51
|
Seranova E, Palhegyi AM, Verma S, Dimova S, Lasry R, Naama M, Sun C, Barrett T, Rosenstock TR, Kumar D, Cohen MA, Buganim Y, Sarkar S. Human Induced Pluripotent Stem Cell Models of Neurodegenerative Disorders for Studying the Biomedical Implications of Autophagy. J Mol Biol 2020; 432:2754-2798. [PMID: 32044344 DOI: 10.1016/j.jmb.2020.01.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022]
Abstract
Autophagy is an intracellular degradation process that is essential for cellular survival, tissue homeostasis, and human health. The housekeeping functions of autophagy in mediating the clearance of aggregation-prone proteins and damaged organelles are vital for post-mitotic neurons. Improper functioning of this process contributes to the pathology of myriad human diseases, including neurodegeneration. Impairment in autophagy has been reported in several neurodegenerative diseases where pharmacological induction of autophagy has therapeutic benefits in cellular and transgenic animal models. However, emerging studies suggest that the efficacy of autophagy inducers, as well as the nature of the autophagy defects, may be context-dependent, and therefore, studies in disease-relevant experimental systems may provide more insights for clinical translation to patients. With the advancements in human stem cell technology, it is now possible to establish disease-affected cellular platforms from patients for investigating disease mechanisms and identifying candidate drugs in the appropriate cell types, such as neurons that are otherwise not accessible. Towards this, patient-derived human induced pluripotent stem cells (hiPSCs) have demonstrated considerable promise in constituting a platform for effective disease modeling and drug discovery. Multiple studies have utilized hiPSC models of neurodegenerative diseases to study autophagy and evaluate the therapeutic efficacy of autophagy inducers in neuronal cells. This review provides an overview of the regulation of autophagy, generation of hiPSCs via cellular reprogramming, and neuronal differentiation. It outlines the findings in various neurodegenerative disorders where autophagy has been studied using hiPSC models.
Collapse
Affiliation(s)
- Elena Seranova
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Adina Maria Palhegyi
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Surbhi Verma
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Simona Dimova
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Rachel Lasry
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, 91120, Israel
| | - Moriyah Naama
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, 91120, Israel
| | - Congxin Sun
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Timothy Barrett
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Tatiana Rosado Rosenstock
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Sciences, São Paulo, SP, 01221-020, Brazil
| | - Dhiraj Kumar
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Malkiel A Cohen
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, 91120, Israel
| | - Sovan Sarkar
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom.
| |
Collapse
|
52
|
Chua BA, Van Der Werf I, Jamieson C, Signer RAJ. Post-Transcriptional Regulation of Homeostatic, Stressed, and Malignant Stem Cells. Cell Stem Cell 2020; 26:138-159. [PMID: 32032524 PMCID: PMC7158223 DOI: 10.1016/j.stem.2020.01.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cellular identity is not driven by differences in genomic content but rather by epigenomic, transcriptomic, and proteomic heterogeneity. Although regulation of the epigenome plays a key role in shaping stem cell hierarchies, differential expression of transcripts only partially explains protein abundance. The epitranscriptome, translational control, and protein degradation have emerged as fundamental regulators of proteome complexity that regulate stem cell identity and function. Here, we discuss how post-transcriptional mechanisms enable stem cell homeostasis and responsiveness to developmental cues and environmental stressors by rapidly shaping the content of their proteome and how these processes are disrupted in pre-malignant and malignant states.
Collapse
Affiliation(s)
- Bernadette A Chua
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA
| | - Inge Van Der Werf
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA; Sanford Stem Cell Clinical Center, La Jolla, CA 92037, USA
| | - Catriona Jamieson
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA; Sanford Stem Cell Clinical Center, La Jolla, CA 92037, USA.
| | - Robert A J Signer
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA.
| |
Collapse
|
53
|
Liu G, David BT, Trawczynski M, Fessler RG. Advances in Pluripotent Stem Cells: History, Mechanisms, Technologies, and Applications. Stem Cell Rev Rep 2020; 16:3-32. [PMID: 31760627 PMCID: PMC6987053 DOI: 10.1007/s12015-019-09935-x] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past 20 years, and particularly in the last decade, significant developmental milestones have driven basic, translational, and clinical advances in the field of stem cell and regenerative medicine. In this article, we provide a systemic overview of the major recent discoveries in this exciting and rapidly developing field. We begin by discussing experimental advances in the generation and differentiation of pluripotent stem cells (PSCs), next moving to the maintenance of stem cells in different culture types, and finishing with a discussion of three-dimensional (3D) cell technology and future stem cell applications. Specifically, we highlight the following crucial domains: 1) sources of pluripotent cells; 2) next-generation in vivo direct reprogramming technology; 3) cell types derived from PSCs and the influence of genetic memory; 4) induction of pluripotency with genomic modifications; 5) construction of vectors with reprogramming factor combinations; 6) enhancing pluripotency with small molecules and genetic signaling pathways; 7) induction of cell reprogramming by RNA signaling; 8) induction and enhancement of pluripotency with chemicals; 9) maintenance of pluripotency and genomic stability in induced pluripotent stem cells (iPSCs); 10) feeder-free and xenon-free culture environments; 11) biomaterial applications in stem cell biology; 12) three-dimensional (3D) cell technology; 13) 3D bioprinting; 14) downstream stem cell applications; and 15) current ethical issues in stem cell and regenerative medicine. This review, encompassing the fundamental concepts of regenerative medicine, is intended to provide a comprehensive portrait of important progress in stem cell research and development. Innovative technologies and real-world applications are emphasized for readers interested in the exciting, promising, and challenging field of stem cells and those seeking guidance in planning future research direction.
Collapse
Affiliation(s)
- Gele Liu
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA.
| | - Brian T David
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| | - Matthew Trawczynski
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| | - Richard G Fessler
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| |
Collapse
|
54
|
Hepburn AC, Sims CHC, Buskin A, Heer R. Engineering Prostate Cancer from Induced Pluripotent Stem Cells-New Opportunities to Develop Preclinical Tools in Prostate and Prostate Cancer Studies. Int J Mol Sci 2020; 21:E905. [PMID: 32019175 PMCID: PMC7036761 DOI: 10.3390/ijms21030905] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/17/2020] [Accepted: 01/28/2020] [Indexed: 12/17/2022] Open
Abstract
One of the key issues hampering the development of effective treatments for prostate cancer is the lack of suitable, tractable, and patient-specific in vitro models that accurately recapitulate this disease. In this review, we address the challenges of using primary cultures and patient-derived xenografts to study prostate cancer. We describe emerging approaches using primary prostate epithelial cells and prostate organoids and their genetic manipulation for disease modelling. Furthermore, the use of human prostate-derived induced pluripotent stem cells (iPSCs) is highlighted as a promising complimentary approach. Finally, we discuss the manipulation of iPSCs to generate 'avatars' for drug disease testing. Specifically, we describe how a conceptual advance through the creation of living biobanks of "genetically engineered cancers" that contain patient-specific driver mutations hold promise for personalised medicine.
Collapse
Affiliation(s)
- Anastasia C. Hepburn
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (C.H.C.S.); (A.B.)
| | - C. H. Cole Sims
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (C.H.C.S.); (A.B.)
| | - Adriana Buskin
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (C.H.C.S.); (A.B.)
| | - Rakesh Heer
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (C.H.C.S.); (A.B.)
- Department of Urology, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| |
Collapse
|
55
|
Lin SL, Chen JS, Ying SY. MiR-302-Mediated Somatic Cell Reprogramming and Method for Generating Tumor-Free iPS Cells Using miR-302. Methods Mol Biol 2020; 2115:199-219. [PMID: 32006403 DOI: 10.1007/978-1-0716-0290-4_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human induced pluripotent stem cells (iPSCs) by four factors have the risks of teratoma formation and potential tumorigenicity. To overcome this major hurdle, we examined the mechanism(s) by which the cell cycle genes of embryonic cells were regulated. Naturally occurring embryonic stem cells (ESCs) possess two unique stemness properties: pluripotent differentiation into all cell types and self-renewal with no risk of tumor formation. Despite overwhelming reports describing iPSC pluripotency, there have been no observations of tumor prevention mechanism that suppresses tumor formation similar to that in naturally occurring ESCs. The ESC-specific microRNA (miRNA), miR-302, regulates human iPSC tumorigenicity through co-suppression of both cyclin E-CDK2 and cyclin D-CDK4/6 cell cycle pathways during G1-S phase transition. MiR-302 also silenced BMI-1, a cancer stem cell marker gene, to promote the expression of two senescence-associated tumor suppressor genes, p16Ink4a and p14/p19Arf. Together, the combinatory effect of reducing G1-S cell cycle transition and increasing p16/p14(p19) expression resulted in a relatively attenuated cell cycle rate similar to that of 2-to-8-cell-stage embryonic cells in early mammalian zygotes (20-24 h/cycle), as compared to the fast proliferation rate of iPSCs induced by four defined factors Oct4-Sox2-Klf4-c-Myc (12-16 h/cycle). In addition to the prevention of stem cell tumorigenicity, the mechanism underlying miR-302-mediated iPSCs also includes the initiation of global genomic DNA methylation, activation of ESC-specific gene expression, and inhibition of developmental signaling. Overall, we have established an effective protocol to express the intronic miR-302 cluster, according to its own natural biogenesis mechanism to generate tumor-free iPSCs for use in biology and therapy.
Collapse
Affiliation(s)
- Shi-Lung Lin
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| | - Jack S Chen
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| | - Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
56
|
Sastre D, Baiochi J, de Souza Lima IM, Canto de Souza F, Corveloni AC, Thomé CH, Faça VM, Schiavinato JLDS, Covas DT, Panepucci RA. Focused screening reveals functional effects of microRNAs differentially expressed in colorectal cancer. BMC Cancer 2019; 19:1239. [PMID: 31864341 PMCID: PMC6925883 DOI: 10.1186/s12885-019-6468-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is still a leading cause of death worldwide. Recent studies have pointed to an important role of microRNAs in carcinogenesis. Several microRNAs are described as aberrantly expressed in CRC tissues and in the serum of patients. However, functional outcomes of microRNA aberrant expression still need to be explored at the cellular level. Here, we aimed to investigate the effects of microRNAs aberrantly expressed in CRC samples in the proliferation and cell death of a CRC cell line. METHODS We transfected 31 microRNA mimics into HCT116 cells. Total number of live propidium iodide negative (PI-) and dead (PI+) cells were measured 4 days post-transfection by using a high content screening (HCS) approach. HCS was further used to evaluate apoptosis (via Annexin V and PI staining), and to discern between intrinsic and extrinsic apoptotic pathways, by detecting cleaved Caspase 9 and 8, respectively. To reveal mRNA targets and potentially involved mechanisms, we performed microarray gene expression and functional pathway enrichment analysis. Quantitative PCR and western blot were used to validate potential mRNA targets. RESULTS Twenty microRNAs altered the proliferation of HCT116 cells in comparison to control. miR-22-3p, miR-24-3p, and miR-101-3p significantly repressed cell proliferation and induced cell death. Interestingly, all anti-proliferative microRNAs in our study had been previously described as poorly expressed in the CRC samples. Predicted miR-101-3p targets that were also downregulated by in our microarray were enriched for genes associated with Wnt and cancer pathways, including MCL-1, a member of the BCL-2 family, involved in apoptosis. Interestingly, miR-101-3p preferentially downregulated the long anti-apoptotic MCL-1 L isoform, and reduced cell survival specifically by activating the intrinsic apoptosis pathway. Moreover, miR-101-3p also downregulated IL6ST, STAT3A/B, and MYC mRNA levels, genes associated with stemness properties of CRC cells. CONCLUSIONS microRNAs upregulated in CRC tend to induce proliferation in vitro, whereas microRNAs poorly expressed in CRC halt proliferation and induce cell death. We provide novel evidence linking preferential inhibition of the anti-apoptotic MCL-1 L isoform by miR-101-3p and consequent activation of the intrinsic apoptotic pathway as potential mechanisms for its antitumoral activity, likely due to the inhibition of the IL-6/JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Danuta Sastre
- Laboratory of Human and Medical Genetics, Federal University of Pará, Rua Augusto Corrêa, 01. Guamá., Belém, Pará CEP 66075-110 Brazil
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - João Baiochi
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Ildercilio Mota de Souza Lima
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Felipe Canto de Souza
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Amanda Cristina Corveloni
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Carolina Hassib Thomé
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Av. Bandeirantes, 3900 - Vila Monte Alegre, Ribeirão Preto, SP 14049-900 Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Av. Bandeirantes, 3900 - Vila Monte Alegre, Ribeirão Preto, SP 14049-900 Brazil
| | - Josiane Lilian dos Santos Schiavinato
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Dimas Tadeu Covas
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Rodrigo Alexandre Panepucci
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| |
Collapse
|
57
|
Bang JS, Choi NY, Lee M, Ko K, Park YS, Ko K. Reprogramming of Cancer Cells into Induced Pluripotent Stem Cells Questioned. Int J Stem Cells 2019; 12:430-439. [PMID: 31474029 PMCID: PMC6881048 DOI: 10.15283/ijsc19067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/26/2019] [Accepted: 07/27/2019] [Indexed: 12/20/2022] Open
Abstract
Background and Objectives Several recent studies have claimed that cancer cells can be reprogrammed into induced pluripotent stem cells (iPSCs). However, in most cases, cancer cells seem to be resistant to cellular reprogramming. Furthermore, the underlying mechanisms of limited reprogramming in cancer cells are largely unknown. Here, we identified the candidate barrier genes and their target genes at the early stage of reprogramming for investigating cancer reprogramming. Methods We tried induction of pluripotency in normal human fibroblasts (BJ) and both human benign (MCF10A) and malignant (MCF7) breast cancer cell lines using a classical retroviral reprogramming method. We conducted RNA-sequencing analysis to compare the transcriptome of the three cell lines at early stage of reprogramming. Results We could generate iPSCs from BJ, whereas we were unable to obtain iPSCs from cancer cell lines. To address the underlying mechanism of limited reprogramming in cancer cells, we identified 29 the candidate barrier genes based on RNA-sequencing data. In addition, we found 40 their target genes using Cytoscape software. Conclusions Our data suggest that these genes might one of the roadblock for cancer cell reprogramming. Furthermore, we provide new insights into application of iPSCs technology in cancer cell field for therapeutic purposes.
Collapse
Affiliation(s)
- Jin Seok Bang
- Department of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Na Young Choi
- Department of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Minseong Lee
- Department of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Kisung Ko
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Yo Seph Park
- Department of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Kinarm Ko
- Department of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Korea.,Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea.,Research Institute of Medical Science, Konkuk University, Seoul, Korea
| |
Collapse
|
58
|
Peskova L, Cerna K, Oppelt J, Mraz M, Barta T. Oct4-mediated reprogramming induces embryonic-like microRNA expression signatures in human fibroblasts. Sci Rep 2019; 9:15759. [PMID: 31673026 PMCID: PMC6823439 DOI: 10.1038/s41598-019-52294-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/16/2019] [Indexed: 12/22/2022] Open
Abstract
Oct4-mediated reprogramming has recently become a novel tool for the generation of various cell types from differentiated somatic cells. Although molecular mechanisms underlying this process are unknown, it is well documented that cells over-expressing Oct4 undergo transition from differentiated state into plastic state. This transition is associated with the acquisition of stem cells properties leading to epigenetically “open” state that is permissive to cell fate switch upon external stimuli. In order to contribute to our understanding of molecular mechanisms driving this process, we characterised human fibroblasts over-expressing Oct4 and performed comprehensive small-RNAseq analysis. Our analyses revealed new interesting aspects of Oct4-mediated cell plasticity induction. Cells over-expressing Oct4 lose their cell identity demonstrated by down-regulation of fibroblast-specific genes and up-regulation of epithelial genes. Interestingly, this process is associated with microRNA expression profile that is similar to microRNA profiles typically found in pluripotent stem cells. We also provide extensive network of microRNA families and clusters allowing us to precisely determine the miRNAome associated with the acquisition of Oct4-induced transient plastic state. Our data expands current knowledge of microRNA and their implications in cell fate alterations and contributing to understanding molecular mechanisms underlying it.
Collapse
Affiliation(s)
- Lucie Peskova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, 625 00, Czech Republic
| | - Katerina Cerna
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic
| | - Jan Oppelt
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
| | - Marek Mraz
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Barta
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, 625 00, Czech Republic.
| |
Collapse
|
59
|
Abstract
Objectives: Firefighters have elevated cancer incidence and mortality rates. MicroRNAs play prominent roles in carcinogenesis, but have not been previously evaluated in firefighters. Methods: Blood from 52 incumbent and 45 new recruit nonsmoking firefighters was analyzed for microRNA expression, and the results adjusted for age, obesity, ethnicity, and multiple comparisons. Results: Nine microRNAs were identified with at least a 1.5-fold significant difference between groups. All six microRNAs with decreased expression in incumbent firefighters have been reported to have tumor suppressor activity or are associated with cancer survival, and two of the three microRNAs with increased expression in incumbent firefighters have activities consistent with cancer promotion, with the remaining microRNA associated with neurological disease. Conclusion: Incumbent firefighters showed differential microRNA expression compared with new recruits, providing potential mechanisms for increased cancer risk in firefighters.
Collapse
|
60
|
Dorfeshan P, Ghaffari Novin M, Salehi M, Farifteh F. Expression of miR-302 in human embryo derived from in-vitro matured oocyte. Int J Reprod Biomed 2019; 17:405-412. [PMID: 31508565 PMCID: PMC6719516 DOI: 10.18502/ijrm.v17i6.4812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 12/12/2017] [Accepted: 11/14/2018] [Indexed: 12/22/2022] Open
Abstract
Background The expression of miR-302 over the period of early embryogenesis could possibly regulate the maternal transcript clearance. Zygotic transcription activation is mostly related to maternal messages degradation. Objective In this study, the effects of in-vitro maturation technique (IVM) on the expression of miR-302 in human embryo produced from immature and mature human oocytes (matured in vitro and in vivo, before sperm exposure) obtained from females under gonadotrophin therapy were evaluated for assisted reproduction. Materials and Methods Immature oocytes were cultured in vitro. The injection of oocytes-producing polar bodies was given using fresh sperm. Then, the embryo quality score was assessed in the IVM group compared with the control group. In both the groups, embryos with normal morphology were included in the molecular study. Only one blastomere was removed from three-day embryos and then the embryos were frozen. The expression of miR-302 in embryos was measured through quantitative real-time polymerase chain reaction. Results Our data showed a significant reduction of miR-302 expression in the IVM group vs. the control group (p = 0.02). The embryo quality score showed a significant difference between the two groups (p = 0.01). Conclusion The present study demonstrated that the IVM process had a negative effect on the expression level of miR-302 in human pre-implantation embryos. Considering the major role of expression miR-302, a reduced potential in miR-302 expression could be related to a decrease in the early embryonic development.
Collapse
Affiliation(s)
- Parvin Dorfeshan
- Department of Social Medicine, Faculty of Medicine, Jundishapure University of Medical Sciences, Ahvaz, Iran.,Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marefat Ghaffari Novin
- Infertility and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Salehi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatane Farifteh
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
61
|
Gong L, Yan Q, Zhang Y, Fang X, Liu B, Guan X. Cancer cell reprogramming: a promising therapy converting malignancy to benignity. Cancer Commun (Lond) 2019; 39:48. [PMID: 31464654 PMCID: PMC6716904 DOI: 10.1186/s40880-019-0393-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
In the past decade, remarkable progress has been made in reprogramming terminally differentiated somatic cells and cancer cells into induced pluripotent cells and cancer cells with benign phenotypes. Recent studies have explored various approaches to induce reprogramming from one cell type to another, including lineage-specific transcription factors-, combinatorial small molecules-, microRNAs- and embryonic microenvironment-derived exosome-mediated reprogramming. These reprogramming approaches have been proven to be technically feasible and versatile to enable re-activation of sequestered epigenetic regions, thus driving fate decisions of differentiated cells. One of the significant utilities of cancer cell reprogramming is the therapeutic potential of retrieving normal cell functions from various malignancies. However, there are several major obstacles to overcome in cancer cell reprogramming before clinical translation, including characterization of reprogramming mechanisms, improvement of reprogramming efficiency and safety, and development of delivery methods. Recently, several insights in reprogramming mechanism have been proposed, and determining progress has been achieved to promote reprogramming efficiency and feasibility, allowing it to emerge as a promising therapy against cancer in the near future. This review aims to discuss recent applications in cancer cell reprogramming, with a focus on the clinical significance and limitations of different reprogramming approaches, while summarizing vital roles played by transcription factors, small molecules, microRNAs and exosomes during the reprogramming process.
Collapse
Affiliation(s)
- Lanqi Gong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Qian Yan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Yu Zhang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Xiaona Fang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Xinyuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China. .,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China.
| |
Collapse
|
62
|
Ahmadalizadeh Khanehsar M, Hoseinbeyki M, Fakhr Taha M, Javeri A. Repression of TGF-β Signaling in Breast Cancer Cells by miR-302/367 Cluster. CELL JOURNAL 2019; 21:444-450. [PMID: 31376326 PMCID: PMC6722449 DOI: 10.22074/cellj.2020.6193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022]
Abstract
Objective Epigenetic alterations of the malignantly transformed cells have increasingly been regarded as an important
event in the carcinogenic development. Induction of some miRNAs such as miR-302/367 cluster has been shown
to induce reprogramming of breast cancer cells and exert a tumor suppressive role by induction of mesenchymal to
epithelial transition, apoptosis and a lower proliferation rate. Here, we aimed to investigate the impact of miR-302/367
overexpression on transforming growth factor-beta (TGF-β) signaling and how this may contribute to tumor suppressive
effects of miR-302/367 cluster.
Materials and Methods In this experimental study, MDA-MB-231 and SK-BR-3 breast cancer cells were cultured and
transfected with miR-302/367 expressing lentivector. The impact of miR-302/367 overexpression on several mediators
of TGF-β signaling and cell cycle was assessed by quantitative real-time polymerase chain reaction (qPCR) and flow
cytometry.
Results Ectopic expression of miR-302/367 cluster downregulated expression of some downstream elements of
TGF-β pathway in MDA-MB-231 and SK-BR-3 breast cancer cell lines. Overexpression of miR-302/367 cluster inhibited
proliferation of the breast cancer cells by suppressing the S-phase of cell cycle which was in accordance with inhibition
of TGF-β pathway.
Conclusion TGF-β signaling is one of the key pathways in tumor progression and a general suppression of TGF-β
mediators by the pleiotropically acting miR-302/367 cluster may be one of the important reasons for its anti-tumor
effects in breast cancer cells.
Collapse
Affiliation(s)
- Mona Ahmadalizadeh Khanehsar
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.,Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Moslem Hoseinbeyki
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Masoumeh Fakhr Taha
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Arash Javeri
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.Electronic Address:
| |
Collapse
|
63
|
Das MK, Evensen HSF, Furu K, Haugen TB. miRNA-302s may act as oncogenes in human testicular germ cell tumours. Sci Rep 2019; 9:9189. [PMID: 31235829 PMCID: PMC6591358 DOI: 10.1038/s41598-019-45573-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Testicular germ cell tumour (TGCT) represents the most common malignancy in young men in large parts of the world, but the aetiology is yet unclear. Multiple TGCT susceptibility loci have been identified, and we have shown that one of these, SPRY4, may act as a TGCT oncogene. Furthermore, many of the loci are in non-coding regions of the genome. miRNAs, a class of non-coding RNAs may play a crucial role in cell proliferation, differentiation, and apoptosis, and alteration in their expression may lead to oncogenesis. Differential expression of miRNAs in TGCT and normal testis has been reported in previous studies. In this study, we used qPCR to analyse, in normal and malignant testis tissue, the expression of the ten miRNAs that we had previously identified by sequencing to be the most upregulated in TGCT. We found high expression of these miRNAs also by qPCR analysis. The levels of miR-302a-3p, miR-302b-3p, and miR-302c-3p were downregulated after treatment of the TGCT cell lines NT2-D1 and 833 K with the chemotherapy drug cisplatin. By using miRNA inhibitor-mediated transient transfection, we inhibited the expression of the three members of miR-302 family (miR-302s). Inhibition of miR-302s resulted in a decreased cell proliferation in NT2-D1 cells, but not in 833 K cells. In both cell lines, inhibition of miR-302s resulted in decreased expression of SPRY4, which we have previously shown to regulate MAPK/ERK and PI3K/Akt signalling pathways in these cells. Inhibition of miR-302b-3p and miR-302c-3p decreased phosphorylation of ERK1/2, whereas inhibition of miR-302a-3p and miR-302b-3p led to decreased expression of the apoptosis inhibitor, survivin. Our findings suggest that miR-302s act as TGCT oncogenes by inducing the expression of SPRY4 and activating MAPK/ERK pathway while inhibiting apoptosis via increased survivin expression.
Collapse
Affiliation(s)
- Mrinal K Das
- Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway.
| | - Herman S F Evensen
- Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Kari Furu
- Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway.,Cancer Registry, Oslo, Norway
| | - Trine B Haugen
- Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
64
|
Bourguignon LYW. Matrix Hyaluronan-CD44 Interaction Activates MicroRNA and LncRNA Signaling Associated With Chemoresistance, Invasion, and Tumor Progression. Front Oncol 2019; 9:492. [PMID: 31293964 PMCID: PMC6598393 DOI: 10.3389/fonc.2019.00492] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor malignancies involve cancer cell growth, issue invasion, metastasis and often drug resistance. A great deal of effort has been placed on searching for unique molecule(s) overexpressed in cancer cells that correlate(s) with tumor cell-specific behaviors. Hyaluronan (HA), one of the major ECM (extracellular matrix) components have been identified as a physiological ligand for surface CD44 isoforms which are frequently overexpressed in malignant tumor cells during cancer progression. The binding interaction between HA and CD44 isoforms often stimulates aberrant cellular signaling processes and appears to be responsible for the induction of multiple oncogenic events required for cancer-specific phenotypes and behaviors. In recent years, both microRNAs (miRNAs) (with ~20–25 nucleotides) and long non-coding RNAs (lncRNAs) (with ~200 nucleotides) have been found to be abnormally expressed in cancer cells and actively participate in numerous oncogenic signaling events needed for tumor cell-specific functions. In this review, I plan to place a special emphasis on HA/CD44-induced signaling pathways and the presence of several novel miRNAs (e.g., miR-10b/miR-302/miR-21) and lncRNAs (e.g., UCA1) together with their target functions (e.g., tumor cell migration, invasion, and chemoresistance) during cancer development and progression. I believe that important information can be obtained from these studies on HA/CD44-activated miRNAs and lncRNA that may be very valuable for the future development of innovative therapeutic drugs for the treatment of matrix HA/CD44-mediated cancers.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- Endocrine Unit (111N2), Department of Medicine, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
65
|
Chen B, Zhu Z, Li L, Ye W, Zeng J, Gao J, Wang S, Zhang L, Huang Z. Effect of overexpression of Oct4 and Sox2 genes on the biological and oncological characteristics of gastric cancer cells. Onco Targets Ther 2019; 12:4667-4682. [PMID: 31417271 PMCID: PMC6592062 DOI: 10.2147/ott.s209734] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 05/07/2019] [Indexed: 11/23/2022] Open
Abstract
Objective: Using the gastric cancer cell line SGC7901, we constructed a cell line that overexpressed octamer-binding protein 4 (Oct4) and SRY-box 2 (Sox2) to explore the stem cell oncological and biological characteristics of these cells and to elucidate the mechanisms of Oct4 and Sox2 in cancer. Methods: A lentiviral vector containing the Sox2 gene was constructed and transfected into a gastric cancer cell line overexpressing Oct4 (SGC7901-Oct4) to obtain a stably transfected cell line (SGC7901-Oct4-Sox2). Oct4 and Sox2 expression was detected by RT-PCR and Western blotting. The proliferation, drug resistance, migration, and invasion abilities of the cells were assessed using in vitro (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium), drug resistance, scratch-wound migration, transwell migration, transwell invasion, and spherical clone formation assays, and their tumorigenic ability was assessed using a tumor formation experiment in mice. Results: Compared with the control group, the expression of Oct4, Sox2, CD44, and E-cadherin was significantly higher in the group that overexpressed Oct4 and Sox2, while the expression of c-Myc and Klf4 did not significantly change. The proliferation, drug resistance, migration, and invasion abilities were significantly enhanced in the overexpression group, and the tumorigenic ability in mice was also significantly enhanced, with significantly increased tumor size and weight. Conclusion: The proliferation, drug resistance, migration, invasion, and tumorigenic abilities of SGC7901 cells overexpressing Oct4 and Sox2 were significantly improved. Oct4 and Sox2 play important roles in the proliferation, migration, invasion, and tumorigenicity of gastric cancer cells, and the two genes may be synergistic to a certain degree.
Collapse
Affiliation(s)
- Borong Chen
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China
| | - Zhipeng Zhu
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China
| | - Lulu Li
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China
| | - Weipeng Ye
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian 350004, People's Republic of China
| | - Junjie Zeng
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China
| | - Jin Gao
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China
| | - Shengjie Wang
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China
| | - Liang Zhang
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China
| | - Zhengjie Huang
- Department of Gastrointestinal Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, People's Republic of China.,Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian 350004, People's Republic of China
| |
Collapse
|
66
|
Groenendyk J, Fan X, Peng Z, Kurgan L, Michalak M. Endoplasmic reticulum and the microRNA environment in the cardiovascular system 1. Can J Physiol Pharmacol 2019; 97:515-527. [PMID: 31063413 DOI: 10.1139/cjpp-2018-0720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stress responses are important to human physiology and pathology, and the inability to adapt to cellular stress leads to cell death. To mitigate cellular stress and re-establish homeostasis, cells, including those in the cardiovascular system, activate stress coping response mechanisms. The endoplasmic reticulum, a component of the cellular reticular network in cardiac cells, mobilizes so-called endoplasmic reticulum stress coping responses, such as the unfolded protein response. MicroRNAs play an important part in the maintenance of cellular and tissue homeostasis, perform a central role in the biology of the cardiac myocyte, and are involved in pathological cardiac function and remodeling. In this paper, we review a link between endoplasmic reticulum homeostasis and microRNA with an emphasis on the impact on stress responses in the cardiovascular system.
Collapse
Affiliation(s)
- Jody Groenendyk
- a Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S7, Canada
| | - Xiao Fan
- b Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Zhenling Peng
- c Center for Applied Mathematics, Tianjin University, Tianjin 300072, China
| | - Lukasz Kurgan
- d Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA.,e Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2V4, Canada
| | - Marek Michalak
- a Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S7, Canada
| |
Collapse
|
67
|
Wu Y, Hu L, Qin Z, Wang X. MicroRNA‑302a upregulation mediates chemo‑resistance in prostate cancer cells. Mol Med Rep 2019; 19:4433-4440. [PMID: 30942454 DOI: 10.3892/mmr.2019.10098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/28/2018] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are post‑transcriptional regulators that mediate the initiation and progression of human cancer. Growing evidence suggests that deregulation of miRNA expression levels underlies chemo‑resistance. To investigate whether miRNA‑302a (miR‑302a) is involved in mediating chemo‑resistance to paclitaxel in prostate cancer, a series of in vitro analyses were performed in paclitaxel‑resistant prostate cancer PC‑3PR cells and non‑resistant prostate cancer PC‑3 cells. It was demonstrated that the expression of miR‑302a was upregulated in PC‑3PR cells. Notably, ectopic expression of miR‑302a also increased resistance to paclitaxel in wild‑type PC‑3 cells. By contrast, silencing of miR‑302a in PC‑3PR cells sensitized the cells to paclitaxel. Gene and protein expression analyses suggested that the miR‑302a target gene breast cancer resistance protein (BCRP) may mediate chemo‑resistance to paclitaxel in PC‑3PR cells. In conclusion, the data suggested that elevated miR‑302a levels, in part, mediate sensitivity to paclitaxel in prostate cancer through the aberrant regulation of its downstream targets, AOF2, BCRP and permeability glycoprotein 1. These data have implications for the development of novel therapeutics in prostate cancer that may improve sensitivity to chemotherapeutics.
Collapse
Affiliation(s)
- Yuqi Wu
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Li Hu
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Zizhen Qin
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Xiangwei Wang
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
68
|
Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Inhibit Endometrial Cancer Cell Proliferation and Migration through Delivery of Exogenous miR-302a. Stem Cells Int 2019; 2019:8108576. [PMID: 31001342 PMCID: PMC6437733 DOI: 10.1155/2019/8108576] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/30/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023] Open
Abstract
MicroRNAs (miRNAs) are potential therapeutic targets in endometrial cancer, but the difficulties associated with their delivery to tumor target cells have hampered their applications. Human umbilical cord mesenchymal stem cells (hUCMSCs) have a well-recognized tumor-homing ability, emphasizing the capacity of tumor-targeted delivery of extracellular vesicles. hUCMSCs release extracellular vesicles rich in miRNAs, which play a vital role in intercellular communication. The purpose of this study was to verify a potential tumor suppressor microRNA, miR-302a, and engineered hUCMSC extracellular vesicles enriched with miR-302a for therapy of endometrial cancer. Here, we observed that miR-302a was significantly downregulated in endometrial cancer tissues when compared with adjacent tissues. Overexpression of miR-302a in endometrial cancer cells robustly suppressed cell proliferation and migration. Meanwhile, the proliferation and migration were significantly inhibited in endometrial cancer cells when cultured with miR-302a-loaded extracellular vesicles derived from hUCMSCs. Importantly, our data showed that engineered extracellular vesicles rich in miR-302 significantly inhibited the expression of cyclin D1 and suppressed AKT signaling pathway in endometrial cancer cells. These results suggested that exogenous miR-302a delivered by hUCMSC-derived extracellular vesicles has exciting potential as an effective anticancer therapy.
Collapse
|
69
|
Wang M, Lv G, Jiang C, Xie S, Wang G. miR-302a inhibits human HepG2 and SMMC-7721 cells proliferation and promotes apoptosis by targeting MAP3K2 and PBX3. Sci Rep 2019; 9:2032. [PMID: 30765768 PMCID: PMC6375964 DOI: 10.1038/s41598-018-38435-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/28/2018] [Indexed: 01/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common liver cancer and has a poor prognosis. miR-302a is an important regulator of tumor occurrence and deterioration, while MAP3K2 and PBX3 genes are involved in cancer cell proliferation and apoptosis. In this study, the expression of miR-302a and MAP3K2/PBX3 were evaluated by qPCR in liver cancer cell lines. Next, the target relationship between miR-302a and MAP3K2/PBX3 was verified using luciferase assays. Meanwhile, the expression correlation between miR-302a and target genes was analyzed in cancer tissue and para-cancerous tissue. In addition, an increased miR-302a level in HepG2 cells and SMMC-7721 cells were achieved through transfection with miR-302a mimics, and the effects on HepG2 cell and SMMC-7721 cell proliferation, apoptosis and MAPK pathways were determined using MTT, flow cytometry, qPCR and western blot assays. The results showed that liver cancer cell lines exhibited low miR-302a expression and MAP3K2 and PBX3 were confirmed to be the target genes of miR-302a. Meanwhile, the HE results showed that cells became enlarged with loose cytoplasm and formed balloon-like lesions in HCC specimens and we found a significant negative correlation between miR-302a and MAP3K2/PBX3 expression. In addition, treatment with miR-302a mimics inhibited HepG2 cells and SMMC-7721 cells proliferation and increased the apoptosis rate. Further research revealed that the MAPK key factors p-p38, p-ERK1/2 and p-JNK were significantly reduced in miR-302a transfected cells and MAP3K2/PBX3 silenced cells. Besides, MAP3K2 and PBX3 overexpression in miR-302a mimics-treated cells exerted the opposite effects. In conclusion, miR-302a inhibited proliferation and promoted apoptosis in human hepatoma cells by targeting MAP3K2 and PBX3.
Collapse
Affiliation(s)
- Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Chao Jiang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Shuli Xie
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Guangyi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
70
|
Chen EYY, Chen JS, Ying SY. The microRNA and the perspectives of miR-302. Heliyon 2019; 5:e01167. [PMID: 30723835 PMCID: PMC6351428 DOI: 10.1016/j.heliyon.2019.e01167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/18/2018] [Accepted: 01/22/2019] [Indexed: 12/24/2022] Open
Abstract
MiRNAs are naturally occurring, small, non-coding RNA molecules that post-transcriptionally regulate the expression of a large number of genes involved in various biological processes, either through mRNA degradation or through translation inhibition. MiRNAs play important roles in many aspects of physiology and pathology throughout the body, particularly in cancer, which have made miRNAs attractive tools and targets for translational research. The types of non-coding RNAs, biogenesis of miRNAs, circulating miRNAs, and direct delivery of miRNA were briefly reviewed. As a case of point, the role and perspective of miR-302, a family of ES-specific miRNA, on cancer, iPSCs, heart disease were presented.
Collapse
Affiliation(s)
- Emily Yen Yu Chen
- Department of Integrative Anatomical Sciences, Keck School of Medicine, BMT-403, University of Southern California, 1333 San Pablo Street, Los Angeles, CA 90033, USA
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA 90670, USA
| | - Jack S. Chen
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA 90670, USA
| | - Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, BMT-403, University of Southern California, 1333 San Pablo Street, Los Angeles, CA 90033, USA
| |
Collapse
|
71
|
MicroRNA-17, MicroRNA-19b, MicroRNA-146a, MicroRNA-302d Expressions in Hepatoblastoma and Clinical Importance. J Pediatr Hematol Oncol 2019; 41:7-12. [PMID: 29889802 DOI: 10.1097/mph.0000000000001234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Hepatoblastoma (HB) is the most common liver malignancy in children. The prognosis changes according to the histologic subtypes of HB. In the present study, we aimed to characterize the expression level of selected microRNAs (miRNAs) in HB as well as in histologic subtypes, and to consider the association with the prognosis. A total of 22 HB tumor samples, subtyped as fetal (n=16) and embryonal (n=6), and 10 nontumorous surrounding liver samples were evaluated in this study. Expressions of miR-17, miR-146a, miR-302d, and miR-19b were analyzed in 22 HB tumor samples and 10 nontumorous surrounding liver samples by quantitative real-time polymerase chain reaction. Lower miRNA-17 expression levels were obtained in tumor samples in comparison with nontumorous surrounding liver samples (P=0.028). Lower miRNA-17 expression was significant for predicting prognosis in HB patients (area under receiver-operator characteristic curve=0.875, P=0.044). A higher-level of miR-19b was found in embryonal samples (P=0.008). Overall and event-free survival was not found to correlate with miRNA expression levels (P>0.05). This research finds miRNA-17 and miRNA-19b expression levels can provide important data on diagnosis and prognosis in HB showing different clinical behaviors.
Collapse
|
72
|
Genadry KC, Pietrobono S, Rota R, Linardic CM. Soft Tissue Sarcoma Cancer Stem Cells: An Overview. Front Oncol 2018; 8:475. [PMID: 30416982 PMCID: PMC6212576 DOI: 10.3389/fonc.2018.00475] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022] Open
Abstract
Soft tissue sarcomas (STSs) are an uncommon group of solid tumors that can arise throughout the human lifespan. Despite their commonality as non-bony cancers that develop from mesenchymal cell precursors, they are heterogeneous in their genetic profiles, histology, and clinical features. This has made it difficult to identify a single target or therapy specific to STSs. And while there is no one cell of origin ascribed to all STSs, the cancer stem cell (CSC) principle—that a subpopulation of tumor cells possesses stem cell-like properties underlying tumor initiation, therapeutic resistance, disease recurrence, and metastasis—predicts that ultimately it should be possible to identify a feature common to all STSs that could function as a therapeutic Achilles' heel. Here we review the published evidence for CSCs in each of the most common STSs, then focus on the methods used to study CSCs, the developmental signaling pathways usurped by CSCs, and the epigenetic alterations critical for CSC identity that may be useful for further study of STS biology. We conclude with discussion of some challenges to the field and future directions.
Collapse
Affiliation(s)
- Katia C Genadry
- Division of Hematology-Oncology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Silvia Pietrobono
- Department of Hematology-Oncology, Bambino Gesù Pediatric Hospital, IRCCS, Rome, Italy
| | - Rossella Rota
- Department of Hematology-Oncology, Bambino Gesù Pediatric Hospital, IRCCS, Rome, Italy
| | - Corinne M Linardic
- Division of Hematology-Oncology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States.,Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
73
|
Kim H, Schaniel C. Modeling Hematological Diseases and Cancer With Patient-Specific Induced Pluripotent Stem Cells. Front Immunol 2018; 9:2243. [PMID: 30323816 PMCID: PMC6172418 DOI: 10.3389/fimmu.2018.02243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022] Open
Abstract
The advent of induced pluripotent stem cells (iPSCs) together with recent advances in genome editing, microphysiological systems, tissue engineering and xenograft models present new opportunities for the investigation of hematological diseases and cancer in a patient-specific context. Here we review the progress in the field and discuss the advantages, limitations, and challenges of iPSC-based malignancy modeling. We will also discuss the use of iPSCs and its derivatives as cellular sources for drug target identification, drug development and evaluation of pharmacological responses.
Collapse
Affiliation(s)
- Huensuk Kim
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christoph Schaniel
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
74
|
Identification of protein kinase inhibitors to reprogram breast cancer cells. Cell Death Dis 2018; 9:915. [PMID: 30206213 PMCID: PMC6133942 DOI: 10.1038/s41419-018-1002-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/09/2018] [Accepted: 05/11/2018] [Indexed: 12/22/2022]
Abstract
Direct reversion of cancers into normal-like tissues is an ideal strategy for cancer treatment. Recent reports have showed that defined transcription factors can induce reprogramming of cancer cells into pluripotent stem cells, supporting this notion. Here, we have developed a reprogramming method that uses a conceptually unique strategy for breast cancer cell treatment. We have screened a kinase inhibitor library and found that Rho-associated protein kinase (ROCK) and mammalian target of rapamycin (mTOR) kinase inhibitors can substitute for all transcription factors to be sufficient to reprogram breast cancer cells into progenitor cells. Furthermore, ROCK–mTOR inhibitors could reprogram breast cancer cells to another terminal lineage-adipogenic cells. Genome-wide transcriptional analysis shows that the induced fat-like cells have a profile different from breast cancer cells and similar to that of normal adipocytes. In vitro and in vivo tumorigenesis assays have shown that induced fat-like cells lose proliferation and tumorigenicity. Moreover, reprogramming treatment with ROCK–mTOR inhibitors prevents breast cancer local recurrence in mice. Currently, ROCK–mTOR inhibitors are already used as antitumor drugs in patients, thus, this reprogramming strategy has significant potential to move rapidly toward clinical trials for breast cancer treatment.
Collapse
|
75
|
Ramezankhani B, Taha MF, Javeri A. Vitamin C counteracts miR-302/367-induced reprogramming of human breast cancer cells and restores their invasive and proliferative capacity. J Cell Physiol 2018; 234:2672-2682. [PMID: 30191953 DOI: 10.1002/jcp.27081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 06/29/2018] [Indexed: 12/21/2022]
Abstract
Epigenetic reprogramming by embryonic stem cell-specific miR-302/367 cluster has shown some tumor suppressive effects in cancer cells of different tissues such as skin, colon, and cervix. Vitamin C has been known as a reprogramming enhancer of human and mouse somatic cells. In this study, first we aimed to investigate whether exogenous induction of miR-302/367 in breast cancer cells shows the same tumor suppressive effects previously observed in other cancer cells lines, and whether vitamin C can enhance reprogramming of breast cancer cells and also improve the tumor suppressive function of miR-302/367 cluster. Overexpression of miR-302/367 cluster in MDA-MB-231 and SK-BR-3 breast cancer cells upregulated expression of miR-302/367 members and also some core pluripotency factors including OCT4A, SOX2 and NANOG, induced mesenchymal to epithelial transition, suppressed invasion, proliferation, and induced apoptosis in the both cell lines. However, treatment of the miR-302/367 transfected cells with vitamin C suppressed the expression of pluripotency factors and augmented the tumorigenicity of the breast cancer cells by restoring their proliferative and invasive capacity and compromising the apoptotic effect of miR-302/367. Supplementing the culture medium with vitamin C downregulated expression of TET1 gene which seems to be the reason behind the negative impact of vitamin C on the reprogramming efficiency of miR-302/367 cluster and its anti-tumor effects. Therefore application of vitamin C may not always serve as a reprogramming enhancer depending on its switching function on TET1. This phenomenon should be carefully considered when considering a reprogramming strategy for tumor suppression.
Collapse
Affiliation(s)
- Bahareh Ramezankhani
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Masoumeh F Taha
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Arash Javeri
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
76
|
Yuan J, Zhang F, Hallahan D, Zhang Z, He L, Wu LG, You M, Yang Q. Reprogramming glioblastoma multiforme cells into neurons by protein kinase inhibitors. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:181. [PMID: 30071868 PMCID: PMC6090992 DOI: 10.1186/s13046-018-0857-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/19/2018] [Indexed: 02/08/2023]
Abstract
Background Reprogramming of cancers into normal-like tissues is an innovative strategy for cancer treatment. Recent reports demonstrate that defined factors can reprogram cancer cells into pluripotent stem cells. Glioblastoma multiforme (GBM) is the most common and aggressive malignant brain tumor in humans. Despite multimodal therapy, the outcome for patients with GBM is still poor. Therefore, developing novel therapeutic strategy is a critical requirement. Methods We have developed a novel reprogramming method that uses a conceptually unique strategy for GBM treatment. We screened a kinase inhibitor library to find which candidate inhibitors under reprogramming condition can reprogram GBM cells into neurons. The induced neurons are identified whether functional and loss of tumorigenicity. Results We have found that mTOR and ROCK kinase inhibitors are sufficient to reprogram GBM cells into neural-like cells and “normal” neurons. The induced neurons expressed neuron-specific proteins, generated action potentials and neurotransmitter receptor-mediated currents. Genome-wide transcriptional analysis showed that the induced neurons had a profile different from GBM cells and were similar to that of control neurons induced by established methods. In vitro and in vivo tumorigenesis assays showed that induced neurons lost their proliferation ability and tumorigenicity. Moreover, reprogramming treatment with ROCK-mTOR inhibitors prevented GBM local recurrence in mice. Conclusion This study indicates that ROCK and mTOR inhibitors-based reprogramming treatment prevents GBM local recurrence. Currently ROCK-mTOR inhibitors are used as anti-tumor drugs in patients, so this reprogramming strategy has significant potential to move rapidly toward clinical trials. Electronic supplementary material The online version of this article (10.1186/s13046-018-0857-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Yuan
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA.,Medical Center of Stomatology, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.,School of Stomatology, Jinan University, Guangzhou, 510630, China
| | - Fan Zhang
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA
| | - Dennis Hallahan
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA
| | - Zhen Zhang
- Synaptic Transmission Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
| | - Liming He
- Synaptic Transmission Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
| | - Ling-Gang Wu
- Synaptic Transmission Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
| | - Meng You
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA
| | - Qin Yang
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA.
| |
Collapse
|
77
|
Hiew MSY, Cheng HP, Huang CJ, Chong KY, Cheong SK, Choo KB, Kamarul T. Incomplete cellular reprogramming of colorectal cancer cells elicits an epithelial/mesenchymal hybrid phenotype. J Biomed Sci 2018; 25:57. [PMID: 30025541 PMCID: PMC6052640 DOI: 10.1186/s12929-018-0461-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023] Open
Abstract
Background Induced pluripotency in cancer cells by ectopic expression of pluripotency-regulating factors may be used for disease modeling of cancers. MicroRNAs (miRNAs) are negative regulators of gene expression that play important role in reprogramming somatic cells. However, studies on the miRNA expression profile and the expression patterns of the mesenchymal-epithelial transition (MET)/epithelial-mesenchymal transition (EMT) genes in induced pluripotent cancer (iPC) cells are lacking. Methods iPC clones were generated from two colorectal cancer (CRC) cell lines by retroviral transduction of the Yamanaka factors. The iPC clones obtained were characterized by morphology, expression of pluripotency markers and the ability to undergo in vitro tri-lineage differentiation. Genome-wide miRNA profiles of the iPC cells were obtained by microarray analysis and bioinformatics interrogation. Gene expression was done by real-time RT-PCR and immuno-staining; MET/EMT protein levels were determined by western blot analysis. Results The CRC-iPC cells showed embryonic stem cell-like features and tri-lineage differentiation abilities. The spontaneously-differentiated post-iPC cells obtained were highly similar to the parental CRC cells. However, down-regulated pluripotency gene expression and failure to form teratoma indicated that the CRC-iPC cells had only attained partial pluripotency. The CRC-iPC cells shared similarities in the genome-wide miRNA expression profiles of both cancer and pluripotent embryonic stem cells. One hundred and two differentially-expressed miRNAs were identified in the CRC-iPC cells, which were predicted by bioinformatics analysis be closely involved in regulating cellular pluripotency and the expression of the MET/EMT genes, possibly via the phosphatidylinositol-3 kinases-protein kinase B (PI3K-Akt) and transforming growth factor beta (TGF-β) signaling pathways. Irregular and inconsistent expression patterns of the EMT vimentin and Snai1 and MET E-cadherin and occludin proteins were observed in the four CRC-iPC clones analyzed, which suggested an epithelial/mesenchymal hybrid phenotype in the partially reprogrammed CRC cells. MET/EMT gene expression was also generally reversed on re-differentiation, also suggesting epigenetic regulation. Conclusions Our data support the elite model for cancer cell-reprogramming in which only a selected subset of cancer may be fully reprogrammed; partial cancer cell reprogramming may also elicit an epithelial-mesenchymal mixed phenotype, and highlight opportunities and challenges in cancer cell-reprogramming. Electronic supplementary material The online version of this article (10.1186/s12929-018-0461-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michele Sook Yuin Hiew
- Centre for Stem Cell Research & Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long campus, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor, Malaysia.,Postgraduate Program, Universiti Tunku Abdul Rahman, Sg. Long, Selangor, Malaysia.,Present address: Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Han Ping Cheng
- Centre for Stem Cell Research & Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long campus, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor, Malaysia.,Postgraduate Program, Universiti Tunku Abdul Rahman, Sg. Long, Selangor, Malaysia
| | - Chiu-Jung Huang
- Department of Animal Science & Graduate Institute of Biotechnology, Chinese Culture University, Taipei, Taiwan
| | - Kowit Yu Chong
- Graduate Institute of Biomedical Sciences, Department of Medical Biotechnology and Laboratory Science & Molecular Medicine Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Soon Keng Cheong
- Centre for Stem Cell Research & Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long campus, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor, Malaysia.,Dean's Office, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sg. Long, Selangor, Malaysia
| | - Kong Bung Choo
- Centre for Stem Cell Research & Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long campus, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor, Malaysia. .,Postgraduate Program, Universiti Tunku Abdul Rahman, Sg. Long, Selangor, Malaysia.
| | - Tunku Kamarul
- National Orthopaedic Centre of Excellence for Research and Learning & Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
78
|
Liu M, Tu J, Gingold JA, Kong CSL, Lee DF. Cancer in a dish: progress using stem cells as a platform for cancer research. Am J Cancer Res 2018; 8:944-954. [PMID: 30034933 PMCID: PMC6048395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 06/08/2023] Open
Abstract
Cancer models derived from patient specimens poorly reflect early-stage cancer development because cancer cells acquire numerous additional molecular alterations before the disease is clinically detectable. Earlier studies have used differentiated cells derived from induced pluripotent cancer cells (iPCCs) to partially mirror cancer disease phenotype, but the highly heterogeneous nature of cancer cells as well as difficulties with reprogramming cancer cells has limited the application of this technique. An alternative approach to modeling cancer in a dish entails reprogramming adult differentiated cells from patients with cancer syndromes to pluripotent stem cells (PSCs), followed by directed differentiation of those PSCs. A directed reprogramming and differentiation strategy has the potential to recapitulate cancer progression and capture the earliest molecular alterations that underlie cancer initiation. The reprogrammed cells share patient-specific genetic and epigenetic traits, offering a new platform to develop personalized therapy for cancer patients. In this review, we will provide an overview of available reprogramming methods of cancer cells and describe how cancer-derived stem cells have been used to characterize effects of defined molecular alterations in specific cell types. We also describe the "disease in a dish" model developed to study genetic cancer syndromes. These approaches highlight recent contributions of stem cell technology to the cancer biology realm.
Collapse
Affiliation(s)
- Mo Liu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at HoustonHouston, TX 77030, USA
| | - Jian Tu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at HoustonHouston, TX 77030, USA
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou 510080, PR China
| | - Julian A Gingold
- Women’s Health Institute, Cleveland Clinic FoundationCleveland, OH 44195, USA
| | - Celine Shuet Lin Kong
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at HoustonHouston, TX 77030, USA
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at HoustonHouston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical SciencesHouston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for The Prevention of Human Diseases, The University of Texas Health Science Center at HoustonHouston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, The University of Texas Health Science Center at HoustonHouston, TX 77030, USA
| |
Collapse
|
79
|
Abstract
Research on stem cells is one of the fastest growing areas of regenerative medicine that paves the way for a comprehensive solution to cell therapy. Today, stem cells are precious assets for generating different types of cells derived from either natural embryonic stem (ES) cells or induced pluripotent stem (iPS) cells. The iPS technology can revolutionize the future of clinics by offering personalized medicine, which will provide the future treatment for curing untreatable diseases. Although iPS cell therapy is now at its infancy, promising research has motivated scientists to pursue this therapeutic approach. In this article, we provide information regarding similarities and differences between ES and iPS cells, and focus on the non-integrating methods of iPS generation via RNA molecules, especially microRNAs with an emphasis on the elucidation of their role and importance in pluripotency.
Collapse
Affiliation(s)
- Abbas Beh-Pajooh
- REBIRTH-Group Translational Hepatology and Stem Cell Biology, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Tobias Cantz
- REBIRTH-Group Translational Hepatology and Stem Cell Biology, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
80
|
Wang N, Zhang Z, Lv J. Fentanyl inhibits proliferation and invasion via enhancing miR-302b expression in esophageal squamous cell carcinoma. Oncol Lett 2018; 16:459-466. [PMID: 29928433 DOI: 10.3892/ol.2018.8616] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/17/2018] [Indexed: 12/21/2022] Open
Abstract
Fentanyl is one of the most commonly used intravenous anesthetic agents during cancer resection surgery, but the effect of fentanyl on esophageal squamous cell carcinoma (ESCC) remains unclear. The aim of the present study was to investigate the involvement of microRNA 302b (miR-302b) in the anti-proliferation and anti-invasion effects of fentanyl in ESCC. In the present study, the effects of fentanyl on cell proliferation, apoptosis and invasion were detected using MTT assays, flow cytometry and Transwell assays in ESCC Eca109 and TE1 cell lines. Subsequently, expression of miR-302b was determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RT-qPCR and western blot analysis were performed in order to evaluate the expression of ErbB4, a target of miR-302b. Furthermore, anti-miR were used to inhibit miR-302b in fentanyl-treated ESCC cells in order to evaluate the role of miR-302b in the effect of fentanyl on malignant behaviors. Fentanyl inhibited the proliferation of Eca109 and TE1 cells in a dose- and time-dependent manner. Following exposure to fentanyl for 48 h, Eca109 and TE1 cells exhibited increased apoptosis and decreased invasion. Furthermore, fentanyl upregulated miR-302b expression, but downregulated ErbB4 expression. Finally, loss of miR-302b using the anti-miR technique reversed the effect of fentanyl on cell proliferation, apoptosis and invasion in the two ESCC cell lines. Taken together, the results of the present study indicated that fentanyl inhibits the proliferation and invasion of ESCC cells through upregulation of miR-302b.
Collapse
Affiliation(s)
- Ning Wang
- Department of Anesthesiology, Second Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710002, P.R. China
| | - Zhenni Zhang
- Department of Anesthesiology, Second Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710002, P.R. China
| | - Jianrui Lv
- Department of Anesthesiology, Second Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710002, P.R. China
| |
Collapse
|
81
|
c-Jun-mediated microRNA-302d-3p induces RPE dedifferentiation by targeting p21 Waf1/Cip1. Cell Death Dis 2018; 9:451. [PMID: 29670082 PMCID: PMC5906557 DOI: 10.1038/s41419-018-0481-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/01/2018] [Accepted: 03/09/2018] [Indexed: 11/08/2022]
Abstract
Dedifferentiation of retinal pigment epithelium (RPE) cells and choroidal neovascularization (CNV) contributes to the pathogenesis of age-related macular degeneration (AMD). MicroRNAs (miRNAs) have crucial roles in AMD onset and progression. We thus aim to investigate the effects of miRNAs on RPE dedifferentiation and endothelium cell (EC) behavior, and analyze its downstream pathways. We have previously identified miR-302d-3p as the most downregulated miRNA signature along with RPE differentiation. Herein, in vitro study supported that miR-302d-3p induces RPE dedifferentiation typified by reduction of RPE characteristic markers, interrupts its phagocytosis, and promotes its migration, proliferation, and cell-cycle progression. c-Jun was identified as a potential upstream transcript factor for MIR302D, which might modulate RPE function by regulating miR-302d-3p expression. P21Waf1/Cip1, a cyclin-dependent kinase inhibitor encoded by the CDKN1A gene, was identified as a downstream target of miR-302d-3p. Our data suggested that p21Waf1/Cip1 could promote RPE differentiation, and inhibit its proliferation, migration, and cell-cycle progression. We also demonstrated that miR-302d-3p suppresses RPE differentiation through directly targeting p21Waf1/Cip1. In addition, the miR-302d-3p/CDKN1A axis was also involved in regulating tube formation of ECs, indicating its potential involvement in CNV formation. Taken together, our study implies that miR-302d-3p, regulated by c-Jun, contributes to the pathogenesis of both atrophic and exudative AMD. MiR-302d-3p promotes RPE dedifferentiation, migration, proliferation and cell-cycle progression, inhibits RPE phagocytosis, and induces abnormal EC behavior by targeting p21Waf1/Cip1. Pharmacological miR-302d-3p inhibitors are prospective therapeutic options for prevention and treatment of AMD.
Collapse
|
82
|
LaMarca EA, Powell SK, Akbarian S, Brennand KJ. Modeling Neuropsychiatric and Neurodegenerative Diseases With Induced Pluripotent Stem Cells. Front Pediatr 2018; 6:82. [PMID: 29666786 PMCID: PMC5891587 DOI: 10.3389/fped.2018.00082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) have revolutionized our ability to model neuropsychiatric and neurodegenerative diseases, and recent progress in the field is paving the way for improved therapeutics. In this review, we discuss major advances in generating hiPSC-derived neural cells and cutting-edge techniques that are transforming hiPSC technology, such as three-dimensional "mini-brains" and clustered, regularly interspersed short palindromic repeats (CRISPR)-Cas systems. We examine specific examples of how hiPSC-derived neural cells are being used to uncover the pathophysiology of schizophrenia and Parkinson's disease, and consider the future of this groundbreaking research.
Collapse
Affiliation(s)
- Elizabeth A. LaMarca
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Samuel K. Powell
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Schahram Akbarian
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J. Brennand
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
83
|
Sun WL, Wang N, Xu Y. Impact of miR-302b on Calcium-phosphorus Metabolism and Vascular Calcification of Rats with Chronic Renal Failure by Regulating BMP-2/Runx2/Osterix Signaling Pathway. Arch Med Res 2018; 49:164-171. [DOI: 10.1016/j.arcmed.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 07/30/2018] [Indexed: 12/22/2022]
|
84
|
Balzano F, Cruciani S, Basoli V, Santaniello S, Facchin F, Ventura C, Maioli M. MiR200 and miR302: Two Big Families Influencing Stem Cell Behavior. Molecules 2018; 23:282. [PMID: 29385685 PMCID: PMC6017081 DOI: 10.3390/molecules23020282] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 01/25/2018] [Accepted: 01/27/2018] [Indexed: 02/08/2023] Open
Abstract
In this review, we described different factors that modulate pluripotency in stem cells, in particular we aimed at following the steps of two large families of miRNAs: the miR-200 family and the miR-302 family. We analyzed some factors tuning stem cells behavior as TGF-β, which plays a pivotal role in pluripotency inhibition together with specific miRNAs, reactive oxygen species (ROS), but also hypoxia, and physical stimuli, such as ad hoc conveyed electromagnetic fields. TGF-β plays a crucial role in the suppression of pluripotency thus influencing the achievement of a specific phenotype. ROS concentration can modulate TGF-β activation that in turns down regulates miR-200 and miR-302. These two miRNAs are usually requested to maintain pluripotency, while they are down-regulated during the acquirement of a specific cellular phenotype. Moreover, also physical stimuli, such as extremely-low frequency electromagnetic fields or high-frequency electromagnetic fields conveyed with a radioelectric asymmetric conveyer (REAC), and hypoxia can deeply influence stem cell behavior by inducing the appearance of specific phenotypes, as well as a direct reprogramming of somatic cells. Unraveling the molecular mechanisms underlying the complex interplay between externally applied stimuli and epigenetic events could disclose novel target molecules to commit stem cell fate.
Collapse
Affiliation(s)
- Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- GUNA ATTRE (Advanced Therapies and Tissue REgeneration), Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Istituto di RicercaGenetica e Biomedica, Consiglio Nazionaledelle Ricerche (CNR), Monserrato, 09042 Cagliari, Italy.
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| |
Collapse
|
85
|
Lin SL. Identification and Isolation of Novel Sugar-Like RNA Protecting Materials: Glycylglycerins from Pluripotent Stem Cells. Methods Mol Biol 2018; 1733:305-316. [PMID: 29435942 DOI: 10.1007/978-1-4939-7601-0_24] [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] [Indexed: 06/08/2023]
Abstract
Pluripotent stem cells are a resourceful treasure box for regenerative medicine. They contain a large variety of novel materials useful for designing and developing new medicines and therapies directed against many aging-associated degenerative disorders, including Alzheimer's disease, Parkinson's disease, stroke, diabetes, osteoporosis, and cancers. Currently, identification of these novel stem cell-specific materials is one of major breakthroughs in the field of stem cell research. Particularly, since the discovery of induced pluripotent stem cells (iPSC) in year 2006, the methods of iPSC derivation further provide an unlimited resource for screening, isolating, and even producing theses novel stem cell-specific materials in vitro. Using iPSCs, we can now prepare high quality and quantity of pure stem cell-specific agents for testing their therapeutic functions in treating various illnesses. These newly found stem cell-specific agents are divided into four major categories, including proteins, saccharides, nucleic acids, and small molecules (chemicals). In this article, we herein disclose one of the methodologies for isolating and purifying glycylglycerins-a group of glycylated sugar alcohols that protect hairpin-like microRNA precursors (pre-miRNA) and some of tRNAs in pluripotent stem cells. In view of such a unique RNA-protecting feature, glycylglycerins may be used to preserve and deliver functional small RNAs, such as pre-miRNAs and small interfering RNAs (siRNA), into human cells for eliciting their specific RNA interference (RNAi) effects, which may greatly advance the use of RNAi technology for treating human diseases.
Collapse
Affiliation(s)
- Shi-Lung Lin
- Division of Regenerative Medicine, WJWU and LYNN Institute for Stem Cell Research, Santa Fe Springs, CA, USA.
| |
Collapse
|
86
|
Ying SY, Fang W, Lin SL. The miR-302-Mediated Induction of Pluripotent Stem Cells (iPSC): Multiple Synergistic Reprogramming Mechanisms. Methods Mol Biol 2018; 1733:283-304. [PMID: 29435941 DOI: 10.1007/978-1-4939-7601-0_23] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pluripotency represents a unique feature of embryonic stem cells (ESCs). To generate ESC-like-induced pluripotent stem cells (iPSCs) derived from somatic cells, the cell genome needs to be reset and reprogrammed to express the ESC-specific transcriptome. Numerous studies have shown that genomic DNA demethylation is required for epigenetic reprogramming of somatic cell nuclei to form iPSCs; yet, the mechanism remains largely unclear. In ESCs, the reprogramming process goes through two critical stages: germline and zygotic demethylation, both of which erase genomic DNA methylation sites and hence allow for different gene expression patterns to be reset into a pluripotent state. Recently, miR-302, an ESC-specific microRNA (miRNA), was found to play an essential role in four aspects of this reprogramming mechanism-(1) initiating global genomic DNA demethylation, (2) activating ESC-specific gene expression, (3) inhibiting developmental signaling, and (4) preventing stem cell tumorigenicity. In this review, we will summarize miR-302 functions in all four reprogramming aspects and further discuss how these findings may improve the efficiency and safety of the current iPSC technology.
Collapse
Affiliation(s)
- Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - William Fang
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shi-Lung Lin
- Division of Regenerative Medicine, WJWU & LYNN (W&L) Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| |
Collapse
|
87
|
Abstract
MicroRNAs (miRNAs), widely distributed, small regulatory RNA genes, target both messenger RNA (mRNA) degradation and suppression of protein translation based on sequence complementarity between the miRNA and its targeted mRNA. Different names have been used to describe various types of miRNA. During evolution, RNA retroviruses or transgenes invaded the eukaryotic genome and were inserted itself in the noncoding regions of DNA, conceivably acting as transposon-like jumping genes, providing defense from viral invasion and fine-tuning of gene expression as a secondary level of gene modulation in eukaryotes. When a transposon is inserted in the intron, it becomes an intronic miRNA, taking advantage of the protein synthesis machinery, i.e., mRNA transcription and splicing, as a means for processing and maturation. MiRNAs have been found to play an important, but not life-threatening, role in embryonic development. They might play a pivotal role in diverse biological systems in various organisms, facilitating a quick response and accurate plotting of body physiology and structures. Based on these unique properties, manufactured intronic miRNAs have been developed for in vitro evaluation of gene function, in vivo gene therapy, and generation of transgenic animal models. The biogenesis of miRNAs, circulating miRNAs, miRNAs and cancer, iPSCs, and heart disease are presented in this chapter, highlighting some recent studies on these topics.
Collapse
Affiliation(s)
- Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Donald C Chang
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| | - Shi-Lung Lin
- Division of Regenerative Medicine, WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| |
Collapse
|
88
|
Mechanism and Method for Generating Tumor-Free iPS Cells Using Intronic MicroRNA miR-302 Induction. Methods Mol Biol 2018; 1733:265-282. [PMID: 29435940 DOI: 10.1007/978-1-4939-7601-0_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Today's researchers generating induced pluripotent stem cells (iPS cells or iPSCs) usually consider their pluripotency rather than potential tumorigenicity. Oncogenic factors such as c-Myc and Klf4 are frequently used to boost the survival and proliferative rates of iPSCs, creating an inevitable problem of tumorigenicity that hinders the therapeutic usefulness of these iPSCs. To prevent stem cell tumorigenicity, we have examined mechanisms by which the cell cycle genes are regulated in embryonic stem cells (ESCs). Naturally, ESCs possess two unique stemness properties: pluripotent differentiation into almost all cell types and unlimited self-renewal without the risk of tumor formation. These two features are also important for the use of ESCs or iPSCs in therapy. Currently, despite overwhelming reports describing iPSC pluripotency, there is no report of any tumor prevention mechanism in either ESCs or iPSCs. To this, our studies have revealed for the first time that an ESC-specific microRNA (miRNA), miR-302, regulates human iPSC tumorigenicity through cosuppression of both cyclin E-CDK2 and cyclin D-CDK4/6 cell cycle pathways during G1-S phase transition. Moreover, miR-302 also silences BMI-1, a cancer stem cell gene marker, to promote the expression of two senescence-associated tumor suppressor genes, p16Ink4a and p14/p19Arf. Together, the combinatory effects of inhibiting G1-S cell cycle transition and increasing p16/p14(p19) expression result in an attenuated cell cycle rate similar to that of 2-to-8-cell-stage embryonic cells in early zygotes (20-24 h/cycle), which is however slower than the fast proliferation rate of iPSCs induced by the four defined factors Oct4-Sox2-Klf4-c-Myc (12-16 h/cycle). These findings provide a means to control iPSC tumorigenicity and improve the safety of iPSCs for the therapeutic use. In this chapter, we review the mechanism underlying miR-302-mediated tumor suppression and then demonstrate how to apply this mechanism to generate tumor-free iPSCs. The same strategy may also be used to prevent ESC tumorigenicity.
Collapse
|
89
|
Bourguignon LYW. Hyaluronan-Mediated CD44 Signaling Activates Cancer Stem Cells in Head and Neck Cancer. CURRENT CANCER RESEARCH 2018:525-544. [DOI: 10.1007/978-3-319-78762-6_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
90
|
Pisal RV, Hrebikova H, Chvatalova J, Soukup T, Stanislav F, Mokry J. Cloning of intronic sequence within DsRed2 increased the number of cells expressing red fluorescent protein. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2017; 161:354-359. [PMID: 28840901 DOI: 10.5507/bp.2017.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/11/2017] [Indexed: 11/23/2022] Open
Abstract
AIM Cloning of artificial intronic sequence within the open reading frame (ORF) of DsRed2 gene. METHOD Splice prediction software was used to analyze DsRed2 sequence to find an ideal site for cloning artificial intronic sequence. Intron was cloned within DsRed2 using cyclic ligation assembly. Flow cytometry was used to quantify the number of cells expressing red fluorescence. RESULT Sequencing data confirmed precise cloning of intron at the desired position using cyclic ligation assembly. Successful expression of red fluorescence after cloning of intron confirmed successful intron recognition and splicing by host cell line. Cloning of intron increased the number of cells expressing red fluorescent protein. CONCLUSION Cloning of intronic sequence within DsRed2 has helped to increase the number of cells expressing red fluorescence by approximately four percent.
Collapse
Affiliation(s)
- Rishikaysh V Pisal
- Department of Histology and Embryology, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, 500 03 Hradec Kralove, Czech Republic
| | - Hana Hrebikova
- Department of Histology and Embryology, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, 500 03 Hradec Kralove, Czech Republic
| | - Jana Chvatalova
- Department of Histology and Embryology, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, 500 03 Hradec Kralove, Czech Republic
| | - Tomas Soukup
- Department of Histology and Embryology, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, 500 03 Hradec Kralove, Czech Republic
| | - Filip Stanislav
- Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, 500 03 Hradec Kralove, Czech Republic
| | - Jaroslav Mokry
- Department of Histology and Embryology, Faculty of Medicine in Hradec Kralove, Charles University, Simkova 870, 500 03 Hradec Kralove, Czech Republic
| |
Collapse
|
91
|
|
92
|
Wang L, Chen Y, Guan C, Zhao Z, Li Q, Yang J, Mo J, Wang B, Wu W, Yang X, Song L, Li J. Using low-risk factors to generate non-integrated human induced pluripotent stem cells from urine-derived cells. Stem Cell Res Ther 2017; 8:245. [PMID: 29096702 PMCID: PMC5667457 DOI: 10.1186/s13287-017-0698-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 09/21/2017] [Accepted: 10/16/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Because the lack of an induced pluripotent stem cell (iPSC) induction system with optimal safety and efficiency limits the application of these cells, development of such a system is important. METHODS To create such an induction system, we screened a variety of reprogrammed plasmid combinations and multiple compounds and then verified the system's feasibility using urine cells from different individuals. We also compared large-scale iPSC chromosomal variations and expression of genes associated with genomic stability between this system and the traditional episomal system using karyotype and quantitative reverse transcription polymerase chain reaction analyses. RESULTS We developed a high-efficiency episomal system, the 6F/BM1-4C system, lacking tumorigenic factors for human urine-derived cell (hUC) reprogramming. This system includes six low-risk factors (6F), Oct4, Glis1, Klf4, Sox2, L-Myc, and the miR-302 cluster. Transfected hUCs were treated with four compounds (4C), inhibitor of lysine-demethylase1, methyl ethyl ketone, glycogen synthase kinase 3 beta, and histone deacetylase, within a short time period. Comparative analysis revealed significantly decreased chromosomal variation in iPSCs and significantly increased Sirt1 expression compared with iPSCs induced using the traditional episomal system. CONCLUSION The 6F/BM1-4C system effectively induces reprogramming of urine cells in samples obtained from different individuals. iPSCs induced using the 6F/BM1-4C system are more stable at the cytogenetic level and have potential value for clinical application.
Collapse
Affiliation(s)
- Linli Wang
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China.
| | - Yuehua Chen
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Chunyan Guan
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Zhiju Zhao
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Qiang Li
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Jianguo Yang
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Jian Mo
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Bin Wang
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Wei Wu
- The Guangdong Key Lab for Shock and Microcirculation Research, Departments of Pathophysiology, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiaohui Yang
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China
| | - Libing Song
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Centre, Guangzhou, 510060, People's Republic of China
| | - Jun Li
- Guangzhou Biocare Institute of Cancer, Building D, Guangzhou International Business Incubator, No. 3, Juquan Road, Guangzhou Science Park, Guangzhou, 510663, Guangdong, People's Republic of China. .,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Yuexiu District, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
93
|
Ariyoshi J, Matsuyama Y, Kobori A, Murakami A, Sugiyama H, Yamayoshi A. Effective Anti-miRNA Oligonucleotides Show High Releasing Rate of MicroRNA from RNA-Induced Silencing Complex. Nucleic Acid Ther 2017; 27:303-308. [PMID: 28876213 DOI: 10.1089/nat.2017.0663] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression by forming RNA-induced silencing complexes (RISCs) and have been considered as promising therapeutic targets. MiRNA is an essential component of RISC for the modulation of gene expression. Therefore, the release of miRNA from RISC is considered as an effective method for the inhibition of miRNA functions. In our previous study, we reported that anti-miRNA oligonucleotides (AMOs), which are composed of the 2'-O-methyl (2'-OMe) RNA, could induce the release of miRNA from RISC. However, the mechanisms underlying the miRNA-releasing effects of chemically modified AMOs, which are conventionally used as anti-cancer drugs, are still unclear. In this study, we investigated the relationship between the miRNA releasing rate from RISC and the inhibitory effect on RISC activity (IC50) using conventional chemically modified AMOs. We demonstrated that the miRNA-releasing effects of AMOs are directly proportional to the IC50 values, and AMOs, which have an ability to promote the release of miRNA from RISC, can effectively inhibit RISC activity in living cells.
Collapse
Affiliation(s)
- Jumpei Ariyoshi
- 1 Department of Biomolecular Engineering, Kyoto Institute of Technology , Kyoto, Japan .,2 Department of Chemistry, Graduate School of Science, Kyoto University , Kyoto, Japan
| | - Yohei Matsuyama
- 1 Department of Biomolecular Engineering, Kyoto Institute of Technology , Kyoto, Japan
| | - Akio Kobori
- 1 Department of Biomolecular Engineering, Kyoto Institute of Technology , Kyoto, Japan
| | - Akira Murakami
- 3 Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University , Kyoto, Japan
| | - Hiroshi Sugiyama
- 2 Department of Chemistry, Graduate School of Science, Kyoto University , Kyoto, Japan .,4 Institute for Integrated Cell-Materials Sciences (iCeMS), Kyoto University , Kyoto, Japan
| | - Asako Yamayoshi
- 2 Department of Chemistry, Graduate School of Science, Kyoto University , Kyoto, Japan .,5 The Hakubi Center for Advanced Research, Kyoto University , Kyoto, Japan
| |
Collapse
|
94
|
Ma G, Li Q, Dai W, Yang X, Sang A. Prognostic Implications of miR-302a/b/c/d in Human Gastric Cancer. Pathol Oncol Res 2017; 23:899-905. [PMID: 28795345 DOI: 10.1007/s12253-017-0282-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/06/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND The microRNA (miR)-302 family consisting four members, miR-302a, miR-302b, miR-302c and miR-302d, plays an important role in diverse biological processes, and regulates many pathological changes, including cancer. However, the involvement of the miR-302 family into human gastric cancer (GC) remains unclear. The aim of this study was to investigate the expression patterns of miR-302a/b/c/d and determine their clinical significance in GC. MATERIALS AND METHODS Expression levels of miR-302a/b/c/d in 160 pairs of human GC and matched normal mucosa tissues were detected by quantitative real-time polymerase chain reaction. Then, the associations of miR-302a/b/c/d expression with various clinicopathological characteristics and patients' prognosis were statistically evaluated. RESULTS The expression levels of miR-302a, miR-302b and miR-302c in GC tissues were all significantly lower than those in matched normal mucosa (all P < 0.001), but miR-302d expression had no significant differences between cancer and normal groups. Additionally, GC patients with low miR-302a, miR-302b and miR-302c expression more frequently had positive lymph node metastasis (all P < 0.05), advanced TNM stage (all P < 0.05) and great depth of invasion (all P < 0.05). More importantly, low miR-302a, miR-302b and miR-302c expression in GC tissues were significantly associated with shorter disease-free and overall survivals of GC patients (all P < 0.05). Further multivariate analysis identified miR-302a, miR-302b and miR-302c as independent prognostic markers for GC patients. CONCLUSIONS GC patients with the decreased expression of miR-302a, miR-302b and miR-302c may had aggressive cancer progression and unfavorable prognosis. Further rigorous validation based on a large cohort of clinical cases should be performed.
Collapse
Affiliation(s)
- Gang Ma
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, People's Republic of China
| | - Qianjun Li
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, People's Republic of China
| | - Weijie Dai
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, People's Republic of China
| | - Xiaozhong Yang
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, People's Republic of China.
| | - Aiyu Sang
- Department of Internal Medicine, Lianshui Third People's Hospital, 12 Gaogouzhen 307 Road South, Lianshui, Jiangsu, 223411, People's Republic of China.
| |
Collapse
|
95
|
Li Y, Li L, Chen ZN, Gao G, Yao R, Sun W. Engineering-derived approaches for iPSC preparation, expansion, differentiation and applications. Biofabrication 2017; 9:032001. [DOI: 10.1088/1758-5090/aa7e9a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
96
|
Chen Q, Hu G. Post-transcriptional regulation of the pluripotent state. Curr Opin Genet Dev 2017; 46:15-23. [PMID: 28654825 DOI: 10.1016/j.gde.2017.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/17/2017] [Accepted: 06/08/2017] [Indexed: 12/20/2022]
Abstract
Pluripotency describes the developmental capacity to give rise to all cell types in the adult body. A comprehensive understanding of the molecular mechanisms that regulate pluripotency is important for both basic and translational research. While earlier studies mostly focused on signaling pathways, transcriptional regulation, and epigenetic modifications, recent investigations showed that RNA binding proteins, RNA processing machineries, and regulatory RNA molecules also play essential roles. Here, we provide a concise review on the latest findings and developments in post-transcriptional regulation of the pluripotent state.
Collapse
Affiliation(s)
- Qing Chen
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, RTP, NC, United States.
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, RTP, NC, United States.
| |
Collapse
|
97
|
Rawat N, Singh MK. Induced pluripotent stem cell: A headway in reprogramming with promising approach in regenerative biology. Vet World 2017; 10:640-649. [PMID: 28717316 PMCID: PMC5499081 DOI: 10.14202/vetworld.2017.640-649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 04/26/2017] [Indexed: 12/17/2022] Open
Abstract
Since the embryonic stem cells have knocked the doorsteps, they have proved themselves in the field of science, research, and medicines, but the hovered restrictions confine their application in human welfare. Alternate approaches used to reprogram the cells to the pluripotent state were not up to par, but the innovation of induced pluripotent stem cells (iPSCs) paved a new hope for the researchers. Soon after the discovery, iPSCs technology is undergoing renaissance day by day, i.e., from the use of genetic material to recombinant proteins and now only chemicals are employed to convert somatic cells to iPSCs. Thus, this technique is moving straightforward and productive at an astonishing pace. Here, we provide a brief introduction to iPSCs, the mechanism and methods for their generation, their prevailing and prospective applications and the future opportunities that can be expected from them.
Collapse
Affiliation(s)
- N Rawat
- Embryo Biotechnology Lab, Animal Biotechnology Centre, ICAR - National Dairy Research Institute, Karnal - 132 001, Haryana, India
| | - M K Singh
- Embryo Biotechnology Lab, Animal Biotechnology Centre, ICAR - National Dairy Research Institute, Karnal - 132 001, Haryana, India
| |
Collapse
|
98
|
Abstract
Stem cells are undifferentiated cells and have multi-lineage differentiation potential. Generally, stem cells are classified into adult stem cells, embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Stem cells have great potential in clinical therapy due to their pluripotency and self-renewal ability. microRNAs (miRNAs) are small non-coding RNAs which are evolutionarily conserved and participate in the pathogenesis of many diseases, cell cycle regulation, apoptosis, aging, cell fate decisions, and different signaling pathways. Different kinds of stem cells possess distinct miRNA expression profiles. Our review summarizes the critical roles of miRNAs in stem cell reprogramming, pluripotency maintenance, and differentiation. In the future, miRNAs may greatly contribute to stem cell clinical therapy and have potential applications in regenerative medicine.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bo Long
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Han
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Shumin Yuan
- Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Kun Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
99
|
Verusingam ND, Yeap SK, Ky H, Paterson IC, Khoo SP, Cheong SK, Ong AHK, Kamarul T. Susceptibility of Human Oral Squamous Cell Carcinoma (OSCC) H103 and H376 cell lines to Retroviral OSKM mediated reprogramming. PeerJ 2017; 5:e3174. [PMID: 28417059 PMCID: PMC5392249 DOI: 10.7717/peerj.3174] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
Although numbers of cancer cell lines have been shown to be successfully reprogrammed into induced pluripotent stem cells (iPSCs), reprogramming Oral Squamous Cell Carcinoma (OSCC) to pluripotency in relation to its cancer cell type and the expression pattern of pluripotent genes under later passage remain unexplored. In our study, we reprogrammed and characterised H103 and H376 oral squamous carcinoma cells using retroviral OSKM mediated method. Reprogrammed cells were characterized for their embryonic stem cells (ESCs) like morphology, pluripotent gene expression via quantitative real-time polymerase chain reaction (RT-qPCR), immunofluorescence staining, embryoid bodies (EB) formation and directed differentiation capacity. Reprogrammed H103 (Rep-H103) exhibited similar ESCs morphologies with flatten cells and clear borders on feeder layer. Reprogrammed H376 (Rep-H376) did not show ESCs morphologies but grow with a disorganized morphology. Critical pluripotency genes Oct4, Sox2 and Nanog were expressed higher in Rep-H103 against the parental counterpart from passage 5 to passage 10. As for Rep-H376, Nanog expression against its parental counterpart showed a significant decrease at passage 5 and although increased in passage 10, the level of expression was similar to the parental cells. Rep-H103 exhibited pluripotent signals (Oct4, Sox2, Nanog and Tra-1-60) and could form EB with the presence of three germ layers markers. Rep-H103 displayed differentiation capacity into adipocytes and osteocytes. The OSCC cell line H103 which was able to be reprogrammed into an iPSC like state showed high expression of Oct4, Sox2 and Nanog at late passage and may provide a potential iPSC model to study multi-stage oncogenesis in OSCC.
Collapse
Affiliation(s)
- Nalini Devi Verusingam
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Swee Keong Yeap
- Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia.,Current affiliation: China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Selangor, Malaysia
| | - Huynh Ky
- College of Agriculture and Applied Science, Cantho University, Vietnam
| | - Ian C Paterson
- Department of Oral Biology & Biomedical Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Suan Phaik Khoo
- School of Dentistry, International Medical University, Kuala Lumpur, Malaysia
| | - Soon Keng Cheong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia.,Majlis Kanser Nasional (MAKNA) Cancer Research Institute, Kuala Lumpur, Malaysia
| | - Alan H K Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Tunku Kamarul
- Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning, Department of Orthopaedic Surgery, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
100
|
Wooten DJ, Quaranta V. Mathematical models of cell phenotype regulation and reprogramming: Make cancer cells sensitive again! Biochim Biophys Acta Rev Cancer 2017; 1867:167-175. [PMID: 28396217 DOI: 10.1016/j.bbcan.2017.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023]
Abstract
A cell's phenotype is the observable actualization of complex interactions between its genome, epigenome, and local environment. While traditional views in cancer have held that cellular and tumor phenotypes are largely functions of genomic instability, increasing attention has recently been given to epigenetic and microenvironmental influences. Such non-genetic factors allow cancer cells to experience intrinsic diversity and plasticity, and at the tumor level can result in phenotypic heterogeneity and treatment evasion. In 2006, Takahashi and Yamanaka exploited the epigenome's plasticity by "reprogramming" differentiated cells into a pluripotent state by inducing expression of a cocktail of four transcription factors. Recent advances in cancer biology have shown not only that cellular reprogramming is possible for malignant cells, but it may provide a foundation for future therapies. Nevertheless, cell reprogramming experiments are frequently plagued by low efficiency, activation of aberrant transcriptional programs, instability, and often rely on expertise gathered from systems which may not translate directly to cancer. Here, we review a theoretical framework tracing back to Waddington's epigenetic landscape which may be used to derive quantitative and qualitative understanding of cellular reprogramming. Implications for tumor heterogeneity, evolution and adaptation are discussed in the context of designing new treatments to re-sensitize recalcitrant tumors. This article is part of a Special Issue entitled: Evolutionary principles - heterogeneity in cancer?, edited by Dr. Robert A. Gatenby.
Collapse
Affiliation(s)
- David J Wooten
- Vanderbilt University School of Medicine, 2220 Pierce Ave., 446B, Nashville, TN 37232, United States
| | - Vito Quaranta
- Vanderbilt University School of Medicine, 2220 Pierce Ave., 446B, Nashville, TN 37232, United States.
| |
Collapse
|