101
|
Liang J, Fan J, Wang M, Niu Z, Zhang Z, Yuan L, Tai Y, Chen Z, Song S, Wang X, Liu X, Huang H, Sun Q. CDKN2A inhibits formation of homotypic cell-in-cell structures. Oncogenesis 2018; 7:50. [PMID: 29904067 PMCID: PMC6002405 DOI: 10.1038/s41389-018-0056-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/07/2018] [Accepted: 04/28/2018] [Indexed: 01/26/2023] Open
Abstract
Cell-in-cell (CIC) structures, characterized by enclosure of one or more cells within another cell, were extensively documented in human cancers. Although elevated CIC formation was found in cancers with CDKN2A inactivation, a causal link between them remains to be established. We reported here that inhibiting CDKN2A expression effectively promoted homotypic CIC formation, whereas ectopic overexpression of p16INK4a or p14ARF, two proteins encoded by CDKN2A gene, significantly suppressed CIC formation in MCF7 cells. The regulation of CIC formation by CDKN2A was tightly correlated with subcellular redistribution of E-cadherin, F-actin rearrangement and reduced phosphorylation of myosin light chain 2 (p-MLC2), consistent with which, CDKN2A expression imparted cells winner/outer identity in competition assay. Moreover, CIC formation negatively correlates with p16INK4a expression in human breast cancers. Thus, our work identifies CDKN2A as the first tumor suppressor whose inactivation promotes homotypic CIC formation in human cancer cells.
Collapse
Affiliation(s)
- Jianqing Liang
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, 10 TIEYI Road, 100038, Beijing, P. R. China
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China
- School of Biological Science and Engineering, South China University of Technology, 510000, Guangzhou, P.R. China
| | - Jie Fan
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, 10 TIEYI Road, 100038, Beijing, P. R. China
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China
- The 307 Hospital, 8 Dongda Street, 100071, Beijing, P. R. China
| | - Manna Wang
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China
| | - Zubiao Niu
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China
| | - Zhengrong Zhang
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China
| | - Long Yuan
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China
| | - Yanhong Tai
- The 307 Hospital, 8 Dongda Street, 100071, Beijing, P. R. China
| | - Zhaolie Chen
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China
| | - Santai Song
- The 307 Hospital, 8 Dongda Street, 100071, Beijing, P. R. China
| | - Xiaoning Wang
- School of Biological Science and Engineering, South China University of Technology, 510000, Guangzhou, P.R. China
| | - Xiaoqing Liu
- The 307 Hospital, 8 Dongda Street, 100071, Beijing, P. R. China.
| | - Hongyan Huang
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, 10 TIEYI Road, 100038, Beijing, P. R. China.
| | - Qiang Sun
- Institute of Biotechnology, 20 Dongda Street, 100071, Beijing, P.R. China.
| |
Collapse
|
102
|
Qin Z, Stoilov P, Zhang X, Xing Y. SEASTAR: systematic evaluation of alternative transcription start sites in RNA. Nucleic Acids Res 2018; 46:e45. [PMID: 29546410 PMCID: PMC5934623 DOI: 10.1093/nar/gky053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 12/30/2017] [Accepted: 03/12/2018] [Indexed: 12/23/2022] Open
Abstract
Alternative first exons diversify the transcriptomes of eukaryotes by producing variants of the 5' Untranslated Regions (5'UTRs) and N-terminal coding sequences. Accurate transcriptome-wide detection of alternative first exons typically requires specialized experimental approaches that are designed to identify the 5' ends of transcripts. We developed a computational pipeline SEASTAR that identifies first exons from RNA-seq data alone then quantifies and compares alternative first exon usage across multiple biological conditions. The exons inferred by SEASTAR coincide with transcription start sites identified directly by CAGE experiments and bear epigenetic hallmarks of active promoters. To determine if differential usage of alternative first exons can yield insights into the mechanism controlling gene expression, we applied SEASTAR to an RNA-seq dataset that tracked the reprogramming of mouse fibroblasts into induced pluripotent stem cells. We observed dynamic temporal changes in the usage of alternative first exons, along with correlated changes in transcription factor expression. Using a combined sequence motif and gene set enrichment analysis we identified N-Myc as a regulator of alternative first exon usage in the pluripotent state. Our results demonstrate that SEASTAR can leverage the available RNA-seq data to gain insights into the control of gene expression and alternative transcript variation in eukaryotic transcriptomes.
Collapse
Affiliation(s)
- Zhiyi Qin
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division TNLIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Peter Stoilov
- Department of Biochemistry and Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division TNLIST / Department of Automation, Tsinghua University, Beijing 100084, China
- School of Life Sciences, and Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
| | - Yi Xing
- Department of Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
103
|
Guan Y, Liu L, Wang Q, Zhao J, Li P, Hu J, Yang Z, Running MP, Sun H, Huang J. Gene refashioning through innovative shifting of reading frames in mosses. Nat Commun 2018; 9:1555. [PMID: 29674719 PMCID: PMC5908804 DOI: 10.1038/s41467-018-04025-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 03/28/2018] [Indexed: 12/20/2022] Open
Abstract
Early-diverging land plants such as mosses are known for their outstanding abilities to grow in various terrestrial habitats, incorporating tremendous structural and physiological innovations, as well as many lineage-specific genes. How these genes and functional innovations evolved remains unclear. In this study, we show that a dual-coding gene YAN/AltYAN in the moss Physcomitrella patens evolved from a pre-existing hemerythrin gene. Experimental evidence indicates that YAN/AltYAN is involved in fatty acid and lipid metabolism, as well as oil body and wax formation. Strikingly, both the recently evolved dual-coding YAN/AltYAN and the pre-existing hemerythrin gene might have similar physiological effects on oil body biogenesis and dehydration resistance. These findings bear important implications in understanding the mechanisms of gene origination and the strategies of plants to fine-tune their adaptation to various habitats. Extant representatives of the earliest land plant lineages adapt to various terrestrial habitats with structural and physiological innovations. Here the authors show a dual-coding gene in the moss Physcomitrella patens evolved from a hemerythrin gene, with effects on oil body biogenesis and dehydration resistance.
Collapse
Affiliation(s)
- Yanlong Guan
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China
| | - Li Liu
- Key Laboratory of Economic Plants and Biotechnology, Yunnan Key Laboratory for Wild Plant Resources, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China
| | - Qia Wang
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China
| | - Jinjie Zhao
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China
| | - Ping Li
- Key Laboratory of Economic Plants and Biotechnology, Yunnan Key Laboratory for Wild Plant Resources, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jinyong Hu
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China
| | - Zefeng Yang
- Jiangsu Key Laboratory of Crop Genetics and Physiology, Co-Innovation Center for Modern Production Technology of Grain Crops, Key Laboratory of Plant Functional Genomics of the Ministry of Education, Yangzhou University, 225009, Yangzhou, China
| | - Mark P Running
- Department of Biology, University of Louisville, Louisville, KY, 40292, USA
| | - Hang Sun
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China
| | - Jinling Huang
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, 650201, Kunming, China. .,Institute of Plant Stress Biology, State Key Laboratory of Cotton Biology, Henan University, 475001, Kaifeng, China. .,Department of Biology, East Carolina University, Greenville, NC, 27858, USA.
| |
Collapse
|
104
|
Brunet MA, Levesque SA, Hunting DJ, Cohen AA, Roucou X. Recognition of the polycistronic nature of human genes is critical to understanding the genotype-phenotype relationship. Genome Res 2018; 28:609-624. [PMID: 29626081 PMCID: PMC5932603 DOI: 10.1101/gr.230938.117] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/27/2018] [Indexed: 12/12/2022]
Abstract
Technological advances promise unprecedented opportunities for whole exome sequencing and proteomic analyses of populations. Currently, data from genome and exome sequencing or proteomic studies are searched against reference genome annotations. This provides the foundation for research and clinical screening for genetic causes of pathologies. However, current genome annotations substantially underestimate the proteomic information encoded within a gene. Numerous studies have now demonstrated the expression and function of alternative (mainly small, sometimes overlapping) ORFs within mature gene transcripts. This has important consequences for the correlation of phenotypes and genotypes. Most alternative ORFs are not yet annotated because of a lack of evidence, and this absence from databases precludes their detection by standard proteomic methods, such as mass spectrometry. Here, we demonstrate how current approaches tend to overlook alternative ORFs, hindering the discovery of new genetic drivers and fundamental research. We discuss available tools and techniques to improve identification of proteins from alternative ORFs and finally suggest a novel annotation system to permit a more complete representation of the transcriptomic and proteomic information contained within a gene. Given the crucial challenge of distinguishing functional ORFs from random ones, the suggested pipeline emphasizes both experimental data and conservation signatures. The addition of alternative ORFs in databases will render identification less serendipitous and advance the pace of research and genomic knowledge. This review highlights the urgent medical and research need to incorporate alternative ORFs in current genome annotations and thus permit their inclusion in hypotheses and models, which relate phenotypes and genotypes.
Collapse
Affiliation(s)
- Marie A Brunet
- Biochemistry Department, Université de Sherbrooke, Quebec J1E 4K8, Canada.,Groupe de recherche PRIMUS, Department of Family and Emergency Medicine, Quebec J1H 5N4, Canada.,PROTEO, Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Quebec G1V 0A6, Canada
| | - Sébastien A Levesque
- Pediatric Department, Centre Hospitalier de l'Université de Sherbrooke, Quebec J1H 5N4, Canada
| | - Darel J Hunting
- Department of Nuclear Medicine & Radiobiology, Université de Sherbrooke, Quebec J1H 5N4, Canada
| | - Alan A Cohen
- Groupe de recherche PRIMUS, Department of Family and Emergency Medicine, Quebec J1H 5N4, Canada
| | - Xavier Roucou
- Biochemistry Department, Université de Sherbrooke, Quebec J1E 4K8, Canada.,PROTEO, Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Quebec G1V 0A6, Canada
| |
Collapse
|
105
|
Inoue K, Fry EA. Aberrant Expression of p14 ARF in Human Cancers: A New Biomarker? TUMOR & MICROENVIRONMENT 2018; 1:37-44. [PMID: 30740529 PMCID: PMC6364748 DOI: 10.4103/tme.tme_24_17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ARF and INK4a genes are located on the CDKN2a locus, both showing tumor suppressive activity. ARF has been shown to monitor potentially harmful oncogenic signalings, making early stage cancer cells undergo senescence or programmed cell death to prevent cancer. Conversely, INK4a detects both aging and incipient cancer cell signals, and thus these two gene functions are different. The efficiency of detection of oncogenic signals is more efficient for the for the former than the latter in the mouse system. Both ARF and INK4a genes are inactivated by gene deletion, promoter methylation, frame shift, aberrant splicing although point mutations for the coding region affect only the latter. Recent studies show the splicing alterations that affect only ARF or both ARF and INK4a genes suggesting that ARF is inactivated in human tumors more frequently than what was previously thought. The ARF gene is activated by E2Fs and Dmp1 transcription factors while it is repressed by Bmi1, Tbx2/3, Twist1, and Pokemon nuclear proteins. It is also regulated at protein levels by Arf ubiquitin ligase named ULF, MKRN1, and Siva1. The prognostic value of ARF overexpression is controversial since it is induced in early stage cancer cells to eliminate pre-malignant cells (better prognosis); however, it may also indicate that the tumor cells have mutant p53 associated with worse prognosis. The ARF tumor suppressive protein can be used as a biomarker to detect early stage cancer cells as well as advanced stage tumors with p53 inactivation.
Collapse
Affiliation(s)
- Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| | - Elizabeth A. Fry
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| |
Collapse
|
106
|
Jiménez-García L, Herranz S, Higueras MA, Luque A, Hortelano S. Tumor suppressor ARF regulates tissue microenvironment and tumor growth through modulation of macrophage polarization. Oncotarget 2018; 7:66835-66850. [PMID: 27572316 PMCID: PMC5341841 DOI: 10.18632/oncotarget.11652] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/24/2016] [Indexed: 12/31/2022] Open
Abstract
Tumor microenvironment has been described to play a key role in tumor growth, progression, and metastasis. Macrophages are a major cellular constituent of the tumor stroma, and particularly tumor associated macrophages (TAMs or M2-like macrophages) exert important immunosuppressive activity and a pro-tumoral role within the tumor microenvironment. Alternative-reading frame (ARF) gene is widely inactivated in human cancer. We have previously demonstrated that ARF deficiency severely impairs inflammatory response establishing a new role for ARF in the regulation of innate immunity. On the basis of these observations, we hypothesized that ARF may also regulates tumor growth through recruitment and modulation of the macrophage phenotype in the tumor microenvironment. Xenograft assays of B16F10 melanoma cells into ARF-deficient mice resulted in increased tumor growth compared to those implanted in WT control mice. Tumors from ARF-deficient mice exhibited significantly increased number of TAMs as well as microvascular density. Transwell assays showed crosstalk between tumor cells and macrophages. On the one hand, ARF-deficient macrophages modulate migratory ability of the tumor cells. And on the other, tumor cells promote the skewing of ARF-/- macrophages toward a M2-type polarization. In conclusion, these results demonstrate that ARF deficiency facilitates the infiltration of macrophages into the tumor mass and favors their polarization towards a M2 phenotype, thus promoting tumor angiogenesis and tumor growth. This work provides novel information about the critical role of ARF in the modulation of tumor microenvironment.
Collapse
Affiliation(s)
- Lidia Jiménez-García
- Unidad de Terapias Farmacológicas. Instituto de Investigaciones de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Herranz
- Unidad de Terapias Farmacológicas. Instituto de Investigaciones de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - María Angeles Higueras
- Unidad de Terapias Farmacológicas. Instituto de Investigaciones de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Luque
- Unidad de Terapias Farmacológicas. Instituto de Investigaciones de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Sonsoles Hortelano
- Unidad de Terapias Farmacológicas. Instituto de Investigaciones de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
107
|
Du Y, Hao X, Liu X. Low expression of long noncoding RNA CDKN2B-AS1 in patients with idiopathic pulmonary fibrosis predicts lung cancer by regulating the p53-signaling pathway. Oncol Lett 2018. [PMID: 29541247 PMCID: PMC5835920 DOI: 10.3892/ol.2018.7910] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The present study aimed to investigate the expression of long non-coding RNA (lncRNA) cyclin dependent kinase inhibitor-2B-antisense RNA 1 CDKN2B-AS1 in patients with peripheral blood of idiopathic pulmonary fibrosis (IPF). A total of 24 patients with IPF and 24 healthy controls were included in the study, four patients with IPF and four healthy controls were selected randomly to extract RNA. There were no other diseases such as hypertension and diabetes in the two groups. RNA from peripheral blood was extracted by high-throughput sequencing and bioinformatics analysis was performed. Based on selected differentially expressed lncRNA and mRNA, gene ontology analysis was performed to screen out the tumor-associated mRNA. A total of 20 samples were chosen to avoid variance due to individual differences. A total of 20 patients with IPF, and 20 controls were further studied, RNA extracted from peripheral blood was used to verify the lncRNA and mRNA levels. A total of 440 lncRNAs were identified to be upregulated and 1,376 downregulated according to the screening results of differential expression. High-throughput sequencing and bioinformatics analysis demonstrated that the expression of CDKN2B-AS1 decreased significantly in patients with IPF compared with healthy controls. The adjacent gene mRNA of CDKN2B-AS1 was identified as CDKN2A, an important anti-oncogene, which is concentrated on the p53 signaling-pathway according to the Kyoto Encyclopedia of Genes and Genomes database. CDKN2A mRNA expression levels were lower in patients with IPF and higher in the control group. The expression of CDKN2B-AS1 and CDKN2A mRNA was significantly lower in IPF group compared with in the control group (P<0.05). The results suggest the expression of the CDKN2B-AS1 and adjacent gene, CDKN2A, are downregulated in the peripheral blood of patients with IPF, which activates the p53-signaling pathway to promote lung cancer formation.
Collapse
Affiliation(s)
- Yufeng Du
- Department of Geriatric Diseases, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiaoyan Hao
- Department of Geriatric Diseases, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xuejun Liu
- Department of Geriatric Diseases, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
108
|
Abstract
The ARF and INK4a genes are located in the same CDKN2a locus, both showing its tumor suppressive activity. ARF has been shown to detect potentially harmful oncogenic signals, making incipient cancer cells undergo senescence or apoptosis. INK4a, on the other hand, responds to signals from aging in a variety of tissues including islets of Langerhans, neuronal cells, and cancer stem cells in general. It also detects oncogenic signals from incipient cancer cells to induce them senescent to prevent neoplastic transformation. Both of these genes are inactivated by gene deletion, promoter methylation, frame shift, and aberrant splicing although mutations changing the amino acid sequences affect only the latter. Recent studies indicated that polycomb gene products EZH2 and BMI1 repressed p16INK4a expression in primary cells, but not in cells deficient for pRB protein function. It was also reported that that p14ARF inhibits the stability of the p16INK4a protein in human cancer cell lines and mouse embryonic fibroblasts through its interaction with regenerating islet-derived protein 3γ. Overexpression of INK4a is associated with better prognosis of cancer when it is associated with human papilloma virus infection. However, it has a worse prognostic value in other tumors since it is an indicator of pRB loss. The p16INK4a tumor suppressive protein can thus be used as a biomarker to detect early stage cancer cells as well as advanced tumor cells with pRB inactivation since it is not expressed in normal cells.
Collapse
Affiliation(s)
- Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| | - Elizabeth A Fry
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| |
Collapse
|
109
|
Ying SY, Fang W, Lin SL. The miR-302-Mediated Induction of Pluripotent Stem Cells (iPSC): Multiple Synergistic Reprogramming Mechanisms. Methods Mol Biol 2018; 1733:283-304. [PMID: 29435941 DOI: 10.1007/978-1-4939-7601-0_23] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pluripotency represents a unique feature of embryonic stem cells (ESCs). To generate ESC-like-induced pluripotent stem cells (iPSCs) derived from somatic cells, the cell genome needs to be reset and reprogrammed to express the ESC-specific transcriptome. Numerous studies have shown that genomic DNA demethylation is required for epigenetic reprogramming of somatic cell nuclei to form iPSCs; yet, the mechanism remains largely unclear. In ESCs, the reprogramming process goes through two critical stages: germline and zygotic demethylation, both of which erase genomic DNA methylation sites and hence allow for different gene expression patterns to be reset into a pluripotent state. Recently, miR-302, an ESC-specific microRNA (miRNA), was found to play an essential role in four aspects of this reprogramming mechanism-(1) initiating global genomic DNA demethylation, (2) activating ESC-specific gene expression, (3) inhibiting developmental signaling, and (4) preventing stem cell tumorigenicity. In this review, we will summarize miR-302 functions in all four reprogramming aspects and further discuss how these findings may improve the efficiency and safety of the current iPSC technology.
Collapse
Affiliation(s)
- Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - William Fang
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shi-Lung Lin
- Division of Regenerative Medicine, WJWU & LYNN (W&L) Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| |
Collapse
|
110
|
Mechanism and Method for Generating Tumor-Free iPS Cells Using Intronic MicroRNA miR-302 Induction. Methods Mol Biol 2018; 1733:265-282. [PMID: 29435940 DOI: 10.1007/978-1-4939-7601-0_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Today's researchers generating induced pluripotent stem cells (iPS cells or iPSCs) usually consider their pluripotency rather than potential tumorigenicity. Oncogenic factors such as c-Myc and Klf4 are frequently used to boost the survival and proliferative rates of iPSCs, creating an inevitable problem of tumorigenicity that hinders the therapeutic usefulness of these iPSCs. To prevent stem cell tumorigenicity, we have examined mechanisms by which the cell cycle genes are regulated in embryonic stem cells (ESCs). Naturally, ESCs possess two unique stemness properties: pluripotent differentiation into almost all cell types and unlimited self-renewal without the risk of tumor formation. These two features are also important for the use of ESCs or iPSCs in therapy. Currently, despite overwhelming reports describing iPSC pluripotency, there is no report of any tumor prevention mechanism in either ESCs or iPSCs. To this, our studies have revealed for the first time that an ESC-specific microRNA (miRNA), miR-302, regulates human iPSC tumorigenicity through cosuppression of both cyclin E-CDK2 and cyclin D-CDK4/6 cell cycle pathways during G1-S phase transition. Moreover, miR-302 also silences BMI-1, a cancer stem cell gene marker, to promote the expression of two senescence-associated tumor suppressor genes, p16Ink4a and p14/p19Arf. Together, the combinatory effects of inhibiting G1-S cell cycle transition and increasing p16/p14(p19) expression result in an attenuated cell cycle rate similar to that of 2-to-8-cell-stage embryonic cells in early zygotes (20-24 h/cycle), which is however slower than the fast proliferation rate of iPSCs induced by the four defined factors Oct4-Sox2-Klf4-c-Myc (12-16 h/cycle). These findings provide a means to control iPSC tumorigenicity and improve the safety of iPSCs for the therapeutic use. In this chapter, we review the mechanism underlying miR-302-mediated tumor suppression and then demonstrate how to apply this mechanism to generate tumor-free iPSCs. The same strategy may also be used to prevent ESC tumorigenicity.
Collapse
|
111
|
Sherr CJ, Sicinski P. The D-Type Cyclins: A Historical Perspective. D-TYPE CYCLINS AND CANCER 2018. [DOI: 10.1007/978-3-319-64451-6_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
112
|
Abstract
Peptides encoded by short open reading frames (sORFs) are usually defined as peptides ≤100 aa long. Usually sORFs were ignored by automatic genome annotation programs due to the high probability of false discovery. However, improved computational tools along with a high-throughput RIBO-seq approach identified a myriad of translated sORFs. Their importance becomes evident as we are gaining experimental validation of their diverse cellular functions. This Review examines various computational and experimental approaches of sORFs identification as well as provides the summary of our current knowledge of their functional roles in cells.
Collapse
Affiliation(s)
- Anastasia Chugunova
- Lomonosov Moscow State University , Department of Chemistry and A.N. Belozersky Institute of Physico-Chemical Biology, Moscow 119992, Russia.,Skolkovo Institute of Science and Technology , Skolkovo, Moscow Region 143025, Russia
| | - Tsimafei Navalayeu
- Lomonosov Moscow State University , Department of Chemistry and A.N. Belozersky Institute of Physico-Chemical Biology, Moscow 119992, Russia
| | - Olga Dontsova
- Lomonosov Moscow State University , Department of Chemistry and A.N. Belozersky Institute of Physico-Chemical Biology, Moscow 119992, Russia.,Skolkovo Institute of Science and Technology , Skolkovo, Moscow Region 143025, Russia
| | - Petr Sergiev
- Lomonosov Moscow State University , Department of Chemistry and A.N. Belozersky Institute of Physico-Chemical Biology, Moscow 119992, Russia.,Skolkovo Institute of Science and Technology , Skolkovo, Moscow Region 143025, Russia
| |
Collapse
|
113
|
Sewastianik T, Jiang M, Sukhdeo K, Patel SS, Roberts K, Kang Y, Alduaij A, Dennis PS, Lawney B, Liu R, Song Z, Xiong J, Zhang Y, Lemieux ME, Pinkus GS, Rich JN, Weinstock DM, Mullighan CG, Sharpless NE, Carrasco RD. Constitutive Ras signaling and Ink4a/Arf inactivation cooperate during the development of B-ALL in mice. Blood Adv 2017; 1:2361-2374. [PMID: 29296886 PMCID: PMC5729631 DOI: 10.1182/bloodadvances.2017012211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/24/2017] [Indexed: 11/20/2022] Open
Abstract
Despite recent advances in treatment, human precursor B-cell acute lymphoblastic leukemia (B-ALL) remains a challenging clinical entity. Recent genome-wide studies have uncovered frequent genetic alterations involving RAS pathway mutations and loss of the INK4A/ARF locus, suggesting their important role in the pathogenesis, relapse, and chemotherapy resistance of B-ALL. To better understand the oncogenic mechanisms by which these alterations might promote B-ALL and to develop an in vivo preclinical model of relapsed B-ALL, we engineered mouse strains with induced somatic KrasG12D pathway activation and/or loss of Ink4a/Arf during early stages of B-cell development. Although constitutive activation of KrasG12D in B cells induced prominent transcriptional changes that resulted in enhanced proliferation, it was not sufficient by itself to induce development of a high-grade leukemia/lymphoma. Instead, in 40% of mice, these engineered mutations promoted development of a clonal low-grade lymphoproliferative disorder resembling human extranodal marginal-zone lymphoma of mucosa-associated lymphoid tissue or lymphoplasmacytic lymphoma. Interestingly, loss of the Ink4a/Arf locus, apart from reducing the number of apoptotic B cells broadly attenuated KrasG12D-induced transcriptional signatures. However, combined Kras activation and Ink4a/Arf inactivation cooperated functionally to induce a fully penetrant, highly aggressive B-ALL phenotype resembling high-risk subtypes of human B-ALL such as BCR-ABL and CRFL2-rearranged. Ninety percent of examined murine B-ALL tumors showed loss of the wild-type Ink4a/Arf locus without acquisition of highly recurrent cooperating events, underscoring the role of Ink4a/Arf in restraining Kras-driven oncogenesis in the lymphoid compartment. These data highlight the importance of functional cooperation between mutated Kras and Ink4a/Arf loss on B-ALL.
Collapse
Affiliation(s)
- Tomasz Sewastianik
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Meng Jiang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
- Department of Surgical Oncology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kumar Sukhdeo
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Sanjay S Patel
- Department of Pathology, Brigham & Women's Hospital, Boston, MA
| | - Kathryn Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Yue Kang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Ahmad Alduaij
- Pathology and Laboratory Medicine Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Peter S Dennis
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Brian Lawney
- Center for Computational Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruiyang Liu
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Zeyuan Song
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Jessie Xiong
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina, Chapel Hill, NC
| | - Yunyu Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; and
| | | | | | - Jeremy N Rich
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Cleveland, OH
| | - David M Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; and
| | | | - Norman E Sharpless
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina, Chapel Hill, NC
| | - Ruben D Carrasco
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pathology, Brigham & Women's Hospital, Boston, MA
| |
Collapse
|
114
|
Casadevall D, Kilian AY, Bellmunt J. The prognostic role of epigenetic dysregulation in bladder cancer: A systematic review. Cancer Treat Rev 2017; 61:82-93. [PMID: 29121502 DOI: 10.1016/j.ctrv.2017.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/14/2017] [Accepted: 10/16/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Despite adequate treatment and follow-up, around one fifth of patients with localized bladder cancer will present with disease progression. Adequate prognostic biomarkers are lacking to define patients who are at risk. Mutations in chromatin remodeling genes are more frequently found in bladder cancer than in any other solid tumor. However, the prognostic relevance of epigenetic dysregulation has not been established and may offer an opportunity for biomarker discovery. METHODS Looking for prognostic epigenetic factors, we performed a comprehensive PubMed search using keywords such as "bladder cancer", "chromatin remodeling", "gene methylation" and "epigenetics". We only included studies reporting on the association of epigenetic markers with prognostic outcomes such as recurrence, progression or survival. RESULTS Of 1113 results, 87 studies met the inclusion criteria, which represented a total of 85 epigenetic markers with potential prognostic relevance. No prospective studies were identified. Seventy-three percent (64/87) of the studies involved mixed cohorts of muscle invasive and non-muscle invasive bladder cancer. Promoter methylation of genes with putative prognostic value affected cellular processes such as cell cycle, apoptosis, cell-adhesion or migration, as well as critical pathways such as MAP-kinase or Wnt. Alteration of chromatin regulatory elements suggest a prognostic relevance alterations leading to a predominantly silenced chromatin state. CONCLUSIONS The prognostic impact of epigenetic alterations in bladder cancer is still unclear. Prospective evaluation of methylation marks and chromatin remodeling gene alterations using consistent methods and criteria is warranted.
Collapse
Affiliation(s)
- David Casadevall
- Cancer Research Program, PSMAR-IMIM (Hospital del Mar Medical Research Institute), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain.
| | | | - Joaquim Bellmunt
- Cancer Research Program, PSMAR-IMIM (Hospital del Mar Medical Research Institute), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain; Dana-Farber Cancer Institute, 450 Brookline Ave, DANA 1230, Boston, MA 02215, USA.
| |
Collapse
|
115
|
Samandi S, Roy AV, Delcourt V, Lucier JF, Gagnon J, Beaudoin MC, Vanderperre B, Breton MA, Motard J, Jacques JF, Brunelle M, Gagnon-Arsenault I, Fournier I, Ouangraoua A, Hunting DJ, Cohen AA, Landry CR, Scott MS, Roucou X. Deep transcriptome annotation enables the discovery and functional characterization of cryptic small proteins. eLife 2017; 6:27860. [PMID: 29083303 PMCID: PMC5703645 DOI: 10.7554/elife.27860] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 10/29/2017] [Indexed: 01/10/2023] Open
Abstract
Recent functional, proteomic and ribosome profiling studies in eukaryotes have concurrently demonstrated the translation of alternative open-reading frames (altORFs) in addition to annotated protein coding sequences (CDSs). We show that a large number of small proteins could in fact be coded by these altORFs. The putative alternative proteins translated from altORFs have orthologs in many species and contain functional domains. Evolutionary analyses indicate that altORFs often show more extreme conservation patterns than their CDSs. Thousands of alternative proteins are detected in proteomic datasets by reanalysis using a database containing predicted alternative proteins. This is illustrated with specific examples, including altMiD51, a 70 amino acid mitochondrial fission-promoting protein encoded in MiD51/Mief1/SMCR7L, a gene encoding an annotated protein promoting mitochondrial fission. Our results suggest that many genes are multicoding genes and code for a large protein and one or several small proteins. Proteins are often referred to as the workhorses of the cell, and these molecules affect all aspects of human health and disease. Thus, deciphering the entire set of proteins made by an organism is often an important challenge for biologists. Genes contain the instructions to make a protein, but first they must be copied into a molecule called an mRNA. The part of the mRNA that actually codes for the protein is referred to as an open reading frame (or ORF for short). For many years, most scientists assumed that, except for in bacteria, each mature mRNA in an organism has just a single functional ORF, and that this was generally the longest possible ORF within the mRNA. Many also assumed that RNAs copied from genes that had been labelled as “non-coding” or as “pseudogenes” did not contain functional ORFs. Yet, new ORFs encoding small proteins were recently discovered in RNAs (or parts of RNA) that had previously been annotated as non-coding. Working out what these small proteins actually do will require scientists being able to find more of these overlooked ORFs. The RNAs produced by many organisms – from humans and mice to fruit flies and yeast – have been catalogued and the data stored in publicly accessible databases. Samandi, Roy et al. have now taken a fresh look at the data for nine different organisms, and identified several thousand examples of possibly overlooked ORFs, which they refer to as “alternative ORFs”. This included more than 180,000 from humans. Further analysis of other datasets that captured details of the proteins actually produced in human cells uncovered thousands of small proteins encoded by the predicted alternative ORFs. Many of the so-called alternative proteins also resembled parts of other proteins that have a known activity or function. Lastly, Samandi, Roy et al. focused on two alternative proteins and showed that they both might affect the activity of the proteins coded within the main ORF in their respective genes. These findings reveal new details about the different proteins encoded within the genes of humans and other organisms, including that many mRNAs encode more that one protein. The implications and applications of this research could be far-reaching, and may help scientists to better understand how genes work in both health and disease.
Collapse
Affiliation(s)
- Sondos Samandi
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada
| | - Annie V Roy
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada
| | - Vivian Delcourt
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada.,INSERM U1192, Laboratoire Protéomique, Réponse Inflammatoire & Spectrométrie de Masse (PRISM) F-59000 Lille, Université de Lille, Lille, France
| | - Jean-François Lucier
- Department of Biology, Université de Sherbrooke, Québec, Canada.,Center for Scientific computing, Information Technologies Services,, Université de Sherbrooke, Québec, Canada
| | - Jules Gagnon
- Department of Biology, Université de Sherbrooke, Québec, Canada.,Center for Scientific computing, Information Technologies Services,, Université de Sherbrooke, Québec, Canada
| | - Maxime C Beaudoin
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada
| | - Benoît Vanderperre
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada
| | - Marc-André Breton
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada
| | - Julie Motard
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada
| | - Jean-François Jacques
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada
| | - Mylène Brunelle
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada
| | - Isabelle Gagnon-Arsenault
- PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada.,Département de biochimie, microbiologie et bioinformatique, Université Laval, Québec, Canada.,IBIS, Université Laval, Québec, Canada
| | - Isabelle Fournier
- INSERM U1192, Laboratoire Protéomique, Réponse Inflammatoire & Spectrométrie de Masse (PRISM) F-59000 Lille, Université de Lille, Lille, France
| | - Aida Ouangraoua
- Department of Computer Science, Université de Sherbrooke, Québec, Canada
| | - Darel J Hunting
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Québec, Canada
| | - Alan A Cohen
- Department of Family Medicine, Université de Sherbrooke, Québec, Canada
| | - Christian R Landry
- PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada.,Département de biochimie, microbiologie et bioinformatique, Université Laval, Québec, Canada.,IBIS, Université Laval, Québec, Canada
| | - Michelle S Scott
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada
| | - Xavier Roucou
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada.,PROTEO, Québec Network for Research on Protein Function, Structure and Engineering, Québec, Canada
| |
Collapse
|
116
|
Lujambio A. To clear, or not to clear (senescent cells)? That is the question. Bioessays 2017; 38 Suppl 1:S56-64. [PMID: 27417123 DOI: 10.1002/bies.201670910] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 12/22/2022]
Abstract
Cellular senescence is an anti-proliferative program that restricts the propagation of cells subjected to different kinds of stress. Cellular senescence was initially described as a cell-autonomous tumor suppressor mechanism that triggers an irreversible cell cycle arrest that prevents the proliferation of damaged cells at risk of neoplastic transformation. However, discoveries during the last decade have established that senescent cells can also impact the surrounding tissue microenvironment and the neighboring cells in a non-cell-autonomous manner. These non-cell-autonomous activities are, in part, mediated by the selective secretion of extracellular matrix degrading enzymes, cytokines, chemokines and immune modulators, which collectively constitute the senescence-associated secretory phenotype. One of the key functions of the senescence-associated secretory phenotype is to attract immune cells, which in turn can orchestrate the elimination of senescent cells. Interestingly, the clearance of senescent cells seems to be critical to dictate the net effects of cellular senescence. As a general rule, the successful elimination of senescent cells takes place in processes that are considered beneficial, such as tumor suppression, tissue remodeling and embryonic development, while the chronic accumulation of senescent cells leads to more detrimental consequences, namely, cancer and aging. Nevertheless, exceptions to this rule may exist. Now that cellular senescence is in the spotlight for both anti-cancer and anti-aging therapies, understanding the precise underpinnings of senescent cell removal will be essential to exploit cellular senescence to its full potential.
Collapse
Affiliation(s)
- Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
117
|
Lambert CA, Garbacki N, Colige AC. Chemotherapy induces alternative transcription and splicing: Facts and hopes for cancer treatment. Int J Biochem Cell Biol 2017; 91:84-97. [PMID: 28433505 DOI: 10.1016/j.biocel.2017.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/04/2017] [Accepted: 04/15/2017] [Indexed: 01/14/2023]
|
118
|
Kastenhuber ER, Lowe SW. Putting p53 in Context. Cell 2017; 170:1062-1078. [PMID: 28886379 DOI: 10.1016/j.cell.2017.08.028] [Citation(s) in RCA: 1350] [Impact Index Per Article: 168.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/09/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
TP53 is the most frequently mutated gene in human cancer. Functionally, p53 is activated by a host of stress stimuli and, in turn, governs an exquisitely complex anti-proliferative transcriptional program that touches upon a bewildering array of biological responses. Despite the many unveiled facets of the p53 network, a clear appreciation of how and in what contexts p53 exerts its diverse effects remains unclear. How can we interpret p53's disparate activities and the consequences of its dysfunction to understand how cell type, mutation profile, and epigenetic cell state dictate outcomes, and how might we restore its tumor-suppressive activities in cancer?
Collapse
Affiliation(s)
- Edward R Kastenhuber
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Scott W Lowe
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, New York, NY 10065, USA.
| |
Collapse
|
119
|
Lin X, Wei F, Whyte P, Tang D. BMI1 reduces ATR activation and signalling caused by hydroxyurea. Oncotarget 2017; 8:89707-89721. [PMID: 29163782 PMCID: PMC5685703 DOI: 10.18632/oncotarget.21111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 09/03/2017] [Indexed: 01/31/2023] Open
Abstract
BMI1 facilitates DNA damage response (DDR) induced by double strand DNA breaks; however, it remains unknown whether BMI1 functions in single strand DNA (ssDNA) lesions-initiated DDR. We report here that BMI1 reduces hydroxyurea-elicited ATR activation, thereby reducing the S-phase checkpoints. Hydroxyurea induces ssDNA lesions, which activate ATR through binding TOPBP1 as evidenced by phosphorylation of ATR at threonine 1989 (ATRpT1989). ATR subsequently phosphorylates H2AX at serine 139 (γH2AX) and CHK1 at serine 345 (CHK1pS345), leading to phosphorylation of CDK1 at tyrosine 15 (CDK1pY15) and S-phase arrest. BMI1 overexpression reduced γH2AX, CHK1pS345, CDK1pY15, S-phase arrest, and ATR activation in HU-treated MCF7 and DU145 cells, whereas BMI1 knockdown enhanced these events. BMI1 contains a ring finger, helix-turn, proline/serine domain and two nuclear localization signals (NLS). Individual deletion of these domains did not abolish BMI1-derived reductions of CHK1pS345 in MCF7 cells following HU exposure, suggesting that these structural features are not essential for BMI1 to attenuate ATR-mediated CHK1pS345. BMI1 interacts with both TOPBP1 and ATR. Furthermore, all of our BMI1 mutants associate with endogenous TOPBP1. It has previously been established that association of TOPBP1 and ATR is required for ATR activation. Thus, our results suggest that BMI1 decreases ATR activation through a mechanism that involves binding to TOPBP1 and/or ATR.
Collapse
Affiliation(s)
- Xiaozeng Lin
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada.,The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - Fengxiang Wei
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada.,The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada.,The Genetics Laboratory, Longgang District Maternity and Child Healthcare Hospital, Longgang District, Shenzhen, Guangdong, P.R. China
| | - Peter Whyte
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Damu Tang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Father Sean O'Sullivan Research Institute, Hamilton, Ontario, Canada.,The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| |
Collapse
|
120
|
Dysfunction of the circadian transcriptional factor CLOCK in mice resists chemical carcinogen-induced tumorigenesis. Sci Rep 2017; 7:9995. [PMID: 28855649 PMCID: PMC5577256 DOI: 10.1038/s41598-017-10599-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/10/2017] [Indexed: 01/03/2023] Open
Abstract
The chronic disruption of circadian rhythms has been implicated in the risk of cancer development in humans and laboratory animals. The gene product CLOCK is a core molecular component of the circadian oscillator, so that mice with a mutated Clock gene (Clk/Clk) exhibit abnormal rhythms in various physiological processes. However, we demonstrated here that Clk/Clk mice resisted chemical carcinogen-induced tumorigenesis by suppressing epidermal growth factor (EGF) receptor-mediated proliferation signals. The repetitive application of 7,12-dimethylbenz[α]anthracene (DMBA) to skin on the back resulted in the significant development of tumors in wild-type mice, whereas chemically-induced tumorigenesis was alleviated in Clk/Clk mice. Although the degree of DMBA-induced DNA damage was not significantly different between wild-type and Clk/Clk mice, EGF receptor-mediated Ras activation was not detected in DMBA-treated Clk/Clk mice. Genetic and biochemical experiments revealed that the suppression of EGF receptor-mediated signal transduction in DMBA-treated Clk/Clk mice was associated with the expression of the cellular senescence factor p16INK4a. These results suggest an uncovered role for CLOCK in the development of chemical carcinogen-induced primary tumors and offers new preventive strategies.
Collapse
|
121
|
Zhang R, Wu X, Xia X, Khanniche A, Song F, Zhang B, Wang Y, Ge H. OVA12 promotes tumor growth by regulating p53 expression in human cancer cells. Oncotarget 2017; 8:52854-52865. [PMID: 28881777 PMCID: PMC5581076 DOI: 10.18632/oncotarget.17501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 04/10/2017] [Indexed: 11/30/2022] Open
Abstract
Ovarian cancer-associated antigen 12 (OVA12) was first identified in an ovarian carcinoma complementary DNA (cDNA) expression library and has been shown to play an important role in tumor growth. Here, we found that overexpression of OVA12 accelerated tumor growth in different tumor cells, whereas OVA12 depletion was associated with the opposite effect. Moreover, knocking down OVA12 led to a significant increase in the protein levels of p53, and the overexpression of OVA12 significantly decreased endogenous p53 levels. In addition, OVA12 stimulated p53 polyubiquitination and degradation by the proteasome and promoted tumor growth at least partially through the p53 pathway. Taken together, these results indicate that OVA12 is a negative regulator of p53 and that inhibition of OVA12 expression might serve as a therapeutic target to restore tumor suppression.
Collapse
Affiliation(s)
- Renfeng Zhang
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xicai Wu
- Clinical Laboratory, People's Hospital of Rizhao, Rizhao, China
| | - Xiangfeng Xia
- Department of Radiology, The Third People's Hospital of Rizhao, Rizhao, China
| | - Asma Khanniche
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feifei Song
- Department of Pathology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Bingchang Zhang
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Ying Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hailiang Ge
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
122
|
Loss of p16 INK4A stimulates aberrant mitochondrial biogenesis through a CDK4/Rb-independent pathway. Oncotarget 2017; 8:55848-55862. [PMID: 28915557 PMCID: PMC5593528 DOI: 10.18632/oncotarget.19862] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/09/2017] [Indexed: 02/06/2023] Open
Abstract
The tumor suppressor p16INK4A (p16) inhibits cell cycle progression through the CDK4/Rb pathway. We have previously shown that p16 regulates cellular oxidative stress, independent of its role in cell cycle control. We investigated whether loss of p16 had a direct impact on the mitochondria. We found that p16-null primary mouse fibroblasts (PMFs) displayed increased mitochondrial mass and expression of mitochondrial respiratory subunit proteins compared to wild-type (WT) PMFs. These findings in p16-null PMFs were associated with increased expression of the mitochondrial biogenesis transcription factors PRC and TFAM. On the other hand, p16-deficient PMFs demonstrated reduced mitochondrial respiration capacity consistent with electron microscopy findings showing that mitochondria in p16-deficient PMFs have abnormal morphology. Consistent with increased mitochondrial mass and reduced respiratory capacity, p16-deficient PMFs generated increased mitochondrial superoxide. One biological consequence of elevated ROS in p16-deficient PMFs was enhanced migration, which was reduced by the ROS scavenger N-acetylcysteine. Finally, p16-deficient PMFs displayed increased mitochondrial membrane potential, which was also required for their enhanced migration. The mitochondrial and migration phenotype was restored in p16-deficient PMFs by forced expression of p16. Similarly, over-expression of p16 in human melanocytes and A375 melanoma cells led to decreased expression of some mitochondrial respiratory proteins, enhanced respiration, and decreased migration. Inhibition of Rb phosphorylation in melanocytes and melanoma cells, either by addition of chemical CDK4 inhibitors or RNAi-mediated knockdown of CDK4, did not mimic the effects of p16 loss. These results suggest that p16 regulates mitochondrial biogenesis and function, which is independent of the canonical CDK4/Rb pathway.
Collapse
|
123
|
Ahmad I, Guroji P, DeBrot AH, Manapragada PP, Katiyar SK, Elmets CA, Yusuf N. Loss of INK4a/Arf gene enhances ultraviolet radiation-induced cutaneous tumor development. Exp Dermatol 2017; 26:1018-1025. [PMID: 28418604 DOI: 10.1111/exd.13356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2017] [Indexed: 12/22/2022]
Abstract
The CDKN2A locus encodes for tumor suppressor genes p16INK4a and p14Arf which are frequently inactivated in human skin tumors. The purpose of this study was to determine the relationship between loss of INK4a/Arf activity and inflammation in the development of ultraviolet (UV) radiation-induced skin tumors. Panels of INK4a/Arf-/- mice and wild-type (WT) mice were treated with a single dose of UVB (200 mJ/cm2 ). For long-term studies, these mice were irradiated with UVB (200 mJ/cm2 ) three times weekly for 30 weeks. At the end of the experiment, tissues were harvested from mice and assayed for inflammatory biomarkers and cytokines. A single dose of UVB resulted in a significant increase in reactive oxygen species (ROS) and 8-dihydroxyguanosine (8-oxo-dG) lesions in INK4a/Arf-/- mice compared to WT mice. When subjected to chronic UVB, we found that 100% of INK4a/Arf-/- mice had tumors, whereas there were no tumors in WT controls after 24 weeks of UVB exposure. The increase in tumor development correlated with a significant increase in nuclear factor (NF)-κB, cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2 ) and its receptors both in UVB-exposed skin and in the tumors. A significant increase was seen in inflammatory cytokines in skin samples of INK4a/Arf-/- mice following treatment with chronic UVB radiation. Furthermore, significantly more CD11b+ Gr1+ myeloid cells were present in UVB-exposed INK4a/Arf-/- mice compared to WT mice. Our data indicate that by targeting UVB-induced inflammation, it may be possible to prevent UVB-induced skin tumors in individuals that carry CDKN2A mutation.
Collapse
Affiliation(s)
- Israr Ahmad
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Purushotham Guroji
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amanda H DeBrot
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Padma P Manapragada
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Santosh K Katiyar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA.,Veteran Affairs Medical Center, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Craig A Elmets
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA.,Veteran Affairs Medical Center, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nabiha Yusuf
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA.,Veteran Affairs Medical Center, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
124
|
Sun H, Shi JX, Zhang HF, Xing MT, Li P, Dai LP, Luo CL, Wang X, Wang P, Ye H, Li LX, Zhang JY. Serum autoantibodies against a panel of 15 tumor-associated antigens in the detection of ovarian cancer. Tumour Biol 2017; 39:1010428317699132. [PMID: 28618923 DOI: 10.1177/1010428317699132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In this study, enzyme-linked immunosorbent assay has been used to examine the frequencies of serum autoantibodies against two candidate tumor-associated antigens intensively selected from the Human Protein Atlas database, in combination with 13 tumor-associated antigens available from our lab in sera from 44 OC patients and 50 normal healthy controls. Conventional evaluation (mean + 3SD as the cutoff value to determine a positive reactivity), receiver operating characteristic curve analyses, and classification tree analysis were further used to evaluate the diagnostic performance of autoantibodies against these tumor-associated antigens (anti-tumor-associated antigens) in ovarian cancer. For single anti-tumor-associated antigen, when the cutoff values were set as mean + 3SD of normal healthy controls, NPM1, MDM2, PLAT, p53, and c-Myc could achieve sensitivity higher than 20% at 98% specificity. Combinational utilization of autoantibodies against MDM2, PLAT, NPM1, 14-3-3 Zeta, p53, and RalA achieved the optimal diagnostic performance with 72.7% sensitivity at 96% specificity. Receiver operating characteristic curve analysis showed that the area under the receiver operating characteristic curves of autoantibodies against c-Myc, NPM1, MDM2, p16, p53, and 14-3-3 Zeta were greater than 0.80. This indicated that these tumor-associated antigens held high potential to serve as diagnostic biomarkers in ovarian cancer detection. Decision tree analysis indicated that anti-c-Myc held high potential in the detection of ovarian cancer. Further studies are warranted to validate the diagnostic performance of these anti-tumor-associated antigens with high area under the receiver operating characteristic curve, including autoantibodies against c-Myc, MDM2, PLAT, NPM1, 14-3-3 Zeta, p53, and RalA.
Collapse
Affiliation(s)
- Hao Sun
- 1 Affiliated Cancer Hospital of Zhengzhou University, College of Public Health, Zhengzhou University, Zhengzhou, China.,2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Jian-Xiang Shi
- 1 Affiliated Cancer Hospital of Zhengzhou University, College of Public Health, Zhengzhou University, Zhengzhou, China.,2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Hong-Fei Zhang
- 1 Affiliated Cancer Hospital of Zhengzhou University, College of Public Health, Zhengzhou University, Zhengzhou, China.,2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Meng-Tao Xing
- 2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Pei Li
- 2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA.,3 Henan Academy of Medical and Pharmaceutical Sciences and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Li-Ping Dai
- 2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA.,3 Henan Academy of Medical and Pharmaceutical Sciences and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Cheng-Lin Luo
- 2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Xiao Wang
- 2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA.,3 Henan Academy of Medical and Pharmaceutical Sciences and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Peng Wang
- 1 Affiliated Cancer Hospital of Zhengzhou University, College of Public Health, Zhengzhou University, Zhengzhou, China.,3 Henan Academy of Medical and Pharmaceutical Sciences and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hua Ye
- 1 Affiliated Cancer Hospital of Zhengzhou University, College of Public Health, Zhengzhou University, Zhengzhou, China.,3 Henan Academy of Medical and Pharmaceutical Sciences and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Liu-Xia Li
- 2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA.,3 Henan Academy of Medical and Pharmaceutical Sciences and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian-Ying Zhang
- 1 Affiliated Cancer Hospital of Zhengzhou University, College of Public Health, Zhengzhou University, Zhengzhou, China.,2 Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA.,3 Henan Academy of Medical and Pharmaceutical Sciences and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
125
|
Small mitochondrial Arf (smArf) protein corrects p53-independent developmental defects of Arf tumor suppressor-deficient mice. Proc Natl Acad Sci U S A 2017; 114:7420-7425. [PMID: 28652370 DOI: 10.1073/pnas.1707292114] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mouse p19Arf (human p14ARF) tumor suppressor protein, encoded in part from an alternative reading frame of the Ink4a (Cdkn2a) gene, inhibits the Mdm2 E3 ubiquitin ligase to activate p53. Arf is not expressed in most normal tissues of young mice but is induced by high thresholds of aberrant hyperproliferative signals, thereby activating p53 in incipient tumor cells that have experienced oncogene activation. The single Arf mRNA encodes two distinct polypeptides, including full-length p19Arf and N-terminally truncated and unstable p15smArf ("small mitochondrial Arf") initiated from an internal in-frame AUG codon specifying methionine-45. Interactions of p19Arf with Mdm2, or separately with nucleophosmin (NPM, B23) that localizes and stabilizes p19Arf within the nucleolus, require p19Arf N-terminal amino acids that are not present within p15smArf We have generated mice that produce either smARF alone or M45A-mutated (smArf-deficient) full-length p19Arf proteins. BCR-ABL-expressing pro/pre-B cells producing smArf alone are as oncogenic as their Arf-null counterparts in generating acute lymphoblastic leukemia when infused into unconditioned syngeneic mice. In contrast, smArf-deficient cells from mice of the ArfM45A strain are as resistant as wild-type Arf+/+ cells to comparable oncogenic challenge and do not produce tumors. Apart from being prone to tumor development, Arf-null mice are blind, and their male germ cells exhibit defects in meiotic maturation and sperm production. Although ArfM45A mice manifest the latter defects, smArf alone remarkably rescues both of these p53-independent developmental phenotypes.
Collapse
|
126
|
Tanaka T, Ochi H, Takahashi S, Ueno N, Taira M. Genes coding for cyclin-dependent kinase inhibitors are fragile in Xenopus. Dev Biol 2017; 426:291-300. [PMID: 27393661 DOI: 10.1016/j.ydbio.2016.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 06/16/2016] [Accepted: 06/16/2016] [Indexed: 11/27/2022]
Abstract
Cell proliferation is strictly regulated by the dosage balance among cell-cycle regulators such as CDK/cyclin complexes and CDK-Inhibitors. Even in the allotetraploid genome of Xenopus laevis, the dosage balance must be maintained for animals to stay alive, and the duplicated homeologous genes seem to have gradually changed, through evolution, resulting in the best genes for them to thrive. In the Xenopus laevis genome, while homeologous gene pairs of CDKs are fundamentally maintained and a few cyclin genes are amplified, homeologous gene pairs of the important CDK-Inhibitors, CDKn1c and CDKn2a, are deleted from chromosomes L and S. Although losses of CDKn1c and CDKn2a can lead to diseases in humans, their loss in X. laevis does not affect the animals' health. Also, another gene coding CDKn1b is lost besides CDKn1c and CDKn2a in the genome of Xenopus tropicalis. These findings suggest a high resistance of Xenopus to diseases. We also found that CDKn2c.S expression is higher than that of CDKn2c.L, and a conserved noncoding sequence (CNS) of CDKn2c genomic loci on X. laevis chromosome S and X. tropicalis has an enhancement activity in regulating the different expression. These findings together indicate a surprising fragility of CDK inhibitor gene loci in the Xenopus genome in spite of their importance, and may suggest that factors other than CDK-inhibitors decelerate cell-cycling in Xenopus.
Collapse
Affiliation(s)
- Toshiaki Tanaka
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan.
| | - Haruki Ochi
- Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, Yamagata 990-9585, Japan
| | - Shuji Takahashi
- Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Naoto Ueno
- National Institute for Basic Biology, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki 444-8585, Aichi, Japan
| | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
127
|
Fantauzzo KA, Soriano P. Generation of an immortalized mouse embryonic palatal mesenchyme cell line. PLoS One 2017; 12:e0179078. [PMID: 28582446 PMCID: PMC5459506 DOI: 10.1371/journal.pone.0179078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/23/2017] [Indexed: 12/17/2022] Open
Abstract
Palatogenesis is a complex morphogenetic process, disruptions in which result in highly prevalent birth defects in humans. In recent decades, the use of model systems such as genetically-modified mice, mouse palatal organ cultures and primary mouse embryonic palatal mesenchyme (MEPM) cultures has provided significant insight into the molecular and cellular defects underlying cleft palate. However, drawbacks in each of these systems have prevented high-throughput, large-scale studies of palatogenesis in vitro. Here, we report the generation of an immortalized MEPM cell line that maintains the morphology, migration ability, transcript expression and responsiveness to exogenous growth factors of primary MEPM cells, with increased proliferative potential over primary cultures. The immortalization method described in this study will facilitate the generation of palatal mesenchyme cells with an unlimited capacity for expansion from a single genetically-modified mouse embryo and enable mechanistic studies of palatogenesis that have not been possible using primary culture.
Collapse
Affiliation(s)
- Katherine A. Fantauzzo
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Philippe Soriano
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
128
|
Ni S, Ye M, Huang T. Short stature homeobox 2 methylation as a potential noninvasive biomarker in bronchial aspirates for lung cancer diagnosis. Oncotarget 2017; 8:61253-61263. [PMID: 28977861 PMCID: PMC5617421 DOI: 10.18632/oncotarget.18056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/25/2017] [Indexed: 12/16/2022] Open
Abstract
Gene methylation has been frequently observed in lung cancer. However, the use of methylated genes in bronchial aspirates of patients with lung cancer remains to be evaluated. The purpose of this study was to analyze whether the detection of genes with aberrant promoter methylation can be useful noninvasive biomarkers in bronchial aspirates from lung cancer. We found that the methylation status of the cyclin-dependent kinase inhibitor 2A (P16), Ras association domain family 1 isoform (RASSF1A), adenomatous polyposis coli (APC) and short stature homeobox 2 (SHOX2) genes was significantly correlated with lung cancer in bronchial aspirates. The P16, RASSF1A and APC methylation had a bad diagnostic effect in bronchial aspirates of patients with lung cancer compared with non-tumor controls (P16: sensitivity = 0.26, specificity = 0.99, area under the curve (AUC) = 0.67; RASSF1A: sensitivity = 0.40, specificity = 0.99, AUC = 0.66; APC: sensitivity = 0.17, specificity = 0.98, AUC = 0.65). The pooled sensitivity, specificity, and AUC of the SHOX2 methylation were 0.75, 0.94, and 0.94, respectively. Moreover, when squamous cell carcinoma (SCC) was compared to adenocarcinoma (AC), the SHOX2 gene had a significantly higher methylation rate in SCC than in AC (P < 0.001). Methylated P16, RASSF1A, APC and retinoic acid receptor beta2 (RARβ2) genes had similar frequencies in these two histotypes (P > 0.1). Our findings suggest that methylated SHOX2 gene could be a specific and potential noninvasive biomarker using bronchial aspirates for lung cancer diagnosis, especially for SCC.
Collapse
Affiliation(s)
- Shumin Ni
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315020, People's Republic of China
| | - Meng Ye
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315020, People's Republic of China
| | - Tao Huang
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315020, People's Republic of China
| |
Collapse
|
129
|
Modeling the response of a tumor-suppressive network to mitogenic and oncogenic signals. Proc Natl Acad Sci U S A 2017; 114:5337-5342. [PMID: 28484034 DOI: 10.1073/pnas.1702412114] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Intrinsic tumor-suppressive mechanisms protect normal cells against aberrant proliferation. Although cellular signaling pathways engaged in tumor repression have been largely identified, how they are orchestrated to fulfill their function still remains elusive. Here, we built a tumor-suppressive network model composed of three modules responsible for the regulation of cell proliferation, activation of p53, and induction of apoptosis. Numerical simulations show a rich repertoire of network dynamics when normal cells are subject to serum stimulation and adenovirus E1A overexpression. We showed that oncogenic signaling induces ARF and that ARF further promotes p53 activation to inhibit proliferation. Mitogenic signaling activates E2F activators and promotes Akt activation. p53 and E2F1 cooperate to induce apoptosis, whereas Akt phosphorylates p21 to repress caspase activation. These prosurvival and proapoptotic signals compete to dictate the cell fate of proliferation, cell-cycle arrest, or apoptosis. The cellular outcome is also impacted by the kinetic mode (ultrasensitivity or bistability) of p53. When cells are exposed to serum deprivation and recovery under fixed E1A, the shortest starvation time required for apoptosis induction depends on the terminal serum concentration, which was interpreted in terms of the dynamics of caspase-3 activation and cytochrome c release. We discovered that caspase-3 can be maintained active at high serum concentrations and that E1A overexpression sensitizes serum-starved cells to apoptosis. This work elucidates the roles of tumor repressors and prosurvival factors in tumor repression based on a dynamic network analysis and provides a framework for quantitatively exploring tumor-suppressive mechanisms.
Collapse
|
130
|
A comprehensive review of paediatric low-grade diffuse glioma: pathology, molecular genetics and treatment. Brain Tumor Pathol 2017; 34:51-61. [DOI: 10.1007/s10014-017-0282-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/08/2017] [Indexed: 12/13/2022]
|
131
|
Tang J, Xie Y, Xu X, Yin Y, Jiang R, Deng L, Tan Z, Gangarapu V, Tang J, Sun B. Bidirectional transcription of Linc00441 and RB1 via H3K27 modification-dependent way promotes hepatocellular carcinoma. Cell Death Dis 2017; 8:e2675. [PMID: 28300839 PMCID: PMC5386573 DOI: 10.1038/cddis.2017.81] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 01/16/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023]
Abstract
The retinoblastoma gene (RB1), a known tumor-suppressor gene (TSG), was decreased in multiple cancers including hepatocellular carcinoma (HCC). Here we focused on the bidirectional transcripted long noncoding RNA (Linc00441) with neighbor gene RB1 to investigate whether Linc00441 is involved in the suppression of RB1 in HCC. We found that aberrant upregulated intranuclear Linc00441 was reversely correlated with RB1 expression in human HCC samples. The gain- and loss-of-function investigation revealed that Linc00441 could promote the proliferation of HCC cells in vitro and in vivo with an apoptosis suppression and cell cycle rearrangement. Furthermore, RNA pull-down assay indicated the decreased level of RB1 induced by Linc00441 was associated with the incidental methylation by DNMT3A recruited by Linc00441. On the contrary, the transcription factor (TCF-4) enhanced H3K27 acetylation and direct transcription factor for Linc00441 was responsible for the upregulation of Linc00441 in HCC. In conclusion, the epigenetic interaction between Linc00441 and bidirectional transcripted neighbor RB1 may be a de novo theory cutting-point for the inactivation of RB1 in HCC and may serve as targeting site for tumor therapy in the future.
Collapse
Affiliation(s)
- Junwei Tang
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yu Xie
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaoliang Xu
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yin Yin
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Runqiu Jiang
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lei Deng
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhongming Tan
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Venkatanarayana Gangarapu
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jinhai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Beicheng Sun
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
132
|
Sheikh BN, Metcalf D, Voss AK, Thomas T. MOZ and BMI1 act synergistically to maintain hematopoietic stem cells. Exp Hematol 2017; 47:83-97.e8. [DOI: 10.1016/j.exphem.2016.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/30/2016] [Accepted: 10/11/2016] [Indexed: 11/25/2022]
|
133
|
Next-Generation Sequencing Reveals Pathway Activations and New Routes to Targeted Therapies in Cutaneous Metastatic Melanoma. Am J Dermatopathol 2017; 39:1-13. [PMID: 28045747 DOI: 10.1097/dad.0000000000000729] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Comprehensive genomic profiling of clinical samples by next-generation sequencing (NGS) can identify one or more therapy targets for the treatment of metastatic melanoma (MM) with a single diagnostic test. METHODS NGS was performed on hybridization-captured, adaptor ligation-based libraries using DNA extracted from 4 formalin-fixed paraffin-embedded sections cut at 10 microns from 30 MM cases. The exons of 182 cancer-related genes were fully sequenced using the Illumina HiSeq 2000 at an average sequencing depth of 1098X and evaluated for genomic alterations (GAs) including point mutations, insertions, deletions, copy number alterations, and select gene fusions/rearrangements. Clinically relevant GAs (CRGAs) were defined as those identifying commercially available targeted therapeutics or therapies in registered clinical trials. RESULTS The 30 American Joint Committee on Cancer Stage IV MM included 17 (57%) male and 13 (43%) female patients with a mean age of 59.5 years (range 41-83 years). All MM samples had at least 1 GA, and an average of 2.7 GA/sample (range 1-7) was identified. The mean number of GA did not differ based on age or sex; however, on average, significantly more GAs were identified in amelanotic and poorly differentiated MM. GAs were most commonly identified in BRAF (12 cases, 40%), CDKN2A (6 cases, 20%), NF1 (8 cases, 26.7%), and NRAS (6 cases, 20%). CRGAs were identified in all patients, and represented 77% of the GA (64/83) detected. The median and mean CRGAs per tumor were 2 and 2.1, respectively (range 1-7). CONCLUSION Comprehensive genomic profiling of MM, using a single diagnostic test, uncovers an unexpectedly high number of CRGA that would not be identified by standard of care testing. Moreover, NGS has the potential to influence therapy selection and can direct patients to enter relevant clinical trials evaluating promising targeted therapies.
Collapse
|
134
|
Laporte AN, Barrott JJ, Yao RJ, Poulin NM, Brodin BA, Jones KB, Underhill TM, Nielsen TO. HDAC and Proteasome Inhibitors Synergize to Activate Pro-Apoptotic Factors in Synovial Sarcoma. PLoS One 2017; 12:e0169407. [PMID: 28056055 PMCID: PMC5215898 DOI: 10.1371/journal.pone.0169407] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/16/2016] [Indexed: 12/29/2022] Open
Abstract
Conventional cytotoxic therapies for synovial sarcoma provide limited benefit, and no drugs specifically targeting its driving SS18-SSX fusion oncoprotein are currently available. Patients remain at high risk for early and late metastasis. A high-throughput drug screen consisting of over 900 tool compounds and epigenetic modifiers, representing over 100 drug classes, was undertaken in a panel of synovial sarcoma cell lines to uncover novel sensitizing agents and targetable pathways. Top scoring drug categories were found to be HDAC inhibitors and proteasomal targeting agents. We find that the HDAC inhibitor quisinostat disrupts the SS18-SSX driving protein complex, thereby reestablishing expression of EGR1 and CDKN2A tumor suppressors. In combination with proteasome inhibition, HDAC inhibitors synergize to decrease cell viability and elicit apoptosis. Quisinostat inhibits aggresome formation in response to proteasome inhibition, and combination treatment leads to elevated endoplasmic reticulum stress, activation of pro-apoptotic effector proteins BIM and BIK, phosphorylation of BCL-2, increased levels of reactive oxygen species, and suppression of tumor growth in a murine model of synovial sarcoma. This study identifies and provides mechanistic support for a particular susceptibility of synovial sarcoma to the combination of quisinostat and proteasome inhibition.
Collapse
Affiliation(s)
- Aimée N. Laporte
- Faculty of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jared J. Barrott
- Department of Orthopaedics, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Ren Jie Yao
- Faculty of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neal M. Poulin
- Faculty of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bertha A. Brodin
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kevin B. Jones
- Department of Orthopaedics, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - T. Michael Underhill
- Department of Cellular and Physiological Sciences, Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Torsten O. Nielsen
- Faculty of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
135
|
Wang Y, Chen G, Cao R, Li J, He L, Guo X, Liang J, Shi P, Zhou Y, Xu B. Allogeneic hematopoietic stem cell transplantation improves the prognosis of p16-deleted adult patients with acute lymphoblastic leukemia. Pharmacogenomics 2017; 18:77-84. [PMID: 27967319 DOI: 10.2217/pgs-2016-0075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The prognostic value of CDKN2A inactivation in adult patients with acute lymphoblastic leukemia (ALL) is still under debate, and the role of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for adult ALL with p16 deletion remains to be evaluated. Materials & methods: This study analyzed the clinical implications of p16 deletion in adult ALL and investigated the efficacy of allo-HSCT in patients with p16 deletion. Results: Deletion of p16 was identified in 38.4% of the adult ALL patients, and the prevalences of hemizygous deletion, homozygous deletion and mixed hemi/homozygous of p16 were 22.1, 11.6 and 5.5%, respectively. The prevalence of p16 deletion was 39.7% in B-lineage ALL and 33.3% in T-lineage ALL. Deletion of p16 was significantly associated with higher white blood cell count (p = 0.032) and lower platelets (p = 0.023) but was not related to age, sex, percentage of bone marrow blasts, hepatosplenomegaly, CNS leukemia rate, first complete remission and relapse rate (p > 0.05). Deletion of p16 was significantly correlated with poor outcome in terms of event-free survival (EFS; p = 0.028) and overall survival (OS; p = 0.033). Twenty-two of the 33 patients with p16 deletion received allo-HSCT treatment. Patients with p16 deletion after allo-HSCT experienced higher EFS and OS than those without (52.9 vs 0%, p < 0.001; 46.8 vs 29.1%, p = 0.01, respectively). Multivariate analysis found CNS leukemia and poor response to induction chemotherapy to be the risk factors for EFS and OS, whereas no deletions of p16 and allo-HSCT were favorable factors. Conclusion: Deletion of p16 is a strong adverse prognostic factor in adult ALL. Testing for p16 alterations at diagnosis may help in risk stratification, and we propose to implement testing for p16 deletion in future treatment protocols.
Collapse
Affiliation(s)
- Yan Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Guoshu Chen
- Department of Hematology, Huizhou Municipal Central Hospital, Huizhou, PR China
| | - Rui Cao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jie Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Lingli He
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Xutao Guo
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jiabao Liang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Pengcheng Shi
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Yong Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen, PR China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen, PR China
| |
Collapse
|
136
|
Neault M, Couteau F, Bonneau É, De Guire V, Mallette FA. Molecular Regulation of Cellular Senescence by MicroRNAs: Implications in Cancer and Age-Related Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:27-98. [DOI: 10.1016/bs.ircmb.2017.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
137
|
Mohan A, Asakura A. CDK inhibitors for muscle stem cell differentiation and self-renewal. ACTA ACUST UNITED AC 2017; 6:65-74. [PMID: 28713664 DOI: 10.7600/jpfsm.6.65] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Regeneration of muscle is undertaken by muscle stem cell populations named satellite cells which are normally quiescent or at the G0 phase of the cell cycle. However, upon signals from damaged muscle, satellite cells lose their quiescence, and enter the G1 cell cycle phase to expand the population of satellite cell progenies termed myogenic precursor cells (MPCs). Eventually, MPCs stop their cell cycle and undergo terminal differentiation to form skeletal muscle fibers. Some MPCs retract to quiescent satellite cells as a self-renewal process. Therefore, cell cycle regulation, consisting of satellite cell activation, proliferation, differentiation and self-renewal, is the key event of muscle regeneration. In this review, we summarize up-to-date progress on research about cell cycle regulation of myogenic progenitor cells and muscle stem cells during embryonic myogenesis and adult muscle regeneration, aging, exercise and muscle diseases including muscular dystrophy and muscle fiber atrophy, especially focusing on cyclin-dependent kinase inhibitors (CDKIs).
Collapse
Affiliation(s)
- Amrudha Mohan
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, 2001 6th Street SE, MTRF 4-220, Minneapolis, MN 55455, USA
| | - Atsushi Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, 2001 6th Street SE, MTRF 4-220, Minneapolis, MN 55455, USA
| |
Collapse
|
138
|
Yang X, Yang L, Dai W, Ye B. Role of p14ARF and p15INK4B promoter methylation in patients with lung cancer: a systematic meta-analysis. Onco Targets Ther 2016; 9:6977-6985. [PMID: 27956841 PMCID: PMC5113922 DOI: 10.2147/ott.s117161] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background The cyclin-dependent kinase inhibitors p14ARF and p15INK4B are tumor suppressor genes that have been reported to be silenced through promoter methylation in many human cancers. However, the strength of association between p14ARF or p15INK4B promoter methylation and lung cancer remains unclear. Thus, we first determined whether p14ARF and p15INK4B promoter methylation played a key role in the carcinogenesis of lung cancer. Methods Eligible studies were selected from the online electronic databases. The pooled odds ratios or hazard ratios and 95% confidence intervals were calculated and summarized. Results Finally, 12 studies with 625 lung cancer samples and 488 nontumor samples were included under the fixed-effects model. The pooled odds ratio showed that p14ARF promoter methylation was observed to be significantly higher in non-small-cell lung cancer (NSCLC) than in nontumor samples (P<0.001). No significant correlation was found between p15INK4B promoter methylation and lung cancer (P=0.27). Subgroup analysis of ethnicity revealed that p14ARF promoter methylation was significantly related to the risk of NSCLC in Asian and Caucasian populations. Subgroup analysis of sample type demonstrated that p14ARF promoter methylation was correlated with the risk of NSCLC in tissue samples (P<0.001), but not in bronchoalveolar lavage fluid and blood samples. P14ARF promoter methylation from one study was not significantly correlated with overall survival of patients with NSCLC. Promoter methylation of p14ARF and p15INK4B was not correlated with clinicopathological characteristics, such as gender status, smoking status, tumor differentiation, and tumor stage (P>0.05). Conclusion Our findings suggested that p14ARF promoter methylation may play an important role in the carcinogenesis of lung cancer, but not p15INK4B promoter methylation. Promoter methylation of p14ARF and p15INK4B was not associated with clinicopathological parameters. However, more extensive large-scale studies are essential to further validate our study.
Collapse
Affiliation(s)
- Xinmei Yang
- Department of Oncology, the First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun
| | - Wanrong Dai
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University, Zhejiang
| | - Bo Ye
- Department of Thoracic Surgery, Hangzhou Red Cross Hospital, Hangzhou, People's Republic of China
| |
Collapse
|
139
|
Chalick M, Jacobi O, Pichinuk E, Garbar C, Bensussan A, Meeker A, Ziv R, Zehavi T, Smorodinsky NI, Hilkens J, Hanisch FG, Rubinstein DB, Wreschner DH. MUC1-ARF-A Novel MUC1 Protein That Resides in the Nucleus and Is Expressed by Alternate Reading Frame Translation of MUC1 mRNA. PLoS One 2016; 11:e0165031. [PMID: 27768738 PMCID: PMC5074479 DOI: 10.1371/journal.pone.0165031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 10/05/2016] [Indexed: 01/26/2023] Open
Abstract
Translation of mRNA in alternate reading frames (ARF) is a naturally occurring process heretofore underappreciated as a generator of protein diversity. The MUC1 gene encodes MUC1-TM, a signal-transducing trans-membrane protein highly expressed in human malignancies. Here we show that an AUG codon downstream to the MUC1-TM initiation codon initiates an alternate reading frame thereby generating a novel protein, MUC1-ARF. MUC1-ARF, like its MUC1-TM 'parent’ protein, contains a tandem repeat (VNTR) domain. However, the amino acid sequence of the MUC1-ARF tandem repeat as well as N- and C- sequences flanking it differ entirely from those of MUC1-TM. In vitro protein synthesis assays and extensive immunohistochemical as well as western blot analyses with MUC1-ARF specific monoclonal antibodies confirmed MUC1-ARF expression. Rather than being expressed at the cell membrane like MUC1-TM, immunostaining showed that MUC1-ARF protein localizes mainly in the nucleus: Immunohistochemical analyses of MUC1-expressing tissues demonstrated MUC1-ARF expression in the nuclei of secretory luminal epithelial cells. MUC1-ARF expression varies in different malignancies. While the malignant epithelial cells of pancreatic cancer show limited expression, in breast cancer tissue MUC1-ARF demonstrates strong nuclear expression. Proinflammatory cytokines upregulate expression of MUC1-ARF protein and co-immunoprecipitation analyses demonstrate association of MUC1-ARF with SH3 domain-containing proteins. Mass spectrometry performed on proteins coprecipitating with MUC1-ARF demonstrated Glucose-6-phosphate 1-dehydrogenase (G6PD) and Dynamin 2 (DNM2). These studies not only reveal that the MUC1 gene generates a previously unidentified MUC1-ARF protein, they also show that just like its ‘parent’ MUC1-TM protein, MUC1-ARF is apparently linked to signaling and malignancy, yet a definitive link to these processes and the roles it plays awaits a precise identification of its molecular functions. Comprising at least 524 amino acids, MUC1-ARF is, furthermore, the longest ARF protein heretofore described.
Collapse
Affiliation(s)
- Michael Chalick
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Oded Jacobi
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Edward Pichinuk
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Christian Garbar
- Department of Biopathology, Institut Jean-Godinot, Reims Cedex, France
| | | | - Alan Meeker
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ravit Ziv
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Tania Zehavi
- Department of Pathology, Meir Medical Center, Kfar Saba, Israel
| | | | - John Hilkens
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, Köln, Germany
| | | | - Daniel H. Wreschner
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
- * E-mail:
| |
Collapse
|
140
|
Li Z, Gonzalez CL, Wang B, Zhang Y, Mejia O, Katsonis P, Lichtarge O, Myers JN, El-Naggar AK, Caulin C. Cdkn2asuppresses metastasis in squamous cell carcinomas induced by the gain-of-function mutantp53R172H. J Pathol 2016; 240:224-34. [DOI: 10.1002/path.4770] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/27/2016] [Accepted: 07/08/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Zhongyou Li
- Department of Head and Neck Surgery; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Cassandra L Gonzalez
- Department of Head and Neck Surgery; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Bingbing Wang
- Department of Head and Neck Surgery; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Yuanyuan Zhang
- Department of Head and Neck Surgery; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Olga Mejia
- Department of Head and Neck Surgery; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Panagiotis Katsonis
- Department of Human and Molecular Genetics; Baylor College of Medicine; Houston TX USA
| | - Olivier Lichtarge
- Department of Human and Molecular Genetics; Baylor College of Medicine; Houston TX USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Adel K El-Naggar
- Department of Pathology; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Carlos Caulin
- Department of Head and Neck Surgery; The University of Texas MD Anderson Cancer Center; Houston TX USA
- Department of Genetics; The University of Texas MD Anderson Cancer Center; Houston TX USA
| |
Collapse
|
141
|
Abstract
Cancer is an evolutionary disease, containing the hallmarks of an asexually reproducing unicellular organism subject to evolutionary paradigms. Pancreatic ductal adenocarcinoma (hereafter referred to as pancreatic cancer) is a particularly robust example of this phenomenon. Genomic features indicate that pancreatic cancer cells are selected for fitness advantages when encountering the geographic and resource-depleted constraints of the microenvironment. Phenotypic adaptations to these pressures help disseminated cells to survive in secondary sites, a major clinical problem for patients with this disease. In this Review we gather the wide-ranging aspects of pancreatic cancer research into a single concept rooted in Darwinian evolution, with the goal of identifying novel insights and opportunities for study.
Collapse
Affiliation(s)
- Alvin Makohon-Moore
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Christine A Iacobuzio-Donahue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
142
|
Saito K, Iioka H, Kojima C, Ogawa M, Kondo E. Peptide-based tumor inhibitor encoding mitochondrial p14(ARF) is highly efficacious to diverse tumors. Cancer Sci 2016; 107:1290-301. [PMID: 27317619 PMCID: PMC5021028 DOI: 10.1111/cas.12991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/31/2022] Open
Abstract
p14ARF is one of the major tumor suppressors conventionally identified both as the mdm2‐binding molecule restoring p53 function in the nucleus, and as a nucleophosmin‐binding partner inside the nucleolous to stabilize ribosomal RNA. However, its recently reported mitochondrial localization has pointed to novel properties as a tumor suppressor. At the same time, functional peptides are gaining much attention in nanomedicine for their in vivo utility as non‐invasive biologics. We previously reported the p14ARF‐specific peptide that restored the sensitivity to gefitinib on the gefitinib‐resistant lung cancer cells. Based on the information of this prototype peptide, here we generated the more powerful anti‐tumor peptide “r9‐CatB‐p14 MIS,” which comprises the minimal inhibitory sequence of the mitochondrial targeting p14ARF protein in combination with the proteolytic cleavage site for cathepsin B, which is activated in various tumor cells, fused with the nine‐polyarginine‐domain for cell penetration, and demonstrated its novel action of regulating mitochondrial function in accordance with localization of endogenous p14ARF. The p14 MIS peptide showed a potent tumor inhibiton in vitro and in vivo against not only lung cancer cells but also tumor cells of diverse lineages, via modulating mitochondrial membrane potential, with minimal cytotoxicity to non‐neoplastic cells and tissues. Hence, this mitochondrially targeted p14 peptide agent provides a novel basis for non‐invasive peptide‐based antitumor therapeutics.
Collapse
Affiliation(s)
- Ken Saito
- Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hidekazu Iioka
- Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Chie Kojima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Osaka, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Eisaku Kondo
- Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
143
|
p18 inhibits reprogramming through inactivation of Cdk4/6. Sci Rep 2016; 6:31085. [PMID: 27484146 PMCID: PMC4971472 DOI: 10.1038/srep31085] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/13/2016] [Indexed: 01/03/2023] Open
Abstract
Pluripotent stem cells (PSCs), including embryonic and induced pluripotent stem cells (iPSCs), show atypical cell cycle regulation characterized by a high proliferation rate and a shorter G1 phase compared with somatic cells. The mechanisms by which somatic cells remodel their cell cycle to achieve the high proliferation rate of PSCs during reprogramming are unclear. Here we identify that the Ink4 protein p18, which is expressed at high levels in somatic cells but at low levels in PSCs, is a roadblock to successful reprogramming. Mild inhibition of p18 expression enhances reprogramming efficiency, while ectopic expression of p18 completely blocks reprogramming. Mechanistic studies show that expression of wild-type p18, but not a p18D68N mutant which cannot inhibit Cdk4/6, down-regulates expression of Cdk4/6 target genes involved in DNA synthesis (TK, TS, DHFR, PCNA) and cell cycle regulation (CDK1 and CCNA2) and thus inhibits reprogramming. These results indicate that p18 blocks reprogramming by targeting Cdk4/6-mediated cell cycle regulation. Taken together, our results define a novel pathway that inhibits somatic cell reprogramming, and provide a new target to enhance reprogramming efficiency.
Collapse
|
144
|
Wan Z, Lu Y, Rui L, Yu X, Li Z. PRDM1 overexpression induce G0/G1 arrest in DF-1 cell line. Gene 2016; 592:119-127. [PMID: 27474451 DOI: 10.1016/j.gene.2016.07.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/04/2016] [Accepted: 07/25/2016] [Indexed: 10/21/2022]
Abstract
PRDM1 (PR domain containing 1) is a transcriptional repressor that affects the expression of numerous genes involved in cell proliferation, differentiation and metabolism. However, the molecular mechanisms underlying PRDM1-regulated gene expression in the DF-1 cell line remain to be elucidated. In this study, we explored the role of PRDM1 in cell proliferation and cell cycle by forced expression of PRDM1 in DF-1 cells. Our results showed an absence of endogenous PRDM1 in this cell line, while exogenous PRDM1 was specifically localized to the nucleus. Ectopic expression of PRDM1 inhibited DF-1 cell proliferation and altered clonal morphology. Furthermore, PRDM1 overexpression caused an increase in the G0/G1 phase population. The levels of p53 mRNA and the p53-regulated p21(WAF1) and MDM2 genes were significantly increased in DF-1 cells transfected with the PRDM1 expression vector. Examination of the Rb pathway further revealed that Rb, E2F-1 and p15(INK4b) alternate reading frame (ARF) mRNA were also significantly increased after transient transfection. Interestingly, the mRNA expression levels of multiple chicken cyclin genes were also increased. These results show that PRDM1 overexpression induced G0/G1 arrest in DF-1 cells through multiple parallel mechanisms, including the p53 and Rb pathways.
Collapse
Affiliation(s)
- Zhiyi Wan
- State key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Beijing 100193, China
| | - Yanan Lu
- State key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Beijing 100193, China
| | - Lei Rui
- State key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Beijing 100193, China
| | - Xiaoxue Yu
- State key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Beijing 100193, China
| | - Zandong Li
- State key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Beijing 100193, China.
| |
Collapse
|
145
|
Kalatskaya I. Overview of major molecular alterations during progression from Barrett's esophagus to esophageal adenocarcinoma. Ann N Y Acad Sci 2016; 1381:74-91. [PMID: 27415609 DOI: 10.1111/nyas.13134] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/13/2016] [Accepted: 05/19/2016] [Indexed: 12/14/2022]
Abstract
Esophageal adenocarcinoma (EAC) develops in the sequential transformation of normal epithelium into metaplastic epithelium, called Barrett's esophagus (BE), then to dysplasia, and finally cancer. BE is a common condition in which normal stratified squamous epithelium of the esophagus is replaced with an intestine-like columnar epithelium, and it is the most prominent risk factor for EAC. This review aims to impartially systemize the knowledge from a large number of publications that describe the molecular and biochemical alterations occurring over this progression sequence. In order to provide an unbiased extraction of the knowledge from the literature, a text-mining methodology was used to select genes that are involved in the BE progression, with the top candidate genes found to be TP53, CDKN2A, CTNNB1, CDH1, GPX3, and NOX5. In addition, sample frequencies across analyzed patient cohorts at each stage of disease progression are summarized. All six genes are altered in the majority of EAC patients, and accumulation of alterations correlates well with the sequential progression of BE to cancer, indicating that the text-mining method is a valid approach for gene prioritization. This review discusses how, besides being cancer drivers, these genes are functionally interconnected and might collectively be considered a central hub of BE progression.
Collapse
Affiliation(s)
- Irina Kalatskaya
- Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario, Canada.
| |
Collapse
|
146
|
De Braekeleer M, Douet-Guilbert N, De Braekeleer E. Prognostic impact ofp15gene aberrations in acute leukemia. Leuk Lymphoma 2016; 58:257-265. [DOI: 10.1080/10428194.2016.1201574] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
147
|
Hylebos M, Van Camp G, van Meerbeeck JP, Op de Beeck K. The Genetic Landscape of Malignant Pleural Mesothelioma: Results from Massively Parallel Sequencing. J Thorac Oncol 2016; 11:1615-26. [PMID: 27282309 DOI: 10.1016/j.jtho.2016.05.020] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/27/2016] [Accepted: 05/22/2016] [Indexed: 12/18/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare yet aggressive tumor that is causally associated with-mostly professional-asbestos exposure. Given the long latency between exposure and disease, and because asbestos is still being used, MPM will remain a global health issue for decades to come. Notwithstanding the increasing incidence of MPM and the fact that patients with MPM face a poor prognosis, currently available treatment options are limited. To enable the development of novel targeted therapies, identification of the genetic alterations underlying MPM will be crucial. The first studies reporting on the genomic background of MPM identified recurrent somatic mutations in a number of tumor suppressor genes (i.e., cyclin-dependent kinase inhibitor 2A gene [CDKN2A], neurofibromin 2 (merlin) gene [NF2], and BRCA1 associated protein 1 gene [BAP1]). More recently, massively parallel sequencing strategies have been used and have provided a more genome-wide view on the genetic landscape of MPM. This review summarizes their results, describing alterations that cluster mainly in four pathways: the tumor protein p53/DNA repair, cell cycle, mitogen-activated protein kinase, and phosphoinisitide 3-kinase (PI3K)/AKT pathways. As these pathways are important during tumor development, they provide interesting candidates for targeting with novel drugs.
Collapse
Affiliation(s)
- Marieke Hylebos
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium; Center for Oncological Research, University of Antwerp, Antwerp, Belgium.
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Jan P van Meerbeeck
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Thoracic Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium; Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
148
|
Nabeshima K, Matsumoto S, Hamasaki M, Hida T, Kamei T, Hiroshima K, Tsujimura T, Kawahara K. Use of p16 FISH for differential diagnosis of mesothelioma in smear preparations. Diagn Cytopathol 2016; 44:774-80. [PMID: 27219841 DOI: 10.1002/dc.23501] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 04/11/2016] [Accepted: 04/18/2016] [Indexed: 12/27/2022]
Abstract
Because most of malignant pleural mesothelioma (MPM) patients first present with pleural effusion, detection of mesothelioma cells on effusion smears is critical for early diagnosis. Recently, accumulating evidence indicating that the cytological diagnosis of MPM supported by ancillary techniques is as reliable as that based on histopathology has led to new guidelines for the cytopathologic diagnosis of MPM. Based on the guidelines, a combination of cytomorphological criteria and verification by ancillary techniques is required for the cytologic diagnosis of MPM. Detection of p16 homozygous deletion by fluorescence in situ hybridization (FISH) is the most reliable ancillary technique for differentiating MPM from reactive mesothelial cells (RMC) because of its relatively high sensitivity and extremely high specificity. We showed that the p16 deletion status of MPM cells in pleural effusions reflected that of the underlying invasive MPM tissues, indicating the usefulness of p16 FISH in effusion smear cytology for MPM diagnosis. Thus, for differentiating MPM from RMC, we propose to perform p16 FISH as often as possible. A positive p16 homozygous deletion supports the diagnosis of MPM. However, a negative result does not rule out the possibility of MPM. In such cases, a morphological assessment is critical. Therefore, we analyzed the morphological characteristics of p16 deletion-positive mesothelioma cells using a combination of virtual microscopy and p16 FISH, and identified three morphological characteristics useful for the differentiation, including cell-in-cell engulfment with or without hump formation, multinucleate cells, and larger berry-like cell aggregates. Diagn. Cytopathol. 2016;44:774-780. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kazuki Nabeshima
- Department of Pathology, Fukuoka University Hospital and School of Medicine, Fukuoka, Japan
| | - Shinji Matsumoto
- Department of Pathology, Fukuoka University Hospital and School of Medicine, Fukuoka, Japan
| | - Makoto Hamasaki
- Department of Pathology, Fukuoka University Hospital and School of Medicine, Fukuoka, Japan
| | - Tomoyuki Hida
- Department of Pathology, Fukuoka University Hospital and School of Medicine, Fukuoka, Japan
| | - Toshiaki Kamei
- Department of Pathology, PCL Japan Fukuoka, Pathology-Cytology Center, Fukuoka, Japan
| | - Kenzo Hiroshima
- Department of Pathology, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo, Japan
| | - Tohru Tsujimura
- Department of Pathology, Hyogo College of Medicine, Hyogo, Japan
| | - Kunimitsu Kawahara
- Department of Pathology, Osaka Prefectural Medical Center for Respiratory and Allergic Disease, Habikino, Japan
| |
Collapse
|
149
|
Zhao R, Choi BY, Lee MH, Bode AM, Dong Z. Implications of Genetic and Epigenetic Alterations of CDKN2A (p16(INK4a)) in Cancer. EBioMedicine 2016; 8:30-39. [PMID: 27428416 PMCID: PMC4919535 DOI: 10.1016/j.ebiom.2016.04.017] [Citation(s) in RCA: 282] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/01/2016] [Accepted: 04/14/2016] [Indexed: 12/12/2022] Open
Abstract
Aberrant gene silencing is highly associated with altered cell cycle regulation during carcinogenesis. In particular, silencing of the CDKN2A tumor suppressor gene, which encodes the p16(INK4a) protein, has a causal link with several different types of cancers. The p16(INK4a) protein plays an executional role in cell cycle and senescence through the regulation of the cyclin-dependent kinase (CDK) 4/6 and cyclin D complexes. Several genetic and epigenetic aberrations of CDKN2A lead to enhanced tumorigenesis and metastasis with recurrence of cancer and poor prognosis. In these cases, the restoration of genetic and epigenetic reactivation of CDKN2A is a practical approach for the prevention and therapy of cancer. This review highlights the genetic status of CDKN2A as a prognostic and predictive biomarker in various cancers.
Collapse
Affiliation(s)
- Ran Zhao
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Bu Young Choi
- Department of Pharmaceutical Science and Engineering, Seowon University, Cheongju 361-742, South Korea
| | - Mee-Hyun Lee
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China.
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China; The Hormel Institute, University of Minnesota, Austin, MN 55912, USA.
| |
Collapse
|
150
|
Bedoya-López A, Estrada K, Sanchez-Flores A, Ramírez OT, Altamirano C, Segovia L, Miranda-Ríos J, Trujillo-Roldán MA, Valdez-Cruz NA. Effect of Temperature Downshift on the Transcriptomic Responses of Chinese Hamster Ovary Cells Using Recombinant Human Tissue Plasminogen Activator Production Culture. PLoS One 2016; 11:e0151529. [PMID: 26991106 PMCID: PMC4798216 DOI: 10.1371/journal.pone.0151529] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/28/2016] [Indexed: 12/30/2022] Open
Abstract
Recombinant proteins are widely used as biopharmaceuticals, but their production by mammalian cell culture is expensive. Hence, improvement of bioprocess productivity is greatly needed. A temperature downshift (TDS) from 37°C to 28–34°C is an effective strategy to expand the productive life period of cells and increase their productivity (qp). Here, TDS in Chinese hamster ovary (CHO) cell cultures, initially grown at 37°C and switched to 30°C during the exponential growth phase, resulted in a 1.6-fold increase in the qp of recombinant human tissue plasminogen activator (rh-tPA). The transcriptomic response using next-generation sequencing (NGS) was assessed to characterize the cellular behavior associated with TDS. A total of 416 (q > 0.8) and 3,472 (q > 0.9) differentially expressed transcripts, with more than a 1.6-fold change at 24 and 48 h post TDS, respectively, were observed in cultures with TDS compared to those at constant 37°C. In agreement with the extended cell survival resulting from TDS, transcripts related to cell growth arrest that controlled cell proliferation without the activation of the DNA damage response, were differentially expressed. Most upregulated genes were related to energy metabolism in mitochondria, mitochondrial biogenesis, central metabolism, and avoidance of apoptotic cell death. The gene coding for rh-tPA was not differentially expressed, but fluctuations were detected in the transcripts encoding proteins involved in the secretory machinery, particularly in glycosylation. Through NGS the dynamic processes caused by TDS were assessed in this biological system.
Collapse
Affiliation(s)
- Andrea Bedoya-López
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Karel Estrada
- Unidad Universitaria de Apoyo Bioinformático, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. México
| | - Alejandro Sanchez-Flores
- Unidad Universitaria de Apoyo Bioinformático, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. México
| | - Octavio T. Ramírez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. México
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Lorenzo Segovia
- Departamento de Ingeniería Celular y Biocatálisis. Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor. México
| | - Juan Miranda-Ríos
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mauricio A. Trujillo-Roldán
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Norma A. Valdez-Cruz
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
- * E-mail:
| |
Collapse
|