1
|
Baker TM, Waise S, Tarabichi M, Van Loo P. Aneuploidy and complex genomic rearrangements in cancer evolution. NATURE CANCER 2024; 5:228-239. [PMID: 38286829 PMCID: PMC7616040 DOI: 10.1038/s43018-023-00711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/14/2023] [Indexed: 01/31/2024]
Abstract
Mutational processes that alter large genomic regions occur frequently in developing tumors. They range from simple copy number gains and losses to the shattering and reassembly of entire chromosomes. These catastrophic events, such as chromothripsis, chromoplexy and the formation of extrachromosomal DNA, affect the expression of many genes and therefore have a substantial effect on the fitness of the cells in which they arise. In this review, we cover large genomic alterations, the mechanisms that cause them and their effect on tumor development and evolution.
Collapse
Affiliation(s)
- Toby M Baker
- The Francis Crick Institute, London, UK
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sara Waise
- The Francis Crick Institute, London, UK
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Maxime Tarabichi
- The Francis Crick Institute, London, UK
- Institute for Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Peter Van Loo
- The Francis Crick Institute, London, UK.
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Platt JL, Cascalho M. Somatic Cell Fusion in Host Defense and Adaptation. Results Probl Cell Differ 2024; 71:213-225. [PMID: 37996680 DOI: 10.1007/978-3-031-37936-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Evidence of fusion of somatic cells has been noted in health and in disease for more than a century. The most obvious but uncertain hallmark has been the presence of multiple nuclei in cells. Although multinucleated cells are found in normal and diseased tissues, the benefit or harm of such cells can be difficult to elucidate. Still more difficult however is the identification of mononuclear cells previously formed by fusion of somatic cells with one or more nuclei disposed. The later process can introduce mutations that promote viral diversification, cancer, and tissue senescence. Less obvious the potential benefits of cell fusion. Recent work in cell biology, immunology, and genomic analysis however makes it possible to postulate benefits and potentially arrive at novel therapeutic agents and approaches that replicate or enhance these benefits.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Departments of Surgery and Microbiology & Immunology University of Michigan, Ann Arbor, MI, USA.
| | - Marilia Cascalho
- Departments of Surgery and Microbiology & Immunology University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
3
|
Noubissi FK, Odubanjo OV, Ogle BM, Tchounwou PB. Mechanisms of Cell Fusion in Cancer. Results Probl Cell Differ 2024; 71:407-432. [PMID: 37996688 PMCID: PMC10893907 DOI: 10.1007/978-3-031-37936-9_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Cell-cell fusion is a normal physiological mechanism that requires a well-orchestrated regulation of intracellular and extracellular factors. Dysregulation of this process could lead to diseases such as osteoporosis, malformation of muscles, difficulties in pregnancy, and cancer. Extensive literature demonstrates that fusion occurs between cancer cells and other cell types to potentially promote cancer progression and metastasis. However, the mechanisms governing this process in cancer initiation, promotion, and progression are less well-studied. Fusogens involved in normal physiological processes such as syncytins and associated factors such as phosphatidylserine and annexins have been observed to be critical in cancer cell fusion as well. Some of the extracellular factors associated with cancer cell fusion include chronic inflammation and inflammatory cytokines, hypoxia, and viral infection. The interaction between these extracellular factors and cell's intrinsic factors potentially modulates actin dynamics to drive the fusion of cancer cells. In this review, we have discussed the different mechanisms that have been identified or postulated to drive cancer cell fusion.
Collapse
Affiliation(s)
- Felicite K Noubissi
- Department of Biology, Jackson State University, Jackson, MS, USA.
- Research Centers in Minority Institutions (RCMI), Center for Health Disparity Research (RCMI-CHDR), Jackson State University, Jackson, MS, USA.
| | - Oluwatoyin V Odubanjo
- Department of Biology, Jackson State University, Jackson, MS, USA
- Research Centers in Minority Institutions (RCMI), Center for Health Disparity Research (RCMI-CHDR), Jackson State University, Jackson, MS, USA
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Paul B Tchounwou
- Department of Biology, Jackson State University, Jackson, MS, USA
- Research Centers in Minority Institutions (RCMI), Center for Health Disparity Research (RCMI-CHDR), Jackson State University, Jackson, MS, USA
| |
Collapse
|
4
|
Faggioli F, Velarde MC, Wiley CD. Cellular Senescence, a Novel Area of Investigation for Metastatic Diseases. Cells 2023; 12:cells12060860. [PMID: 36980201 PMCID: PMC10047218 DOI: 10.3390/cells12060860] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Metastasis is a systemic condition and the major challenge among cancer types, as it can lead to multiorgan vulnerability. Recently, attention has been drawn to cellular senescence, a complex stress response condition, as a factor implicated in metastatic dissemination and outgrowth. Here, we examine the current knowledge of the features required for cells to invade and colonize secondary organs and how senescent cells can contribute to this process. First, we describe the role of senescence in placentation, itself an invasive process which has been linked to higher rates of invasive cancers. Second, we describe how senescent cells can contribute to metastatic dissemination and colonization. Third, we discuss several metabolic adaptations by which senescent cells could promote cancer survival along the metastatic journey. In conclusion, we posit that targeting cellular senescence may have a potential therapeutic efficacy to limit metastasis formation.
Collapse
Affiliation(s)
- Francesca Faggioli
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR) uos Milan, Via Fantoli 15/16, 20090 Milan, Italy
- Correspondence: ; Tel.: +39-02-82245211
| | - Michael C. Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City PH 1101, Philippines
| | - Christopher D. Wiley
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA 02111, USA
- School of Medicine, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
5
|
Generation of Cancer Stem/Initiating Cells by Cell-Cell Fusion. Int J Mol Sci 2022; 23:ijms23094514. [PMID: 35562905 PMCID: PMC9101717 DOI: 10.3390/ijms23094514] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
CS/ICs have raised great expectations in cancer research and therapy, as eradication of this key cancer cell type is expected to lead to a complete cure. Unfortunately, the biology of CS/ICs is rather complex, since no common CS/IC marker has yet been identified. Certain surface markers or ALDH1 expression can be used for detection, but some studies indicated that cancer cells exhibit a certain plasticity, so CS/ICs can also arise from non-CS/ICs. Another problem is intratumoral heterogeneity, from which it can be inferred that different CS/IC subclones must be present in the tumor. Cell–cell fusion between cancer cells and normal cells, such as macrophages and stem cells, has been associated with the generation of tumor hybrids that can exhibit novel properties, such as an enhanced metastatic capacity and even CS/IC properties. Moreover, cell–cell fusion is a complex process in which parental chromosomes are mixed and randomly distributed among daughter cells, resulting in multiple, unique tumor hybrids. These, if they have CS/IC properties, may contribute to the heterogeneity of the CS/IC pool. In this review, we will discuss whether cell–cell fusion could also lead to the origin of different CS/ICs that may expand the overall CS/IC pool in a primary tumor.
Collapse
|
6
|
Lazebnik Y. Cell fusion as a link between the SARS-CoV-2 spike protein, COVID-19 complications, and vaccine side effects. Oncotarget 2021; 12:2476-2488. [PMID: 34917266 PMCID: PMC8664391 DOI: 10.18632/oncotarget.28088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/24/2021] [Indexed: 12/23/2022] Open
Abstract
A distinctive feature of the SARS-CoV-2 spike protein is its ability to efficiently fuse cells, thus producing syncytia found in COVID-19 patients. This commentary proposes how this ability enables spike to cause COVID-19 complications as well as side effects of COVID-19 vaccines, and suggests how these effects can be prevented.
Collapse
|
7
|
Dittmar T, Weiler J, Luo T, Hass R. Cell-Cell Fusion Mediated by Viruses and HERV-Derived Fusogens in Cancer Initiation and Progression. Cancers (Basel) 2021; 13:5363. [PMID: 34771528 PMCID: PMC8582398 DOI: 10.3390/cancers13215363] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Cell fusion is a well-known, but still scarcely understood biological phenomenon, which might play a role in cancer initiation, progression and formation of metastases. Although the merging of two (cancer) cells appears simple, the entire process is highly complex, energy-dependent and tightly regulated. Among cell fusion-inducing and -regulating factors, so-called fusogens have been identified as a specific type of proteins that are indispensable for overcoming fusion-associated energetic barriers and final merging of plasma membranes. About 8% of the human genome is of retroviral origin and some well-known fusogens, such as syncytin-1, are expressed by human (cancer) cells. Likewise, enveloped viruses can enable and facilitate cell fusion due to evolutionarily optimized fusogens, and are also capable to induce bi- and multinucleation underlining their fusion capacity. Moreover, multinucleated giant cancer cells have been found in tumors derived from oncogenic viruses. Accordingly, a potential correlation between viruses and fusogens of human endogenous retroviral origin in cancer cell fusion will be summarized in this review.
Collapse
Affiliation(s)
- Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58448 Witten, Germany;
| | - Julian Weiler
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58448 Witten, Germany;
| | - Tianjiao Luo
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| | - Ralf Hass
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
8
|
Hybrid Formation and Fusion of Cancer Cells In Vitro and In Vivo. Cancers (Basel) 2021; 13:cancers13174496. [PMID: 34503305 PMCID: PMC8431460 DOI: 10.3390/cancers13174496] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cell fusion as a fundamental biological process is required for various physiological processes, including fertilization, placentation, myogenesis, osteoclastogenesis, and wound healing/tissue regeneration. However, cell fusion is also observed during pathophysiological processes like tumor development. Mesenchymal stroma/stem-like cells (MSC) which play an important role within the tumor microenvironment like other cell types such as macrophages can closely interact and hybridize with cancer cells. The formation of cancer hybrid cells can involve various different mechanisms whereby the genomic parts of the hybrid cells require rearrangement to form a new functional hybrid cell. The fusion of cancer cells with neighboring cell types may represent an important mechanism during tumor development since cancer hybrid cells are detectable in various tumor tissues. During this rare event with resulting genomic instability the cancer hybrid cells undergo a post-hybrid selection process (PHSP) to reorganize chromosomes of the two parental nuclei whereby the majority of the hybrid population undergoes cell death. The remaining cancer hybrid cells survive by displaying altered properties within the tumor tissue. Abstract The generation of cancer hybrid cells by intra-tumoral cell fusion opens new avenues for tumor plasticity to develop cancer stem cells with altered properties, to escape from immune surveillance, to change metastatic behavior, and to broaden drug responsiveness/resistance. Genomic instability and chromosomal rearrangements in bi- or multinucleated aneuploid cancer hybrid cells contribute to these new functions. However, the significance of cell fusion in tumorigenesis is controversial with respect to the low frequency of cancer cell fusion events and a clonal advantage of surviving cancer hybrid cells following a post-hybrid selection process. This review highlights alternative processes of cancer hybrid cell development such as entosis, emperipolesis, cannibalism, therapy-induced polyploidization/endoreduplication, horizontal or lateral gene transfer, and focusses on the predominant mechanisms of cell fusion. Based upon new properties of cancer hybrid cells the arising clinical consequences of the subsequent tumor heterogeneity after cancer cell fusion represent a major therapeutic challenge.
Collapse
|
9
|
Haas OA. Somatic Sex: On the Origin of Neoplasms With Chromosome Counts in Uneven Ploidy Ranges. Front Cell Dev Biol 2021; 9:631946. [PMID: 34422788 PMCID: PMC8373647 DOI: 10.3389/fcell.2021.631946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 06/22/2021] [Indexed: 01/09/2023] Open
Abstract
Stable aneuploid genomes with nonrandom numerical changes in uneven ploidy ranges define distinct subsets of hematologic malignancies and solid tumors. The idea put forward herein suggests that they emerge from interactions between diploid mitotic and G0/G1 cells, which can in a single step produce all combinations of mono-, di-, tri-, tetra- and pentasomic paternal/maternal homologue configurations that define such genomes. A nanotube-mediated influx of interphase cell cytoplasm into mitotic cells would thus be responsible for the critical nondisjunction and segregation errors by physically impeding the proper formation of the cell division machinery, whereas only a complete cell fusion can simultaneously generate pentasomies, uniparental trisomies as well as biclonal hypo- and hyperdiploid cell populations. The term "somatic sex" was devised to accentuate the similarities between germ cell and somatic cell fusions. A somatic cell fusion, in particular, recapitulates many processes that are also instrumental in the formation of an abnormal zygote that involves a diploid oocyte and a haploid sperm, which then may further develop into a digynic triploid embryo. Despite their somehow deceptive differences and consequences, the resemblance of these two routes may go far beyond of what has hitherto been appreciated. Based on the arguments put forward herein, I propose that embryonic malignancies of mesenchymal origin with these particular types of aneuploidies can thus be viewed as the kind of flawed somatic equivalent of a digynic triploid embryo.
Collapse
Affiliation(s)
- Oskar A Haas
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| |
Collapse
|
10
|
Mittal K, Kaur J, Jaczko M, Wei G, Toss MS, Rakha EA, Janssen EAM, Søiland H, Kucuk O, Reid MD, Gupta MV, Aneja R. Centrosome amplification: a quantifiable cancer cell trait with prognostic value in solid malignancies. Cancer Metastasis Rev 2021; 40:319-339. [PMID: 33106971 PMCID: PMC7897259 DOI: 10.1007/s10555-020-09937-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Numerical and/or structural centrosome amplification (CA) is a hallmark of cancers that is often associated with the aberrant tumor karyotypes and poor clinical outcomes. Mechanistically, CA compromises mitotic fidelity and leads to chromosome instability (CIN), which underlies tumor initiation and progression. Recent technological advances in microscopy and image analysis platforms have enabled better-than-ever detection and quantification of centrosomal aberrancies in cancer. Numerous studies have thenceforth correlated the presence and the degree of CA with indicators of poor prognosis such as higher tumor grade and ability to recur and metastasize. We have pioneered a novel semi-automated pipeline that integrates immunofluorescence confocal microscopy with digital image analysis to yield a quantitative centrosome amplification score (CAS), which is a summation of the severity and frequency of structural and numerical centrosome aberrations in tumor samples. Recent studies in breast cancer show that CA increases across the disease progression continuum, while normal breast tissue exhibited the lowest CA, followed by cancer-adjacent apparently normal, ductal carcinoma in situ and invasive tumors, which showed the highest CA. This finding strengthens the notion that CA could be evolutionarily favored and can promote tumor progression and metastasis. In this review, we discuss the prevalence, extent, and severity of CA in various solid cancer types, the utility of quantifying amplified centrosomes as an independent prognostic marker. We also highlight the clinical feasibility of a CA-based risk score for predicting recurrence, metastasis, and overall prognosis in patients with solid cancers.
Collapse
Affiliation(s)
- Karuna Mittal
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Jaspreet Kaur
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Meghan Jaczko
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Guanhao Wei
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Michael S Toss
- Department of Pathology, University of Nottingham and Nottingham University Hospitals, Nottingham, UK
| | - Emad A Rakha
- Department of Pathology, University of Nottingham and Nottingham University Hospitals, Nottingham, UK
| | | | - Håvard Søiland
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, Stavanger, Norway
| | - Omer Kucuk
- Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University Hospital, Atlanta, GA, USA
| | | | | | - Ritu Aneja
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA.
| |
Collapse
|
11
|
Genome remodeling upon mesenchymal tumor cell fusion contributes to tumor progression and metastatic spread. Oncogene 2020; 39:4198-4211. [PMID: 32242148 DOI: 10.1038/s41388-020-1276-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/17/2022]
Abstract
Cell fusion in tumor progression mostly refers to the merging of a cancer cell with a cell that has migration and immune escape capabilities such as macrophages. Here we show that spontaneous hybrids made from the fusion of transformed mesenchymal cells with partners from the same lineage undergo nonrecurrent large-scale genomic rearrangements, leading to the creation of highly aneuploid cells with novel phenotypic traits, including metastatic spreading capabilities. Moreover, in contrast to their parents, hybrids were the only cells able to recapitulate in vivo all features of human pleomorphic sarcomas, a rare and genetically complex mesenchymal tumor. Hybrid tumors not only displayed specific mesenchymal markers, but also combined a complex genetic profile with a highly metastatic behavior, like their human counterparts. Finally, we provide evidence that patient-derived pleomorphic sarcoma cells are inclined to spontaneous cell fusion. The resulting hybrids also gain in aggressiveness, exhibiting superior growth capacity in mouse models. Altogether, these results indicate that cell fusion has the potential to promote cancer progression by increasing growth and/or metastatic capacities, regardless of the nature of the companion cell. Moreover, such events likely occur upon sarcoma development, paving the way for better understanding of the biology, and aggressiveness of these tumors.
Collapse
|
12
|
Dörnen J, Sieler M, Weiler J, Keil S, Dittmar T. Cell Fusion-Mediated Tissue Regeneration as an Inducer of Polyploidy and Aneuploidy. Int J Mol Sci 2020; 21:E1811. [PMID: 32155721 PMCID: PMC7084716 DOI: 10.3390/ijms21051811] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022] Open
Abstract
The biological phenomenon of cell fusion plays a crucial role in several physiological processes, including wound healing and tissue regeneration. Here, it is assumed that bone marrow-derived stem cells (BMSCs) could adopt the specific properties of a different organ by cell fusion, thereby restoring organ function. Cell fusion first results in the production of bi- or multinucleated hybrid cells, which either remain as heterokaryons or undergo ploidy reduction/heterokaryon-to-synkaryon transition (HST), thereby giving rise to mononucleated daughter cells. This process is characterized by a merging of the chromosomes from the previously discrete nuclei and their subsequent random segregation into daughter cells. Due to extra centrosomes concomitant with multipolar spindles, the ploidy reduction/HST could also be associated with chromosome missegregation and, hence, induction of aneuploidy, genomic instability, and even putative chromothripsis. However, while the majority of such hybrids die or become senescent, aneuploidy and genomic instability appear to be tolerated in hepatocytes, possibly for stress-related adaption processes. Likewise, cell fusion-induced aneuploidy and genomic instability could also lead to a malignant conversion of hybrid cells. This can occur during tissue regeneration mediated by BMSC fusion in chronically inflamed tissue, which is a cell fusion-friendly environment, but is also enriched for mutagenic reactive oxygen and nitrogen species.
Collapse
Affiliation(s)
| | | | | | | | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58448 Witten, Germany; (J.D.); (M.S.); (J.W.); (S.K.)
| |
Collapse
|
13
|
Abstract
This review by Levine and Holland reviews the sources of mitotic errors in human tumors and their effect on cell fitness and transformation. They discuss new findings that suggest that chromosome missegregation can produce a proinflammatory environment and impact tumor responsiveness to immunotherapy and survey the vulnerabilities exposed by cell division errors and how they can be exploited therapeutically. Mitosis is a delicate event that must be executed with high fidelity to ensure genomic stability. Recent work has provided insight into how mitotic errors shape cancer genomes by driving both numerical and structural alterations in chromosomes that contribute to tumor initiation and progression. Here, we review the sources of mitotic errors in human tumors and their effect on cell fitness and transformation. We discuss new findings that suggest that chromosome missegregation can produce a proinflammatory environment and impact tumor responsiveness to immunotherapy. Finally, we survey the vulnerabilities exposed by cell division errors and how they can be exploited therapeutically.
Collapse
Affiliation(s)
- Michelle S Levine
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
14
|
Vittoria MA, Shenk EM, O'Rourke KP, Bolgioni AF, Lim S, Kacprzak V, Quinton RJ, Ganem NJ. A genome-wide microRNA screen identifies regulators of tetraploid cell proliferation. Mol Biol Cell 2018; 29:1682-1692. [PMID: 29791254 PMCID: PMC6080710 DOI: 10.1091/mbc.e18-02-0141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tetraploid cells, which are most commonly generated by errors in cell division, are genomically unstable and have been shown to promote tumorigenesis. Recent genomic studies have estimated that ∼40% of all solid tumors have undergone a genome-doubling event during their evolution, suggesting a significant role for tetraploidy in driving the development of human cancers. To safeguard against the deleterious effects of tetraploidy, nontransformed cells that fail mitosis and become tetraploid activate both the Hippo and p53 tumor suppressor pathways to restrain further proliferation. Tetraploid cells must therefore overcome these antiproliferative barriers to ultimately drive tumor development. However, the genetic routes through which spontaneously arising tetraploid cells adapt to regain proliferative capacity remain poorly characterized. Here, we conducted a comprehensive gain-of-function genome-wide screen to identify microRNAs (miRNAs) that are sufficient to promote the proliferation of tetraploid cells. Our screen identified 23 miRNAs whose overexpression significantly promotes tetraploid proliferation. The vast majority of these miRNAs facilitate tetraploid growth by enhancing mitogenic signaling pathways (e.g., miR-191-3p); however, we also identified several miRNAs that impair the p53/p21 pathway (e.g., miR-523-3p), and a single miRNA (miR-24-3p) that potently inactivates the Hippo pathway via down-regulation of the tumor suppressor gene NF2. Collectively, our data reveal several avenues through which tetraploid cells may regain the proliferative capacity necessary to drive tumorigenesis.
Collapse
Affiliation(s)
- Marc A Vittoria
- Department of Pharmacology and Experimental Therapeutics, University School of Medicine, Boston, MA 02118
| | - Elizabeth M Shenk
- Department of Pharmacology and Experimental Therapeutics, University School of Medicine, Boston, MA 02118.,Department of Biomedical Engineering, Boston University, Boston, MA 02118
| | - Kevin P O'Rourke
- Weill Cornell Medicine/Rockefeller University/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065
| | - Amanda F Bolgioni
- Department of Pharmacology and Experimental Therapeutics, University School of Medicine, Boston, MA 02118
| | - Sanghee Lim
- Department of Pharmacology and Experimental Therapeutics, University School of Medicine, Boston, MA 02118
| | - Victoria Kacprzak
- Department of Pharmacology and Experimental Therapeutics, University School of Medicine, Boston, MA 02118
| | - Ryan J Quinton
- Department of Pharmacology and Experimental Therapeutics, University School of Medicine, Boston, MA 02118
| | - Neil J Ganem
- Department of Pharmacology and Experimental Therapeutics, University School of Medicine, Boston, MA 02118.,Division of Hematology and Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
15
|
Cho MG, Ahn JH, Choi HS, Lee JH. DNA double-strand breaks and Aurora B mislocalization induced by exposure of early mitotic cells to H 2O 2 appear to increase chromatin bridges and resultant cytokinesis failure. Free Radic Biol Med 2017; 108:129-145. [PMID: 28343997 DOI: 10.1016/j.freeradbiomed.2017.03.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 01/14/2023]
Abstract
Aneuploidy, an abnormal number of chromosomes that is a hallmark of cancer cells, can arise from tetraploid/binucleated cells through a failure of cytokinesis. Reactive oxygen species (ROS) have been implicated in various diseases, including cancer. However, the nature and role of ROS in cytokinesis progression and related mechanisms has not been clearly elucidated. Here, using time-lapse analysis of asynchronously growing cells and immunocytochemical analyses of synchronized cells, we found that hydrogen peroxide (H2O2) treatment at early mitosis (primarily prometaphase) significantly induced cytokinesis failure. Cytokinesis failure and the resultant formation of binucleated cells containing nucleoplasmic bridges (NPBs) seemed to be caused by increases in DNA double-strand breaks (DSBs) and subsequent unresolved chromatin bridges. We further found that H2O2 induced mislocalization of Aurora B during mitosis. All of these effects were attenuated by pretreatment with N-acetyl-L-cysteine (NAC) or overexpression of Catalase. Surprisingly, the PARP inhibitor PJ34 also reduced H2O2-induced Aurora B mislocalization and binucleated cell formation. Results of parallel experiments with etoposide, a topoisomerase IIα inhibitor that triggers DNA DSBs, suggested that both DNA DSBs and Aurora B mislocalization contribute to chromatin bridge formation. Aurora B mislocalization also appeared to weaken the "abscission checkpoint". Finally, we showed that KRAS-induced binucleated cell formation appeared to be also H2O2-dependent. In conclusion, we propose that a ROS, mainly H2O2 increases binucleation through unresolved chromatin bridges caused by DNA damage and mislocalization of Aurora B, the latter of which appears to augment the effect of DNA damage on chromatin bridge formation.
Collapse
Affiliation(s)
- Min-Guk Cho
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 443-721, South Korea; Genomic Instability Research Center, Ajou University School of Medicine, Suwon 443-721, South Korea; Department of Biomedical Science, Graduate School of Ajou university, Suwon 443-721, South Korea.
| | - Ju-Hyun Ahn
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 443-721, South Korea; Genomic Instability Research Center, Ajou University School of Medicine, Suwon 443-721, South Korea; Department of Biomedical Science, Graduate School of Ajou university, Suwon 443-721, South Korea.
| | - Hee-Song Choi
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 443-721, South Korea; Genomic Instability Research Center, Ajou University School of Medicine, Suwon 443-721, South Korea; Department of Biomedical Science, Graduate School of Ajou university, Suwon 443-721, South Korea.
| | - Jae-Ho Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 443-721, South Korea; Genomic Instability Research Center, Ajou University School of Medicine, Suwon 443-721, South Korea; Department of Biomedical Science, Graduate School of Ajou university, Suwon 443-721, South Korea.
| |
Collapse
|
16
|
Liao Z, Muth DC, Eitan E, Travers M, Learman LN, Lehrmann E, Witwer KW. Serum extracellular vesicle depletion processes affect release and infectivity of HIV-1 in culture. Sci Rep 2017; 7:2558. [PMID: 28566772 PMCID: PMC5451420 DOI: 10.1038/s41598-017-02908-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/20/2017] [Indexed: 01/07/2023] Open
Abstract
Extracellular vesicles (EVs) are involved in intercellular communication and affect processes including immune and antiviral responses. Blood serum, a common cell culture medium component, is replete with EVs and must be depleted prior to EV-related experiments. The extent to which depletion processes deplete non-EV particles is incompletely understood, but depleted serum is associated with reduced viability and growth in cell culture. Here, we examined whether serum depleted by two methods affected HIV-1 replication. In cell lines, including HIV-1 latency models, increased HIV-1 production was observed, along with changes in cell behavior and viability. Add-back of ultracentrifuge pellets (enriched in EVs but possibly other particles) rescued baseline HIV-1 production. Primary cells were less sensitive to serum depletion processes. Virus produced under processed serum conditions was more infectious. Finally, changes in cellular metabolism, surface markers, and gene expression, but not miRNA profiles, were associated with depleted serum culture. In conclusion, depleted serum conditions have a substantial effect on HIV-1 production and infectivity. Dependence of cell cultures on “whole serum” must be examined carefully along with other experimental variables, keeping in mind that the effects of EVs may be accompanied by or confused with those of closely associated or physically similar particles.
Collapse
Affiliation(s)
- Zhaohao Liao
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dillon C Muth
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Erez Eitan
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, United States
| | - Meghan Travers
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lisa N Learman
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elin Lehrmann
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, United States
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States. .,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States. .,Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
17
|
Bach DH, Hong JY, Park HJ, Lee SK. The role of exosomes and miRNAs in drug-resistance of cancer cells. Int J Cancer 2017; 141:220-230. [PMID: 28240776 DOI: 10.1002/ijc.30669] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/08/2017] [Accepted: 02/20/2017] [Indexed: 12/14/2022]
Abstract
Chemotherapy, one of the principal approaches for cancer patients, plays a crucial role in controlling tumor progression. Clinically, tumors reveal a satisfactory response following the first exposure to the chemotherapeutic drugs in treatment. However, most tumors sooner or later become resistant to even chemically unrelated anticancer agents after repeated treatment. The reduced drug accumulation in tumor cells is considered one of the significant mechanisms by decreasing drug permeability and/or increasing active efflux (pumping out) of the drugs across the cell membrane. The mechanisms of treatment failure of chemotherapeutic drugs have been investigated, including drug efflux, which is mediated by extracellular vesicles (EVs). Exosomes, a subset of EVs with a size range of 40-150 nm and a lipid bilayer membrane, can be released by all cell types. They mediate specific cell-to-cell interactions and activate signaling pathways in cells they either fuse with or interact with, including cancer cells. Exosomal RNAs are heterogeneous in size but enriched in small RNAs, such as miRNAs. In the primary tumor microenvironment, cancer-secreted exosomes and miRNAs can be internalized by other cell types. MiRNAs loaded in these exosomes might be transferred to recipient niche cells to exert genome-wide regulation of gene expression. How exosomal miRNAs contribute to the development of drug resistance in the context of the tumor microenvironment has not been fully described. In this review, we will highlight recent studies regarding EV-mediated microRNA delivery in formatting drug resistance. We also suggest the use of EVs as an advancing method in antiresistance treatment.
Collapse
Affiliation(s)
- Duc-Hiep Bach
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Ji-Young Hong
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hyen Joo Park
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Do M, Kwak IH, Ahn JH, Lee IJ, Lee JH. Survivin protects fused cancer cells from cell death. BMB Rep 2017; 50:361-366. [PMID: 28193315 PMCID: PMC5584743 DOI: 10.5483/bmbrep.2017.50.7.185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Indexed: 01/06/2023] Open
Abstract
Tetraploidy, a potential precursor of cancer-associated aneuploidy, is produced either by cell fusion or failure of cytokinesis. In this study, low p53-expressing HeLa cells were used to address the fate of cancer cells after fusion. We found that massive cell death or growth arrest occurred a few days after fusion. Interestingly, cells with larger nuclei preferentially died after fusion, suggesting that a larger deviation of DNA content is a strong inducer of apoptosis. Notably, a fraction of cells escaped cell death. Also, the stability of survivin increased, and its localization changed preferentially to the cytosol in fused cells. Knockdown of survivin decreased the survival of fused cells, more than observed in unfused cells, showing increased dependency of fused cells on survivin. Collectively, after cancer cell fusion, some fused cells avoid the apoptotic crisis partly owing to survivin, and continue to proliferate, a process that contributes to human cancer progression.
Collapse
Affiliation(s)
- Mihyang Do
- Department of Biochemistry and Molecular Biology, and Genomic instability Research Center, Ajou University School of Medicine; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon 16499, Korea
| | - In-Hae Kwak
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea
| | - Ju-Hyun Ahn
- Department of Biochemistry and Molecular Biology, and Genomic instability Research Center, Ajou University School of Medicine; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon 16499, Korea
| | - In Jeong Lee
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea
| | - Jae-Ho Lee
- Department of Biochemistry and Molecular Biology, and Genomic instability Research Center, Ajou University School of Medicine; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon 16499, Korea
| |
Collapse
|
19
|
Potapova TA, Seidel CW, Box AC, Rancati G, Li R. Transcriptome analysis of tetraploid cells identifies cyclin D2 as a facilitator of adaptation to genome doubling in the presence of p53. Mol Biol Cell 2016; 27:3065-3084. [PMID: 27559130 PMCID: PMC5063615 DOI: 10.1091/mbc.e16-05-0268] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/16/2016] [Indexed: 01/12/2023] Open
Abstract
Tetraploidization, or genome doubling, is a prominent event in tumorigenesis, primarily because cell division in polyploid cells is error-prone and produces aneuploid cells. This study investigates changes in gene expression evoked in acute and adapted tetraploid cells and their effect on cell-cycle progression. Acute polyploidy was generated by knockdown of the essential regulator of cytokinesis anillin, which resulted in cytokinesis failure and formation of binucleate cells, or by chemical inhibition of Aurora kinases, causing abnormal mitotic exit with formation of single cells with aberrant nuclear morphology. Transcriptome analysis of these acute tetraploid cells revealed common signatures of activation of the tumor-suppressor protein p53. Suppression of proliferation in these cells was dependent on p53 and its transcriptional target, CDK inhibitor p21. Rare proliferating tetraploid cells can emerge from acute polyploid populations. Gene expression analysis of single cell-derived, adapted tetraploid clones showed up-regulation of several p53 target genes and cyclin D2, the activator of CDK4/6/2. Overexpression of cyclin D2 in diploid cells strongly potentiated the ability to proliferate with increased DNA content despite the presence of functional p53. These results indicate that p53-mediated suppression of proliferation of polyploid cells can be averted by increased levels of oncogenes such as cyclin D2, elucidating a possible route for tetraploidy-mediated genomic instability in carcinogenesis.
Collapse
Affiliation(s)
| | | | - Andrew C Box
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Giulia Rancati
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
20
|
Yant L, Bomblies K. Genome management and mismanagement--cell-level opportunities and challenges of whole-genome duplication. Genes Dev 2016; 29:2405-19. [PMID: 26637526 PMCID: PMC4691946 DOI: 10.1101/gad.271072.115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Whole-genome duplication (WGD) doubles the DNA content in the nucleus and leads to polyploidy. In this review, Yant and Bomblies discuss both the adaptive potential and problems associated with WGD, focusing primarily on cellular effects. Whole-genome duplication (WGD) doubles the DNA content in the nucleus and leads to polyploidy. In whole-organism polyploids, WGD has been implicated in adaptability and the evolution of increased genome complexity, but polyploidy can also arise in somatic cells of otherwise diploid plants and animals, where it plays important roles in development and likely environmental responses. As with whole organisms, WGD can also promote adaptability and diversity in proliferating cell lineages, although whether WGD is beneficial is clearly context-dependent. WGD is also sometimes associated with aging and disease and may be a facilitator of dangerous genetic and karyotypic diversity in tumorigenesis. Scaling changes can affect cell physiology, but problems associated with WGD in large part seem to arise from problems with chromosome segregation in polyploid cells. Here we discuss both the adaptive potential and problems associated with WGD, focusing primarily on cellular effects. We see value in recognizing polyploidy as a key player in generating diversity in development and cell lineage evolution, with intriguing parallels across kingdoms.
Collapse
Affiliation(s)
- Levi Yant
- John Innes Centre, Colney, Norwich NR4 7UH, United Kingdom
| | | |
Collapse
|
21
|
Shinn-Thomas JH, del Campo JJ, Wang J, Mohler WA. The EFF-1A Cytoplasmic Domain Influences Hypodermal Cell Fusions in C. elegans But Is Not Dependent on 14-3-3 Proteins. PLoS One 2016; 11:e0146874. [PMID: 26800457 PMCID: PMC4723337 DOI: 10.1371/journal.pone.0146874] [Citation(s) in RCA: 8] [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: 10/21/2014] [Accepted: 12/25/2015] [Indexed: 12/19/2022] Open
Abstract
Background Regulatory and biophysical mechanisms of cell-cell fusion are largely unknown despite the fundamental requirement for fused cells in eukaryotic development. Only two cellular fusogens that are not of clear recent viral origin have been identified to date, both in nematodes. One of these, EFF-1, is necessary for most cell fusions in Caenorhabditis elegans. Unregulated EFF-1 expression causes lethality due to ectopic fusion between cells not developmentally programmed to fuse, highlighting the necessity of tight fusogen regulation for proper development. Identifying factors that regulate EFF-1 and its paralog AFF-1 could lead to discovery of molecular mechanisms that control cell fusion upstream of the action of a membrane fusogen. Bioinformatic analysis of the EFF-1A isoform’s predicted cytoplasmic domain (endodomain) previously revealed two motifs that have high probabilities of interacting with 14-3-3 proteins when phosphorylated. Mutation of predicted phosphorylation sites within these motifs caused measurable loss of eff-1 gene function in cell fusion in vivo. Moreover, a human 14-3-3 isoform bound to EFF-1::GFP in vitro. We hypothesized that the two 14-3-3 proteins in C. elegans, PAR-5 and FTT-2, may regulate either localization or fusion-inducing activity of EFF-1. Methodology/Principal Findings Timing of fusion events was slightly but significantly delayed in animals unable to produce full-length EFF-1A. Yet, mutagenesis and live imaging showed that phosphoserines in putative 14-3-3 binding sites are not essential for EFF-1::GFP accumulation at the membrane contact between fusion partner cells. Moreover, although the EFF-1A endodomain was required for normal rates of eff-1-dependent epidermal cell fusions, reduced levels of FTT-2 and PAR-5 did not visibly affect the function of wild-type EFF-1 in the hypodermis. Conclusions/Significance Deletion of the EFF-1A endodomain noticeably affects the timing of hypodermal cell fusions in vivo. However, prohibiting phosphorylation of candidate 14-3-3-binding sites does not impact localization of the fusogen. Hypodermal membrane fusion activity persists when 14-3-3 expression levels are reduced.
Collapse
Affiliation(s)
- Jessica H. Shinn-Thomas
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
- * E-mail: (WAM); (JHST)
| | - Jacob J. del Campo
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
| | - Jianjun Wang
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
| | - William A. Mohler
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
- * E-mail: (WAM); (JHST)
| |
Collapse
|
22
|
Choudhary A, Zachek B, Lera RF, Zasadil LM, Lasek A, Denu RA, Kim H, Kanugh C, Laffin JJ, Harter JM, Wisinski KB, Saha S, Weaver BA, Burkard ME. Identification of Selective Lead Compounds for Treatment of High-Ploidy Breast Cancer. Mol Cancer Ther 2015; 15:48-59. [PMID: 26586723 DOI: 10.1158/1535-7163.mct-15-0527] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/09/2015] [Indexed: 11/16/2022]
Abstract
Increased ploidy is common in tumors but treatments for tumors with excess chromosome sets are not available. Here, we characterize high-ploidy breast cancers and identify potential anticancer compounds selective for the high-ploidy state. Among 354 human breast cancers, 10% have mean chromosome copy number exceeding 3, and this is most common in triple-negative and HER2-positive types. Women with high-ploidy breast cancers have higher risk of recurrence and death in two patient cohorts, demonstrating that it represents an important group for improved treatment. Because high-ploidy cancers are aneuploid, rather than triploid or tetraploid, we devised a two-step screen to identify selective compounds. The screen was designed to assure both external validity on diverse karyotypic backgrounds and specificity for high-ploidy cell types. This screen identified novel therapies specific to high-ploidy cells. First, we discovered 8-azaguanine, an antimetabolite that is activated by hypoxanthine phosphoribosyltransferase 1 (HPRT1), suggesting an elevated gene-dosage of HPRT1 in high-ploidy tumors can control sensitivity to this drug. Second, we discovered a novel compound, 2,3-diphenylbenzo[g]quinoxaline-5,10-dione (DPBQ). DPBQ activates p53 and triggers apoptosis in a polyploid-specific manner, but does not inhibit topoisomerase or bind DNA. Mechanistic analysis demonstrates that DPBQ elicits a hypoxia gene signature and its effect is replicated, in part, by enhancing oxidative stress. Structure-function analysis defines the core benzo[g]quinoxaline-5,10 dione as being necessary for the polyploid-specific effects of DPBQ. We conclude that polyploid breast cancers represent a high-risk subgroup and that DPBQ provides a functional core to develop polyploid-selective therapy. Mol Cancer Ther; 15(1); 48-59. ©2015 AACR.
Collapse
Affiliation(s)
- Alka Choudhary
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Brittany Zachek
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Robert F Lera
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lauren M Zasadil
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Amber Lasek
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ryan A Denu
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Hyunjung Kim
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Craig Kanugh
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, Wisconsin
| | - Jennifer J Laffin
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, Wisconsin. Department of Pediatrics, University of Wisconsin, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Josephine M Harter
- Department of Pathology, University of Wisconsin, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kari B Wisinski
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sandeep Saha
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Beth A Weaver
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mark E Burkard
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Hematology/Oncology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
23
|
Targeting the Mitotic Catastrophe Signaling Pathway in Cancer. Mediators Inflamm 2015; 2015:146282. [PMID: 26491220 PMCID: PMC4600505 DOI: 10.1155/2015/146282] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/30/2015] [Indexed: 12/14/2022] Open
Abstract
Mitotic catastrophe, as defined in 2012 by the International Nomenclature Committee on Cell Death, is a bona fide intrinsic oncosuppressive mechanism that senses mitotic failure and responds by driving a cell to an irreversible antiproliferative fate of death or senescence. Thus, failed mitotic catastrophe can promote the unrestrained growth of defective cells, thereby representing a major gateway to tumour development. Furthermore, the activation of mitotic catastrophe offers significant therapeutic advantage which has been exploited in the action of conventional and targeted anticancer agents. Yet, despite its importance in tumour prevention and treatment, the molecular mechanism of mitotic catastrophe is not well understood. A better understanding of the signals that determine cell fate following failed or defective mitosis will reveal new opportunities to selectively target and enhance the programme for therapeutic benefit and reveal biomarkers to predict patient response. This review is focused on the molecular mechanism of mitotic catastrophe induction and signalling and highlights current strategies to exploit the process in cancer therapy.
Collapse
|
24
|
Lazebnik Y. The shock of being united and symphiliosis. Another lesson from plants? Cell Cycle 2015; 13:2323-9. [PMID: 25483182 DOI: 10.4161/cc.29704] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Yuri Lazebnik
- a Yale Cardiovascular Research Center; New Haven, CT USA
| |
Collapse
|
25
|
Kaur E, Rajendra J, Jadhav S, Shridhar E, Goda JS, Moiyadi A, Dutt S. Radiation-induced homotypic cell fusions of innately resistant glioblastoma cells mediate their sustained survival and recurrence. Carcinogenesis 2015; 36:685-95. [PMID: 25863126 DOI: 10.1093/carcin/bgv050] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/23/2015] [Indexed: 12/13/2022] Open
Abstract
Understanding of molecular events underlying resistance and relapse in glioblastoma (GBM) is hampered due to lack of accessibility to resistant cells from patients undergone therapy. Therefore, we mimicked clinical scenario in an in vitro cellular model developed from five GBM grade IV primary patient samples and two cell lines. We show that upon exposure to lethal dose of radiation, a subpopulation of GBM cells, innately resistant to radiation, survive and transiently arrest in G2/M phase via inhibitory pCdk1(Y15). Although arrested, these cells show multinucleated and giant cell phenotype (MNGC). Significantly, we demonstrate that these MNGCs are not pre-existing giant cells from parent population but formed via radiation-induced homotypic cell fusions among resistant cells. Furthermore, cell fusions induce senescence, high expression of senescence-associated secretory proteins (SASPs) and activation of pro-survival signals (pAKT, BIRC3 and Bcl-xL) in MNGCs. Importantly, following transient non-proliferation, MNGCs escape senescence and despite having multiple spindle poles during mitosis, they overcome mitotic catastrophe to undergo normal cytokinesis forming mononucleated relapse population. This is the first report showing radiation-induced homotypic cell fusions as novel non-genetic mechanism in radiation-resistant cells to sustain survival. These data also underscore the importance of non-proliferative phase in resistant glioma cells. Accordingly, we show that pushing resistant cells into premature mitosis by Wee1 kinase inhibitor prevents pCdk1(Y15)-mediated cell cycle arrest and relapse. Taken together, our data provide novel molecular insights into a multistep process of radiation survival and relapse in GBM that can be exploited for therapeutic interventions.
Collapse
Affiliation(s)
- Ekjot Kaur
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Jacinth Rajendra
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Shailesh Jadhav
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Epari Shridhar
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Jayant Sastri Goda
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Aliasgar Moiyadi
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Shilpee Dutt
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| |
Collapse
|
26
|
He X, Li B, Shao Y, Zhao N, Hsu Y, Zhang Z, Zhu L. Cell fusion between gastric epithelial cells and mesenchymal stem cells results in epithelial-to-mesenchymal transition and malignant transformation. BMC Cancer 2015; 15:24. [PMID: 25633122 PMCID: PMC4318156 DOI: 10.1186/s12885-015-1027-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 01/20/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The discovery of cancer stem cells and tumor heterogeneity prompted the exploration of additional mechanisms aside from genetic mutations for carcinogenesis and cancer progression. The aim of the present study was to investigate the effect of cell fusion between mesenchymal stem cells and the gastric epithelial cells in tumorigenesis. METHODS Cell fusion between cord blood mesenchymal stem cells and human gastric epithelial cells was performed in vitro. Cell scratch and transwell assays were performed to determine migration and invasion abilities of the hybrids. The expressions of epithelial-mesenchymal transition-related proteins and genes were analyzed by immunocytochemistry and real time quantitative PCR. Tumorigenesis of the hybrids was evaluated through in vivo inoculation in nude mice. RESULTS Hybrids expressed the phenotypes of both donor cells. Aneuploidy was observed in 84.1% of cells. The hybrids showed increased proliferation, migration and invasion abilities compared with the parental cells. In addition, the expression of N-cadherin and vimentin in the hybrids was significantly higher than that of the epithelial cells, and the mRNA expression of the epithelial-mesenchymal transition-related genes, Twist and Slug, in the hybrids was also increased compared with that of the parental epithelial cells. Furthermore, the hybrids formed masses of epithelial origin with glandular structures in BALB/c nude mice. CONCLUSIONS These findings suggest that cell fusion between gastric epithelial cells and mesenchymal stem cells may result in epithelial to mesenchymal transition and malignant transformation.
Collapse
Affiliation(s)
- Xianghui He
- Department of General Surgery, Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Baosong Li
- Department of Anorectal Surgery, Affiliated Hospital of Binzhou Medical University, Shandong, 256603, China.
| | - Yang Shao
- Department of Surgery, Feixian Hospital, Shandong, 256603, China.
| | - Na Zhao
- Department of General Surgery, Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Yiling Hsu
- Department of General Surgery, Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Zhixiang Zhang
- Department of General Surgery, Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Liwei Zhu
- Department of General Surgery, Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
27
|
|
28
|
Panopoulos A, Pacios-Bras C, Choi J, Yenjerla M, Sussman MA, Fotedar R, Margolis RL. Failure of cell cleavage induces senescence in tetraploid primary cells. Mol Biol Cell 2014; 25:3105-18. [PMID: 25143403 PMCID: PMC4196863 DOI: 10.1091/mbc.e14-03-0844] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Induction of tetraploidy through cleavage failure induces G1 arrest and senescence in primary mammalian cells but not in immortal cells. Induction of senescence occurs without DNA damage, and the capacity to become senescent appears to be a prerequisite of tetraploid arrest. Tetraploidy can arise from various mitotic or cleavage defects in mammalian cells, and inheritance of multiple centrosomes induces aneuploidy when tetraploid cells continue to cycle. Arrest of the tetraploid cell cycle is therefore potentially a critical cellular control. We report here that primary rat embryo fibroblasts (REF52) and human foreskin fibroblasts become senescent in tetraploid G1 after drug- or small interfering RNA (siRNA)-induced failure of cell cleavage. In contrast, T-antigen–transformed REF52 and p53+/+ HCT116 tumor cells rapidly become aneuploid by continuing to cycle after cleavage failure. Tetraploid primary cells quickly become quiescent, as determined by loss of the Ki-67 proliferation marker and of the fluorescent ubiquitination-based cell cycle indicator/late cell cycle marker geminin. Arrest is not due to DNA damage, as the γ-H2AX DNA damage marker remains at control levels after tetraploidy induction. Arrested tetraploid cells finally become senescent, as determined by SA-β-galactosidase activity. Tetraploid arrest is dependent on p16INK4a expression, as siRNA suppression of p16INK4a bypasses tetraploid arrest, permitting primary cells to become aneuploid. We conclude that tetraploid primary cells can become senescent without DNA damage and that induction of senescence is critical to tetraploidy arrest.
Collapse
Affiliation(s)
- Andreas Panopoulos
- Tumor Initiation and Maintenance Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Cristina Pacios-Bras
- Department of Immunology and Oncology, Centro Nacional de Biotecnologia, Consejo Superior de Investigaciones Científicas, E-28049 Madrid, Spain
| | - Justin Choi
- Tumor Initiation and Maintenance Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Mythili Yenjerla
- Tumor Initiation and Maintenance Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Mark A Sussman
- San Diego Heart Research Institute and Department of Biology; San Diego State University, San Diego, CA 92182
| | - Rati Fotedar
- Tumor Initiation and Maintenance Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Robert L Margolis
- Tumor Initiation and Maintenance Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| |
Collapse
|
29
|
Maiato H, Logarinho E. Mitotic spindle multipolarity without centrosome amplification. Nat Cell Biol 2014; 16:386-94. [DOI: 10.1038/ncb2958] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Avanzi S, Alvisi G, Ripalti A. How virus persistence can initiate the tumorigenesis process. World J Virol 2013; 2:102-9. [PMID: 24175234 PMCID: PMC3785046 DOI: 10.5501/wjv.v2.i2.102] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 04/04/2013] [Accepted: 04/10/2013] [Indexed: 02/05/2023] Open
Abstract
Human oncogenic viruses are defined as necessary but not sufficient to initiate cancer. Experimental evidence suggests that the oncogenic potential of a virus is effective in cells that have already accumulated a number of genetic mutations leading to cell cycle deregulation. Current models for viral driven oncogenesis cannot explain why tumor development in carriers of tumorigenic viruses is a very rare event, occurring decades after virus infection. Considering that viruses are mutagenic agents per se and human oncogenic viruses additionally establish latent and persistent infections, we attempt here to provide a general mechanism of tumor initiation both for RNA and DNA viruses, suggesting viruses could be both necessary and sufficient in triggering human tumorigenesis initiation. Upon reviewing emerging evidence on the ability of viruses to induce DNA damage while subverting the DNA damage response and inducing epigenetic disturbance in the infected cell, we hypothesize a general, albeit inefficient hit and rest mechanism by which viruses may produce a limited reservoir of cells harboring permanent damage that would be initiated when the virus first hits the cell, before latency is established. Cells surviving virus generated damage would consequently become more sensitive to further damage mediated by the otherwise insufficient transforming activity of virus products expressed in latency, or upon episodic reactivations (viral persistence). Cells with a combination of genetic and epigenetic damage leading to a cancerous phenotype would emerge very rarely, as the probability of such an occurrence would be dependent on severity and frequency of consecutive hit and rest cycles due to viral reinfections and reactivations.
Collapse
|
31
|
Abstract
Cellular defects that impair the fidelity of mitosis promote chromosome missegregation and aneuploidy. Increasing evidence reveals that errors in mitosis can also promote the direct and indirect acquisition of DNA damage and chromosome breaks. Consequently, deregulated cell division can devastate the integrity of the normal genome and unleash a variety of oncogenic stimuli that may promote transformation. Recent work has shed light on the mechanisms that link abnormal mitosis with the development of DNA damage, how cells respond to such affronts, and the potential impact on tumorigenesis.
Collapse
Affiliation(s)
- Neil J Ganem
- Howard Hughes Medical Institute, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Children's Hospital, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
32
|
Palma J, Yaddanapudi SC, Pigati L, Havens MA, Jeong S, Weiner GA, Weimer KME, Stern B, Hastings ML, Duelli DM. MicroRNAs are exported from malignant cells in customized particles. Nucleic Acids Res 2012; 40:9125-38. [PMID: 22772984 PMCID: PMC3467054 DOI: 10.1093/nar/gks656] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are released from cells in association with proteins or microvesicles. We previously reported that malignant transformation changes the assortment of released miRNAs by affecting whether a particular miRNA species is released or retained by the cell. How this selectivity occurs is unclear. Here we report that selectively exported miRNAs, whose release is increased in malignant cells, are packaged in structures that are different from those that carry neutrally released miRNAs (n-miRNAs), whose release is not affected by malignancy. By separating breast cancer cell microvesicles, we find that selectively released miRNAs associate with exosomes and nucleosomes. However, n-miRNAs of breast cancer cells associate with unconventional exosomes, which are larger than conventional exosomes and enriched in CD44, a protein relevant to breast cancer metastasis. Based on their large size, we call these vesicles L-exosomes. Contrary to the distribution of miRNAs among different microvesicles of breast cancer cells, normal cells release all measured miRNAs in a single type of vesicle. Our results suggest that malignant transformation alters the pathways through which specific miRNAs are exported from cells. These changes in the particles and their miRNA cargo could be used to detect the presence of malignant cells in the body.
Collapse
Affiliation(s)
- Jaime Palma
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Losing balance: the origin and impact of aneuploidy in cancer. EMBO Rep 2012; 13:501-14. [PMID: 22565320 DOI: 10.1038/embor.2012.55] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/03/2012] [Indexed: 02/06/2023] Open
Abstract
Most solid human tumours are aneuploid, that is, they contain an abnormal number of chromosomes. Paradoxically, however, aneuploidy has been reported to induce a stress response that suppresses cellular proliferation in vitro. Here, we review the progress in our understanding of the causes and effects of aneuploidy in cancer and discuss how, in specific contexts, aneuploidy can provide a growth advantage and facilitate cellular transformation. We also explore the emerging possibilities for targeting the cause or consequences of aneuploidy therapeutically.
Collapse
|
34
|
Skinner AM, Grompe M, Kurre P. Intra-hematopoietic cell fusion as a source of somatic variation in the hematopoietic system. J Cell Sci 2012; 125:2837-43. [PMID: 22393240 DOI: 10.1242/jcs.100123] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cell fusion plays a well-recognized, physiological role during development. Bone-marrow-derived hematopoietic cells have been shown to fuse with non-hematopoietic cells in a wide variety of tissues. Some organs appear to resolve the changes in ploidy status, generating functional and mitotically-competent events. However, cell fusion exclusively involving hematopoietic cells has not been reported. Indeed, genomic copy number variation in highly replicative hematopoietic cells is widely considered a hallmark of malignant transformation. Here we show that cell fusion occurs between cells of the hematopoietic system under injury as well as non-injury conditions. Experiments reveal the acquisition of genetic markers in fusion products, their tractable maintenance during hematopoietic differentiation and long-term persistence after serial transplantation. Fusion events were identified in clonogenic progenitors as well as differentiated myeloid and lymphoid cells. These observations provide a new experimental model for the study of non-pathogenic somatic diversity in the hematopoietic system.
Collapse
Affiliation(s)
- Amy M Skinner
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | | | | |
Collapse
|
35
|
Chan JY. A clinical overview of centrosome amplification in human cancers. Int J Biol Sci 2011; 7:1122-44. [PMID: 22043171 PMCID: PMC3204404 DOI: 10.7150/ijbs.7.1122] [Citation(s) in RCA: 279] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 10/06/2011] [Indexed: 01/11/2023] Open
Abstract
The turn of the 21st century had witnessed a surge of interest in the centrosome and its causal relation to human cancer development - a postulate that has existed for almost a century. Centrosome amplification (CA) is frequently detected in a growing list of human cancers, both solid and haematological, and is a candidate "hallmark" of cancer cells. Several lines of evidence support the progressive involvement of CA in the transition from early to advanced stages of carcinogenesis, being also found in pre-neoplastic lesions and even in histopathologically-normal tissue. CA constitutes the major mechanism leading to chromosomal instability and aneuploidy, via the formation of multipolar spindles and chromosomal missegregation. Clinically, CA may translate to a greater risk for initiation of malignant transformation, tumour progression, chemoresistance and ultimately, poor patient prognosis. As mechanisms underlying CA are progressively being unravelled, the centrosome has emerged as a novel candidate target for cancer treatment. This Review summarizes mainly the clinical studies performed to date focusing on the mechanisms underlying CA in human neoplasia, and highlights the potential utility of centrosomes in the diagnosis, prognosis and treatment of human cancers.
Collapse
|
36
|
A non-genetic route to aneuploidy in human cancers. Nat Cell Biol 2011; 13:324-30. [PMID: 21336303 DOI: 10.1038/ncb2174] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 12/17/2010] [Indexed: 12/18/2022]
Abstract
Aneuploidy is common in human tumours and is often indicative of aggressive disease. Aneuploidy can result from cytokinesis failure, which produces binucleate cells that generate aneuploid offspring with subsequent divisions. In cancers, disruption of cytokinesis is known to result from genetic perturbations to mitotic pathways or checkpoints. Here we describe a non-genetic mechanism of cytokinesis failure that occurs as a direct result of cell-in-cell formation by entosis. Live cells internalized by entosis, which can persist through the cell cycle of host cells, disrupt formation of the contractile ring during host cell division. As a result, cytokinesis frequently fails, generating binucleate cells that produce aneuploid cell lineages. In human breast tumours, multinucleation is associated with cell-in-cell structures. These data define a previously unknown mechanism of cytokinesis failure and aneuploid cell formation that operates in human cancers.
Collapse
|
37
|
Cell Fusion and Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:161-75. [DOI: 10.1007/978-94-007-0763-4_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
New Insights into the Mechanisms and Roles of Cell–Cell Fusion. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 289:149-209. [DOI: 10.1016/b978-0-12-386039-2.00005-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
39
|
Pigati L, Yaddanapudi SCS, Iyengar R, Kim DJ, Hearn SA, Danforth D, Hastings ML, Duelli DM. Selective release of microRNA species from normal and malignant mammary epithelial cells. PLoS One 2010; 5:e13515. [PMID: 20976003 PMCID: PMC2958125 DOI: 10.1371/journal.pone.0013515] [Citation(s) in RCA: 461] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 09/14/2010] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) in body fluids are candidate diagnostics for a variety of conditions and diseases, including breast cancer. One premise for using extracellular miRNAs to diagnose disease is the notion that the abundance of the miRNAs in body fluids reflects their abundance in the abnormal cells causing the disease. As a result, the search for such diagnostics in body fluids has focused on miRNAs that are abundant in the cells of origin. Here we report that released miRNAs do not necessarily reflect the abundance of miRNA in the cell of origin. We find that release of miRNAs from cells into blood, milk and ductal fluids is selective and that the selection of released miRNAs may correlate with malignancy. In particular, the bulk of miR-451 and miR-1246 produced by malignant mammary epithelial cells was released, but the majority of these miRNAs produced by non-malignant mammary epithelial cells was retained. Our findings suggest the existence of a cellular selection mechanism for miRNA release and indicate that the extracellular and cellular miRNA profiles differ. This selective release of miRNAs is an important consideration for the identification of circulating miRNAs as biomarkers of disease.
Collapse
Affiliation(s)
- Lucy Pigati
- Department of Pathology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Sree C. S. Yaddanapudi
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- The Bligh Cancer Research Center, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Ravi Iyengar
- Department of Pathology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Dong-Ja Kim
- Department of Pathology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Steven A. Hearn
- Cold Spring Harbor Laboratory Microscopy Facility, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - David Danforth
- Surgery Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Michelle L. Hastings
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Dominik M. Duelli
- Department of Pathology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
40
|
Boveri revisited: chromosomal instability, aneuploidy and tumorigenesis. Nat Rev Mol Cell Biol 2009; 10:478-87. [PMID: 19546858 DOI: 10.1038/nrm2718] [Citation(s) in RCA: 643] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mitotic checkpoint is a major cell cycle control mechanism that guards against chromosome missegregation and the subsequent production of aneuploid daughter cells. Most cancer cells are aneuploid and frequently missegregate chromosomes during mitosis. Indeed, aneuploidy is a common characteristic of tumours, and, for over 100 years, it has been proposed to drive tumour progression. However, recent evidence has revealed that although aneuploidy can increase the potential for cellular transformation, it also acts to antagonize tumorigenesis in certain genetic contexts. A clearer understanding of the tumour suppressive function of aneuploidy might reveal new avenues for anticancer therapy.
Collapse
|
41
|
Sariyer IK, Safak M, Gordon J, Khalili K. Generation and characterization of JCV permissive hybrid cell lines. J Virol Methods 2009; 159:122-6. [PMID: 19442856 DOI: 10.1016/j.jviromet.2009.02.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 02/10/2009] [Accepted: 02/19/2009] [Indexed: 11/30/2022]
Abstract
JC virus (JCV) is a human neurotropic polyomavirus whose replication in the central nervous system induces the fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML). JCV particles have been detected primarily in oligodendrocytes and astrocytes of the brains of patients with PML and in the laboratory its propagation is limited to primary cultures of human fetal glial cells. In this short communication, the development of a new cell culture system is described through the fusion of primary human fetal astrocytes with the human glioblastoma cell line, U-87MG. The new hybrid cell line obtained from this fusion has the capacity to support efficiently expression of JCV and replication of viral DNA in vitro up to 16 passages. This cell line can serve as a reliable culture system to study the biology of JCV host-cell interaction, determine the mechanisms involved in cell type specific replication of JCV, and provide a convenient cell culture system for high throughput screening of anti-viral agents.
Collapse
Affiliation(s)
- Ilker K Sariyer
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, 1900 North 12th Street, 015-96, Room 203, Philadelphia, PA 19122, USA
| | | | | | | |
Collapse
|
42
|
Abstract
Polyploidy, an increased number of chromosome sets, is a surprisingly common phenomenon in nature, particularly in plants and fungi. In humans, polyploidy often occurs in specific tissues as part of terminal differentiation. Changes in ploidy can also result from pathophysiological events that are caused by viral-induced cell fusion or erroneous cell division. Tetraploidization can initiate chromosomal instability (CIN), probably owing to supernumerary centrosomes and the doubled chromosome mass. CIN, in turn, might persist or soon give way to a stably propagating but aneuploid karyotype. Both CIN and stable aneuploidy are commonly observed in cancers. Recently, it has been proposed that an increased number of chromosome sets can promote cell transformation and give rise to an aneuploid tumor. Here, we review how tetraploidy can occur and describe the cellular responses to increased ploidy. Furthermore, we discuss how the specific physiological changes that are triggered by polyploidization might be used as novel targets for cancer therapy.
Collapse
Affiliation(s)
- Zuzana Storchova
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| | | |
Collapse
|
43
|
Chan YW, On KF, Chan WM, Wong W, Siu HO, Hau PM, Poon RYC. The kinetics of p53 activation versus cyclin E accumulation underlies the relationship between the spindle-assembly checkpoint and the postmitotic checkpoint. J Biol Chem 2008; 283:15716-23. [PMID: 18400748 DOI: 10.1074/jbc.m800629200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Although cells can exit mitotic block aberrantly by mitotic slippage, they are prevented from becoming tetraploids by a p53-dependent postmitotic checkpoint. Intriguingly, disruption of the spindle-assembly checkpoint also compromises the postmitotic checkpoint. The precise mechanism of the interplay between these two pivotal checkpoints is not known. We found that after prolonged nocodazole exposure, the postmitotic checkpoint was facilitated by p53. We demonstrated that although disruption of the mitotic block by a MAD2-binding protein promoted slippage, it did not influence the activation of p53. Both p53 and its downstream target p21(CIP1/WAF1) were activated at the same rate irrespective of whether the spindle-assembly checkpoint was enforced or not. The accelerated S phase entry, as reflected by the premature accumulation of cyclin E relative to the activation of p21(CIP1/WAF1), is the reason for the uncoupling of the postmitotic checkpoint. In support of this hypothesis, forced premature mitotic exit with a specific CDK1 inhibitor triggered DNA replication without affecting the kinetics of p53 activation. Finally, replication after checkpoint bypass was boosted by elevating the level of cyclin E. These observations indicate that disruption of the spindle-assembly checkpoint does not directly influence p53 activation, but the shortening of the mitotic arrest allows cyclin E-CDK2 to be activated before the accumulation of p21(CIP1/WAF1). These data underscore the critical relationship between the spindle-assembly checkpoint and the postmitotic checkpoint in safeguarding chromosomal stability.
Collapse
Affiliation(s)
- Ying Wai Chan
- Department of Biochemistry, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Cell fusions are important to fertilization, placentation, development of skeletal muscle and bone, calcium homeostasis and the immune defense system. Additionally, cell fusions participate in tissue repair and may be important to cancer development and progression. A large number of factors appear to regulate cell fusions, including receptors and ligands, membrane domain organizing proteins, proteases, signaling molecules and fusogenic proteins forming alpha-helical bundles that bring membranes close together. The syncytin family of proteins represent true fusogens and the founding member, syncytin-1, has been documented to be involved in fusions between placental trophoblasts, between cancer cells and between cancer cells and host cells. We review the literature with emphasis on the syncytin family and propose that syncytins may represent universal fusogens in primates and rodents, which work together with a number of other proteins to regulate the cell fusion machinery.
Collapse
|
45
|
Parris GE. 2-Deoxy-d-glucose as a potential drug against fusogenic viruses including HIV. Med Hypotheses 2008; 70:776-82. [DOI: 10.1016/j.mehy.2007.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 08/05/2007] [Indexed: 01/08/2023]
|
46
|
Vitale I, Galluzzi L, Vivet S, Nanty L, Dessen P, Senovilla L, Olaussen KA, Lazar V, Prudhomme M, Golsteyn RM, Castedo M, Kroemer G. Inhibition of Chk1 kills tetraploid tumor cells through a p53-dependent pathway. PLoS One 2007; 2:e1337. [PMID: 18159231 PMCID: PMC2131784 DOI: 10.1371/journal.pone.0001337] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2007] [Accepted: 11/16/2007] [Indexed: 11/19/2022] Open
Abstract
Tetraploidy constitutes an adaptation to stress and an intermediate step between euploidy and aneuploidy in oncogenesis. Tetraploid cells are particularly resistant against genotoxic stress including radiotherapy and chemotherapy. Here, we designed a strategy to preferentially kill tetraploid tumor cells. Depletion of checkpoint kinase-1 (Chk1) by siRNAs, transfection with dominant-negative Chk1 mutants or pharmacological Chk1 inhibition killed tetraploid colon cancer cells yet had minor effects on their diploid counterparts. Chk1 inhibition abolished the spindle assembly checkpoint and caused premature and abnormal mitoses that led to p53 activation and cell death at a higher frequency in tetraploid than in diploid cells. Similarly, abolition of the spindle checkpoint by knockdown of Bub1, BubR1 or Mad2 induced p53-dependent apoptosis of tetraploid cells. Chk1 inhibition reversed the cisplatin resistance of tetraploid cells in vitro and in vivo, in xenografted human cancers. Chk1 inhibition activated p53-regulated transcripts including Puma/BBC3 in tetraploid but not in diploid tumor cells. Altogether, our results demonstrate that, in tetraploid tumor cells, the inhibition of Chk1 sequentially triggers aberrant mitosis, p53 activation and Puma/BBC3-dependent mitochondrial apoptosis.
Collapse
Affiliation(s)
- Ilio Vitale
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Lorenzo Galluzzi
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Sonia Vivet
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Lisa Nanty
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
| | - Philippe Dessen
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
- Centre National de la Recherche Scientifique (CNRS), FRE2939, Villejuif, France
- Unité de Génomique Fonctionnelle, Institut Gustave Roussy,Villejuif, France
| | - Laura Senovilla
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Ken A. Olaussen
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Vladimir Lazar
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
- Centre National de la Recherche Scientifique (CNRS), FRE2939, Villejuif, France
- Unité de Génomique Fonctionnelle, Institut Gustave Roussy,Villejuif, France
| | - Michelle Prudhomme
- Université Blaise Pascal, Synthèse et Etude de Systèmes à Intérêt Biologique, UMR 6504 Centre National de la Recherche Scientifique (CNRS), Aubière, France
| | | | - Maria Castedo
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Guido Kroemer
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| |
Collapse
|
47
|
Abstract
The ability to fuse cells is shared by many viruses, including common human pathogens and several endogenous viruses. Here we will discuss how cell fusion can link viruses to cancer, what types of cancers it can affect, how the existence of this link can be tested and how the hypotheses that we propose might affect the search for human oncogenic viruses. In particular, we will focus on the ability of cell fusion that is caused by viruses to induce chromosomal instability, a common affliction of cancer cells that has been thought to underlie the malignant properties of cancerous tumours.
Collapse
Affiliation(s)
- Dominik Duelli
- Department of Pathology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, USA.
| | | |
Collapse
|
48
|
Jacobsen BM, Harrell JC, Jedlicka P, Borges VF, Varella-Garcia M, Horwitz KB. Spontaneous fusion with, and transformation of mouse stroma by, malignant human breast cancer epithelium. Cancer Res 2007; 66:8274-9. [PMID: 16912208 DOI: 10.1158/0008-5472.can-06-1456] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adenocarcinoma cells from the pleural effusion of a patient with breast cancer were injected into the mammary glands of nude mice and grown into solid tumors. A cell line derived from these tumors expressed alpha-smooth muscle actin but not human cytokeratin 7, indicating "activated" stroma of mouse origin. Cells in mitosis exhibited mainly polyploid mouse karyotypes, but 30% had mixed mouse and human chromosomes, among which 8% carried mouse/human translocations. Nuclei of interphase cells were 64% hybrid. Hybrid mouse/human nuclei were also detected in the primary xenograft. Thus, synkaryons formed in the solid tumor by spontaneous fusion between the malignant human epithelium and the surrounding normal host mouse stroma. The transformed stroma-derived cells are tumorigenic with histopathologic features of malignancy, suggesting a new mechanism for tumor progression.
Collapse
Affiliation(s)
- Britta M Jacobsen
- Department of Medicine, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Hastings ML, Allemand E, Duelli DM, Myers MP, Krainer AR. Control of pre-mRNA splicing by the general splicing factors PUF60 and U2AF(65). PLoS One 2007; 2:e538. [PMID: 17579712 PMCID: PMC1888729 DOI: 10.1371/journal.pone.0000538] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 05/22/2007] [Indexed: 11/19/2022] Open
Abstract
Pre-mRNA splicing is a crucial step in gene expression, and accurate recognition of splice sites is an essential part of this process. Splice sites with weak matches to the consensus sequences are common, though it is not clear how such sites are efficiently utilized. Using an in vitro splicing-complementation approach, we identified PUF60 as a factor that promotes splicing of an intron with a weak 3′ splice-site. PUF60 has homology to U2AF65, a general splicing factor that facilitates 3′ splice-site recognition at the early stages of spliceosome assembly. We demonstrate that PUF60 can functionally substitute for U2AF65in vitro, but splicing is strongly stimulated by the presence of both proteins. Reduction of either PUF60 or U2AF65 in cells alters the splicing pattern of endogenous transcripts, consistent with the idea that regulation of PUF60 and U2AF65 levels can dictate alternative splicing patterns. Our results indicate that recognition of 3′ splice sites involves different U2AF-like molecules, and that modulation of these general splicing factors can have profound effects on splicing.
Collapse
Affiliation(s)
- Michelle L. Hastings
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Eric Allemand
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Dominik M. Duelli
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Michael P. Myers
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Adrian R. Krainer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
50
|
Inducing instability. Nat Rev Microbiol 2007. [DOI: 10.1038/nrmicro1646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|