (CNV), especially recurrent CNV at 20q11.21, may contribute genomic instability of hPSCs. Furthermore, the effects of CNVs in hPSCs at the whole-transcriptome scale are poorly understood. This study aimed to examine the functional in vivo and in vitro effects of frequently detected CNVs at 20q11.21 during early-stage differentiation of hPSCs. Comprehensive transcriptome profiling of abnormal hPSCs revealed that the differential gene expression patterns had a negative effect on differentiation potential. Transcriptional heterogeneity identified by single-cell RNA sequencing (scRNA-seq) of embryoid bodies from two different isogenic lines of hPSCs revealed alterations in differentiated cell distributions compared with that of normal cells. RNA-seq analysis of 22 teratomas identified several differentially expressed lineage-specific markers in hPSCs with CNVs, consistent with the histological results of the altered ecto/meso/endodermal ratio due to CNVs. Our results suggest that CNV amplification contributes to cell proliferation, apoptosis, and cell fate specification. This work shows the functional consequences of recurrent genetic abnormalities and thereby provides evidence to support the development of cell-based applications.
Collapse
|
58
|
Lu J, Wei W. Considerations on chemistry, manufacturing, and control of stem cell products for Investigational New Drug application in China. Biologicals 2020; 68:3-8. [PMID: 33097376 DOI: 10.1016/j.biologicals.2020.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/09/2020] [Indexed: 11/18/2022] Open
Abstract
Tremendous progress has been made in recent years to produce functional cells for cell therapy products. Hundreds of clinical trials of stem cell products (SCPs) have shown promising therapeutic potential worldwide, including the products derived from human pluripotent stem cells (hPSCs), adult stem cells and mesenchymal stem cells (MSC). Before starting a clinical trial, comprehensive chemistry, manufacturing and control (CMC) study is required to assure the safety and quality consistency of SCPs. The heterogeneity of stem cell products arises from the variability in the donor tissues, isolation of cells and differentiation processes, and appropriate testing approaches are needed to characterize and release SCPs. Here we summarize the regulatory considerations of CMC study in Investigational New Drug (IND) application of SCPs in China based on the current knowledge, and they will be updated in the future with the advance of stem cell biology and regulatory science.
Collapse
Affiliation(s)
- Jiaqi Lu
- Center for Drug Evaluation (CDE), National Medical Products Administration, Beijing, 100022, China.
| | - Wei Wei
- Center for Drug Evaluation (CDE), National Medical Products Administration, Beijing, 100022, China
| |
Collapse
|
59
|
Halliwell J, Barbaric I, Andrews PW. Acquired genetic changes in human pluripotent stem cells: origins and consequences. Nat Rev Mol Cell Biol 2020; 21:715-728. [DOI: 10.1038/s41580-020-00292-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2020] [Indexed: 12/14/2022]
|
60
|
Role of Nrf2 and mitochondria in cancer stem cells; in carcinogenesis, tumor progression, and chemoresistance. Biochimie 2020; 179:32-45. [PMID: 32946993 DOI: 10.1016/j.biochi.2020.09.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 08/05/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) are rare sub-population in tumor mass with self-renewal and differentiation abilities; CSCs are considered as the main cells which are responsible for tumor metastasis, cancer recurrence, and chemo/radio-resistance. CSCs are believed to contain low mitochondria in quantity, high concentration of nuclear factor erythroid 2-related factor 2 (Nrf2), and low reactive oxygen species (ROS) levels. Mitochondria regulate certain cellular functions, including controlling of cellular energetics, calcium signaling, cell growth and cell differentiation, cell cycle regulation, and cell death. Also, mitochondria are the main sources of intrinsic ROS production. Dysfunction of CSCs mitochondria due to oxidative phosphorylation is reported in several pathological conditions, including metabolic disorders, age-related diseases, and various types of cancers. ROS levels play a significant role in cellular signal transduction and CSCs' identity and differentiation capability. Nrf2 is a master transcription factor that plays critical functions in maintaining cellular redox hemostasis by regulating several antioxidant and detoxification pathways. Recently, the critical function of Nrf2 in CSCs has been revealed by several studies. Nrf2 is an essential molecule in the maintenance of CSCs' stemness and self-renewal in response to different oxidative stresses such as chemotherapy-induced elevation of ROS. Nrf2 enables these cells to recover from chemotherapy damages, and promotes establishment of invasion and dissemination. In this study, we have summarized the role of Nrf2 and mitochondria function CSCs, which promote cancer development. The significant role of Nrf2 in the regulation of mitochondrial function and ROS levels suggests this molecule as a potential target to eradicate CSCs.
Collapse
|
61
|
Mehrjardi NZ, Molcanyi M, Hatay FF, Timmer M, Shahbazi E, Ackermann JP, Herms S, Heilmann-Heimbach S, Wunderlich TF, Prochnow N, Haghikia A, Lampert A, Hescheler J, Neugebauer EAM, Baharvand H, Šarić T. Acquisition of chromosome 1q duplication in parental and genome-edited human-induced pluripotent stem cell-derived neural stem cells results in their higher proliferation rate in vitro and in vivo. Cell Prolif 2020; 53:e12892. [PMID: 32918782 PMCID: PMC7574866 DOI: 10.1111/cpr.12892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023] Open
Abstract
Objectives Genetic engineering of human‐induced pluripotent stem cell‐derived neural stem cells (hiPSC‐NSC) may increase the risk of genomic aberrations. Therefore, we asked whether genetic modification of hiPSC‐NSCs exacerbates chromosomal abnormalities that may occur during passaging and whether they may cause any functional perturbations in NSCs in vitro and in vivo. Materials and Methods The transgenic cassette was inserted into the AAVS1 locus, and the genetic integrity of zinc‐finger nuclease (ZFN)‐modified hiPSC‐NSCs was assessed by the SNP‐based karyotyping. The hiPSC‐NSC proliferation was assessed in vitro by the EdU incorporation assay and in vivo by staining of brain slices with Ki‐67 antibody at 2 and 8 weeks after transplantation of ZFN‐NSCs with and without chromosomal aberration into the striatum of immunodeficient rats. Results During early passages, no chromosomal abnormalities were detected in unmodified or ZFN‐modified hiPSC‐NSCs. However, at higher passages both cell populations acquired duplication of the entire long arm of chromosome 1, dup(1)q. ZNF‐NSCs carrying dup(1)q exhibited higher proliferation rate than karyotypically intact cells, which was partly mediated by increased expression of AKT3 located on Chr1q. Compared to karyotypically normal ZNF‐NSCs, cells with dup(1)q also exhibited increased proliferation in vivo 2 weeks, but not 2 months, after transplantation. Conclusions These results demonstrate that, independently of ZFN‐editing, hiPSC‐NSCs have a propensity for acquiring dup(1)q and this aberration results in increased proliferation which might compromise downstream hiPSC‐NSC applications.
Collapse
Affiliation(s)
- Narges Zare Mehrjardi
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marek Molcanyi
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Firuze Fulya Hatay
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Marco Timmer
- Department of Neurosurgery, University Hospital Cologne, Cologne, Germany
| | - Ebrahim Shahbazi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Justus P Ackermann
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Stefan Herms
- Department of Genomics, Life & Brain Center, Institute for Human Genetics, University of Bonn, Bonn, Germany.,Department of Biomedicine, Medical Genetics, Research Group Genomics, University Hospital Basel, Basel, Switzerland
| | - Stefanie Heilmann-Heimbach
- Department of Genomics, Life & Brain Center, Institute for Human Genetics, University of Bonn, Bonn, Germany
| | - Thomas F Wunderlich
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Max Planck Institute for Metabolism Research and Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Nora Prochnow
- Clinic for Neurology, St. Josef-Hospital, Clinic of the Ruhr-University Bochum, Bochum, Germany
| | - Aiden Haghikia
- Clinic for Neurology, St. Josef-Hospital, Clinic of the Ruhr-University Bochum, Bochum, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik, RWTH Aachen University, Aachen, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Edmund A M Neugebauer
- Medizinische Hochschule Brandenburg Theodor Fontane, Campus Neuruppin, Neuruppin, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
62
|
Hoff AM, Kraggerud SM, Alagaratnam S, Berg KCG, Johannessen B, Høland M, Nilsen G, Lingjærde OC, Andrews PW, Lothe RA, Skotheim RI. Frequent copy number gains of SLC2A3 and ETV1 in testicular embryonal carcinomas. Endocr Relat Cancer 2020; 27:457-468. [PMID: 32580154 PMCID: PMC7424350 DOI: 10.1530/erc-20-0064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/10/2020] [Indexed: 01/03/2023]
Abstract
Testicular germ cell tumours (TGCTs) appear as different histological subtypes or mixtures of these. They show similar, multiple DNA copy number changes, where gain of 12p is pathognomonic. However, few high-resolution analyses have been performed and focal DNA copy number changes with corresponding candidate target genes remain poorly described for individual subtypes. We present the first high-resolution DNA copy number aberration (CNA) analysis on the subtype embryonal carcinomas (ECs), including 13 primary ECs and 5 EC cell lines. We identified recurrent gains and losses and allele-specific CNAs. Within these regions, we nominate 30 genes that may be of interest to the EC subtype. By in silico analysis of data from 150 TGCTs from The Cancer Genome Atlas (TCGA), we further investigated CNAs, RNA expression, somatic mutations and fusion transcripts of these genes. Among primary ECs, ploidy ranged between 2.3 and 5.0, and the most common aberrations were DNA copy number gains at chromosome (arm) 7, 8, 12p, and 17, losses at 4, 10, 11, and 18, replicating known TGCT genome characteristics. Gain of whole or parts of 12p was found in all samples, including a highly amplified 100 kbp segment at 12p13.31, containing SLC2A3. Gain at 7p21, encompassing ETV1, was the second most frequent aberration. In conclusion, we present novel CNAs and the genes located within these regions, where the copy number gain of SLC2A3 and ETV1 are of interest, and which copy number levels also correlate with expression in TGCTs.
Collapse
Affiliation(s)
- Andreas M Hoff
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sigrid M Kraggerud
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sharmini Alagaratnam
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kaja C G Berg
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Maren Høland
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gro Nilsen
- Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Ole C Lingjærde
- Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Peter W Andrews
- The Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield, UK
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Correspondence should be addressed to R A Lothe or R I Skotheim: or
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Correspondence should be addressed to R A Lothe or R I Skotheim: or
| |
Collapse
|
63
|
Zhang M, Xiao R, Liu G, Huang Y. Genotoxins exaggerate the stressed state of aneuploid embryonic stem cells via activation of autophagy. SCIENCE CHINA. LIFE SCIENCES 2020; 63:1026-1036. [PMID: 31872377 DOI: 10.1007/s11427-019-9666-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/01/2019] [Indexed: 01/19/2023]
Abstract
The cellular consequences of aneuploidy are largely dependent on the cell types examined. Aneuploid yeasts and mouse embryonic fibroblasts exhibit cell proliferation defects and can be selectively inhibited by compounds that cause proteotoxic or energy stress. By contrast, most aneuploid pluripotent stem cells proliferate rapidly and reach higher saturation densities. The responses of aneuploid pluripotent stem cells to the stress-inducing compounds remain uncharacterized. Here, we tested the response of aneuploid embryonic stem cells to several compounds that caused proteotoxic, energy and genotoxic stress using previously established mouse embryonic stem cell lines trisomic for chromosome 6, 8, 11, or 15. Not all trisomic embryonic stem cells were selectively inhibited by compounds that cause proteotoxic or energy stress. However, most of these cells exhibited increased sensitivity to genotoxins. They displayed elevated DNA damage response as characterized by increased γH2A.X foci under genotoxic stress. Further investigations indicated that elevated autophagy levels might contribute to the increased cytotoxic effects of genotoxins on trisomic embryonic stem cells. Our study laid the foundation for eliminating aneuploidy that might be an effective approach for controlling cancer progression.
Collapse
Affiliation(s)
- Meili Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China. .,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| | - Rong Xiao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Guang Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Yue Huang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China. .,Department of Medical Genetics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
64
|
The mutational impact of culturing human pluripotent and adult stem cells. Nat Commun 2020; 11:2493. [PMID: 32427826 PMCID: PMC7237696 DOI: 10.1038/s41467-020-16323-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/20/2020] [Indexed: 01/07/2023] Open
Abstract
Genetic changes acquired during in vitro culture pose a risk for the successful application of stem cells in regenerative medicine. To assess the genetic risks induced by culturing, we determined all mutations in individual human stem cells by whole genome sequencing. Individual pluripotent, intestinal, and liver stem cells accumulate 3.5 ± 0.5, 7.2 ± 1.1 and 8.3 ± 3.6 base substitutions per population doubling, respectively. The annual in vitro mutation accumulation rate of adult stem cells is nearly 40-fold higher than the in vivo mutation accumulation rate. Mutational signature analysis reveals that in vitro induced mutations are caused by oxidative stress. Reducing oxygen tension in culture lowers the mutational load. We use the mutation rates, spectra, and genomic distribution to model the accumulation of oncogenic mutations during typical in vitro expansion, manipulation or screening experiments using human stem cells. Our study provides empirically defined parameters to assess the mutational risk of stem cell based therapies. Genetic changes acquired during in vitro culture pose a challenge to application of stem cells. Here the authors use whole genome sequencing to show that cultured human adult and pluripotent stem cells have a high mutational load caused by oxidative stress and reduced oxygen tension in culture lowers mutation rates.
Collapse
|
65
|
Modulation of Wnt and Activin/Nodal supports efficient derivation, cloning and suspension expansion of human pluripotent stem cells. Biomaterials 2020; 249:120015. [PMID: 32311594 DOI: 10.1016/j.biomaterials.2020.120015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 03/12/2020] [Accepted: 03/27/2020] [Indexed: 01/09/2023]
Abstract
Various culture systems have been used to derive and maintain human pluripotent stem cells (hPSCs), but they are inefficient in sustaining cloning and suspension expansion of hPSCs. Through systematically modulating Wnt and Activin/Nodal signaling, we developed a defined medium (termed AIC), which enables efficient cloning and long-term expansion of hPSCs (AIC-hPSCs) through single-cell passage on feeders, matrix or in suspension (25-fold expansion in 4 days) and maintains genomic stability of hPSCs over extensive expansion. Moreover, the AIC medium supports efficient derivation of hPSCs from blastocysts or somatic cells under feeder-free conditions. Compared to conventional hPSCs, AIC-hPSCs have similar gene expression profiles but down-regulated differentiation genes and display higher metabolic activity. Additionally, the AIC medium shows a good compatibility for different hPSC lines under various culture conditions. Our study provides a robust culture system for derivation, cloning and suspension expansion of high-quality hPSCs that benefits GMP production and processing of therapeutic hPSC products.
Collapse
|
66
|
Rehakova D, Souralova T, Koutna I. Clinical-Grade Human Pluripotent Stem Cells for Cell Therapy: Characterization Strategy. Int J Mol Sci 2020; 21:E2435. [PMID: 32244538 PMCID: PMC7177280 DOI: 10.3390/ijms21072435] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells have the potential to change the way in which human diseases are cured. Clinical-grade human embryonic stem cells and human induced pluripotent stem cells have to be created according to current good manufacturing practices and regulations. Quality and safety must be of the highest importance when humans' lives are at stake. With the rising number of clinical trials, there is a need for a consensus on hPSCs characterization. Here, we summarize mandatory and 'for information only' characterization methods with release criteria for the establishment of clinical-grade hPSC lines.
Collapse
Affiliation(s)
- Daniela Rehakova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 656 91 Brno, Czech Republic;
| | - Tereza Souralova
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 656 91 Brno, Czech Republic;
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 625 00 Brno, Czech Republic
| | - Irena Koutna
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 656 91 Brno, Czech Republic;
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 625 00 Brno, Czech Republic
| |
Collapse
|
67
|
Thompson O, von Meyenn F, Hewitt Z, Alexander J, Wood A, Weightman R, Gregory S, Krueger F, Andrews S, Barbaric I, Gokhale PJ, Moore HD, Reik W, Milo M, Nik-Zainal S, Yusa K, Andrews PW. Low rates of mutation in clinical grade human pluripotent stem cells under different culture conditions. Nat Commun 2020; 11:1528. [PMID: 32251294 PMCID: PMC7089967 DOI: 10.1038/s41467-020-15271-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
The occurrence of repetitive genomic changes that provide a selective growth advantage in pluripotent stem cells is of concern for their clinical application. However, the effect of different culture conditions on the underlying mutation rate is unknown. Here we show that the mutation rate in two human embryonic stem cell lines derived and banked for clinical application is low and not substantially affected by culture with Rho Kinase inhibitor, commonly used in their routine maintenance. However, the mutation rate is reduced by >50% in cells cultured under 5% oxygen, when we also found alterations in imprint methylation and reversible DNA hypomethylation. Mutations are evenly distributed across the chromosomes, except for a slight increase on the X-chromosome, and an elevation in intergenic regions suggesting that chromatin structure may affect mutation rate. Overall the results suggest that pluripotent stem cells are not subject to unusually high rates of genetic or epigenetic alterations.
Collapse
Affiliation(s)
- Oliver Thompson
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Ferdinand von Meyenn
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
- Department of Medical & Molecular Genetics, King's College London, London, SE1 9RT, UK
- Institute of Food, Nutrition and Health, ETH Zurich, 8603, Schwerzenbach, Switzerland
| | - Zoe Hewitt
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - John Alexander
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Andrew Wood
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Richard Weightman
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Sian Gregory
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Felix Krueger
- Bioinformatics Group, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Simon Andrews
- Bioinformatics Group, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Ivana Barbaric
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Paul J Gokhale
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Harry D Moore
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Wolf Reik
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Marta Milo
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Serena Nik-Zainal
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Academic Laboratory of Medical Genetics, Cambridge University Hospitals NHS Foundation Trust, Box 238, Lv6 Addenbrooke' Treatment Centre, Cambridge Biomedical Research Campus, Cambridge, CB2 0QQ, UK
- MRC Cancer Unit, University of Cambridge, Hutchinson/MRC Research Centre, Box 1297, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Kosuke Yusa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.
| | - Peter W Andrews
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
68
|
Yoshihara M, Oguchi A, Murakawa Y. Genomic Instability of iPSCs and Challenges in Their Clinical Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:23-47. [PMID: 31898780 DOI: 10.1007/978-3-030-31206-0_2] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Generation of human-induced pluripotent stem cells (iPSCs) from somatic cells has opened the possibility to design novel therapeutic approaches. In 2014, the first-in-human clinical trial of iPSC-based therapy was conducted. However, the transplantation for the second patient was discontinued at least in part due to genetic aberrations detected in iPSCs. Moreover, many studies have reported genetic aberrations in iPSCs with the rapid progress in genomic technologies. The presence of genomic instability raises serious safety concerns and can hamper the advancement of iPSC-based therapies. Here, we summarize our current knowledge on genomic instability of iPSCs and challenges in their clinical applications. In view of the recent expansion of stem cell therapies, it is crucial to gain deeper mechanistic insights into the genetic aberrations, ranging from chromosomal aberrations, copy number variations to point mutations. On the basis of their origin, these genetic aberrations in iPSCs can be classified as (i) preexisting mutations in parental somatic cells, (ii) reprogramming-induced mutations, and (iii) mutations that arise during in vitro culture. However, it is still unknown whether these genetic aberrations in iPSCs can be an actual risk factor for adverse effects. Intersection of the genomic data on iPSCs with the patients' clinical follow-up data will help to produce evidence-based criteria for clinical application. Furthermore, we discuss novel approaches to generate iPSCs with fewer genetic aberrations. Better understanding of iPSCs from both basic and clinical aspects will pave the way for iPSC-based therapies.
Collapse
Affiliation(s)
- Masahito Yoshihara
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Akiko Oguchi
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yasuhiro Murakawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.
- IFOM, The FIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
69
|
Embryonic Stem Cells in Clinical Trials: Current Overview of Developments and Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1312:19-37. [PMID: 33159303 DOI: 10.1007/5584_2020_592] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The first isolation of human embryonic stem cells (hESC) reported in the late 90s opened a new window to promising possibilities in the fields of human developmental biology and regenerative medicine. Subsequently, the differentiation of hESC lines into different precursor cells showed their potential in treating different incurable diseases. However, this promising field has consistently had remarkable ethical and experimental limitations. This paper is a review of clinical trial studies dealing with hESC and their advantages, limitations, and other specific concerns. Some of the hESC limitations have been solved, and several clinical trial studies are ongoing so that recent clinical trials have strived to improve the clinical applications of hESC, especially in macular degeneration and neurodegenerative diseases. However, regarding hESC-based therapy, several important issues need more research and discussion. Despite considerable studies to Date, hESC-based therapy is not available for conventional clinical applications, and more studies and data are needed to overcome current clinical and ethical limitations. When all the limitations of Embryonic stem cells (ESC) are wholly resolved, perhaps hESC can become superior to the existing stem cell sources. This overview will be beneficial for understanding the standard and promising applications of cell and tissue-based therapeutic approaches and for developing novel therapeutic applications of hESC.
Collapse
|
70
|
Abstract
PURPOSE OF REVIEW The emergence of cell-based therapies has brought much excitement to the field of orthopedic sports medicine. However, the significant inconsistency of reporting has led to the poor understanding, misinformation, and false expectations for patients and clinicians alike. In this paper, we aim to clarify the available cell-therapy treatments and summarize some of the latest research. RECENT FINDINGS Although this technology is in early development, our understanding of cell biology has grown significantly over the last decade. Furthermore, it is becoming evident that tissue specificity may play a significant role in determining the effectiveness and overall clinical benefit attributed to cell therapy. Cell therapy is an emerging field with tremendous potential for clinically significant benefit. However, in its current state, clinical application of these treatments is limited by federal regulations, variability in formulation, and limited understanding of the biologic activity of various cell formulations.
Collapse
Affiliation(s)
- Bijan Dehghani
- Hospital for Special Surgery, 525 East 71 Street, 1st floor, New York, NY 10021 USA
| | - Scott Rodeo
- Hospital for Special Surgery, 525 East 71 Street, 1st floor, New York, NY 10021 USA
| |
Collapse
|
71
|
Nikitina TV, Kashevarova AA, Lebedev IN. Chromosomal Instability and Karyotype Correction in Human Induced Pluripotent Stem Cells. RUSS J GENET+ 2019. [DOI: 10.1134/s1022795419100090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
72
|
Uncovering low-level mosaicism in human embryonic stem cells using high throughput single cell shallow sequencing. Sci Rep 2019; 9:14844. [PMID: 31619727 PMCID: PMC6796059 DOI: 10.1038/s41598-019-51314-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/25/2019] [Indexed: 01/05/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) have significant levels of low-grade genetic mosaicism, which commonly used techniques fail to detect in bulk DNA. These copy number variations remain a hurdle for the clinical translation of hPSC, as their effect in vivo ranges from unknown to dangerous, and the ability to detect them will be necessary as the field advances. As such there is need for techniques which can efficiently analyse genetic content in single cells with higher throughput and lower costs. We report here on the use of the Fluidigm C1 single cell WGA platform in combination with shallow whole genome sequencing to analyse the genetic content of single hPSCs. From a hPSC line carrying an isochromosome 20, 56 single cells were analysed and found to carry a total of 50 aberrations, across 23% of cells, which could not be detected by bulk analysis. Aberrations were predominantly segmental gains, with a fewer number of segmental losses and aneuploidies. Interestingly, 40% of the breakpoints seen here correspond to known DNA fragile sites. Our results therefore demonstrate the feasibility of single cell shallow sequencing of hPSC and further expand upon the biological importance and frequency of single cell mosaicism in hPSC.
Collapse
|
73
|
Avior Y, Eggan K, Benvenisty N. RETRACTED: Cancer-Related Mutations Identified in Primed and Naive Human Pluripotent Stem Cells. Cell Stem Cell 2019; 25:456-461. [DOI: 10.1016/j.stem.2019.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
74
|
Singh R, Fazal Z, Freemantle SJ, Spinella MJ. Mechanisms of cisplatin sensitivity and resistance in testicular germ cell tumors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:580-594. [PMID: 31538140 PMCID: PMC6752046 DOI: 10.20517/cdr.2019.19] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Testicular germ cell tumors (TGCTs) are a cancer pharmacology success story with a majority of patients cured even in the highly advanced and metastatic setting. Successful treatment of TGCTs is primarily due to the exquisite responsiveness of this solid tumor to cisplatin-based therapy. However, a significant percentage of patients are, or become, refractory to cisplatin and die from progressive disease. Mechanisms for both clinical hypersensitivity and resistance have largely remained a mystery despite the promise of applying lessons to the majority of solid tumors that are not curable in the metastatic setting. Recently, this promise has been heightened by the realization that distinct (and perhaps pharmacologically replicable) epigenetic states, rather than fixed genetic alterations, may play dominant roles in not only TGCT etiology and progression but also their curability with conventional chemotherapies. In this review, it discusses potential mechanisms of TGCT cisplatin sensitivity and resistance to conventional chemotherapeutics.
Collapse
Affiliation(s)
- Ratnakar Singh
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Zeeshan Fazal
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sarah J Freemantle
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Michael J Spinella
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,The Carle Illinois College of Medicine , University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,The Cancer Center of Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
75
|
Yang X, Wang R, Wang X, Cai G, Qian Y, Feng S, Tan F, Chen K, Tang K, Huang X, Jing N, Qiao Y. TGFβ signaling hyperactivation-induced tumorigenicity during the derivation of neural progenitors from mouse ESCs. J Mol Cell Biol 2019; 10:216-228. [PMID: 29481611 DOI: 10.1093/jmcb/mjy013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/20/2018] [Indexed: 02/06/2023] Open
Abstract
Clinical therapies of pluripotent stem cells (PSCs)-based transplantation have been hindered by frequent development of teratomas or tumors in animal models and clinical patients. Therefore, clarifying the mechanism of carcinogenesis in stem cell therapy is of great importance for reducing the risk of tumorigenicity. Here we differentiate Oct4-GFP mouse embryonic stem cells (mESCs) into neural progenitor cells (NPCs) and find that a minority of Oct4+ cells are continuously sustained at Oct4+ state. These cells can be enriched and proliferated in a standard ESC medium. Interestingly, the differentiation potential of these enriched cells is tightly restricted with much higher tumorigenic activity, which are thus defined as differentiation-resistant ESCs (DR-ESCs). Transcriptomic and epigenomic analyses show that DR-ESCs are characterized by primordial germ cell-like gene signatures (Dazl, Rec8, Stra8, Blimp1, etc.) and specific epigenetic patterns distinct from mESCs. Moreover, the DR-ESCs possess germ cell potential to generate Sycp3+ haploid cells and are able to reside in sperm-free spermaduct induced by busulfan. Finally, we find that TGFβ signaling is overactivated in DR-ESCs, and inhibition of TGFβ signaling eliminates the tumorigenicity of mESC-derived NPCs by inducing the full differentiation of DR-ESCs. These data demonstrate that these TGFβ-hyperactivated germ cell-like DR-ESCs are the main contributor for the tumorigenicity of ESCs-derived target cell therapy and that inhibition of TGFβ signaling in ESC-derived NPC transplantation could drastically reduce the risk of tumor development.
Collapse
Affiliation(s)
- Xianfa Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ran Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Xiongjun Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Guoqing Cai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Yun Qian
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Su Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Fangzhi Tan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kun Chen
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Ke Tang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yunbo Qiao
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| |
Collapse
|
76
|
Jaroy EG, Acosta-Jimenez L, Hotta R, Goldstein AM, Emblem R, Klungland A, Ougland R. "Too much guts and not enough brains": (epi)genetic mechanisms and future therapies of Hirschsprung disease - a review. Clin Epigenetics 2019; 11:135. [PMID: 31519213 PMCID: PMC6743154 DOI: 10.1186/s13148-019-0718-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/29/2019] [Indexed: 12/21/2022] Open
Abstract
Hirschsprung disease is a neurocristopathy, characterized by aganglionosis in the distal bowel. It is caused by failure of the enteric nervous system progenitors to migrate, proliferate, and differentiate in the gut. Development of an enteric nervous system is a tightly regulated process. Both the neural crest cells and the surrounding environment are regulated by different genes, signaling pathways, and morphogens. For this process to be successful, the timing of gene expression is crucial. Hence, alterations in expression of genes specific for the enteric nervous system may contribute to the pathogenesis of Hirschsprung’s disease. Several epigenetic mechanisms contribute to regulate gene expression, such as modifications of DNA and RNA, histone modifications, and microRNAs. Here, we review the current knowledge of epigenetic and epitranscriptomic regulation in the development of the enteric nervous system and its potential significance for the pathogenesis of Hirschsprung’s disease. We also discuss possible future therapies and how targeting epigenetic and epitranscriptomic mechanisms may open new avenues for novel treatment.
Collapse
Affiliation(s)
- Emilie G Jaroy
- Clinic for Diagnostics and Intervention and Institute of Medical Microbiology, Oslo University Hospital, Rikshospitalet, 0027, Oslo, Norway.,Department of Pediatric Surgery, Oslo University Hospital, Rikshospitalet, 0424, Oslo, Norway.,Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Lourdes Acosta-Jimenez
- Clinic for Diagnostics and Intervention and Institute of Medical Microbiology, Oslo University Hospital, Rikshospitalet, 0027, Oslo, Norway.,Department of Pediatric Surgery, Oslo University Hospital, Rikshospitalet, 0424, Oslo, Norway.,Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ragnhild Emblem
- Department of Pediatric Surgery, Oslo University Hospital, Rikshospitalet, 0424, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0317, Oslo, Norway
| | - Arne Klungland
- Clinic for Diagnostics and Intervention and Institute of Medical Microbiology, Oslo University Hospital, Rikshospitalet, 0027, Oslo, Norway.,Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Rune Ougland
- Clinic for Diagnostics and Intervention and Institute of Medical Microbiology, Oslo University Hospital, Rikshospitalet, 0027, Oslo, Norway. .,Department of Surgery, Baerum Hospital, Vestre Viken Hospital Trust, 3004, Drammen, Norway.
| |
Collapse
|
77
|
Abstract
Human germ cell tumours (GCTs) are derived from stem cells of the early embryo and the germ line. They occur in the gonads (ovaries and testes) and also in extragonadal sites, where migrating primordial germ cells are located during embryogenesis. This group of heterogeneous neoplasms is unique in that their developmental potential is in effect determined by the latent potency state of their cells of origin, which are reprogrammed to omnipotent, totipotent or pluripotent stem cells. Seven GCT types, defined according to their developmental potential, have been identified, each with distinct epidemiological and (epi)genomic features. Heritable predisposition factors affecting the cells of origin and their niches likely explain bilateral, multiple and familial occurrences of the different types of GCTs. Unlike most other tumour types, GCTs are rarely caused by somatic driver mutations, but arise through failure to control the latent developmental potential of their cells of origin, resulting in their reprogramming. Consistent with their non-mutational origin, even the malignant tumours of the group are characterized by wild-type TP53 and high sensitivity for DNA damage. However, tumour progression and the rare occurrence of treatment resistance are driven by embryonic epigenetic state, specific (sub)chromosomal imbalances and somatic mutations. Thus, recent progress in understanding GCT biology supports a comprehensive developmental pathogenetic model for the origin of all GCTs, and provides new biomarkers, as well as potential targets for treatment of resistant disease.
Collapse
Affiliation(s)
- J Wolter Oosterhuis
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, Netherlands.
| | - Leendert H J Looijenga
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
78
|
Ouyang X, Telli ML, Wu JC. Induced Pluripotent Stem Cell-Based Cancer Vaccines. Front Immunol 2019; 10:1510. [PMID: 31338094 PMCID: PMC6628907 DOI: 10.3389/fimmu.2019.01510] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Over a century ago, it was reported that immunization with embryonic/fetal tissue could lead to the rejection of transplanted tumors in animals. Subsequent studies demonstrated that vaccination of embryonic materials in animals induced cellular and humoral immunity against transplantable tumors and carcinogen-induced tumors. Therefore, it has been hypothesized that the shared antigens between tumors and embryonic/fetal tissues (oncofetal antigens) are the key to anti-tumor immune responses in these studies. However, early oncofetal antigen-based cancer vaccines usually utilize xenogeneic or allogeneic embryonic stem cells or tissues, making it difficult to tease apart the anti-tumor immunity elicited by the oncofetal antigens vs. graft-vs.-host responses. Recently, one oncofetal antigen-based cancer vaccine using autologous induced pluripotent stem cells (iPSCs) demonstrated marked prophylactic and therapeutic potential, suggesting critical roles of oncofetal antigens in inducing anti-tumor immunity. In this review, we present an overview of recent studies in the field of oncofetal antigen-based cancer vaccines, including single peptide-based cancer vaccines, embryonic stem cell (ESC)- and iPSC-based whole-cell vaccines, and provide insights on future directions.
Collapse
Affiliation(s)
- Xiaoming Ouyang
- Cardiovascular Institute, School of Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, United States
| | - Melinda L Telli
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Joseph C Wu
- Cardiovascular Institute, School of Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, United States.,Department of Medicine, Stanford University, Stanford, CA, United States.,Department of Radiology, Stanford University, Stanford, CA, United States
| |
Collapse
|
79
|
Brás R, Sunkel CE, Resende LP. Tissue stem cells: the new actors in the aneuploidy field. Cell Cycle 2019; 18:1813-1823. [PMID: 31242809 DOI: 10.1080/15384101.2019.1635867] [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: 10/26/2022] Open
Abstract
The development of multicellular organisms and the maintenance of its tissues relies on mitosis. However, this process represents a major challenge for genomic stability as each time a cell division occurs there are multiple steps where errors can lead to an abnormal chromosomal content in daughter cells - aneuploidy. Aneuploidy was first postulated to act as a tumour promoting agent over one century ago. Since then, we have learned to appreciate the complexity involving the cellular responses to aneuploidy and to value the importance of models where aneuploidy is induced in vivo and in a cell-type specific manner. Recent data suggests that stem cells evolved a distinct response to aneuploidy, being able to survive and proliferate as aneuploid. Since stem cells are the main cells responsible for tissue renewal, it is of the utmost importance to place the spotlight on stem cells within the aneuploidy field. Here, we briefly review some of the biological mechanisms implicated in aneuploidy, the relationship between aneuploidy and tissue pathologies, and summarize the most recent findings in Drosophila on how tissue stem cells respond to aneuploidy. Once we understand how stem cell behavior is impacted by aneuploidy, we might be able to better describe the complicated link between aneuploidy and tumourigenesis.
Collapse
Affiliation(s)
- Rita Brás
- a Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal.,b IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto , Porto , Portugal
| | - Claudio E Sunkel
- a Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal.,c ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto , Porto , Portugal
| | - Luís Pedro Resende
- a Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal.,b IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto , Porto , Portugal
| |
Collapse
|
80
|
Markouli C, Couvreu De Deckersberg E, Regin M, Nguyen HT, Zambelli F, Keller A, Dziedzicka D, De Kock J, Tilleman L, Van Nieuwerburgh F, Franceschini L, Sermon K, Geens M, Spits C. Gain of 20q11.21 in Human Pluripotent Stem Cells Impairs TGF-β-Dependent Neuroectodermal Commitment. Stem Cell Reports 2019; 13:163-176. [PMID: 31178415 PMCID: PMC6627003 DOI: 10.1016/j.stemcr.2019.05.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023] Open
Abstract
Gain of 20q11.21 is one of the most common recurrent genomic aberrations in human pluripotent stem cells. Although it is known that overexpression of the antiapoptotic gene Bcl-xL confers a survival advantage to the abnormal cells, their differentiation capacity has not been fully investigated. RNA sequencing of mutant and control hESC lines, and a line transgenically overexpressing Bcl-xL, shows that overexpression of Bcl-xL is sufficient to cause most transcriptional changes induced by the gain of 20q11.21. Moreover, the differentially expressed genes in mutant and Bcl-xL overexpressing lines are enriched for genes involved in TGF-β- and SMAD-mediated signaling, and neuron differentiation. Finally, we show that this altered signaling has a dramatic negative effect on neuroectodermal differentiation, while the cells maintain their ability to differentiate to mesendoderm derivatives. These findings stress the importance of thorough genetic testing of the lines before their use in research or the clinic. Bcl-xL overexpression drives the transcriptomic profile of 20q11.21 mutant lines 20q11.21 mutant lines downregulate CHCHD2, a known TGF-β pathway modulator Mutant lines differentially express genes involved in TGF-β and SMAD signaling Mutant lines show impaired ectoderm commitment due to TGF-β signaling deregulation
Collapse
Affiliation(s)
- C Markouli
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - E Couvreu De Deckersberg
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - M Regin
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - H T Nguyen
- Center for Molecular Biology, Institute of Research and Development, Duy Tan University, K7/25 Quang Trung, Danang 550000, Vietnam
| | - F Zambelli
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium; Clínica EUGIN, Travessera de les Corts 322, 08029 Barcelona, Spain
| | - A Keller
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - D Dziedzicka
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - J De Kock
- Department of In Vitro Toxicology & Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - L Tilleman
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - F Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - L Franceschini
- Laboratory of Molecular & Cellular Therapy, Department of Immunology - Physiology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - K Sermon
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - M Geens
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - C Spits
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
81
|
Bar S, Benvenisty N. Epigenetic aberrations in human pluripotent stem cells. EMBO J 2019; 38:embj.2018101033. [PMID: 31088843 DOI: 10.15252/embj.2018101033] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/13/2019] [Accepted: 03/15/2019] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are being increasingly utilized worldwide in investigating human development, and modeling and discovering therapies for a wide range of diseases as well as a source for cellular therapy. Yet, since the first isolation of human embryonic stem cells (hESCs) 20 years ago, followed by the successful reprogramming of human-induced pluripotent stem cells (hiPSCs) 10 years later, various studies shed light on abnormalities that sometimes accumulate in these cells in vitro Whereas genetic aberrations are well documented, epigenetic alterations are not as thoroughly discussed. In this review, we highlight frequent epigenetic aberrations found in hPSCs, including alterations in DNA methylation patterns, parental imprinting, and X chromosome inactivation. We discuss the potential origins of these abnormalities in hESCs and hiPSCs, survey the different methods for detecting them, and elaborate on their potential consequences for the different utilities of hPSCs.
Collapse
Affiliation(s)
- Shiran Bar
- Department of Genetics, The Azrieli Center for Stem Cells and Genetic Research, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | - Nissim Benvenisty
- Department of Genetics, The Azrieli Center for Stem Cells and Genetic Research, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
82
|
Xie X, Wu H, Li M, Chen X, Xu X, Ni W, Lu C, Ni R, Bao B, Xiao M. Progress in the application of exosomes as therapeutic vectors in tumor-targeted therapy. Cytotherapy 2019; 21:509-524. [DOI: 10.1016/j.jcyt.2019.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022]
|
83
|
Schumann GG, Fuchs NV, Tristán-Ramos P, Sebe A, Ivics Z, Heras SR. The impact of transposable element activity on therapeutically relevant human stem cells. Mob DNA 2019; 10:9. [PMID: 30899334 PMCID: PMC6408843 DOI: 10.1186/s13100-019-0151-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
Human stem cells harbor significant potential for basic and clinical translational research as well as regenerative medicine. Currently ~ 3000 adult and ~ 30 pluripotent stem cell-based, interventional clinical trials are ongoing worldwide, and numbers are increasing continuously. Although stem cells are promising cell sources to treat a wide range of human diseases, there are also concerns regarding potential risks associated with their clinical use, including genomic instability and tumorigenesis concerns. Thus, a deeper understanding of the factors and molecular mechanisms contributing to stem cell genome stability are a prerequisite to harnessing their therapeutic potential for degenerative diseases. Chemical and physical factors are known to influence the stability of stem cell genomes, together with random mutations and Copy Number Variants (CNVs) that accumulated in cultured human stem cells. Here we review the activity of endogenous transposable elements (TEs) in human multipotent and pluripotent stem cells, and the consequences of their mobility for genomic integrity and host gene expression. We describe transcriptional and post-transcriptional mechanisms antagonizing the spread of TEs in the human genome, and highlight those that are more prevalent in multipotent and pluripotent stem cells. Notably, TEs do not only represent a source of mutations/CNVs in genomes, but are also often harnessed as tools to engineer the stem cell genome; thus, we also describe and discuss the most widely applied transposon-based tools and highlight the most relevant areas of their biomedical applications in stem cells. Taken together, this review will contribute to the assessment of the risk that endogenous TE activity and the application of genetically engineered TEs constitute for the biosafety of stem cells to be used for substitutive and regenerative cell therapies.
Collapse
Affiliation(s)
- Gerald G Schumann
- 1Division of Medical Biotechnology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str.51-59, 63225 Langen, Germany
| | - Nina V Fuchs
- 2Host-Pathogen Interactions, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Pablo Tristán-Ramos
- 3GENYO. Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada-Avenida de la Ilustración, 114, 18016 Granada, Spain.,4Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus Universitario de Cartuja, 18071 Granada, Spain
| | - Attila Sebe
- 1Division of Medical Biotechnology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str.51-59, 63225 Langen, Germany
| | - Zoltán Ivics
- 1Division of Medical Biotechnology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str.51-59, 63225 Langen, Germany
| | - Sara R Heras
- 3GENYO. Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada-Avenida de la Ilustración, 114, 18016 Granada, Spain.,4Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus Universitario de Cartuja, 18071 Granada, Spain
| |
Collapse
|
84
|
Tubuloids derived from human adult kidney and urine for personalized disease modeling. Nat Biotechnol 2019; 37:303-313. [PMID: 30833775 DOI: 10.1038/s41587-019-0048-8] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 01/23/2019] [Indexed: 01/10/2023]
Abstract
Adult stem cell-derived organoids are three-dimensional epithelial structures that recapitulate fundamental aspects of their organ of origin. We describe conditions for the long-term growth of primary kidney tubular epithelial organoids, or 'tubuloids'. The cultures are established from human and mouse kidney tissue and can be expanded for at least 20 passages (>6 months) while retaining a normal number of chromosomes. In addition, cultures can be established from human urine. Human tubuloids represent proximal as well as distal nephron segments, as evidenced by gene expression, immunofluorescence and tubular functional analyses. We apply tubuloids to model infectious, malignant and hereditary kidney diseases in a personalized fashion. BK virus infection of tubuloids recapitulates in vivo phenomena. Tubuloids are established from Wilms tumors. Kidney tubuloids derived from the urine of a subject with cystic fibrosis allow ex vivo assessment of treatment efficacy. Finally, tubuloids cultured on microfluidic organ-on-a-chip plates adopt a tubular conformation and display active (trans-)epithelial transport function.
Collapse
|
85
|
Laing O, Halliwell J, Barbaric I. Rapid PCR Assay for Detecting Common Genetic Variants Arising in Human Pluripotent Stem Cell Cultures. ACTA ACUST UNITED AC 2019; 49:e83. [PMID: 30821932 DOI: 10.1002/cpsc.83] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Human pluripotent stem cells (hPSCs) are prone to acquiring genetic changes upon prolonged culture. Particularly common are copy number changes, including gains of chromosomes 1q, 12p, 17q, and 20q, and/or loss of chromosomes 10p and 18q. The variant cells harboring common genetic changes display altered behaviors compared to their diploid counterparts, thus potentially impacting upon the validity of experimental results and safety of hPSC-derived cellular therapies. Hence, a critical quality attribute in hPSC maintenance should include frequent monitoring for genetic changes arising in cultures. This in turn places large demands on the genotyping assays for detection of genetic changes. Traditional methods for screening cells entail specialized cytogenetic analyses, but their high costs and a lengthy turnaround time make them impractical for high-throughput analyses and routine laboratory use. Here, we detail a protocol for a rapid, accessible, and affordable PCR-based method for detection of frequently occurring copy number changes in hPSCs. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Owen Laing
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield, United Kingdom
| | - Jason Halliwell
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield, United Kingdom
| | - Ivana Barbaric
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
86
|
Kohutova A, Raška J, Kruta M, Seneklova M, Barta T, Fojtik P, Jurakova T, Walter CA, Hampl A, Dvorak P, Rotrekl V. Ligase 3–mediated end‐joining maintains genome stability of human embryonic stem cells. FASEB J 2019; 33:6778-6788. [DOI: 10.1096/fj.201801877rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Aneta Kohutova
- Department of BiologyMasaryk UniversityBrnoCzech Republic
- International Clinical Research Center (ICRC)St. Anne's University HospitalBrnoCzech Republic
| | - Jan Raška
- Department of BiologyMasaryk UniversityBrnoCzech Republic
| | - Miriama Kruta
- Department of BiologyMasaryk UniversityBrnoCzech Republic
| | | | - Tomas Barta
- Department of BiologyMasaryk UniversityBrnoCzech Republic
| | - Petr Fojtik
- Department of BiologyMasaryk UniversityBrnoCzech Republic
| | | | - Christi A. Walter
- Department of Cell Systems and AnatomyThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Ales Hampl
- Department of Histology and EmbryologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research Center (ICRC)St. Anne's University HospitalBrnoCzech Republic
| | - Petr Dvorak
- Department of BiologyMasaryk UniversityBrnoCzech Republic
- International Clinical Research Center (ICRC)St. Anne's University HospitalBrnoCzech Republic
| | - Vladimir Rotrekl
- Department of BiologyMasaryk UniversityBrnoCzech Republic
- International Clinical Research Center (ICRC)St. Anne's University HospitalBrnoCzech Republic
| |
Collapse
|
87
|
Henry MP, Hawkins JR, Boyle J, Bridger JM. The Genomic Health of Human Pluripotent Stem Cells: Genomic Instability and the Consequences on Nuclear Organization. Front Genet 2019; 9:623. [PMID: 30719030 PMCID: PMC6348275 DOI: 10.3389/fgene.2018.00623] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are increasingly used for cell-based regenerative therapies worldwide, with embryonic and induced pluripotent stem cells as potential treatments for debilitating and chronic conditions, such as age-related macular degeneration, Parkinson's disease, spinal cord injuries, and type 1 diabetes. However, with the level of genomic anomalies stem cells generate in culture, their safety may be in question. Specifically, hPSCs frequently acquire chromosomal abnormalities, often with gains or losses of whole chromosomes. This review discusses how important it is to efficiently and sensitively detect hPSC aneuploidies, to understand how these aneuploidies arise, consider the consequences for the cell, and indeed the individual to whom aneuploid cells may be administered.
Collapse
Affiliation(s)
- Marianne P Henry
- Advanced Therapies Division, National Institute for Biological Standards and Control, Potters Bar, United Kingdom.,Laboratory of Nuclear and Genomic Health, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - J Ross Hawkins
- Advanced Therapies Division, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | - Jennifer Boyle
- Advanced Therapies Division, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | - Joanna M Bridger
- Laboratory of Nuclear and Genomic Health, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| |
Collapse
|
88
|
Chemically Defined Neural Conversion of Human Pluripotent Stem Cells. Methods Mol Biol 2019. [PMID: 30656621 DOI: 10.1007/978-1-4939-9007-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Human pluripotent stem cells (hPSCs) are characterized by their ability to self-renew and differentiate into any cell type of the human body. To fully utilize the potential of hPSCs for translational research and clinical applications, it is critical to develop rigorous cell differentiation protocols under feeder-free conditions that are efficient, reproducible, and scalable for high-throughput projects. Focusing on neural conversion of hPSCs, here we describe robust small molecule-based procedures that generate neural stem cells (NSCs) in less than a week under chemically defined conditions. These protocols can be used to dissect the mechanisms of neural lineage entry and to further develop systematic protocols that produce the cellular diversity of the central nervous system at industrial scale.
Collapse
|
89
|
Chunduri NK, Storchová Z. The diverse consequences of aneuploidy. Nat Cell Biol 2019; 21:54-62. [PMID: 30602769 DOI: 10.1038/s41556-018-0243-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/31/2018] [Indexed: 12/25/2022]
Abstract
Aneuploidy, or imbalanced chromosome number, has profound effects on eukaryotic cells. In humans, aneuploidy is associated with various pathologies, including cancer, which suggests that it mediates a proliferative advantage under these conditions. Here, we discuss physiological changes triggered by aneuploidy, such as altered cell growth, transcriptional changes, proteotoxic stress, genomic instability and response to interferons, and how cancer cells adapt to the changing aneuploid genome.
Collapse
Affiliation(s)
| | - Zuzana Storchová
- Department of Molecular Genetics, TU Kaiserslautern, Kaiserslautern, Germany.
| |
Collapse
|
90
|
Morrison LC, Tatari N, Werbowetski-Ogilvie TE. Embryonic Stem Cell Models of Human Brain Tumors. Methods Mol Biol 2019; 1869:127-142. [PMID: 30324520 DOI: 10.1007/978-1-4939-8805-1_12] [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: 12/23/2022]
Abstract
Utilization of human embryonic stem cells (hESCs) as a model system to study highly malignant pediatric cancers has led to significant insight into the molecular mechanisms governing tumor progression and has revealed novel therapeutic targets for these devastating diseases. Here, we describe a method for generating heterogeneous populations of neural precursors from both normal and neoplastic hESCs and the subsequent injection of neoplastic human embryonic neural cells (hENs) into intracerebellar or intracranial xenograft models. Histopathologically, neural tumors derived from neoplastic hENs exhibit features similar to more aggressive medulloblastoma, the most common malignant primary pediatric brain tumor. In this chapter, we will outline the detailed methods for culturing normal and neoplastic neural precursor cells in both adherent and tumorsphere format and the full characterization of the brain tumors generated from these cells in non-obese diabetic severe combined immunodeficiency (NOD SCID) mice.
Collapse
Affiliation(s)
- Ludivine Coudière Morrison
- Regenerative Medicine Program, Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Regenerative Medicine Program, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Nazanin Tatari
- Regenerative Medicine Program, Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Regenerative Medicine Program, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.,McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Tamra E Werbowetski-Ogilvie
- Regenerative Medicine Program, Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada. .,Regenerative Medicine Program, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
91
|
Shilina MA, Grinchuk TM, Anatskaya OV, Vinogradov AE, Alekseenko LL, Elmuratov AU, Nikolsky NN. Cytogenetic and Transcriptomic Analysis of Human Endometrial MSC Retaining Proliferative Activity after Sublethal Heat Shock. Cells 2018; 7:cells7110184. [PMID: 30366433 PMCID: PMC6262560 DOI: 10.3390/cells7110184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/19/2018] [Accepted: 10/23/2018] [Indexed: 12/14/2022] Open
Abstract
Temperature is an important exogenous factor capable of leading to irreversible processes in the vital activity of cells. However, the long-term effects of heat shock (HS) on mesenchymal stromal cells (MSC) remain unstudied. We investigated the karyotype and DNA repair drivers and pathways in the human endometrium MSC (eMSC) survived progeny at passage 6 after sublethal heat stress (sublethal heat stress survived progeny (SHS-SP)). G-banding revealed an outbreak of random karyotype instability caused by chromosome breakages and aneuploidy. Molecular karyotyping confirmed the random nature of this instability. Transcriptome analysis found homologous recombination (HR) deficiency that most likely originated from the low thermostability of the AT-rich HR driving genes. SHS-SP protection from transformation is provided presumably by low oncogene expression maintained by tight co-regulation between thermosensitive HR drivers BRCA, ATM, ATR, and RAD51 (decreasing expression after SHS), and oncogenes mTOR, MDM2, KRAS, and EGFR. The cancer-related transcriptomic features previously identified in hTERT transformed MSC in culture were not found in SHS-SP, suggesting no traits of malignancy in them. The entrance of SHS-SP into replicative senescence after 25 passages confirms their mortality and absence of transformation features. Overall, our data indicate that SHS may trigger non-tumorigenic karyotypic instability due to HR deficiency and decrease of oncogene expression in progeny of SHS-survived MSC. These data can be helpful for the development of new therapeutic approaches in personalized medicine.
Collapse
Affiliation(s)
- Mariia A Shilina
- Institute of Cytology, Russian Academy of Sciences, Tikhoretskay Ave 4, St. 194064 Petersburg, Russia.
| | - Tatiana M Grinchuk
- Institute of Cytology, Russian Academy of Sciences, Tikhoretskay Ave 4, St. 194064 Petersburg, Russia.
| | - Olga V Anatskaya
- Institute of Cytology, Russian Academy of Sciences, Tikhoretskay Ave 4, St. 194064 Petersburg, Russia.
| | - Alexander E Vinogradov
- Institute of Cytology, Russian Academy of Sciences, Tikhoretskay Ave 4, St. 194064 Petersburg, Russia.
| | - Larisa L Alekseenko
- Institute of Cytology, Russian Academy of Sciences, Tikhoretskay Ave 4, St. 194064 Petersburg, Russia.
| | - Artem U Elmuratov
- Institute of Biomedical Chemistry (IBMC) of Russian Academy of Sciences, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia.
- Medical Genetics Centre Genotek, Nastavnichesky Alley 17-1-15, 10510 Moscow, Russia.
| | - Nikolai N Nikolsky
- Institute of Cytology, Russian Academy of Sciences, Tikhoretskay Ave 4, St. 194064 Petersburg, Russia.
| |
Collapse
|
92
|
Al-Ani A, Toms D, Kondro D, Thundathil J, Yu Y, Ungrin M. Oxygenation in cell culture: Critical parameters for reproducibility are routinely not reported. PLoS One 2018; 13:e0204269. [PMID: 30325922 PMCID: PMC6191109 DOI: 10.1371/journal.pone.0204269] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/04/2018] [Indexed: 01/01/2023] Open
Abstract
Mammalian cell culture is foundational to biomedical research, and the reproducibility of research findings across the sciences is drawing increasing attention. While many components contribute to reproducibility, the reporting of factors that impact oxygen delivery in the general biomedical literature has the potential for both significant impact, and immediate improvement. The relationship between the oxygen consumption rate of cells and the diffusive delivery of oxygen through the overlying medium layer means parameters such as medium depth and cell type can cause significant differences in oxygenation for cultures nominally maintained under the same conditions. While oxygenation levels are widely understood to significantly impact the phenotype of cultured cells in the abstract, in practise the importance of the above parameters does not appear to be well recognized in the non-specialist research community. On analyzing two hundred articles from high-impact journals we find a large majority missing at least one key piece of information necessary to ensure consistency in replication. We propose that explicitly reporting these values should be a requirement for publication.
Collapse
Affiliation(s)
- Abdullah Al-Ani
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Derek Toms
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Douglas Kondro
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Jarin Thundathil
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Yang Yu
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mark Ungrin
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- * E-mail:
| |
Collapse
|
93
|
Cho SJ, Kim KT, Jeong HC, Park JC, Kwon OS, Song YH, Shin JG, Kang S, Kim W, Shin HD, Lee MO, Moon SH, Cha HJ. Selective Elimination of Culture-Adapted Human Embryonic Stem Cells with BH3 Mimetics. Stem Cell Reports 2018; 11:1244-1256. [PMID: 30293852 PMCID: PMC6235677 DOI: 10.1016/j.stemcr.2018.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/05/2023] Open
Abstract
The selective survival advantage of culture-adapted human embryonic stem cells (hESCs) is a serious safety concern for their clinical application. With a set of hESCs with various passage numbers, we observed that a subpopulation of hESCs at late passage numbers was highly resistant to various cell death stimuli, such as YM155, a survivin inhibitor. Transcriptome analysis from YM155-sensitive (YM155S) and YM155-resistant (YM155R) hESCs demonstrated that BCL2L1 was highly expressed in YM155R hESCs. By matching the gene signature of YM155R hESCs with the Cancer Therapeutics Response Portal dataset, BH3 mimetics were predicted to selectively ablate these cells. Indeed, short-course treatment with a sub-optimal dose of BH3 mimetics induced the spontaneous death of YM155R, but not YM155S hESCs by disrupting the mitochondrial membrane potential. YM155S hESCs remained pluripotent following BH3 mimetics treatment. Therefore, the use of BH3 mimetics is a promising strategy to specifically eliminate hESCs with a selective survival advantage. Culture-adapted hESCs against YM155/genotoxic agents mediated by high BCL-xL expression Selective cell death of culture-adapted hPSCs by BH3 mimetics Pluripotency maintenance of normal hESCs after exposure to BH3 mimetics
Collapse
Affiliation(s)
- Seung-Ju Cho
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Keun-Tae Kim
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Ho-Chang Jeong
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Ju-Chan Park
- School of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Ok-Seon Kwon
- School of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Yun-Ho Song
- Department of Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Joong-Gon Shin
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Seungmin Kang
- Ewha Research Center for Systems Biology, Division of Molecular & Life Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Wankyu Kim
- Ewha Research Center for Systems Biology, Division of Molecular & Life Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hyoung Doo Shin
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Mi-Ok Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Sung-Hwan Moon
- Department of Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Hyuk-Jin Cha
- School of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
94
|
Gao W, Wu D, Wang Y, Wang Z, Zou C, Dai Y, Ng CF, Teoh JYC, Chan FL. Development of a novel and economical agar-based non-adherent three-dimensional culture method for enrichment of cancer stem-like cells. Stem Cell Res Ther 2018; 9:243. [PMID: 30257704 PMCID: PMC6158801 DOI: 10.1186/s13287-018-0987-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/07/2018] [Accepted: 08/17/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Non-adherent or ultra-low attachment three-dimensional (3D) culture, also called sphere formation assay, has been widely used to assess the malignant phenotype and stemness potential of transformed or cancer cells. This method is also popularly used to isolate the cancer stem-like cells (CSCs) or tumor-initiating cells based on their unique anchorage-independent growth or anoikis-resistant capacity. Different non-adhesive coating agents, such as poly-2-hydroxyethyl methacrylate (poly-HEMA) and synthetic hydrogels, have been used in this non-adherent 3D culture. However, preparation of non-adherent culture-ware is labor-intensive and technically demanding, and also costs of commercial non-adherent culture-ware prepared with various coating agents are relatively expensive and the culture-ware cannot be used repeatedly. METHODS In this study, we developed a non-adherent 3D culture method based on agar coating for growing tumor spheres derived from various cancer cell lines and primary prostate cancer tissues under a non-adherent and serum-free condition. The tumor spheres generated by this 3D culture method were analyzed on their expression profiles of CSC-associated markers by reverse transcription quantitative polymerase chain reaction, presence and relative proportion of CSCs by fluorescence-activated cell sorting (CD133+/CD44+ cell sorting) and also a CSC-visualizing reporter system responsive to OCT4 and SOX2 (SORE6), and in vivo tumorigenicity. The repeated use of agar-coated plates for serial passages of tumor spheres was also evaluated. RESULTS Our results validated that the multicellular tumor spheres generated by this culture method were enriched of CSCs, as evidenced by their enhanced expression profiles of CSC markers, presence of CD133+/CD44+ or SORE6+ cells, enhanced self-renewal capacity, and in vivo tumorigenicity, indicating its usefulness in isolation and enrichment of CSCs. The agar-coated plates could be used multiple times in serial passages of tumor spheres. CONCLUSIONS The described agar-based 3D culture method offers several advantages as compared with other methods in isolation of CSCs, including its simplicity and low-cost and repeated use of agar-coated plates for continuous passages of CSC-enriched spheres.
Collapse
Affiliation(s)
- Weijie Gao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Dinglan Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China. .,Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518110, China.
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Zhu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518000, China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518000, China
| | - Chi-Fai Ng
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeremy Yuen-Chun Teoh
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
95
|
Nguyen PK, Neofytou E, Rhee JW, Wu JC. Potential Strategies to Address the Major Clinical Barriers Facing Stem Cell Regenerative Therapy for Cardiovascular Disease: A Review. JAMA Cardiol 2018; 1:953-962. [PMID: 27579998 DOI: 10.1001/jamacardio.2016.2750] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Importance Although progress continues to be made in the field of stem cell regenerative medicine for the treatment of cardiovascular disease, significant barriers to clinical implementation still exist. Objectives To summarize the current barriers to the clinical implementation of stem cell therapy in patients with cardiovascular disease and to discuss potential strategies to overcome them. Evidence Review Information for this review was obtained through a search of PubMed and the Cochrane database for English-language studies published between January 1, 2000, and July 25, 2016. Ten randomized clinical trials and 8 systematic reviews were included. Findings One of the major clinical barriers facing the routine implementation of stem cell therapy in patients with cardiovascular disease is the limited and inconsistent benefit observed thus far. Reasons for this finding are unclear but may be owing to poor cell retention and survival, as suggested by numerous preclinical studies and a small number of human studies incorporating imaging to determine cell fate. Additional studies in humans using imaging to determine cell fate are needed to understand how these factors contribute to the limited efficacy of stem cell therapy. Treatment strategies to address poor cell retention and survival are under investigation and include the following: coadministration of immunosuppressive and prosurvival agents, delivery of cardioprotective factors packaged in exosomes rather than the cells themselves, and use of tissue-engineering strategies to provide structural support for cells. If larger grafts are achieved using these strategies, it will be imperative to carefully monitor for the potential risks of tumorigenicity, immunogenicity, and arrhythmogenicity. Conclusions and Relevance Despite important achievements to date, stem cell therapy is not yet ready for routine clinical implementation. Significant research is still needed to address the clinical barriers outlined herein before the next wave of large clinical trials is under way.
Collapse
Affiliation(s)
- Patricia K Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California3Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Evgenios Neofytou
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - June-Wha Rhee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California2Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California4Department of Radiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
96
|
Rohani L, Johnson AA, Naghsh P, Rancourt DE, Ulrich H, Holland H. Concise Review: Molecular Cytogenetics and Quality Control: Clinical Guardians for Pluripotent Stem Cells. Stem Cells Transl Med 2018; 7:867-875. [PMID: 30218497 PMCID: PMC6265634 DOI: 10.1002/sctm.18-0087] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 07/07/2018] [Indexed: 12/13/2022] Open
Abstract
Now that induced pluripotent stem cell (iPSC)‐based transplants have been performed in humans and organizations have begun producing clinical‐grade iPSCs, it is imperative that strict quality control standards are agreed upon. This is essential as both ESCs and iPSCs have been shown to accumulate genomic aberrations during long‐term culturing. These aberrations can include copy number variations, trisomy, amplifications of chromosomal regions, deletions of chromosomal regions, loss of heterozygosity, and epigenetic abnormalities. Moreover, although the differences between iPSCs and ESCs appear largely negligible when a high enough n number is used for comparison, the reprogramming process can generate further aberrations in iPSCs, including copy number variations and deletions in tumor‐suppressor genes. If mutations or epigenetic signatures are present in parental cells, these can also be carried over into iPSCs. To maximize patient safety, we recommend a set of standards to be utilized when preparing iPSCs for clinical use. Reprogramming methods that do not involve genomic integration should be used. Cultured cells should be grown using feeder‐free and serum‐free systems to avoid animal contamination. Karyotyping, whole‐genome sequencing, gene expression analyses, and standard sterility tests should all become routine quality control tests. Analysis of mitochondrial DNA integrity, whole‐epigenome analyses, as well as single‐cell genome sequencing of large cell populations may also prove beneficial. Furthermore, clinical‐grade stem cells need to be produced under accepted regulatory good manufacturing process standards. The creation of haplobanks that provide major histocompatibility complex matching is also recommended to improve allogeneic stem cell engraftment. Stem Cells Translational Medicine2018;7:867–875
Collapse
Affiliation(s)
- Leili Rohani
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany.,Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Adiv A Johnson
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Pooyan Naghsh
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Heidrun Holland
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| |
Collapse
|
97
|
Porokh V, Vaňhara P, Bárta T, Jurečková L, Bohačiaková D, Pospíšilová V, Mináriková D, Holubcová Z, Pelková V, Souček K, Hampl A. Soluble Cripto-1 Induces Accumulation of Supernumerary Centrosomes and Formation of Aberrant Mitoses in Human Embryonic Stem Cells. Stem Cells Dev 2018; 27:1077-1084. [PMID: 29882484 DOI: 10.1089/scd.2018.0017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Chromosomal instability evoked by abnormalities in centrosome numbers has been traditionally considered as a hallmark of aberrant, typically cancerous or senescent cells. We have reported previously that pristine human embryonic stem cells (hESC) suffer from high frequency of supernumerary centrosomes and hence may be prone to undergo abnormal mitotic divisions. We have also unraveled that this phenomenon of multicentrosomal mitoses vanishes with prolonged time in culture and with initiation of differentiation, and it is strongly affected by the culture substratum. In this study, we report for the first time that Cripto-1 protein (teratocarcinoma-derived growth factor 1, epidermal growth factor-Cripto/FRL-1/Cryptic) produced by hESC represents a factor capable of inducing formation of supernumerary centrosomes in cultured hESC. Elimination of Cripto-1 signaling on the other hand restores the normal number of centrosomes in hESC. Linking the secretory phenotype of hESC to the centrosomal metabolism may help to develop better strategies for propagation of stable and safe bioindustrial and clinical grade cultures of hESC. From a broader point of view, it may lead to unravelling Cripto-1 as a micro-environmental factor contributing to adverse cell behaviors in vivo.
Collapse
Affiliation(s)
- Volodymyr Porokh
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Petr Vaňhara
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
- 2 Center of Biomolecular and Cellular Engineering (CBCE), International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Tomáš Bárta
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Lucie Jurečková
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Dáša Bohačiaková
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Veronika Pospíšilová
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Daniela Mináriková
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Zuzana Holubcová
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Vendula Pelková
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
| | - Karel Souček
- 2 Center of Biomolecular and Cellular Engineering (CBCE), International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- 3 Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Aleš Hampl
- 1 Department of Histology and Embryology, Faculty of Medicine, Masaryk University , Brno, Czech Republic
- 2 Center of Biomolecular and Cellular Engineering (CBCE), International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
98
|
Zacchigna S, Giacca M. The global role of biotechnology for non communicable disorders. J Biotechnol 2018; 283:115-119. [PMID: 30077584 DOI: 10.1016/j.jbiotec.2018.07.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/06/2018] [Accepted: 07/31/2018] [Indexed: 01/07/2023]
Abstract
The World Health Organization (WHO) has tagged non-communicable diseases (NCDs) as one of the twenty-first century's major development challenges. NCDs account for over 15 million deaths annually and over 80% of those deaths occur in developing countries and among the poorest populations. Biotechnology presents unique opportunities to improve the early diagnosis and the treatment of NCDs. This review describes the major applications of biotechnology for a better clinical management of NCDs, i.e. the implementation of innovative diagnostic approaches and the production of innovative treatments, including those based on monoclonal antibodies, recombinant proteins, regulatory nucleic acids and cell-based therapies for regenerative medicine. In this context, it also examines the major challenges faced by biotechnology in developing countries.
Collapse
Affiliation(s)
- Serena Zacchigna
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
| | - Mauro Giacca
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| |
Collapse
|
99
|
Jiménez G, Hackenberg M, Catalina P, Boulaiz H, Griñán-Lisón C, García MÁ, Perán M, López-Ruiz E, Ramírez A, Morata-Tarifa C, Carrasco E, Aguilera M, Marchal JA. Mesenchymal stem cell's secretome promotes selective enrichment of cancer stem-like cells with specific cytogenetic profile. Cancer Lett 2018; 429:78-88. [DOI: 10.1016/j.canlet.2018.04.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022]
|
100
|
Bobba S, Di Girolamo N, Munsie M, Chen F, Pébay A, Harkin D, Hewitt AW, O'Connor M, McLenachan S, Shadforth AMA, Watson SL. The current state of stem cell therapy for ocular disease. Exp Eye Res 2018; 177:65-75. [PMID: 30029023 DOI: 10.1016/j.exer.2018.07.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/16/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022]
Abstract
Herein, we review the safety, efficacy, regulatory standards and ethical implications of the use of stem cells in ocular disease. A literature review was conducted, registered clinical trials reviewed, and expert opinions sought. Guidelines and codes of conduct from international societies and professional bodies were also reviewed. Collated data is presented on current progress in the field of ocular regenerative medicine, future challenges, the clinical trial process and ethical considerations in stem cell therapy. A greater understanding of the function and location of ocular stem cells has led to rapid advances in possible therapeutic applications. However, in the context of significant technical challenges and potential long-term complications, it is imperative that stem cell practices operate within formal clinical trial frameworks. While there remains broad scope for innovation, ongoing evidence-based review of potential interventions and the development of standardized protocols are necessary to ensure patient safety and best practice in ophthalmic care.
Collapse
Affiliation(s)
- Samantha Bobba
- Prince of Wales Hospital Clinical School, High Street, Randwick, Sydney, New South Wales, 2031, Australia.
| | - Nick Di Girolamo
- School of Medical Sciences, University of New South Wales, Kensington, Sydney, New South Wales, 2052, Australia
| | - Megan Munsie
- Centre for Stem Cell Systems, School of Biomedical Sciences, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Fred Chen
- Lions Eye Institute, 2 Verdun Street, Nedlands, Western Australia, 6009, Australia
| | - Alice Pébay
- Centre for Stem Cell Systems, School of Biomedical Sciences, University of Melbourne, Parkville, Victoria, 3010, Australia; Centre for Eye Research Australia, Level 7/32 Gisborne Street, East Melbourne, Victoria, 3002, Australia
| | - Damien Harkin
- School of Biomedical Sciences, Queensland University of Technology, 2 George Street, Brisbane, Queensland, 4000, Australia
| | - Alex W Hewitt
- Centre for Eye Research Australia, Level 7/32 Gisborne Street, East Melbourne, Victoria, 3002, Australia; School of Medicine, University of Tasmania, Churchill Avenue, Hobart, Tasmania, 7005, Australia
| | - Michael O'Connor
- School of Medicine, Western Sydney University, Victoria Road Parramatta, New South Wales, Parramatta, 2150, Australia
| | - Samuel McLenachan
- Centre for Ophthalmology and Visual Science, University of Western Australia, 35 Stirling Highway, Crawley, Western Australia, 6009, Australia
| | - Audra M A Shadforth
- School of Biomedical Sciences, Queensland University of Technology, 2 George Street, Brisbane, Queensland, 4000, Australia
| | - Stephanie L Watson
- Prince of Wales Hospital Clinical School, High Street, Randwick, Sydney, New South Wales, 2031, Australia; Save Sight Institute, University of Sydney, 8 Macquarie Street, Sydney, New South Wales, 2000, Australia; Sydney Eye Hospital, 8 Macquarie Street, Sydney, New South Wales, 2000, Australia.
| |
Collapse
|