51
|
Trosko JE. The Concept of "Cancer Stem Cells" in the Context of Classic Carcinogenesis Hypotheses and Experimental Findings. Life (Basel) 2021; 11:1308. [PMID: 34947839 PMCID: PMC8708536 DOI: 10.3390/life11121308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
In this Commentary, the operational definition of cancer stem cells or cancer initiating cells includes the ability of certain cells, found in a heterogeneous mixture of cells within a tumor, which are able to sustain growth of that tumor. However, that concept of cancer stem cells does not resolve the age-old controversy of two opposing hypotheses of the origin of the cancer, namely the stem cell hypothesis versus the de-differentiation or re-programming hypothesis. Moreover, this cancer stem concept has to take into account classic experimental observations, techniques, and concepts, such as the multi-stage, multi-mechanism process of carcinogenesis; roles of mutagenic, cytotoxic and epigenetic mechanisms; the important differences between errors of DNA repair and errors of DNA replication in forming mutations; biomarkers of known characteristics of normal adult organ-specific stem cells and of cancer stem cells; and the characteristics of epigenetic mechanisms involved in the carcinogenic process. In addition, vague and misleading terms, such as carcinogens, immortal and normal cells have to be clarified in the context of current scientific facts. The ultimate integration of all of these historic factors to provide a current understanding of the origin and characteristics of a cancer stem cell, which is required for a rational strategy for prevention and therapy for cancer, does not follow a linear path. Lastly, it will be speculated that there exists evidence of two distinct types of cancer stem cells, one that has its origin in an organ-specific adult stem cell that is 'initiated' in the stem cell stage, expressing the Oct4A gene and not expressing any connexin gene or having functional gap junctional intercellular communication (GJIC). The other cancer stem cell is derived from a stem cell that is initiated early after the Oct4A gene is suppressed and the connexin gene is expressed, which starts early differentiation, but it is blocked from terminal differentiation.
Collapse
Affiliation(s)
- James E Trosko
- Department of Pediatrics/Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 048864, USA
| |
Collapse
|
52
|
Trosko JE. In Search of a Unifying Concept in Human Diseases. Diseases 2021; 9:68. [PMID: 34698126 PMCID: PMC8544458 DOI: 10.3390/diseases9040068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Throughout the history of biological/medicine sciences, there has been opposing strategies to find solutions to complex human disease problems. Both empirical and deductive approaches have led to major insights and concepts that have led to practical preventive and therapeutic benefits for the human population. The classic definitions of "science" (to know) has been paired with the classic definition of technology (to do). One knew more as the technology developed, and that development was often based on science. In other words, one could do more if science could improve the technology. In turn, this made possible to know more science with improved technology. However, with the development of new technologies of today in biology and medicine, major advances have been made, such as the information from the Human Genome Project, genetic engineering techniques and the use of bioinformatic uses of sophisticated computer analyses. This has led to the renewed idea that Precision Medicine, while raising some serious ethical concerns, also raises the expectation of improved potential of risk predictions for prevention and treatment of various genetically and environmentally influenced human diseases. This new field Artificial Intelligence, as a major handmaiden to Precision Medicine, is significantly altering the fundamental means of biological discovery. However, can today's fundamental premise of "Artificial Intelligence", based on identifying DNA, as the primary nexus of human health and disease, provide the practical solutions to complex human diseases that involve the interaction of those genes with the broad spectrum of "environmental factors"? Will it be "precise" enough to provide practical solutions for prevention and treatments of diseases? In this "Commentary", with the example of human carcinogenesis, it will be challenged that, without the integration of mechanistic and hypothesis-driven approaches with the "unbiased" empirical analyses of large numbers of data, the Artificial Intelligence approach with fall short.
Collapse
Affiliation(s)
- James Edward Trosko
- Department of Pediatrics/Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
53
|
Atkinson SP. A Preview of Selected Articles. Stem Cells 2021. [DOI: 10.1002/stem.3437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
54
|
Qin S, Mao Y, Wang H, Duan Y, Zhao L. The interplay between m6A modification and non-coding RNA in cancer stemness modulation: mechanisms, signaling pathways, and clinical implications. Int J Biol Sci 2021; 17:2718-2736. [PMID: 34345203 PMCID: PMC8326131 DOI: 10.7150/ijbs.60641] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer stemness, mainly consisting of chemo-resistance, radio-resistance, tumorigenesis, metastasis, tumor self-renewal, cancer metabolism reprogramming, and tumor immuno-microenvironment remodeling, play crucial roles in the cancer progression process and has become the hotspot of cancer research field in recent years. Nowadays, the exact molecular mechanisms of cancer stemness have not been fully understood. Extensive studies have recently implicated that non-coding RNA (ncRNA) plays vital roles in modulating cancer stemness. Notably, N6-methyladenosine (m6A) modification is of crucial importance for RNAs to exert their biological functions, including RNA splicing, stability, translation, degradation, and export. Emerging evidence has revealed that m6A modification can govern the expressions and functions of ncRNAs, consequently controlling cancer stemness properties. However, the interaction mechanisms between ncRNAs and m6A modification in cancer stemness modulation are rarely investigated. In this review, we elucidate the recent findings on the relationships of m6A modification, ncRNAs, and cancer stemness. We also focus on some key signaling pathways such as Wnt/β-catenin signaling, MAPK signaling, Hippo signaling, and JAK/STAT3 signaling to illustrate the underlying interplay mechanisms between m6A modification and ncRNAs in cancer stemness. In particular, we briefly highlight the clinical potential of ncRNAs and m6A modifiers as promising biomarkers and therapeutic targets for indicating cancer stemness properties and improving the diagnostic precision for a wide variety of cancers.
Collapse
Affiliation(s)
- Sha Qin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China; and Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yitao Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haofan Wang
- Department of Interventional Radiology, The 3rd Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yingxing Duan
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luqing Zhao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China; and Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
55
|
Islam Z, Ali AM, Naik A, Eldaw M, Decock J, Kolatkar PR. Transcription Factors: The Fulcrum Between Cell Development and Carcinogenesis. Front Oncol 2021; 11:681377. [PMID: 34195082 PMCID: PMC8236851 DOI: 10.3389/fonc.2021.681377] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022] Open
Abstract
Higher eukaryotic development is a complex and tightly regulated process, whereby transcription factors (TFs) play a key role in controlling the gene regulatory networks. Dysregulation of these regulatory networks has also been associated with carcinogenesis. Transcription factors are key enablers of cancer stemness, which support the maintenance and function of cancer stem cells that are believed to act as seeds for cancer initiation, progression and metastasis, and treatment resistance. One key area of research is to understand how these factors interact and collaborate to define cellular fate during embryogenesis as well as during tumor development. This review focuses on understanding the role of TFs in cell development and cancer. The molecular mechanisms of cell fate decision are of key importance in efforts towards developing better protocols for directed differentiation of cells in research and medicine. We also discuss the dysregulation of TFs and their role in cancer progression and metastasis, exploring TF networks as direct or indirect targets for therapeutic intervention, as well as specific TFs' potential as biomarkers for predicting and monitoring treatment responses.
Collapse
Affiliation(s)
- Zeyaul Islam
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Ameena Mohamed Ali
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Adviti Naik
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Mohamed Eldaw
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Julie Decock
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Prasanna R. Kolatkar
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| |
Collapse
|
56
|
Mohan A, Raj Rajan R, Mohan G, Kollenchery Puthenveettil P, Maliekal TT. Markers and Reporters to Reveal the Hierarchy in Heterogeneous Cancer Stem Cells. Front Cell Dev Biol 2021; 9:668851. [PMID: 34150761 PMCID: PMC8209516 DOI: 10.3389/fcell.2021.668851] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
A subpopulation within cancer, known as cancer stem cells (CSCs), regulates tumor initiation, chemoresistance, and metastasis. At a closer look, CSCs show functional heterogeneity and hierarchical organization. The present review is an attempt to assign marker profiles to define the functional heterogeneity and hierarchical organization of CSCs, based on a series of single-cell analyses. The evidences show that analogous to stem cell hierarchy, self-renewing Quiescent CSCs give rise to the Progenitor CSCs with limited proliferative capacity, and later to a Progenitor-like CSCs, which differentiates to Proliferating non-CSCs. Functionally, the CSCs can be tumor-initiating cells (TICs), drug-resistant CSCs, or metastasis initiating cells (MICs). Although there are certain marker profiles used to identify CSCs of different cancers, molecules like CD44, CD133, ALDH1A1, ABCG2, and pluripotency markers [Octamer binding transcriptional factor 4 (OCT4), SOX2, and NANOG] are used to mark CSCs of a wide range of cancers, ranging from hematological malignancies to solid tumors. Our analysis of the recent reports showed that a combination of these markers can demarcate the heterogeneous CSCs in solid tumors. Reporter constructs are widely used for easy identification and quantification of marker molecules. In this review, we discuss the suitability of reporters for the widely used CSC markers that can define the heterogeneous CSCs. Since the CSC-specific functions of CD44 and CD133 are regulated at the post-translational level, we do not recommend the reporters for these molecules for the detection of CSCs. A promoter-based reporter for ABCG2 may also be not relevant in CSCs, as the expression of the molecule in cancer is mainly regulated by promoter demethylation. In this context, a dual reporter consisting of one of the pluripotency markers and ALDH1A1 will be useful in marking the heterogeneous CSCs. This system can be easily adapted to high-throughput platforms to screen drugs for eliminating CSCs.
Collapse
Affiliation(s)
- Amrutha Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Manipal Academy of Higher Education, Manipal, India
| | - Reshma Raj Rajan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Gayathri Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | | |
Collapse
|
57
|
Trosko JE. On the potential origin and characteristics of cancer stem cells. Carcinogenesis 2021; 42:905-912. [PMID: 34014276 DOI: 10.1093/carcin/bgab042] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/08/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
The 'cancer stem cell' hypothesis has pointed to a specific target for new cancer therapies. The hypothesis is based on the observation that only the 'cancer stem cell' among the other heterogeneous cancer cells can sustain the growth of the cancer. The goal is to identify biomarkers of 'cancer stem cells' to distinguish them from the 'cancer non-stem cells' and normal adult tissue-specific stem cells. This analyst posits a hypothesis that, although all cancers originated from a single cell, there exist two types of 'cancer stem cells' either by the 'Stem Cell hypothesis' or from the 'De-differentiation hypothesis'. It is proposed that there exist two different 'cancer stem cells'. Some 'cancer stem cells' (a) lack the expression of connexins or gap junction genes and lack any form of gap junctional intercellular communication (GJIC) or (b) they have the expressed connexin-coded proteins for functional GJIC but are dysfunctional by some expressed oncogene. This is consistent with the Loewenstein hypothesis that a universal characteristic of cancer cells is they do not have growth control, nor terminally differentiate. This review speculates the normal organ-specific adult stem cell, that is 'initiated', is the origin of the 'cancer stem cells' with expressed Oct4A gene and no expressed connexin genes; whereas the other cancer stem cell has no expressed Oct4A genes but expressed connexin gene, whose coded protein is dysfunctional. Hence. both types of 'cancer stem cells' lack GJIC, for two different reasons, the selective therapies have to be different for these different cell types.
Collapse
Affiliation(s)
- James E Trosko
- Department of Pediatrics/Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
58
|
Le P, Romano G, Nana-Sinkam P, Acunzo M. Non-Coding RNAs in Cancer Diagnosis and Therapy: Focus on Lung Cancer. Cancers (Basel) 2021; 13:cancers13061372. [PMID: 33803619 PMCID: PMC8003033 DOI: 10.3390/cancers13061372] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Over the last several decades, clinical evaluation and treatment of lung cancers have largely improved with the classification of genetic drivers of the disease, such as EGFR, ALK, and ROS1. There are numerous regulatory factors that exert cellular control over key oncogenic pathways involved in lung cancers. In particular, non-coding RNAs (ncRNAs) have a diversity of regulatory roles in lung cancers such that they have been shown to be involved in inducing proliferation, suppressing apoptotic pathways, increasing metastatic potential of cancer cells, and acquiring drug resistance. The dysregulation of various ncRNAs in human cancers has prompted preclinical studies examining the therapeutic potential of restoring and/or inhibiting these ncRNAs. Furthermore, ncRNAs demonstrate tissue-specific expression in addition to high stability within biological fluids. This makes them excellent candidates as cancer biomarkers. This review aims to discuss the relevance of ncRNAs in cancer pathology, diagnosis, and therapy, with a focus on lung cancer.
Collapse
|
59
|
Maiuthed A, Prakhongcheep O, Chanvorachote P. Microarray-based Analysis of Genes, Transcription Factors, and Epigenetic Modifications in Lung Cancer Exposed to Nitric Oxide. Cancer Genomics Proteomics 2021; 17:401-415. [PMID: 32576585 DOI: 10.21873/cgp.20199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIM Nitric oxide (NO) is recognized as an important biological mediator that exerts several human physiological functions. As its nature is an aqueous soluble gas that can diffuse through cells and tissues, NO can affect cell signaling, the phenotype of cancer and modify surrounding cells. The variety of effects of NO on cancer cell biology has convinced researchers to determine the defined mechanisms of these effects and how to control this mediator for a better understanding as well as for therapeutic gain. MATERIALS AND METHODS We used bioinformatics and pharmacological experiments to elucidate the potential regulation and underlying mechanisms of NO in non-small a lung cancer cell model. RESULTS Using microarrays, we identified a total of 151 NO-regulated genes (80 up-regulated genes, 71 down-regulated genes) with a strong statistically significant difference compared to untreated controls. Among these, the genes activated by a factor of more than five times were: DCBLD2, MGC24975, RAB40AL, PER3, RCN1, MRPL51, PTTG1, KLF5, NFIX. On the other hand, the expression of RBMS2, PDP2, RBAK, ORMDL2, GRPEL2, ZNF514, MTHFD2, POLR2D, RCBTB1, JOSD1, RPS27, GPR4 genes were significantly decreased by a factor of more than five times. Bioinformatics further revealed that NO exposure of lung cancer cells resulted in a change in transcription factors (TFs) and epigenetic modifications (histone modification and miRNA). Interestingly, NO treatment was shown to potentiate cancer stem cell-related genes and transcription factors Oct4, Klf4, and Myc. CONCLUSION Through this comprehensive approach, the present study illustrated the scheme of how NO affects molecular events in lung cancer cells.
Collapse
Affiliation(s)
- Arnatchai Maiuthed
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Ornjira Prakhongcheep
- Cell-based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.,Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Pithi Chanvorachote
- Cell-based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand .,Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
60
|
Zhang Q, Han Z, Zhu Y, Chen J, Li W. The Role and Specific Mechanism of OCT4 in Cancer Stem Cells: A Review. Int J Stem Cells 2020; 13:312-325. [PMID: 32840233 PMCID: PMC7691851 DOI: 10.15283/ijsc20097] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Recently, evidences show that cancer stem cells (CSCs) are a type of cancer cell group with self-renewal and play a huge role in tumor recurrence, metastasis, and drug resistance. Finding new treatment directions and targets for cancer prognosis and reducing mortality has become a top priority. OCT4, as a transcription factor, participates in maintaining the stem characteristics of CSCs, but the mechanism of OCT4 is often overlooked. In this review, we try to illustrate the mechanism by which OCT4 plays a role in CSCs from the perspective of genetic modification of OCT4, non-coding RNA, complexes and signaling pathways associated with OCT4. Our ultimate goal is to provide new targets for cancer treatment to prolong the survival of cancer patients.
Collapse
Affiliation(s)
- Qi Zhang
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zhenzhen Han
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yanbo Zhu
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jingcheng Chen
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Wei Li
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
61
|
Embryonic stem cell-like subpopulations are present within Schwannoma. J Clin Neurosci 2020; 81:201-209. [PMID: 33222917 DOI: 10.1016/j.jocn.2020.09.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/10/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND There is accumulating evidence of the presence of embryonic stem cell (ESC)-like cells in benign tumors. AIM This study aimed to identify ESC-like cells in Schwannoma using the induced-pluripotent stem cell (iPSC) markers OCT4, SOX2, NANOG, KLF4 and c-MYC. METHODS Immunohistochemical (IHC) staining (n = 20) and RT-qPCR (n = 6) were performed on Schwannoma tissue samples (STS) to investigate protein and mRNA expression of these iPSC markers, respectively. Immunofluorescence (IF) staining was performed to investigate co-localization of the iPSC markers with CD34, α-SMA and CD133. RESULTS IHC staining and RT-qPCR demonstrated protein and mRNA expression of all five iPSC markers, respectively. IF staining showed expression of SOX2, KLF4 and c-MYC on the tumor cells and the endothelium of the tumor microvessels which also expressed OCT4, while NANOG was exclusively expressed on the endothelium of the tumor microvessels. The OCT4+/CD34+ endothelium expressed CD133. CONCLUSION We have identified a putative OCT4+/SOX2+/NANOG+/KLF4+/c-MYC+/CD133+ ESC-like subpopulation on the endothelium of tumor microvessels and an OCT4-/SOX2+/NANOG-/KLF4+/c-MYC+/CD133+ ESC-like subpopulation, within Schwannoma.
Collapse
|
62
|
Ford E, Pearlman J, Ruan T, Manion J, Waller M, Neely GG, Caron L. Human Pluripotent Stem Cells-Based Therapies for Neurodegenerative Diseases: Current Status and Challenges. Cells 2020; 9:E2517. [PMID: 33233861 PMCID: PMC7699962 DOI: 10.3390/cells9112517] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases are characterized by irreversible cell damage, loss of neuronal cells and limited regeneration potential of the adult nervous system. Pluripotent stem cells are capable of differentiating into the multitude of cell types that compose the central and peripheral nervous systems and so have become the major focus of cell replacement therapies for the treatment of neurological disorders. Human embryonic stem cell (hESC) and human induced pluripotent stem cell (hiPSC)-derived cells have both been extensively studied as cell therapies in a wide range of neurodegenerative disease models in rodents and non-human primates, including Parkinson's disease, stroke, epilepsy, spinal cord injury, Alzheimer's disease, multiple sclerosis and pain. In this review, we discuss the latest progress made with stem cell therapies targeting these pathologies. We also evaluate the challenges in clinical application of human pluripotent stem cell (hPSC)-based therapies including risk of oncogenesis and tumor formation, immune rejection and difficulty in regeneration of the heterogeneous cell types composing the central nervous system.
Collapse
Affiliation(s)
- Elizabeth Ford
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Jodie Pearlman
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Travis Ruan
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| | - John Manion
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA
- Departments of Surgery and Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew Waller
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| | - Gregory G. Neely
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| | - Leslie Caron
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, University of Sydney, Camperdown, NSW 2006, Australia; (E.F.); (J.P.); (T.R.); (J.M.); (M.W.); (G.G.N.)
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
63
|
Fang W, Ni M, Zhang M, Chen H. Prognostic value of OCT4 in colorectal cancer: analysis using immunohistochemistry and bioinformatics validation. Biomark Med 2020; 14:1473-1484. [PMID: 33185466 DOI: 10.2217/bmm-2020-0069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aim: This study was first performed to investigate the role of octamer-binding transcription factor 4 (OCT4) in colorectal cancer (CRC). Methods: The electronic databases were searched for the eligible studies. Odds ratios and hazard ratios were calculated. Functional analysis of OCT4 was examined. Results: Eight studies with 1480 CRC cases were identified. OCT4 expression was correlated with advanced clinical stage, tumor grade, lymph node metastasis, lymphatic invasion, and distal metastasis. OCT4 was an independent prognostic biomarker for predicting worse disease-specific survival and overall survival in CRC. The functional analyses demonstrated that OCT4 was involved in multiple functions, such as cell adhesion, phosphoinositide 3-kinase/Akt signaling, and regulating pluripotency of stem cells. Conclusion: OCT4 may be correlated with disease progression and metastasis, and could predict prognosis in CRC.
Collapse
Affiliation(s)
- Wenjia Fang
- Department of Gastroenterology, Ningbo Yinzhou no. 2 Hospital, Ningbo, Zhejiang 315100, China
| | - Meilin Ni
- Ningbo Customs District Technology Center, Ningbo, Zhejiang 315000, China
| | - Mingming Zhang
- Department of Gastroenterology, Ningbo Yinzhou no. 2 Hospital, Ningbo, Zhejiang 315100, China
| | - Hanqing Chen
- Department of Gastroenterology, Ningbo Yinzhou no. 2 Hospital, Ningbo, Zhejiang 315100, China
| |
Collapse
|
64
|
Wei-Hua W, Ning Z, Qian C, Dao-Wen J. ZIC2 promotes cancer stem cell traits via up-regulating OCT4 expression in lung adenocarcinoma cells. J Cancer 2020; 11:6070-6080. [PMID: 32922547 PMCID: PMC7477430 DOI: 10.7150/jca.44367] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Accumulating evidence has revealed the importance of cancer stem cells (CSCs) in self-renewal and chemoresistance. Previous studies reported high expression of ZIC2 was closely associated with tumorigenesis and CSC traits. However, the role of ZIC2 as a crucial factor for regulating CSC properties in lung adenocarcinoma (LAC) remains elusive. Methods: RT-PCR and WB assay were employed to assess ZIC2 expression in 20 LAC tumor tissues and the matched non-cancerous tissues. The role of ZIC2 in LAC CSC were analyzed by evaluation of CSC-related markers expression and spheroid formation in vitro. Cisplatin and paclitaxel resistance capacities were evaluated by CCK8 assay, colony formation assay, and flow cytometry analysis. Subcutaneous NOD/SCID mice models were generated to assess in vivo CSC features. Results: High expression of ZIC2 was found in LAC tumor tissues and indicated a poor overall survival in LAC patients. ZIC2 upregulated an array of CSCs-related genes, including EpCAM, OCT4, SOX2, NANOG, C-Myc and Bmi-1. Knockdown of ZIC2 inhibited sphere-forming capacity and decreased cisplatin and paclitaxel resistance. However, overexpression of ZIC2 achieved opposite effects. Mechanically, ZIC2 acts upstream of OCT4 to promote its expression, resulting in enhancement of CSC traits in LAC. Conclusion: Our results demonstrated that ZIC2 was crucial for promoting CSC traits in LAC cells, and served as a potential biomarker for predicting prognosis. The ZIC2-OCT4 network will facilitate the evaluation of the potential therapeutic efficacy of chemotherapy and predict patient sensitivity to treatment.
Collapse
Affiliation(s)
- Wang Wei-Hua
- Department of thoracic surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Zhou Ning
- Department of thoracic surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Chen Qian
- Department of general surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Jiang Dao-Wen
- Department of thoracic surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| |
Collapse
|
65
|
Kilmister EJ, Patel J, van Schaijik B, Bockett N, Brasch HD, Paterson E, Sim D, Davis PF, Roth IM, Itinteang T, Tan ST. Cancer Stem Cell Subpopulations Are Present Within Metastatic Head and Neck Cutaneous Squamous Cell Carcinoma. Front Oncol 2020; 10:1091. [PMID: 32850316 PMCID: PMC7406827 DOI: 10.3389/fonc.2020.01091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) have been identified in many cancer types including primary head and neck cutaneous squamous cell carcinoma (HNcSCC). This study aimed to identify and characterize CSCs in metastatic HNcSCC (mHNcSCC). Immunohistochemical staining performed on mHNcSCC samples from 15 patients demonstrated expression of the induced pluripotent stem cell (iPSC) markers OCT4, SOX2, NANOG, KLF4, and c-MYC in all 15 samples. In situ hybridization and RT-qPCR performed on four of these mHNcSCC tissue samples confirmed transcript expression of all five iPSC markers. Immunofluorescence staining performed on three of these mHNcSCC samples demonstrated expression of c-MYC on cells within the tumor nests (TNs) and the peri-tumoral stroma (PTS) that also expressed KLF4. OCT4 was expressed on the SOX2+/NANOG+/KLF4+ cells within the TNs, and the SOX2+/NANOG+/KLF4+ cells within the PTS. RT-qPCR demonstrated transcript expression of all five iPSC markers in all three mHNcSCC-derived primary cell lines, except for SOX2 in one cell line. Western blotting showed the presence of SOX2, KLF4, and c-MYC but not OCT4 and NANOG in the three mHNcSCC-derived primary cell lines. All three cell lines formed tumorspheres, at the first passage. We demonstrated an OCT4+/NANOG+/SOX2+/KLF4+/c-MYC+ CSC subpopulation and an OCT4+/NANOG-/SOX2+/KLF4+/c-MYC+ subpopulation within the TNs, and an OCT4+/NANOG+/SOX2+/KLF4+/c-MYC+ subpopulation within the PTS of mHNcSCC.
Collapse
Affiliation(s)
| | - Josie Patel
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | | | | | - Helen D Brasch
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Erin Paterson
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Dalice Sim
- Biostatistical Group/Dean's Department, University of Otago, Wellington, New Zealand
| | - Paul F Davis
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Imogen M Roth
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | | | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand.,Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Wellington, New Zealand.,Department of Surgery, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
66
|
Huang T, Song X, Xu D, Tiek D, Goenka A, Wu B, Sastry N, Hu B, Cheng SY. Stem cell programs in cancer initiation, progression, and therapy resistance. Am J Cancer Res 2020; 10:8721-8743. [PMID: 32754274 PMCID: PMC7392012 DOI: 10.7150/thno.41648] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Over the past few decades, substantial evidence has convincingly revealed the existence of cancer stem cells (CSCs) as a minor subpopulation in cancers, contributing to an aberrantly high degree of cellular heterogeneity within the tumor. CSCs are functionally defined by their abilities of self-renewal and differentiation, often in response to cues from their microenvironment. Biological phenotypes of CSCs are regulated by the integrated transcriptional, post-transcriptional, metabolic, and epigenetic regulatory networks. CSCs contribute to tumor progression, therapeutic resistance, and disease recurrence through their sustained proliferation, invasion into normal tissue, promotion of angiogenesis, evasion of the immune system, and resistance to conventional anticancer therapies. Therefore, elucidation of the molecular mechanisms that drive cancer stem cell maintenance, plasticity, and therapeutic resistance will enhance our ability to improve the effectiveness of targeted therapies for CSCs. In this review, we highlight the key features and mechanisms that regulate CSC function in tumor initiation, progression, and therapy resistance. We discuss factors for CSC therapeutic resistance, such as quiescence, induction of epithelial-to-mesenchymal transition (EMT), and resistance to DNA damage-induced cell death. We evaluate therapeutic approaches for eliminating therapy-resistant CSC subpopulations, including anticancer drugs that target key CSC signaling pathways and cell surface markers, viral therapies, the awakening of quiescent CSCs, and immunotherapy. We also assess the impact of new technologies, such as single-cell sequencing and CRISPR-Cas9 screening, on the investigation of the biological properties of CSCs. Moreover, challenges remain to be addressed in the coming years, including experimental approaches for investigating CSCs and obstacles in therapeutic targeting of CSCs.
Collapse
|
67
|
Bodhankar K, Bansal S, Jashnani K, Desai RS. Immunohistochemical expression of stem cell markers OCT-4 and SOX-2 in giant cell tumor, central giant cell granuloma, and peripheral giant cell granuloma. Oral Surg Oral Med Oral Pathol Oral Radiol 2020; 130:78-84. [PMID: 32493681 DOI: 10.1016/j.oooo.2020.03.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/25/2020] [Accepted: 03/31/2020] [Indexed: 10/24/2022]
Abstract
OBJECTIVES This study aimed to evaluate and compare the immunohistochemical expression of OCT-4 and SOX-2 and to determine their use in differentiating giant cell tumor (GCT) from central giant cell granuloma (CGCG) and peripheral giant cell granuloma (PGCG). STUDY DESIGN Formalin-fixed, paraffin-embedded tissue blocks of 10 histopathologically diagnosed cases of GCT, CGCG, or PGCG were examined for anti-OCT-4 and anti-SOX-2 antibodies. Nuclear staining of stromal mononuclear cells and multinucleated giant cells was considered positive for OCT-4 and SOX-2 expression. RESULTS Nuclear immunoexpression of OCT-4 in stromal mononuclear cells was observed in 80% (8 of 10) of GCT cases, whereas none of the CGCG and PGCG cases showed OCT-4 immunoreactivity. SOX-2 immunoreactivity was negative in GCT, CGCG, and PGCG. CONCLUSIONS OCT-4 immunopositivity in GCT can be used as a cancer stem cell marker to differentiate GCT from CGCG and PGCG. The presence of OCT-4 in GCT versus its complete absence in CGCG and PGCG suggests that these three conditions are separate entities. The absence of stem cell marker OCT-4 and SOX-2 raises questions regarding their role in the pathogenesis of CGCG and PGCG.
Collapse
Affiliation(s)
- Kshitija Bodhankar
- Post-graduate Student, Department of Oral Pathology and Microbiology, Nair Hospital Dental College, Mumbai, India
| | - Shivani Bansal
- Professor (Additional), Department of Oral Pathology and Microbiology, Nair Hospital Dental College, Mumbai, India.
| | - Kusum Jashnani
- Professor and Head, Department of Pathology, BYL Nair Charitable Hospital and T.N Medical College, Mumbai, India
| | - Rajiv S Desai
- Professor and Head, Department of Oral Pathology and Microbiology, Nair Hospital Dental College, Mumbai, India
| |
Collapse
|
68
|
Swain N, Thakur M, Pathak J, Swain B. SOX2, OCT4 and NANOG: The core embryonic stem cell pluripotency regulators in oral carcinogenesis. J Oral Maxillofac Pathol 2020; 24:368-373. [PMID: 33456249 PMCID: PMC7802841 DOI: 10.4103/jomfp.jomfp_22_20] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Embryonic stem cells provide their major contribution to embryogenesis through formation of germ layers as they have pluripotency potential and capacity for self-renewal. Retention of pluripotency of these stem cells depends on expression/level of transcription factors, i.e., SOX2, OCT4 and NANOG. During organogenesis, the altered expression of the molecules also influences these stem cells to lose their pluripotency and turn toward the lineage selection. As the differentiation progresses, the maintenance of the somatic cells including the oral squamous cells also depends on differential expression of the transcription factors to some extent. Recently, many experimental and observational studies documented the significant contribution in carcinogenesis of various human cancers. In this review, we have attempted to summarize the evidences indicating about the putative role of these master pluripotency regulators in various phases of oral carcinogenesis i.e. initiation , progression and prognosis of oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Niharika Swain
- Department of Oral Pathology, MGM Dental College and Hospital, MGM Institute of Health Sciences, Navi Mumbai, Maharashtra, India
| | - Mansee Thakur
- Department of Medical Biotechnology, MGM School of Biomedical Sciences, MGM Institute of Health Sciences, Navi Mumbai, Maharashtra, India
| | - Jigna Pathak
- Department of Oral Pathology, MGM Dental College and Hospital, MGM Institute of Health Sciences, Navi Mumbai, Maharashtra, India
| | - Biswaranjan Swain
- Department of Electronics and Communications Engineering, Institute of Technical Education and Research, S’O’A Deemed to be University, Bhubaneswar, Odisha, India
| |
Collapse
|
69
|
The effect of Candida cell wall beta-glucan on treatment-resistant LL/2 cancer cell line: in vitro evaluation. Mol Biol Rep 2020; 47:3653-3661. [PMID: 32323263 DOI: 10.1007/s11033-020-05459-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/11/2020] [Indexed: 01/06/2023]
Abstract
Candida albicans (C. albicans) cell wall beta-glucan has been considered as a potential agent in the treatment of cancers due to its anti-tumor properties. Therefore, in the present study, we investigated the anti-cancer effects of Candida cell wall beta-glucan on Lewis lung carcinoma cell line (LL/2) cells. Beta-glucan of C. albicans cell wall was extracted. LL/2 cell line was cultured, then sphere cells and parental cells were exposed to the different concentrations of beta-glucan extracted from C. albicans (10-6000 μg/ml), for 24, 48 and 72 h. Cytotoxicity of beta-glucan was assayed by MTT test, then RNA extracted from cells population (treated and untreated cells), cDNA synthetized and expression level of Sox2, Oct4, C-myc, Nanog genes were also investigated using Real-time methods. At optimal concentrations of 800 and 1000 μg/ml, the extracted beta-glucan showed a significant cytotoxic effect on both parental and sphere cell populations (p < 0.05). Real-time PCR analysis revealed a decreased expression of Oct4 and Sox2 genes in treatment of cells with beta-glucan compared with control group. Since the extracted beta-glucan showed an inhibitory effect on the expression of Oct4 and Sox2 genes involved in LL/2 metastasis, therefore, beta-glucan can be considered as an anti-tumor agent because of its anti-metastatic properties, however, more in vitro and in vivo studies are needed to provide further evidence on this topic in the future.
Collapse
|
70
|
Cellular Functions of OCT-3/4 Regulated by Ubiquitination in Proliferating Cells. Cancers (Basel) 2020; 12:cancers12030663. [PMID: 32178477 PMCID: PMC7139964 DOI: 10.3390/cancers12030663] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/18/2022] Open
Abstract
Octamer-binding transcription factor 3/4 (OCT-3/4), which is involved in the tumorigenesis of somatic cancers, has diverse functions during cancer development. Overexpression of OCT-3/4 has been detected in various human somatic tumors, indicating that OCT-3/4 activation may contribute to the development and progression of cancers. Stem cells can undergo self-renewal, pluripotency, and reprogramming with the help of at least four transcription factors, OCT-3/4, SRY box-containing gene 2 (SOX2), Krüppel-like factor 4 (KLF4), and c-MYC. Of these, OCT-3/4 plays a critical role in maintenance of undifferentiated state of embryonic stem cells (ESCs) and in production of induced pluripotent stem cells (iPSCs). Stem cells can undergo partitioning through mitosis and separate into specific cell types, three embryonic germ layers: the endoderm, the mesoderm, and the trophectoderm. It has been demonstrated that the stability of OCT-3/4 is mediated by the ubiquitin-proteasome system (UPS), which is one of the key cellular mechanisms for cellular homeostasis. The framework of the mechanism is simple, but the proteolytic machinery is complicated. Ubiquitination promotes protein degradation, and ubiquitination of OCT-3/4 leads to regulation of cellular proliferation and differentiation. Therefore, it is expected that OCT-3/4 may play a key role in proliferation and differentiation of proliferating cells.
Collapse
|
71
|
Cosio T, Mazzilli S, Bianchi L, Campione E. The Dark Side of Gefitinib: Reflectance Confocal Microscopy Applied to Hair Hyperpigmentation. Skin Appendage Disord 2020; 6:44-47. [PMID: 32021862 DOI: 10.1159/000503758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 09/26/2019] [Indexed: 11/19/2022] Open
Abstract
Gefitinib is a multi-target tyrosine kinase inhibitor used for the treatment of non-small cell lung cancer. Papulo-pustular and/or paronychia, abnormalities in hair growth, itching, and dryness due to epidermal growth factor inhibitors - i.e., PRIDE Complex - are a common effect of tyrosine kinase inhibitors. We report a case of hair and eyebrow hyperpigmentation after 7 months of treatment with gefitinib. In the literature, we found no data regarding rapid pigmentation of hair due to treatment with any multi-target tyrosine kinase inhibitors. To our knowledge, this is the first case reporting both hair and eyebrow hyperpigmentation. We hypothesize the role of different mechanisms linked to rapid hair hyperpigmentation.
Collapse
Affiliation(s)
- Terenzio Cosio
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sara Mazzilli
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luca Bianchi
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Elena Campione
- Dermatologic Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
72
|
Shi YB, Li J, Lai XN, Jiang R, Zhao RC, Xiong LX. Multifaceted Roles of Caveolin-1 in Lung Cancer: A New Investigation Focused on Tumor Occurrence, Development and Therapy. Cancers (Basel) 2020; 12:cancers12020291. [PMID: 31991790 PMCID: PMC7073165 DOI: 10.3390/cancers12020291] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/13/2020] [Accepted: 01/22/2020] [Indexed: 12/26/2022] Open
Abstract
Lung cancer is one of the most common and malignant cancers with extremely high morbidity and mortality in both males and females. Although traditional lung cancer treatments are fast progressing, there are still limitations. Caveolin-1 (Cav-1), a main component of caveolae, participates in multiple cellular events such as immune responses, endocytosis, membrane trafficking, cellular signaling and cancer progression. It has been found tightly associated with lung cancer cell proliferation, migration, apoptosis resistance and drug resistance. In addition to this, multiple bioactive molecules have been confirmed to target Cav-1 to carry on their anti-tumor functions in lung cancers. Cav-1 can also be a predictor for lung cancer patients’ prognosis. In this review, we have summarized the valuable research on Cav-1 and lung cancer in recent years and discussed the multifaceted roles of Cav-1 on lung cancer occurrence, development and therapy, hoping to provide new insights into lung cancer treatment.
Collapse
Affiliation(s)
- Yu-Bo Shi
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Y.-B.S.); (J.L.); (X.-N.L.); (R.-C.Z.)
- Queen Mary School, Jiangxi Medical College of Nanchang University, Nanchang 330006, China;
| | - Jun Li
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Y.-B.S.); (J.L.); (X.-N.L.); (R.-C.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Xing-Ning Lai
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Y.-B.S.); (J.L.); (X.-N.L.); (R.-C.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Rui Jiang
- Queen Mary School, Jiangxi Medical College of Nanchang University, Nanchang 330006, China;
| | - Rui-Chen Zhao
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Y.-B.S.); (J.L.); (X.-N.L.); (R.-C.Z.)
- Queen Mary School, Jiangxi Medical College of Nanchang University, Nanchang 330006, China;
| | - Li-Xia Xiong
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Y.-B.S.); (J.L.); (X.-N.L.); (R.-C.Z.)
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China
- Correspondence: ; Tel.: +86-791-8636-0556
| |
Collapse
|
73
|
Zhao B, Zheng X, Tan X, Ke K, Wang F, Wang Y, Xing X, Zhang C, Hu P, Lan S, Li Q, Huang A, Liu X. Ku80 negatively regulates the expression of OCT4 via competitive binding to SALL4 and promoting lysosomal degradation of OCT4. Int J Biochem Cell Biol 2020; 118:105664. [PMID: 31816404 DOI: 10.1016/j.biocel.2019.105664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/29/2019] [Accepted: 12/05/2019] [Indexed: 10/25/2022]
Abstract
SALL4 and OCT4, along with other pluripotency-associated transcription factors, play critical roles in maintaining embryonic stem cell pluripotency and self-renewal. Ku80 is a component of the protein complex called DNA-dependent protein kinase, which mainly involved in DNA double-strand break repair. In this study, we show evidence that Ku80 physically interacted with SALL4. The interaction competitively disrupts the SALL4-OCT4 complex and result in OCT4 lysosomal degradation. Finally, Ku80 inhibits self-renewal and metastasis of hepatocellular carcinoma cells through breaking the SALL4-OCT4 interactions and down-regulating the expression of OCT4. Our study reveal novel function of Ku80 in stemness maintaining of cancer stem cells via its interaction with SALL4 and highlight the double-sidedness of Ku80 as an anti-cancer target.
Collapse
Affiliation(s)
- Bixing Zhao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Xiaoyuan Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Xionghong Tan
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Kun Ke
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Fei Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Xiaohua Xing
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Cuilin Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Ping Hu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350108, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Shubing Lan
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Qin Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China
| | - Aimin Huang
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350108, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China.
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, PR China; Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, PR China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, PR China.
| |
Collapse
|
74
|
Du R, Shen W, Liu Y, Gao W, Zhou W, Li J, Zhao S, Chen C, Chen Y, Liu Y, Sun P, Xiang R, Shi Y, Luo Y. TGIF2 promotes the progression of lung adenocarcinoma by bridging EGFR/RAS/ERK signaling to cancer cell stemness. Signal Transduct Target Ther 2019; 4:60. [PMID: 31871777 PMCID: PMC6908606 DOI: 10.1038/s41392-019-0098-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
TGF-β-induced factor homeobox 2 (TGIF2) is a transcription regulator that plays essential roles in the regulation of development and cell fate decisions. Aberrant expression of TGIF family proteins has been observed in several cancers, including ovarian, esophageal, and colorectal cancers. Here, we report that TGIF2 mediates the EGFR-RAS-ERK signaling pathway to enhance the stemness of lung adenocarcinoma (LUAD) cells and, therefore, promote the progression and metastasis of LUAD. We found that high TGIF2 expression was closely correlated with tumor growth, lymph node metastasis, and survival of patients with LUAD. Mice bearing TGIF2-silenced H1299 xenografts developed smaller tumors and fewer lung metastases. Importantly, silencing TGIF2 decreased the cancer stem cell (CSC)-like properties in A549 and H1299 cells. Furthermore, we identified that TGIF2 binding to the OCT4 promoter promotes its expression. In both LUAD cells and in vivo LUAD mouse models, we revealed that EGFR-RAS-ERK signaling phosphorylated TGIF2 and increased its stability, which was important for TGIF2-promoted LUAD stemness since phosphorylation-deficient TGIF2 mutants lost these functions. Thus, our study revealed that an important factor, TGIF2, bridges EGFR signaling to the CSC characteristics of LUAD cells, which can be utilized as an effective target for LUAD therapy.
Collapse
Affiliation(s)
- Renle Du
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Wenzhi Shen
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining, 272067 China
| | - Yi Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Wenjuan Gao
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Wei Zhou
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Jun Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Shuangtao Zhao
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Chong Chen
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science, School of Basic Medicine, Peking Union Medical College, Beijing, 100005 China
| | - Yanan Chen
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin, 300071 China
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Tianjin, 300071 China
| | - Yanhua Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin, 300071 China
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Tianjin, 300071 China
| | - Peiqing Sun
- Department of Cancer Biology, School of Medicine, Wake Forest University, Winston-Salem, NC 27157 USA
| | - Rong Xiang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin, 300071 China
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Tianjin, 300071 China
| | - Yi Shi
- Department of Immunology, School of Medicine, Nankai University, Tianjin, 300071 China
- 2011 Project Collaborative Innovation Center for Biotherapy of Ministry of Education, Tianjin, 300071 China
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Tianjin, 300071 China
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science, School of Basic Medicine, Peking Union Medical College, Beijing, 100005 China
| |
Collapse
|
75
|
Gzil A, Zarębska I, Bursiewicz W, Antosik P, Grzanka D, Szylberg Ł. Markers of pancreatic cancer stem cells and their clinical and therapeutic implications. Mol Biol Rep 2019; 46:6629-6645. [PMID: 31486978 DOI: 10.1007/s11033-019-05058-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/31/2019] [Indexed: 12/17/2022]
Abstract
Pancreatic cancer (PC) is the fourth most common cause of death among all cancers. Poor prognosis of PC may be caused by a prevalence of cancer stem cells (CSCs). CSCs are a population of cancer cells showing stem cell-like characteristics. CSCs have the ability to self-renew and may initiate tumorigenesis. PC CSCs express markers such as CD133, CD24, CD44, DCLK1, CXCR4, ESA, Oct4 and ABCB1. There is a wide complexity of interaction and relationships between CSC markers in PC. These markers are negative prognostic factors and are connected with tumor recurrence and clinical progression. Additionally, PC CSCs are resistant to treatment with gemcitabine. Thus, most current therapies for PC are ineffective. Numerous studies have shown, that targeting of these proteins may increase both disease-free and overall survival in PC.
Collapse
Affiliation(s)
- Arkadiusz Gzil
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Sklodowskiej-Curie Str. 9, 85-094, Bydgoszcz, Poland.
| | - Izabela Zarębska
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Sklodowskiej-Curie Str. 9, 85-094, Bydgoszcz, Poland
| | - Wiktor Bursiewicz
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Sklodowskiej-Curie Str. 9, 85-094, Bydgoszcz, Poland
| | - Paulina Antosik
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Sklodowskiej-Curie Str. 9, 85-094, Bydgoszcz, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Sklodowskiej-Curie Str. 9, 85-094, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Sklodowskiej-Curie Str. 9, 85-094, Bydgoszcz, Poland
- Department of Pathomorphology, Military Clinical Hospital, Bydgoszcz, Poland
| |
Collapse
|
76
|
Srivastava Y, Tan DS, Malik V, Weng M, Javed A, Cojocaru V, Wu G, Veerapandian V, Cheung LWT, Jauch R. Cancer-associated missense mutations enhance the pluripotency reprogramming activity of OCT4 and SOX17. FEBS J 2019; 287:122-144. [PMID: 31569299 DOI: 10.1111/febs.15076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/26/2019] [Accepted: 09/29/2019] [Indexed: 12/21/2022]
Abstract
The functional consequences of cancer-associated missense mutations are unclear for the majority of proteins. We have previously demonstrated that the activity of SOX and Pit-Oct-Unc (POU) family factors during pluripotency reprogramming can be switched and enhanced with rationally placed point mutations. Here, we interrogated cancer mutation databases and identified recurrently mutated positions at critical structural interfaces of the DNA-binding domains of paralogous SOX and POU family transcription factors. Using the conversion of mouse embryonic fibroblasts to induced pluripotent stem cells as functional readout, we identified several gain-of-function mutations that enhance pluripotency reprogramming by SOX2 and OCT4. Wild-type SOX17 cannot support reprogramming but the recurrent missense mutation SOX17-V118M is capable of inducing pluripotency. Furthermore, SOX17-V118M promotes oncogenic transformation, enhances thermostability and elevates cellular protein levels of SOX17. We conclude that the mutational profile of SOX and POU family factors in cancer can guide the design of high-performance reprogramming factors. Furthermore, we propose cellular reprogramming as a suitable assay to study the functional impact of cancer-associated mutations.
Collapse
Affiliation(s)
- Yogesh Srivastava
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Medical University, China.,Genome Regulation Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Daisylyn Senna Tan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Vikas Malik
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Medical University, China.,Genome Regulation Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mingxi Weng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Asif Javed
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Vlad Cojocaru
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Guangming Wu
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Veeramohan Veerapandian
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Medical University, China.,Genome Regulation Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lydia W T Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ralf Jauch
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Medical University, China.,Genome Regulation Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
77
|
Siddiqui Z, Srivastava AN, Sankhwar SN, Zaidi N, Fatima N, Singh S, Yusuf M. Oct-4: a prognostic biomarker of urinary bladder cancer in North India. Ther Adv Urol 2019; 11:1756287219875576. [PMID: 31632462 PMCID: PMC6769204 DOI: 10.1177/1756287219875576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/19/2019] [Indexed: 12/29/2022] Open
Abstract
Background The objective of this study was to evaluate Octamer-binding transcription factor 4 (Oct-4), neutrophil to lymphocyte ratio (NLR) and body mass index (BMI) as independent prognostic biomarkers for prediction of urinary bladder cancer (UBC) outcomes. With the advancement in prognostic biomarker discovery, tumor recurrence is difficult to accurately predict in UBC. UBC is costly to treat due to the requirement of frequent invasive follow-up sessions. Therefore, it is of utmost importance to evaluate good prognostic biomarkers for UBC surveillance. Methods We studied 39 UBC tissue samples. Oct-4 protein expression was evaluated semiquantitatively by immunohistochemistry (IHC). Complete blood count data and body weight as well as the height of the patients were retrieved and recorded before the date of the first transurethral resection of bladder tumor (TURBT). The follow-up period was 48 months for recurrence-free survival (RFS), progression-free survival (PFS), and overall survival (OS). Results Oct-4 expression profile was found to be significantly associated with gender (p = 0.028), tumor grade (p = 0.038), tumor stage (p = 0.003), lymph node status (p = 0.029), recurrence (p = 0.004), progression (p = 0.011), and treatment modality (p = 0.016). Tumor grade and progression were found significant with NLR values (tumor grade, p = 0.006; progression, p = 0.038) and BMI (tumor grade, p = 0.036; progression, p = 0.014). Moreover, BMI was also significantly associated with UBC recurrence (p = 0.014). Kaplan-Meier survival analysis showed poor prognosis with both high Oct-4 expression (RFS, p = 0.001; PFS, p = 0.004; OS, p = 0.014) and high NLR values (RFS, p = 0.049; PFS, p = 0.004; OS, p = 0.005). Patients with high BMI too had poor RFS (p = 0.025) and poor PFS (p = 0.032). Furthermore, multivariate Cox regression analysis, indicated Oct-4 as an independent prognostic biomarker for RFS (HR = 0.240, 95% CI, 0.072-0.804, p = 0.021). Conclusions We conclude that the expression profile of Oct-4 will be beneficial in prediction of UBC recurrence, and could have profound implications on the development of new therapeutic targets for UBC treatment.
Collapse
Affiliation(s)
- Zainab Siddiqui
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India; Department of Biotechnology, Dr. A. P. J. Abdul Kalam Technical University, Lucknow, India
| | - Anand N Srivastava
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, Uttar Pradesh, India
| | - Satya N Sankhwar
- Department of Urology, King George's Medical University, Lucknow, India
| | - Noorin Zaidi
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India
| | - Naseem Fatima
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India
| | - Shivangi Singh
- Research Metabolic Unit, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India
| | - Mohd Yusuf
- Department of Pathology, King George's Medical University, Lucknow, India
| |
Collapse
|
78
|
Long W, Zhao W, Ning B, Huang J, Chu J, Li L, Ma Q, Xing C, Wang HY, Liu Q, Wang RF. PHF20 collaborates with PARP1 to promote stemness and aggressiveness of neuroblastoma cells through activation of SOX2 and OCT4. J Mol Cell Biol 2019; 10:147-160. [PMID: 29452418 DOI: 10.1093/jmcb/mjy007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/09/2018] [Indexed: 12/22/2022] Open
Abstract
The differentiation status of neuroblastoma (NB) strongly correlates with its clinical outcomes; however, the molecular mechanisms driving maintenance of stemness and differentiation remain poorly understood. Here, we show that plant homeodomain finger-containing protein 20 (PHF20) functions as a critical epigenetic regulator in sustaining stem cell-like phenotype of NB by using CRISPR/Cas9-based targeted knockout (KO) for high-throughput screening of gene function in NB cell differentiation. The expression of PHF20 in NB was significantly associated with high aggressiveness of the tumor and poor outcomes for NB patients. Deletion of PHF20 inhibited NB cell proliferation, invasive migration, and stem cell-like traits. Mechanistically, PHF20 interacts with poly(ADP-ribose) polymerase 1 (PARP1) and directly binds to promoter regions of octamer-binding transcription factor 4 (OCT4) and sex determining region Y-box 2 (SOX2) to modulate a histone mark associated with active transcription, trimethylation of lysine 4 on histone H3 protein subunit (H3K4me3). Overexpression of OCT4 and SOX2 restored growth and progression of PHF20 KO tumor cells. Consistently, OCT4 and SOX2 protein levels in clinical NB specimens were positively correlated with PHF20 expression. Our results establish PHF20 as a key driver of NB stem cell-like properties and aggressive behaviors, with implications for prognosis and therapy.
Collapse
Affiliation(s)
- Wenyong Long
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha 410008, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Wei Zhao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Key Laboratory of Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Bo Ning
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Institute Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Jing Huang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Junjun Chu
- Key Laboratory of Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Linfeng Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Qianquan Ma
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha 410008, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Changsheng Xing
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Helen Y Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Qing Liu
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.,Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
79
|
Villodre ES, Felipe KB, Oyama MZ, Oliveira FHD, Lopez PLDC, Solari C, Sevlever G, Guberman A, Lenz G. Silencing of the transcription factors Oct4, Sox2, Klf4, c-Myc or Nanog has different effect on teratoma growth. Biochem Biophys Res Commun 2019; 517:324-329. [DOI: 10.1016/j.bbrc.2019.07.064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 10/26/2022]
|
80
|
Yi J, Zhou LY, Yi YY, Zhu X, Su XY, Zhao Q, Lin J, Qian J, Deng ZQ. Low Expression of Pseudogene POU5F1B Affects Diagnosis and Prognosis in Acute Myeloid Leukemia (AML). Med Sci Monit 2019; 25:4952-4959. [PMID: 31271156 PMCID: PMC6625577 DOI: 10.12659/msm.914352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background The transcription factor Oct-4 is necessary for maintaining pluripotency and self-renewal of embryonic stem cells, and POU5F1B is a processed pseudogene of Oct-4 with coding capacity. The purpose of this study is to evaluate the expression and clinical implication of POU5F1B in AML. Material/Methods The expression of the POU5F1B transcript was evaluated in 175 newly diagnosed AML patients and 39 healthy controls by use of real-time quantitative PCR (RQ-PCR). Results POU5F1B was underexpressed in AML compared with controls (P<0.001). The receiver operating characteristic (ROC) curve revealed that the POU5F1B transcript level was able to differentiate AML patients from healthy individuals (AUC=0.682). In non-APL AML patients, the POU5F1Blow group had significantly higher WBC than the POU5F1Bhigh group (20.2×109vs. 4.6×109 L−1, P=0.021). Among whole-cohort AML, non-APL AML, and intermediate-risk AML, POU5F1Bhigh patients had obviously higher complete remission (CR) rates than POU5F1Blow patients (P=0.012, P=0.012 and P=0.027). In addition, Kaplan-Meier analysis demonstrated better overall survival (OS, P=0.019, P=0.007 and P=0.046, respectively) in POU5F1Bhigh patients compared with POU5F1Blow patients. Furthermore, in multivariate survival analysis, POU5F1B was independently associated with OS in non-APL AML patients and intermediate-risk AML as a favorable prognostic factor. Conclusions POU5F1B was frequently underexpressed in AML, and might contribute to the diagnosis and prognosis of AML.
Collapse
Affiliation(s)
- Jing Yi
- Department of Laboratory Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| | - Ling-Yu Zhou
- Department of Emergency Medicine, Huashan Hospital, Fudan University, Shanghai, China (mainland)
| | - Yun-Yun Yi
- Department of Laboratory Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| | - Xin Zhu
- Department of Laboratory Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| | - Xiao-Yu Su
- Department of Laboratory Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| | - Qian Zhao
- Department of Laboratory Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| | - Jiang Lin
- Department of Laboratory Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| | - Jun Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| | - Zhao-Qun Deng
- Department of Laboratory Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China (mainland)
| |
Collapse
|
81
|
Chen Y, Wu Z, Zhu X, Zhang M, Zang X, Li X, Xu Y. OCT4B-190 protects against ischemic stroke by modulating GSK-3β/HDAC6. Exp Neurol 2019; 316:52-62. [DOI: 10.1016/j.expneurol.2019.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/25/2019] [Accepted: 04/10/2019] [Indexed: 01/09/2023]
|
82
|
Organista-Nava J, Gómez-Gómez Y, Garibay-Cerdenares OL, Leyva-Vázquez MA, Illades-Aguiar B. Cervical cancer stem cell-associated genes: Prognostic implications in cervical cancer. Oncol Lett 2019; 18:7-14. [PMID: 31289465 PMCID: PMC6540231 DOI: 10.3892/ol.2019.10307] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 03/18/2019] [Indexed: 12/14/2022] Open
Abstract
Cervical cancer is the fourth most common type of gynecological malignancy to affect females, worldwide. Although high-risk human papillomavirus (HR-HPV) infection is the primary etiologic agent associated with the development of cervical cancer, cancer stem cells (CSCs) also serve a prominent role in the development, metastasis, recurrence and prognosis of the disease. CSCs are a small subpopulation of cells that have the ability to self-renew and are present in the majority of tumors, including cervical cancer. Studies describing the phenotype of cervical CSCs (CCSCs) vary in their definition of the expression pattern of principal biomarkers, including Musashi-1, aldehyde dehydrogenase 1, Oct3/4, Sox2 and CD49f. However, these markers are not observed in all cancers, although several may be present in multiple tumor types. The present review describes the potential biomarkers of CSCs in cervical cancer. These CCSC biomarkers may serve as molecular targets to enhance the efficacy and reduce the side effects associated with chemotherapeutic treatment in HR-HPV-positive cervical cancer.
Collapse
Affiliation(s)
- Jorge Organista-Nava
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero 39090, Mexico
| | - Yazmín Gómez-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero 39090, Mexico
| | - Olga Lilia Garibay-Cerdenares
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero 39090, Mexico.,Consejo Nacional de Ciencia y Tecnología, Mexico City 03940, Mexico
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero 39090, Mexico
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero 39090, Mexico
| |
Collapse
|
83
|
Jin X, Li Y, Guo Y, Jia Y, Qu H, Lu Y, Song P, Zhang X, Shao Y, Qi D, Xu W, Quan C. ERα is required for suppressing OCT4-induced proliferation of breast cancer cells via DNMT1/ISL1/ERK axis. Cell Prolif 2019; 52:e12612. [PMID: 31012189 PMCID: PMC6668970 DOI: 10.1111/cpr.12612] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/04/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Objective POU5F1 (OCT4) is implicated in cancer stem cell self‐renewal. Currently, some studies have shown that OCT4 has a dual function in suppressing or promoting cancer progression. However, the precise molecular mechanism of OCT4 in breast cancer progression remains unclear. Materials and Methods RT‐PCR and Western blot were utilized to investigate OCT4 expression in breast cancer tissues and cells. Cell proliferation assays and mouse models were applied to determine the effects of OCT4 on breast cancer cell proliferation. DNMT1 inhibitors, ChIP, CoIP, IHC and ERα inhibitors were used to explore the molecular mechanism of OCT4 in breast cancer. Results OCT4 was down‐regulated in breast cancer tissues, and the overexpression of OCT4 promoted MDA‐MB‐231 cell proliferation and inhibited the proliferation of MCF‐7 cells in vitro and in vivo, respectively. Two DNMT1 inhibitors (5‐aza‐dC and zebularine) suppressed OCT4‐induced MDA‐MB‐231 cell proliferation through Ras/Raf1/ERK inactivation by targeting ISL1, which is the downstream of DNMT1. In contrast, OCT4 interacted with ERα, decreased DNMT1 expression and inactivated the Ras/Raf1/ERK signalling pathway in MCF‐7 cells. Moreover, ERα inhibitor (AZD9496) reversed the suppression of OCT4‐induced proliferation in MCF‐7 cells via the activation of ERK signalling pathway. Conclusions OCT4 is dependent on ERα to suppress the proliferation of breast cancer cells through DNMT1/ISL1/ERK axis.
Collapse
Affiliation(s)
- Xiangshu Jin
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yanru Li
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yantong Guo
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yiyang Jia
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Huinan Qu
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yan Lu
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Peiye Song
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Xiaoli Zhang
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yijia Shao
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Da Qi
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Wenhong Xu
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Chengshi Quan
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| |
Collapse
|
84
|
Lettnin AP, Wagner EF, Carrett-Dias M, Dos Santos Machado K, Werhli A, Cañedo AD, Trindade GS, de Souza Votto AP. Silencing the OCT4-PG1 pseudogene reduces OCT-4 protein levels and changes characteristics of the multidrug resistance phenotype in chronic myeloid leukemia. Mol Biol Rep 2019; 46:1873-1884. [PMID: 30721421 DOI: 10.1007/s11033-019-04639-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022]
Abstract
Cancer stem cells show epigenetic plasticity and intrinsic resistance to anti-cancer therapy, rendering capable of initiating cancer relapse and progression. Transcription factor OCT-4 regulates various pathways in stem cells, but its expression can be regulated by pseudogenes. This work evaluated how OCT4-PG1 pseudogene can affect OCT-4 expression and mechanisms related to the multidrug resistance (MDR) phenotype in FEPS cells. Considering that OCT-4 protein is a transcription factor that regulates expression of ABC transporters, level of gene expression, activity of ABC proteins and cell sensitivity to chemotherapy were evaluated after OCT4-PG1 silencing. Besides we set up a STRING network. Results showed that after OCT4-PG1 silencing, cells expressed OCT-4 gene and protein to a lesser extent than mock cells. The gene and protein expression of ABCB1, as well as its activity were reduced. On the other hand, ALOX5 and ABCC1 genes was increased even as the activity of this transporter. Moreover, the silencing cells become sensitive to two chemotherapics tested. The network structure demonstrated that OCT4-PG1 protein interacts directly with OCT-4, SOX2, and NANOG and indirectly with ABC transporters. We conclude that OCT4-PG1 pseudogene plays a key role in the regulation OCT-4 transcription factor, which alters MDR phenotype in the FEPS cell line.
Collapse
Affiliation(s)
- Aline Portantiolo Lettnin
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil.,Laboratory of Cell Culture, Institute of Biological Sciences - ICB, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Eduardo Felipe Wagner
- Laboratory of Cell Culture, Institute of Biological Sciences - ICB, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Michele Carrett-Dias
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Karina Dos Santos Machado
- Center of Computational Sciences - C3, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Adriano Werhli
- Center of Computational Sciences - C3, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Andrés Delgado Cañedo
- Federal University of Pampa - UNIPAMPA, Avenue Antônio Trilha, 1847, São Gabriel, RS, Zip Code 97300-000, Brazil
| | - Gilma Santos Trindade
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Ana Paula de Souza Votto
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil. .,Laboratory of Cell Culture, Institute of Biological Sciences - ICB, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil. .,Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Avenida Itália, Km 8, s/n, Rio Grande, RS, Zip Code 96203-900, Brazil.
| |
Collapse
|
85
|
Guo T, Wen XZ, Li ZY, Han HB, Zhang CG, Bai YH, Xing XF, Cheng XJ, Du H, Hu Y, Wang XH, Jia YN, Nie ML, Xie M, Li QD, Ji JF. ISL1 predicts poor outcomes for patients with gastric cancer and drives tumor progression through binding to the ZEB1 promoter together with SETD7. Cell Death Dis 2019; 10:33. [PMID: 30674889 PMCID: PMC6393520 DOI: 10.1038/s41419-018-1278-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 02/06/2023]
Abstract
ISL1, a LIM-homeodomain transcription factor, serves as a biomarker of metastasis in multiple tumors. However, the function and underlying mechanisms of ISL1 in gastric cancer (GC) have not been fully elucidated. Here we found that ISL1 was frequently overexpressed in GC FFPE samples (104/196, 53.06%), and associated with worse clinical outcomes. Furthermore, the overexpression of ISL1 and loss-of-function of ISL1 influenced cell proliferation, invasion and migration in vitro and in vivo, including GC patient-derived xenograft models. We used ChIP-seq and RNA-seq to identify that ISL1 influenced the regulation of H3K4 methylation and bound to ZEB1, a key regulator of the epithelial–mesenchymal transition (EMT). Meanwhile, we validated ISL1 as activating ZEB1 promoter through influencing H3K4me3. We confirmed that a complex between ISL1 and SETD7 (a histone H3K4-specific methyltransferase) can directly bind to the ZEB1 promoter to activate its expression in GC cells by immunoprecipitation, mass spectrometry, and ChIP-re-ChIP. Moreover, ZEB1 expression was significantly positively correlated with ISL1 and was positively associated with a worse outcome in primary GC specimens. Our paper uncovers a molecular mechanism of ISL1 promoting metastasis of GC through binding to the ZEB1 promoter together with co-factor SETD7. ISL1 might be a potential prognostic biomarker of GC.
Collapse
Affiliation(s)
- Ting Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xian-Zi Wen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zi-Yu Li
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hai-Bo Han
- The Tissue Bank, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chen-Guang Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan-Hua Bai
- Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiao-Fang Xing
- The Tissue Bank, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiao-Jing Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ying Hu
- The Tissue Bank, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiao-Hong Wang
- The Tissue Bank, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yong-Ning Jia
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng-Lin Nie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qing-Da Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jia-Fu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China. .,Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
86
|
Monferrer E, Burgos-Panadero R, Blanquer-Maceiras M, Cañete A, Navarro S, Noguera R. High Oct4 expression: implications in the pathogenesis of neuroblastic tumours. BMC Cancer 2019; 19:1. [PMID: 30606139 PMCID: PMC6318846 DOI: 10.1186/s12885-018-5219-3] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/13/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Neuroblastic tumours (NBTs) are paediatric solid tumours derived from embryonic neural crest cells which harbour their own cancer stem cells (CSC). There is evidence indicating that CSC may be responsible for tumour progression, chemotherapy resistance and recurrence in NBTs. Oct4 is a transcription factor which plays a key role in mammal embryonic development and stem cell fate regulation. The aim of the study is to elucidate the clinical significance of Oct4 in NBTs. METHODS We studied Oct4 expression in 563 primary NBTs using digital image quantification. Chi-square test was applied to analyse the correlation between histopathology and the Oct4+ cell percentage. Survival analysis was carried out with Kaplan-Meier curves and log-rank test. Additionally, a multivariate Cox regression analysis with the stepwise backwards (Wald) method was undertaken to calculate the impact of Oct4 expression level on survival. RESULTS We found that tumours with a high proportion of cells expressing Oct4 correlated statistically with undifferentiated and poorly differentiated neuroblastoma / nodular ganglioneuroblastoma, and that Oct4 expression was not present in ganglioneuroma (p < 0.05). Statistical analysis also indicated a relationship between high Oct4 expression levels, high-risk patients according to the International Neuroblastoma Risk Group pre-treatment classification parameters, larger blood vessels and low survival rates. CONCLUSIONS These results suggest that the Oct4 gene may regulate NBT pathogenic differentiation pathways, and should thus be considered as a target for knockdown when developing novel therapies for high-risk NBT patients.
Collapse
Affiliation(s)
- Ezequiel Monferrer
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Rebeca Burgos-Panadero
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Maite Blanquer-Maceiras
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- CIBERONC, Madrid, Spain
| | - Adela Cañete
- Pediatric Oncology Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Samuel Navarro
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- CIBERONC, Madrid, Spain
| | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- CIBERONC, Madrid, Spain
| |
Collapse
|
87
|
Klec C, Prinz F, Pichler M. Involvement of the long noncoding RNA NEAT1 in carcinogenesis. Mol Oncol 2018; 13:46-60. [PMID: 30430751 PMCID: PMC6322192 DOI: 10.1002/1878-0261.12404] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
Altered expression levels of the long noncoding RNA (lncRNA) nuclear‐enriched abundant transcript 1 (NEAT1) have been reported in different types of cancer. More than half of the NEAT1 studies in cancer have been published within the last 2 years. In this review, we discuss very recent developments and insights into NEAT1 contribution to carcinogenesis. Summarizing the literature, it becomes obvious that NEAT1 is a lncRNA highly de‐/upregulated in a variety of cancer entities, in which it primarily acts as a competing endogenous RNA (ceRNA) which sponges tumor‐suppressive microRNA (miRNA). The sponged miRNA lose their ability to degrade, silence, or hamper translation of their downstream—mostly oncogenic—target transcripts, ultimately promoting carcinogenesis. This role of NEAT1 function in tumorigenesis suggests it may be a prognostic biomarker as well as potential therapeutic target, pending the completion of further studies into the underlying mechanisms.
Collapse
Affiliation(s)
- Christiane Klec
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria
| | - Felix Prinz
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
88
|
Saha SK, Jeong Y, Cho S, Cho SG. Systematic expression alteration analysis of master reprogramming factor OCT4 and its three pseudogenes in human cancer and their prognostic outcomes. Sci Rep 2018; 8:14806. [PMID: 30287838 PMCID: PMC6172215 DOI: 10.1038/s41598-018-33094-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
OCT4 is a master transcription factor that regulates the pluripotency of pluripotent stem cells and cancer stem cells along with other factors, including SOX2, KLF4, and C-MYC. Three different transcripts, OCT4A, OCT4B, and OCT4B1, are known to be generated by alternative splicing and eight OCT4 pseudogenes have been found in the human genome. Among them, we examined OCT4 and three pseudogenes (POU5F1P1, POU5F1P3, and POU5F1P4) because of their high expression possibility in cancer. In addition, previous studies indicated that OCT4 expression is augmented in cervical cancer and associated with poor prognosis, whereas OCT4 is down-regulated and correlated with good clinical outcomes in breast cancer. Because of these conflicting reports, we systematically evaluated whether expression of OCT4 and its pseudogenes can serve as oncogenic markers in various human cancers using the Oncomine database. Moreover, copy number alterations and mutations in OCT4 gene and its pseudogenes were analyzed using cBioPortal and the relationship between expression of OCT4 and pseudogenes and survival probability of cancer patients were explored using Kaplan-Meier plotter, OncoLnc, PROGgeneV2, and PrognoScan databases. Multivariate survival analysis was further conducted to determine the risk of the expression of the occurrence of OCT4 and its pseudogenes on certain cancer types using data from the Kaplan-Meier plotter. Overall, an association between expression of OCT4 and pseudogenes and cancer prognosis were established, which may serve as a therapeutic target for various human cancers.
Collapse
Affiliation(s)
- Subbroto Kumar Saha
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul, 05029, Republic of Korea
| | - Yeojin Jeong
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul, 05029, Republic of Korea
| | - Sungha Cho
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
89
|
Chao HM, Chern E. Patient-derived induced pluripotent stem cells for models of cancer and cancer stem cell research. J Formos Med Assoc 2018; 117:1046-1057. [PMID: 30172452 DOI: 10.1016/j.jfma.2018.06.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 05/28/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are embryonic stem cell-like cells reprogrammed from somatic cells by four transcription factors, OCT4, SOX2, KLF4 and c-MYC. iPSCs derived from cancer cells (cancer-iPSCs) could be a novel strategy for studying cancer. During cancer cell reprogramming, the epigenetic status of the cancer cell may be altered, such that it acquires stemness and pluripotency. The cellular behavior of the reprogrammed cells exhibits dynamic changes during the different stages of reprogramming. The cells may acquire the properties of cancer stem cells (CSCs) during the process of reprogramming, and lose their carcinogenic properties during reprogramming into a cancer-iPSCs. Differentiation of cancer-iPSCs by teratoma formation or organoid culturing could mimic the process of tumorigenesis. Some of the molecular mechanisms associated with cancer progression could be elucidated using the cancer-iPSC model. Furthermore, cancer-iPSCs could be expanded in culture system or bioreactors, and serve as cell sources for research, and as personal disease models for therapy and drug screening. This article introduces cancer studies that used the cell reprogramming strategy.
Collapse
Affiliation(s)
- Hsiao-Mei Chao
- niChe Lab for Stem Cell and Regenerative Medicine, Department of Biochemical Science and Technology, National Taiwan University, Taiwan; Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taiwan
| | - Edward Chern
- niChe Lab for Stem Cell and Regenerative Medicine, Department of Biochemical Science and Technology, National Taiwan University, Taiwan.
| |
Collapse
|
90
|
Liu F, Li L, Li Y, Ma X, Bian X, Liu X, Wang G, Zhang D. Overexpression of SENP1 reduces the stemness capacity of osteosarcoma stem cells and increases their sensitivity to HSVtk/GCV. Int J Oncol 2018; 53:2010-2020. [PMID: 30226577 PMCID: PMC6192779 DOI: 10.3892/ijo.2018.4537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma stem cells are able to escape treatment with conventional chemotherapeutic drugs, as the majority of them are in a quiescent state. Recent reports have suggested that small ubiquitin-like modifiers (SUMOs) serve important roles in the maintenance of cancer stem cell stemness. Therefore, a potential strategy to increase the effectiveness of chemotherapeutic agents is to interfere with SUMO modification of proteins associated with the maintenance of stemness in osteosarcoma stem cells. The present study revealed a significant decrease in the expression of SUMO1 specific peptidase 1 (SENP1) in osteosarcoma tissues and osteosarcoma cell lines, and SENP1 expression was much lower in osteosarcoma stem cells than in non-cancer stem cells. Further experiments indicated that the low levels of SENP1 were essential for maintenance of stemness in osteosarcoma stem cells. Overexpression of SENP1 resulted in a marked decrease in the maintenance of stemness, but only slightly induced apoptosis of osteosarcoma cells, which is crucial to reduce the side effects of drugs on normal precursor cells. Finally, SENP1 overexpression led to a significant increase in the sensitivity of osteosarcoma stem cells to the herpes simplex virus 1 thymidine kinase gene in combination with ganciclovir in vitro and in vivo. In conclusion, the present study described a novel method to increase the sensitivity of osteosarcoma stem cells to chemotherapeutic drugs. Notably, this approach may significantly reduce the required dose of conventional chemotherapeutic drugs and reduce side effects.
Collapse
Affiliation(s)
- Fengting Liu
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Lili Li
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Yanxia Li
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Xiaofang Ma
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Xiyun Bian
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Xiaozhi Liu
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Dianying Zhang
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing 100044, P.R. China
| |
Collapse
|
91
|
Dittmer J. Breast cancer stem cells: Features, key drivers and treatment options. Semin Cancer Biol 2018; 53:59-74. [PMID: 30059727 DOI: 10.1016/j.semcancer.2018.07.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
The current view is that breast cancer is a stem cell disease characterized by the existence of cancer cells with stem-like features and tumor-initiating potential. These cells are made responsible for tumor dissemination and metastasis. Common therapies by chemotherapeutic drugs fail to eradicate these cells and rather increase the pool of cancer stem cells in tumors, an effect that may increase the likelyhood of recurrence. Fifteen years after the first evidence for a small stem-like subpopulation playing a major role in breast cancer initiation has been published a large body of knowledge has been accumulated regarding the signaling cascades and proteins involved in maintaining stemness in breast cancer. Differences in the stem cell pool size and in mechanisms regulating stemness in the different breast cancer subtypes have emerged. Overall, this knowledge offers new approaches to intervene with breast cancer stem cell activity. New options are particularly needed for the treatment of triple-negative breast cancer subtype, which is particularly rich in cancer stem cells and is also the subtype for which specific therapies are still not available.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
92
|
Xiang X, Deng L, Xiong R, Xiao D, Chen Z, Yang F, Liu K, Feng G. Tex10 is upregulated and promotes cancer stem cell properties and chemoresistance in hepatocellular carcinoma. Cell Cycle 2018; 17:1310-1318. [PMID: 30045663 DOI: 10.1080/15384101.2018.1480208] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Testis expressed 10 (Tex10), a new core component of the pluripotency circuitry, has been reported to positively regulate embryonic stem cell (ESC) super-enhancers to promote ESC self-renewal; however, the expression and function of Tex10 in hepatocellular carcinoma (HCC) and liver cancer stem cells remains unclear. The present study was designed to investigate the expression patterns of Tex10 with immunohistochemistry, western blotting and RT-qPCR in samples from HCC patients and HCC cell lines. The results obtained show that Tex10 was highly expressed in HCC tissues, and elevated Tex10 protein levels positively correlate with the poorly differentiated carcinoma. Likewise, we found that Tex10 expression in the high-metastasis HCCLM3 potential cell line was higher than that in the low-metastasis HepG2 potential cell line, and Tex10 expression in liver cancer stem cells was also higher than that in adhered HCC cells. In addition, Tex10 knockdown decreased stem cell marker expression and drug resistance. Tex10 promoted cancer stemness through activation of the STAT3 signaling pathway. Taken together, our study demonstrates that Tex10 plays a potent carcinogenic role in HCC tumorigenesis by maintaining cancer stem cell properties through activation of the STAT3 signaling pathway and promoting chemo-resistance. Thus, targeting Tex10 may provide a novel and effective therapeutic strategy to suppress the tumorigenicity of advanced HCC.
Collapse
Affiliation(s)
- Xiaocong Xiang
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| | - Li Deng
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| | - Rong Xiong
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| | - Dongqin Xiao
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| | - Zhu Chen
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| | - Fei Yang
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| | - Kang Liu
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| | - Gang Feng
- a Institute of Tissue Engineering and Stem Cells , Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College , Nanchong , China
| |
Collapse
|
93
|
Klein RH, Tung PY, Somanath P, Fehling HJ, Knoepfler PS. Genomic functions of developmental pluripotency associated factor 4 (Dppa4) in pluripotent stem cells and cancer. Stem Cell Res 2018; 31:83-94. [PMID: 30031967 PMCID: PMC6133722 DOI: 10.1016/j.scr.2018.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/20/2022] Open
Abstract
Developmental pluripotency associated factor 4 (Dppa4) is a highly specific marker of pluripotent cells, and is also overexpressed in certain cancers, but its function in either of these contexts is poorly understood. In this study, we use ChIP-Seq to identify Dppa4 binding genome-wide in three distinct cell types: mouse embryonic stem cells (mESC), embryonal carcinoma cells, and 3T3 fibroblasts ectopically expressing Dppa4. We find a core set of Dppa4 binding sites shared across cell types, and also a substantial number of sites unique to each cell type. Across cell types Dppa4 shows a preference for binding to regions with active chromatin signatures, and can influence chromatin modifications at target genes. In 3T3 fibroblasts with enforced Dppa4 expression, Dppa4 represses the cell cycle inhibitor Cdkn2c and activates Ets family transcription factor Etv4, leading to alterations in the cell cycle that likely contribute to the oncogenic phenotype. Dppa4 also directly regulates Etv4 in mESC but represses it in this context, and binds with Oct4 to a set of shared targets that are largely independent of Sox2 and Nanog, indicating that Dppa4 functions independently of the core pluripotency network in stem cells. Together these data provide novel insights into Dppa4 function in both pluripotent and oncogenic contexts.
Collapse
Affiliation(s)
- Rachel Herndon Klein
- Department of Cell Biology and Human Anatomy, University of California, Davis, 1 Shields Ave, Davis, CA 95616, United States.; Institute of Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States
| | - Po-Yuan Tung
- Department of Cell Biology and Human Anatomy, University of California, Davis, 1 Shields Ave, Davis, CA 95616, United States.; Institute of Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States
| | - Priyanka Somanath
- Department of Cell Biology and Human Anatomy, University of California, Davis, 1 Shields Ave, Davis, CA 95616, United States.; Institute of Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States
| | | | - Paul S Knoepfler
- Department of Cell Biology and Human Anatomy, University of California, Davis, 1 Shields Ave, Davis, CA 95616, United States.; Institute of Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States.
| |
Collapse
|
94
|
OCT4 Potentiates Radio-Resistance and Migration Activity of Rectal Cancer Cells by Improving Epithelial-Mesenchymal Transition in a ZEB1 Dependent Manner. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3424956. [PMID: 30112378 PMCID: PMC6077687 DOI: 10.1155/2018/3424956] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/31/2018] [Accepted: 06/14/2018] [Indexed: 01/20/2023]
Abstract
Radiotherapy is an important strategy for rectal cancer patient treatment. However, the efficiency of radiation is usually poor, especially in patients with advanced stage rectal cancer due to the radio-resistance developed. At the present study, OCT4 was found to play a critical role in radio-resistance development in human rectal cancer cells by improving the epithelial-mesenchymal transition process (EMT). Endogenous OCT4 expression could confer resistant phonotype on human rectal cancer cells, which was supported by the data from clonogenic forming assay and cell cycle arrest recovering experiment. EMT related transcription factor ZEB1 might take part in the radio-resistance induced by OCT4, as its expression could be upregulated by OCT4 and its silence could reverse the OCT4 induced resistance to radiation in SW480 cells. More interestingly, CHK1 was also upregulated in OCT4/ZEB1 dependent manner conferring stronger DNA damage repair activity on cancer cells, which might explain the underlying mechanisms why OCT4/ZEB1 axis could promote the resistance of human rectal cancer cell to radiation. Taken together, our results provided a novel mechanism for radio-resistance development in human rectal cancer cells and a new target to overcome this resistance.
Collapse
|
95
|
Maiuthed A, Bhummaphan N, Luanpitpong S, Mutirangura A, Aporntewan C, Meeprasert A, Rungrotmongkol T, Rojanasakul Y, Chanvorachote P. Nitric oxide promotes cancer cell dedifferentiation by disrupting an Oct4:caveolin-1 complex: A new regulatory mechanism for cancer stem cell formation. J Biol Chem 2018; 293:13534-13552. [PMID: 29986880 DOI: 10.1074/jbc.ra117.000287] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 06/19/2018] [Indexed: 01/11/2023] Open
Abstract
Cancer stem cells (CSCs) are unique populations of cells that can self-renew and generate different cancer cell lineages. Although CSCs are believed to be a promising target for novel therapies, the specific mechanisms by which these putative therapeutics could intervene are less clear. Nitric oxide (NO) is a biological mediator frequently up-regulated in tumors and has been linked to cancer aggressiveness. Here, we search for targets of NO that could explain its activity. We find that it directly affects the stability and function of octamer-binding transcription factor 4 (Oct4), known to drive the stemness of lung cancer cells. We demonstrated that NO promotes the CSC-regulatory activity of Oct4 through a mechanism that involves complex formation between Oct4 and the scaffolding protein caveolin-1 (Cav-1). In the absence of NO, Oct4 forms a molecular complex with Cav-1, which promotes the ubiquitin-mediated proteasomal degradation of Oct4. NO promotes Akt-dependent phosphorylation of Cav-1 at tyrosine 14, disrupting the Cav-1:Oct4 complex. Site-directed mutagenesis and computational modeling studies revealed that the hydroxyl moiety at tyrosine 14 of Cav-1 is crucial for its interaction with Oct4. Both removal of the hydroxyl via mutation to phenylalanine and phosphorylation lead to an increase in binding free energy (ΔGbind) between Oct4 and Cav-1, destabilizing the complex. Together, these results unveiled a novel mechanism of CSC regulation through NO-mediated stabilization of Oct4, a key stem cell transcription factor, and point to new opportunities to design CSC-related therapeutics.
Collapse
Affiliation(s)
- Arnatchai Maiuthed
- From the Department of Pharmacology and Physiology.,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences
| | - Narumol Bhummaphan
- Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences.,the Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sudjit Luanpitpong
- the Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700 Thailand, and
| | - Apiwat Mutirangura
- the Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, and
| | | | - Arthitaya Meeprasert
- Structural and Computational Biology Research Group, and Department of Biochemistry, Faculty of Science
| | - Thanyada Rungrotmongkol
- Structural and Computational Biology Research Group, and Department of Biochemistry, Faculty of Science.,Ph.D. Program in Bioinformatics and Computational Biology
| | - Yon Rojanasakul
- WVU Cancer Institute, West Virginia University, Morgantown, West Virginia 26506
| | - Pithi Chanvorachote
- From the Department of Pharmacology and Physiology, .,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences
| |
Collapse
|
96
|
Xiang Y, Zhou X. Octamer-binding transcription factor 4 correlates with complex karyotype, FLT3-ITD mutation and poorer risk stratification, and predicts unfavourable prognosis in patients with acute myeloid leukaemia. Hematology 2018; 23:721-728. [PMID: 29950146 DOI: 10.1080/10245332.2018.1482050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yongsheng Xiang
- Department of Hematology, The First People’s Hospital of Jingmen, Jingmen, People’s Republic of China
| | - Xiaofen Zhou
- Department of Hematology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People’s Republic of China
| |
Collapse
|
97
|
Wefers C, Schreibelt G, Massuger LFAG, de Vries IJM, Torensma R. Immune Curbing of Cancer Stem Cells by CTLs Directed to NANOG. Front Immunol 2018; 9:1412. [PMID: 29971070 PMCID: PMC6018198 DOI: 10.3389/fimmu.2018.01412] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) have been identified as the source of tumor growth and disease recurrence. Eradication of CSCs is thus essential to achieve durable responses, but CSCs are resistant to current anti-tumor therapies. Novel therapeutic approaches that specifically target CSCs will, therefore, be crucial to improve patient outcome. Immunotherapies, which boost the body's own immune system to eliminate cancerous cells, could be an alternative approach to target CSCs. Vaccines of dendritic cells (DCs) loaded with tumor antigens can evoke highly specific anti-tumor T cell responses. Importantly, DC vaccination also promotes immunological memory formation, paving the way for long-term cancer control. Here, we propose a DC vaccination that specifically targets CSCs. DCs loaded with NANOG peptides, a protein required for maintaining stem cell properties, could evoke a potent anti-tumor immune response against CSCs. We hypothesize that the resulting immunological memory will also control newly formed CSCs, thereby preventing disease recurrence.
Collapse
Affiliation(s)
- Christina Wefers
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
- Department of Obstetrics and Gynecology, Radboudumc, Nijmegen, Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| | | | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| | - Ruurd Torensma
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| |
Collapse
|
98
|
Choi J, Baek KH. Cellular functions of stem cell factors mediated by the ubiquitin-proteasome system. Cell Mol Life Sci 2018; 75:1947-1957. [PMID: 29423528 PMCID: PMC11105287 DOI: 10.1007/s00018-018-2770-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/12/2018] [Accepted: 02/01/2018] [Indexed: 12/16/2022]
Abstract
Stem cells undergo partitioning through mitosis and separate into specific cells of each of the three embryonic germ layers: endoderm, mesoderm, and ectoderm. Pluripotency, reprogramming, and self-renewal are essential elements of embryonic stem cells (ESCs), and it is becoming evident that regulation of protein degradation mediated by the ubiquitin-proteasome system (UPS) is one of the key cellular mechanisms in ESCs. Although the framework of that mechanism may seem simple, it involves complicated proteolytic machinery. The UPS controls cell development, survival, differentiation, lineage commitment, migration, and homing processes. This review is centered on the connection between stem cell factors NANOG, OCT-3/4, SOX2, KLF4, C-MYC, LIN28, FAK, and telomerase and the UPS. Herein, we summarize recent findings and discuss potential UPS mechanisms involved in pluripotency, reprogramming, differentiation, and self-renewal. Interactions between the UPS and stem cell transcription factors can apply to various human diseases which can be treated by generating more efficient iPSCs. Such complexes may permit the design of novel therapeutics and the establishment of biomarkers that may be used in diagnosis and prognosis development. Therefore, the UPS is an important target for stem cell therapeutic product research.
Collapse
Affiliation(s)
- Jihye Choi
- Department of Biomedical Science, CHA Stem Cell Institute, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA Stem Cell Institute, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea.
| |
Collapse
|
99
|
Chalajour F, Siyahian A, Hanley FL. The Role of Microenvironment in Preserving the Potency of Adult Porcine Pulmonary Valve Stem Cells In Vitro. Int J Stem Cells 2018; 11:121-130. [PMID: 29843194 PMCID: PMC5984066 DOI: 10.15283/ijsc18020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 03/07/2018] [Accepted: 04/15/2018] [Indexed: 11/09/2022] Open
Abstract
Background and Objective The potency of tissue resident stem cells is regulated primarily by inputs from the local microenvironment. Isolation of stem cells through enzymatic digestion of tissue may affect epigenetic regulation of cell fate and performance. Here we employ a non-enzymatic method to harvest and investigate tissue resident stem cells from the adult porcine pulmonary valve. Methods and Results The presence of c-Kit+ stem cells within the valve tissue was confirmed by immunohistochemistry. An in vitro culture of minced valve leaflets was developed under the standard conditions (37°C with 5% CO2). The viability of the cellular outgrowths was evaluated over the subsequent 12 weeks. Under this culture condition, we identified a population of non-adherent c-Kit+ cells and multiple cellular structures mimicking the phenotype of embryonic stem cells at different stages of development. Formation of multinucleated cells through cell fusion provided an active niche area for homing and interaction of the non-adherent c-Kit+ cells. Expression of pluripotency markers Oct-4 and Nanog was detected in the newly formed multinucleated cells but not in mature colonies. Partial cell fusion was shown by fluorescent live-cell tracking, which confirmed intercellular molecular exchange between donor and recipient cells, resulting in altered cytoplasmic protein expression by the recipient cell. Conclusions These results suggest a role for the microenvironment in decrypting the potential of the valve somatic stem cells in vitro. In addition, our data provide evidence for cell fusion, which may play a critical role in reversing somatic cell fate and spontaneous cellular reprogramming.
Collapse
Affiliation(s)
- Fariba Chalajour
- Department of Cardiothoracic Surgery, Division of Pediatric Cardiovascular Surgery, Stanford University, California, USA
| | - Arpi Siyahian
- Department of Cardiothoracic Surgery, Division of Pediatric Cardiovascular Surgery, Stanford University, California, USA
| | - Frank L Hanley
- Department of Cardiothoracic Surgery, Division of Pediatric Cardiovascular Surgery, Stanford University, California, USA
| |
Collapse
|
100
|
Zhou Y, Chen X, Kang B, She S, Zhang X, Chen C, Li W, Chen W, Dan S, Pan X, Liu X, He J, Zhao Q, Zhu C, Peng L, Wang H, Yao H, Cao H, Li L, Herlyn M, Wang YJ. Endogenous authentic OCT4A proteins directly regulate FOS/AP-1 transcription in somatic cancer cells. Cell Death Dis 2018; 9:585. [PMID: 29789579 PMCID: PMC5964179 DOI: 10.1038/s41419-018-0606-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/14/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022]
Abstract
OCT4A is well established as a master transcription factor for pluripotent stem cell (PSC) self-renewal and a pioneer factor for initiating somatic cell reprogramming, yet its presence and functionality in somatic cancer cells remain controversial and obscure. By combining the CRISPR-Cas9-based gene editing with highly specific PCR assays, highly sensitive immunoassays, and mass spectrometry, we provide unequivocal evidence here that full-length authentic OCT4A transcripts and proteins were both present in somatic cancer cells, and OCT4A proteins were heterogeneously expressed in the whole cell population and when expressed, they are predominantly localized in cell nucleus. Despite their extremely low abundance (approximately three orders of magnitude lower than in PSCs), OCT4A proteins bound to the promoter/enhancer regions of the AP-1 transcription factor subunit c-FOS gene and critically regulated its transcription. Knocking out OCT4A in somatic cancer cells led to dramatic reduction of the c-FOS protein level, aberrant AP-1 signaling, dampened self-renewal capacity, deficient cell migration that were associated with cell growth retardation in vitro and in vivo, and their enhanced sensitivity to anticancer drugs. Taken together, we resolve the long-standing controversy and uncertainty in the field, and reveal a fundamental role of OCT4A protein in regulating FOS/AP-1 signaling-centered genes that mediate the adhesion, migration, and propagation of somatic cancer cells.
Collapse
Affiliation(s)
- Yanwen Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
- Department of Infectious Diseases, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xinyu Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Bo Kang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Shiqi She
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Xiaobing Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Cheng Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Wenxin Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Wenjie Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Songsong Dan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Xiaoyun Pan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Xiaoli Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Jianqin He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Qingwei Zhao
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Chenggang Zhu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Ling Peng
- Department of Radiotherapy, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Ying-Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|