51
|
Du M, Zhang S, Liu X, Xu C, Zhang X. Ploidy Status of Ovarian Cancer Cell Lines and Their Association with Gene Expression Profiles. Biomolecules 2023; 13:biom13010092. [PMID: 36671477 PMCID: PMC9855421 DOI: 10.3390/biom13010092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
As a cancer type potentially dominated by copy number variations, ovarian cancer shows hyperploid karyotypes and large-scale chromosome alterations, which might be promising biomarkers correlated with tumor metastasis and chemoresistance. Experimental studies have provided more information about the roles of aneuploids and polyploids in ovarian cancer. However, ploidy evaluation of ovarian cancer cell lines is still limited, even in some ploidy-related research. Herein, the ploidy landscape of 51 ovarian cancer cell lines from the Cancer Cell Line Encyclopedia (CCLE) were analyzed, and the ploidy statuses of 13 human ovarian cancer cell lines and 2 murine cell lines were evaluated using G-banding and flow cytometry. Most human ovarian cancer cell lines were aneuploid, with modal numbers of 52-86 and numerical complexity ranging from 5 to 12. A2780, COV434 and TOV21G were screened as diploid cell lines, with a modal number of 46, a low aneuploid score and a near-diploid ploidy value. Two murine cell lines, both OV2944-HM1 and ID-8, were near-tetraploid. Integrated information on karyotypes, aneuploid score and ploidy value supplied references for a nondiploid model construction and a parallel analysis of diploid versus aneuploid. Moreover, the gene expression profiles were compared between diploid and aneuploid cell lines. The functions of differentially expressed genes were mainly enriched in terms of protein function regulation, TGF-β signaling and cell adhesion molecules. Genes downregulated in the aneuploid group were mainly related to metabolism and protein function regulation, and genes upregulated in the aneuploid group were mainly involved in immune regulation. Differentially expressed genes were randomly distributed on all chromosomes, while chromosome 1 alteration might contribute to immune-related alterations in aneuploid cell lines. Chromosome 19 alteration might be potentially significant for aneuploid ovarian cancer cell lines and patients, which needs further verification in ploidy research.
Collapse
Affiliation(s)
- Ming Du
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Shuo Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Xiaoxia Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Congjian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
- Correspondence: (C.X.); (X.Z.)
| | - Xiaoyan Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
- Correspondence: (C.X.); (X.Z.)
| |
Collapse
|
52
|
Zamkova MA, Persiyantseva NA, Tatarskiy VV, Shtil AA. Therapy-Induced Tumor Cell Senescence: Mechanisms and Circumvention. BIOCHEMISTRY (MOSCOW) 2023; 88:86-104. [PMID: 37068872 DOI: 10.1134/s000629792301008x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Plasticity of tumor cells (multitude of molecular regulation pathways) allows them to evade cytocidal effects of chemo- and/or radiation therapy. Metabolic adaptation of the surviving cells is based on transcriptional reprogramming. Similarly to the process of natural cell aging, specific features of the survived tumor cells comprise the therapy-induced senescence phenotype. Tumor cells with this phenotype differ from the parental cells since they become less responsive to drugs and form aggressive progeny. Importance of the problem is explained by the general biological significance of transcriptional reprogramming as a mechanism of adaptation to stress, and by the emerging potential of its pharmacological targeting. In this review we analyze the mechanisms of regulation of the therapy-induced tumor cell senescence, as well as new drug combinations aimed to prevent this clinically unfavorable phenomenon.
Collapse
Affiliation(s)
- Maria A Zamkova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
| | - Nadezhda A Persiyantseva
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Alexander A Shtil
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
- Institute of Cyber Intelligence Systems, National Research Nuclear University MEPHI, Moscow, 115409, Russia
| |
Collapse
|
53
|
Cheng T, Zhang S, Xia T, Zhang Y, Ji Y, Pan S, Xie H, Ren Q, You Y, You B. EBV promotes vascular mimicry of dormant cancer cells by potentiating stemness and EMT. Exp Cell Res 2022; 421:113403. [PMID: 36336028 DOI: 10.1016/j.yexcr.2022.113403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/15/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022]
Abstract
Vascular mimicry (VM) is defined as a vascular channel-like structure composed of tumor cells that correlates with the growth of cancer cells by providing blood circulation. However, whether VM can be formed in dormant cancer cells remains unclear. Our previous research revealed that polyploid giant cancer cells (PGCCs) are specific dormant cells related to the poor prognosis of head and neck cancer. Here, we demonstrated that EBV could promote VM formation by PGCCs in vivo and in vitro. Furthermore, we revealed that the activation of the ERK pathway partly mediated by LMP2A is responsible for stemness, and the acquisition of the stemness phenotype is crucial to the malignant biological behavior of PGCCs. The epithelial-to-mesenchymal transition (EMT) process plays a considerable role in PGCCs, and EMT progression is vital for EBV-positive PGCCs to form VM. This is the first study to reveal that EBV creates plasticity in PGCC-VM and provide a new strategy for targeted anti-tumor therapy.
Collapse
Affiliation(s)
- Tianyi Cheng
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Department of Otorhinolaryngology Head and Neck Surgery, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Siyu Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China
| | - Tian Xia
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China
| | - Yanshu Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Yan Ji
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China
| | - Si Pan
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China
| | - Haijing Xie
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China
| | - Qianqian Ren
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China.
| | - Bo You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China; Medical College of Nantong University, Nantong 226019, Jiangsu Province, China. Department of Pathology, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong, 226019, Jiangsu Province, China.
| |
Collapse
|
54
|
Vainshelbaum NM, Giuliani A, Salmina K, Pjanova D, Erenpreisa J. The Transcriptome and Proteome Networks of Malignant Tumours Reveal Atavistic Attractors of Polyploidy-Related Asexual Reproduction. Int J Mol Sci 2022; 23:ijms232314930. [PMID: 36499258 PMCID: PMC9736112 DOI: 10.3390/ijms232314930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/18/2022] [Accepted: 11/26/2022] [Indexed: 12/02/2022] Open
Abstract
The expression of gametogenesis-related (GG) genes and proteins, as well as whole genome duplications (WGD), are the hallmarks of cancer related to poor prognosis. Currently, it is not clear if these hallmarks are random processes associated only with genome instability or are programmatically linked. Our goal was to elucidate this via a thorough bioinformatics analysis of 1474 GG genes in the context of WGD. We examined their association in protein-protein interaction and coexpression networks, and their phylostratigraphic profiles from publicly available patient tumour data. The results show that GG genes are upregulated in most WGD-enriched somatic cancers at the transcriptome level and reveal robust GG gene expression at the protein level, as well as the ability to associate into correlation networks and enrich the reproductive modules. GG gene phylostratigraphy displayed in WGD+ cancers an attractor of early eukaryotic origin for DNA recombination and meiosis, and one relative to oocyte maturation and embryogenesis from early multicellular organisms. The upregulation of cancer-testis genes emerging with mammalian placentation was also associated with WGD. In general, the results suggest the role of polyploidy for soma-germ transition accessing latent cancer attractors in the human genome network, which appear as pre-formed along the whole Evolution of Life.
Collapse
Affiliation(s)
- Ninel M. Vainshelbaum
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia
- Faculty of Biology, The University of Latvia, LV-1586 Riga, Latvia
- Correspondence: (N.M.V.); (J.E.)
| | - Alessandro Giuliani
- Environmen and Health Department, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Kristine Salmina
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia
| | - Dace Pjanova
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia
| | - Jekaterina Erenpreisa
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia
- Correspondence: (N.M.V.); (J.E.)
| |
Collapse
|
55
|
Ma Z, Zhang F, Xiong J, Zhang H, Lin HK, Liu C. Activation of embryonic/germ cell-like axis links poor outcomes of gliomas. Cancer Cell Int 2022; 22:371. [DOI: 10.1186/s12935-022-02792-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 11/14/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
It is unclear which core events drive the malignant progression of gliomas. Earlier studies have revealed that the embryonic stem (ES) cell/early PGC state is associated with tumourigenicity. This study was designed to investigate the role of ES/PGC state in poor outcomes of gliomas.
Methods
Crispr-Cas9 technology, RT–PCR and animal experiments were used to investigate whether PGC-like cell formation play crucial roles in the tumorigenicity of human glioma cells. Bioinformatic analysis was used to address the link between ES/PGC developmental axis and glioma overall outcomes.
Results
Here, our findings showed that germ cell-like cells were present in human gliomas and cultured glioma cells and that the formation of germ cell-like cells was essential for glioma tumours. Bioinformatic analysis showed that the mRNA levels of genes related to embryonic/germ cell development could be detected in most gliomas. Our findings showed that the activation of genes related to reprogramming or the germ cell-like state alone seemed to be insufficient to lead to a malignant prognosis, whereas increased mRNA levels of genes related to the activation of the embryonic/germ cell-like cycle (somatic PGC-EGC-like cycle and somatic parthenogenetic embryo-like cycle) were positively correlated with malignant prognoses and poor clinical outcomes of gliomas. Genes related to the embryonic/germ cell cycle alone or in combination with the WHO grade or 1p19q codeletion status could be used to subdivide gliomas with distinct clinical behaviours.
Conclusion
Together, our findings indicated that a crucial role of germ cell-like cell formation in glioma initiation as well as activation of genes related with the parthenogenetic embryo-like cycle and PGC-EGC-like cycle link to the malignant prognosis and poor outcomes of gliomas, which might provide a novel way to better understand the nature of and develop targeted therapies for gliomas as well as important markers for predicting clinical outcomes in gliomas.
Collapse
|
56
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
57
|
Zhou X, Zhou M, Zheng M, Tian S, Yang X, Ning Y, Li Y, Zhang S. Polyploid giant cancer cells and cancer progression. Front Cell Dev Biol 2022; 10:1017588. [PMID: 36274852 PMCID: PMC9581214 DOI: 10.3389/fcell.2022.1017588] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/21/2022] [Indexed: 12/02/2022] Open
Abstract
Polyploid giant cancer cells (PGCCs) are an important feature of cellular atypia, the detailed mechanisms of their formation and function remain unclear. PGCCs were previously thought to be derived from repeated mitosis/cytokinesis failure, with no intrinsic ability to proliferate and divide. However, recently, PGCCs have been confirmed to have cancer stem cell (CSC)-like characteristics, and generate progeny cells through asymmetric division, which express epithelial-mesenchymal transition-related markers to promote invasion and migration. The formation of PGCCs can be attributed to multiple stimulating factors, including hypoxia, chemotherapeutic reagents, and radiation, can induce the formation of PGCCs, by regulating the cell cycle and cell fusion-related protein expression. The properties of CSCs suggest that PGCCs can be induced to differentiate into non-tumor cells, and produce erythrocytes composed of embryonic hemoglobin, which have a high affinity for oxygen, and thereby allow PGCCs survival from the severe hypoxia. The number of PGCCs is associated with metastasis, chemoradiotherapy resistance, and recurrence of malignant tumors. Targeting relevant proteins or signaling pathways related with the formation and transdifferentiation of adipose tissue and cartilage in PGCCs may provide new strategies for solid tumor therapy.
Collapse
Affiliation(s)
- Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Mingming Zhou
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Shiwu Zhang,
| |
Collapse
|
58
|
Borkowska A, Olszewska A, Skarzynska W, Marciniak M, Skrzeszewski M, Kieda C, Was H. High Hemin Concentration Induces Escape from Senescence of Normoxic and Hypoxic Colon Cancer Cells. Cancers (Basel) 2022; 14:cancers14194793. [PMID: 36230727 PMCID: PMC9564005 DOI: 10.3390/cancers14194793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary High red-meat consumption as well as bleeding or bruising can promote oxidative stress and, in consequence, cancer development. However, the mechanism of that phenomenon is not understood. The induction of therapy-induced senescence (TIS) might also be induced by oxidative stress. Recently, TIS cells, despite their inhibited proliferation potential, have been identified as one of the sources of tumor re-growth. Here, with the use of molecular analyses, we found that oxidative stress, promoted by high doses of hemin or H2O2, can trigger TIS escape and cell re-population. It is closely related to the activity of antioxidative enzymes, especially heme oxygenase-1. Hypoxia might accelerate these effects. Therefore, we propose that the prevention of excessive oxidative stress could be a potential target in senolytic therapies. Abstract Hemoglobin from either red meat or bowel bleeding may promote oxidative stress and increase the risk of colorectal cancer (CRC). Additionally, solid cancers or their metastases may be present with localized bruising. Escape from therapy-induced senescence (TIS) might be one of the mechanisms of tumor re-growth. Therefore, we sought to study whether hemin can cause escape from TIS in CRC. To induce senescence, human colon cancer cells were exposed to a chemotherapeutic agent irinotecan (IRINO). Cells treated with IRINO exhibited common hallmarks of TIS. To mimic bleeding, colon cancer cells were additionally treated with hemin. High hemin concentration activated heme oxygenase-1 (HO-1), induced escape from TIS and epithelial-to-mesenchymal transition, and augmented progeny production. The effect was even stronger in hypoxic conditions. Similar results were obtained when TIS cells were treated with another prooxidant agent, H2O2. Silencing of antioxidative enzymes such as catalase (CAT) or glutathione peroxidase-1 (GPx-1) maintained colon cancer cells in a senescent state. Our study demonstrates that a high hemin concentration combined with an increased activity of antioxidative enzymes, especially HO-1, leads to escape from the senescence of colon cancer cells. Therefore, our observations could be used in targeted anti-cancer therapy.
Collapse
Affiliation(s)
- Agata Borkowska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, 04-141 Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091 Warsaw, Poland
| | - Aleksandra Olszewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, 04-141 Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091 Warsaw, Poland
| | - Weronika Skarzynska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, 04-141 Warsaw, Poland
| | - Marta Marciniak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, 04-141 Warsaw, Poland
| | - Maciej Skrzeszewski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, 04-141 Warsaw, Poland
- Doctoral School of Translational Medicine, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, 04-141 Warsaw, Poland
- Centre for Molecular Biophysics, UPR CNRS 4301, CEDEX 2, 45071 Orléans, France
| | - Halina Was
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, 04-141 Warsaw, Poland
- Correspondence:
| |
Collapse
|
59
|
Du M, Zhang S, Liu X, Xu C, Zhang X. Nondiploid cancer cells: Stress, tolerance and therapeutic inspirations. Biochim Biophys Acta Rev Cancer 2022; 1877:188794. [PMID: 36075287 DOI: 10.1016/j.bbcan.2022.188794] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022]
Abstract
Aberrant ploidy status is a prominent characteristic in malignant neoplasms. Approximately 90% of solid tumors and 75% of haematopoietic malignancies contain aneuploidy cells, and 30%-60% of tumors undergo whole-genome doubling, indicating that nondiploidy might be a prevalent genomic aberration in cancer. Although the role of aneuploid and polyploid cells in cancer remains to be elucidated, recent studies have suggested that nondiploid cells might be a dangerous minority that severely challenges cancer management. Ploidy shifts cause multiple fitness coasts for cancer cells, mainly including genomic, proteotoxic, metabolic and immune stresses. However, nondiploid comprises a well-adopted subpopulation, with many tolerance mechanisms evident in cells along with ploidy shifts. Aneuploid and polyploid cells elegantly maintain an autonomous balance between the stress and tolerance during adaptive evolution in cancer. Breaking the balance might provide some inspiration for ploidy-selective cancer therapy and alleviation of ploidy-related chemoresistance. To understand of the complex role and therapeutic potential of nondiploid cells better, we reviewed the survival stresses and adaptive tolerances within nondiploid cancer cells and summarized therapeutic ploidy-selective alterations for potential use in developing future cancer therapy.
Collapse
Affiliation(s)
- Ming Du
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, People's Republic of China
| | - Shuo Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, People's Republic of China
| | - Xiaoxia Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, People's Republic of China
| | - Congjian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, People's Republic of China.
| | - Xiaoyan Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, People's Republic of China.
| |
Collapse
|
60
|
Polyploidy and Myc Proto-Oncogenes Promote Stress Adaptation via Epigenetic Plasticity and Gene Regulatory Network Rewiring. Int J Mol Sci 2022; 23:ijms23179691. [PMID: 36077092 PMCID: PMC9456078 DOI: 10.3390/ijms23179691] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Polyploid cells demonstrate biological plasticity and stress adaptation in evolution; development; and pathologies, including cardiovascular diseases, neurodegeneration, and cancer. The nature of ploidy-related advantages is still not completely understood. Here, we summarize the literature on molecular mechanisms underlying ploidy-related adaptive features. Polyploidy can regulate gene expression via chromatin opening, reawakening ancient evolutionary programs of embryonality. Chromatin opening switches on genes with bivalent chromatin domains that promote adaptation via rapid induction in response to signals of stress or morphogenesis. Therefore, stress-associated polyploidy can activate Myc proto-oncogenes, which further promote chromatin opening. Moreover, Myc proto-oncogenes can trigger polyploidization de novo and accelerate genome accumulation in already polyploid cells. As a result of these cooperative effects, polyploidy can increase the ability of cells to search for adaptive states of cellular programs through gene regulatory network rewiring. This ability is manifested in epigenetic plasticity associated with traits of stemness, unicellularity, flexible energy metabolism, and a complex system of DNA damage protection, combining primitive error-prone unicellular repair pathways, advanced error-free multicellular repair pathways, and DNA damage-buffering ability. These three features can be considered important components of the increased adaptability of polyploid cells. The evidence presented here contribute to the understanding of the nature of stress resistance associated with ploidy and may be useful in the development of new methods for the prevention and treatment of cardiovascular and oncological diseases.
Collapse
|
61
|
Polyploidy as an Adaptation against Loss of Heterozygosity in Cancer. Int J Mol Sci 2022; 23:ijms23158528. [PMID: 35955663 PMCID: PMC9369199 DOI: 10.3390/ijms23158528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 12/13/2022] Open
Abstract
Polyploidy is common in cancer cells and has implications for tumor progression and resistance to therapies, but it is unclear whether it is an adaptation of the tumor or the non-adaptive effect of genomic instability. I discuss the possibility that polyploidy reduces the deleterious effects of loss of heterozygosity, which arises as a consequence of mitotic recombination, and which in diploid cells leads instead to the rapid loss of complementation of recessive deleterious mutations. I use computational predictions of loss of heterozygosity to show that a population of diploid cells dividing by mitosis with recombination can be easily invaded by mutant polyploid cells or cells that divide by endomitosis, which reduces loss of complementation, or by mutant cells that occasionally fuse, which restores heterozygosity. A similar selective advantage of polyploidy has been shown for the evolution of different types of asexual reproduction in nature. This provides an adaptive explanation for cyclical ploidy, mitotic slippage and cell fusion in cancer cells.
Collapse
|
62
|
Stochastic models of Mendelian and reverse transcriptional inheritance in state-structured cancer populations. Sci Rep 2022; 12:13079. [PMID: 35906318 PMCID: PMC9338039 DOI: 10.1038/s41598-022-17456-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/26/2022] [Indexed: 11/08/2022] Open
Abstract
Recent evidence suggests that a polyaneuploid cancer cell (PACC) state may play a key role in the adaptation of cancer cells to stressful environments and in promoting therapeutic resistance. The PACC state allows cancer cells to pause cell division and to avoid DNA damage and programmed cell death. Transition to the PACC state may also lead to an increase in the cancer cell’s ability to generate heritable variation (evolvability). One way this can occur is through evolutionary triage. Under this framework, cells gradually gain resistance by scaling hills on a fitness landscape through a process of mutation and selection. Another way this can happen is through self-genetic modification whereby cells in the PACC state find a viable solution to the stressor and then undergo depolyploidization, passing it on to their heritably resistant progeny. Here, we develop a stochastic model to simulate both of these evolutionary frameworks. We examine the impact of treatment dosage and extent of self-genetic modification on eco-evolutionary dynamics of cancer cells with aneuploid and PACC states. We find that under low doses of therapy, evolutionary triage performs better whereas under high doses of therapy, self-genetic modification is favored. This study generates predictions for teasing apart these biological hypotheses, examines the implications of each in the context of cancer, and provides a modeling framework to compare Mendelian and non-traditional forms of inheritance.
Collapse
|
63
|
Liu Y, Shi Y, Wu M, Liu J, Wu H, Xu C, Chen L. Hypoxia-induced polypoid giant cancer cells in glioma promote the transformation of tumor-associated macrophages to a tumor-supportive phenotype. CNS Neurosci Ther 2022; 28:1326-1338. [PMID: 35762580 PMCID: PMC9344088 DOI: 10.1111/cns.13892] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 11/28/2022] Open
Abstract
Aims Polypoid giant cancer cells (PGCCs) represent a unique subgroup of stem‐like cells, acting as a critical factor in promoting the recurrence of various solid tumors. The effect of PGCCs on the tumor malignancy of glioma and its immune microenvironment remains unclear. Methods Bioinformatic analysis was performed to investigate the relationship between M2 tumor‐associated macrophages (TAMs) infiltration and survival of glioblastoma (GBM) patients. The spatial location of M2 TAMs in GBM was also investigated using the Ivy Glioblastoma Atlas Project (Ivy GAP) database. PGCCs were quantified in glioma of different grades. CoCl2 was used to induce PGCCs in cultures of A172 cells. PGCCs, and their progeny cells in cultures were further evaluated for morphological features, tumorsphere formation, and TAMs activation. Results The magnitude of M2 TAMs infiltration is significantly correlated with poor survival in GBM patients. M2 TAMs were enriched in the perinecrotic zone (PNZ) of GBM and positively correlated with hypoxic levels. Increased PGCCs were detected in glioma specimens of higher grades. CoCl2 induced hypoxia and the transformation of A172 cultures into PGCCs, producing the progeny cells, PGCCs‐Dau, through asymmetric division. PGCCs and PGCCs‐Dau possessed tumor stem cell‐like features, while PGCCs‐Dau enhanced the polarization of TAMs into an M2 phenotype with relevance to immunosuppression and malignancy in GBM. Conclusions PGCCs promote malignancy and immune‐suppressive microenvironment in GBM. PGCCs or their progeny cells may be a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Yuyang Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ying Shi
- School of Medicine, University of Electronic science and Technology of China, Chengdu, China.,Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Mengwan Wu
- School of Medicine, University of Electronic science and Technology of China, Chengdu, China.,Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Jialin Liu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Hong Wu
- Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Chuan Xu
- Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Ling Chen
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
64
|
Yart L, Bastida-Ruiz D, Allard M, Dietrich PY, Petignat P, Cohen M. Linking unfolded protein response to ovarian cancer cell fusion. BMC Cancer 2022; 22:622. [PMID: 35672715 PMCID: PMC9172076 DOI: 10.1186/s12885-022-09648-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Background Polyploid giant cancer cells (PGCCs) have been observed in epithelial ovarian tumors. They can resist antimitotic drugs, thus participating in tumor maintenance and recurrence. Although their origin remains unclear, PGCC formation seems to be enhanced by conditions that trigger the unfolded protein response (UPR) such as hypoxia or chemotherapeutic drugs like paclitaxel. Hypoxia has been shown to promote the formation of ovarian PGCCs by cell fusion. We thus hypothesized that the UPR could be involved in EOC cell fusion, possibly explaining the occurrence of PGCCs and the aggressiveness of EOC. Methods The UPR was induced in two ovarian cancer cell lines (SKOV3 and COV318). The UPR activation was assessed by Western blot and polyploidy indexes were calculated. Then, to confirm the implication of cell fusion in PGCC formation, two populations of SKOV3 cells were transfected with plasmids encoding for two distinct nuclear fluorescent proteins (GFP and mCherry) associated with different antibiotic resistance genes, and the two cell populations were mixed in co-culture. The co-culture was submitted to a double-antibiotic selection. The resulting cell population was characterized for its morphology, cyclicity, and proliferative and tumorigenic capacities, in addition to transcriptomic characterization. Results We demonstrated that cell fusion could be involved in the generation of ovarian PGCCs and this process was promoted by paclitaxel and the UPR activation. Double-antibiotic treatment of PGCCs led to the selection of a pure population of cells containing both GFP- and mCherry-positive nuclei. Interestingly, after 3 weeks of selection, we observed that these cells were no longer polynucleated but displayed a single nucleus positive for both fluorescent proteins, suggesting that genetic material mixing had occurred. These cells had reinitiated their normal cell cycles, acquired an increased invasive capacity, and could form ovarian tumors in ovo. Conclusions The UPR activation increased the in vitro formation of PGCCs by cell fusion, with the newly generated cells further acquiring new properties. The UPR modulation in ovarian cancer patients could represent an interesting therapeutic strategy to avoid the formation of PGCCs and therefore limit cancer relapse and drug resistance. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09648-4.
Collapse
Affiliation(s)
- Lucile Yart
- Center for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland.,Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland
| | - Daniel Bastida-Ruiz
- Center for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland.,Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland
| | - Mathilde Allard
- Center for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland.,Present address: Research Center of Cancerology and Immunology Nantes-Angers, Department of Biology, University of Nantes, FR-44035, Nantes, France
| | - Pierre-Yves Dietrich
- Center for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland
| | - Patrick Petignat
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland
| | - Marie Cohen
- Center for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland. .,Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, CH-1206, Geneva, Switzerland.
| |
Collapse
|
65
|
Nehme Z, Pasquereau S, Haidar Ahmad S, El Baba R, Herbein G. Polyploid giant cancer cells, EZH2 and Myc upregulation in mammary epithelial cells infected with high-risk human cytomegalovirus. EBioMedicine 2022; 80:104056. [PMID: 35596973 PMCID: PMC9121245 DOI: 10.1016/j.ebiom.2022.104056] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 02/08/2023] Open
Abstract
Background Human cytomegalovirus (HCMV) infection has been actively implicated in complex neoplastic processes. Beyond oncomodulation, the molecular mechanisms that might underlie HCMV-induced oncogenesis are being extensively studied. Polycomb repressive complex 2 (PRC2) proteins, in particular enhancer of zeste homolog 2 (EZH2) are associated with cancer progression. Nevertheless, little is known about EZH2 activation in the context of HCMV infection and breast oncogenesis. Methods Herein, we identified EZH2 as a downstream target for HCMV-induced Myc upregulation upon acute and chronic infection with high-risk strains using a human mammary epithelial model. Findings We detected polyploidy and CMV-transformed HMECs (CTH) cells harboring HCMV and dynamically undergoing the giant cells cycle. Acquisition of embryonic stemness markers positively correlated with EZH2 and Myc expression. EZH2 inhibitors curtail sustained CTH cells’ malignant phenotype. Besides harboring polyploid giant cancer cells (PGCCs), tumorigenic breast biopsies were characterized by an enhanced EZH2 and Myc expression, with a strong positive correlation between EZH2 and Myc expression, and between PGCC count and EZH2/Myc expression in the presence of HCMV. Further, we isolated two HCMV strains from EZH2HighMycHigh basal-like tumors which replicate in MRC5 cells and transform HMECs toward CTH cells after acute infection. Interpretation Our data establish a potential link between HCMV-induced Myc activation, the subsequent EZH2 upregulation, and polyploidy induction. These data support the proposed tumorigenesis properties of EZH2/Myc, and allow the isolation of two oncogenic HCMV strains from EZH2HighMycHigh basal breast tumors while identifying EZH2 as a potential therapeutic target in the management of breast cancer, particularly upon HCMV infection. Funding This work was supported by grants from the University of Franche-Comté (UFC) (CR3300), the Région Franche-Comté (2021-Y-08292 and 2021-Y-08290) and the Ligue contre le Cancer (CR3304) to Georges Herbein. Zeina Nehme is a recipient of a doctoral scholarship from the municipality of Habbouch. Sandy Haidar Ahmad is recipient of a doctoral scholarship from Lebanese municipality. Ranim El Baba is a recipient of a doctoral scholarship from Hariri foundation for sustainable human development.
Collapse
Affiliation(s)
- Zeina Nehme
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Sébastien Pasquereau
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Sandy Haidar Ahmad
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Ranim El Baba
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France
| | - Georges Herbein
- Department Pathogens and Inflammation-EPILAB, EA4266, Université de Franche-Comté, Université Bourgogne Franche-Comté (UBFC), 16 route de Gray, Besançon F-25030, France; Department of Virology, CHU Besançon, Besançon, France.
| |
Collapse
|
66
|
Liu C, Moten A, Ma Z, Lin HK. The foundational framework of tumors: Gametogenesis, p53, and cancer. Semin Cancer Biol 2022; 81:193-205. [PMID: 33940178 PMCID: PMC9382687 DOI: 10.1016/j.semcancer.2021.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022]
Abstract
The completion-of-tumor hypothesis involved in the dynamic interplay between the initiating oncogenic event and progression is essential to better recognize the foundational framework of tumors. Here we review and extend the gametogenesis-related hypothesis of tumors, because high embryonic/germ cell traits are common in tumors. The century-old gametogenesis-related hypothesis of tumors postulated that tumors arise from displaced/activated trophoblasts, displaced (lost) germ cells, and the reprogramming/reactivation of gametogenic program in somatic cells. Early primordial germ cells (PGCs), embryonic stem (ES) cells, embryonic germ cells (EGCs), and pre-implantation embryos at the stage from two-cell stage to blastocysts originating from fertilization or parthenogenesis have the potential to develop teratomas/teratocarcinomas. In addition, the teratomas/teratocarcinomas/germ cells occur in gonads and extra-gonads. Undoubtedly, the findings provide strong support for the hypothesis. However, it was thought that these tumor types were an exception rather than verification. In fact, there are extensive similarities between somatic tumor types and embryonic/germ cell development, such as antigens, migration, invasion, and immune escape. It was documented that embryonic/germ cell genes play crucial roles in tumor behaviors, e.g. tumor initiation and metastasis. Of note, embryonic/germ cell-like tumor cells at different developmental stages including PGC and oocyte to the early embryo-like stage were identified in diverse tumor types by our group. These embryonic/germ cell-like cancer cells resemble the natural embryonic/germ cells in morphology, gene expression, the capability of teratoma formation, and the ability to undergo the process of oocyte maturation and parthenogenesis. These embryonic/germ cell-like cancer cells are derived from somatic cells and contribute to tumor formation, metastasis, and drug resistance, establishing asexual meiotic embryonic life cycle. p53 inhibits the reactivation of embryonic/germ cell state in somatic cells and oocyte-like cell maturation. Based on earlier and our recent studies, we propose a novel model to complete the gametogenesis-related hypothesis of tumors, which can be applied to certain somatic tumors. That is, tumors tend to establish a somatic asexual meiotic embryonic cycle through the activation of somatic female gametogenesis and parthenogenesis in somatic tumor cells during the tumor progression, thus passing on corresponding embryonic/germ cell traits leading to the malignant behaviors and enhancing the cells' independence. This concept may be instrumental to better understand the nature and evolution of tumors. We rationalize that targeting the key events of somatic pregnancy is likely a better therapeutic strategy for cancer treatment than directly targeting cell mitotic proliferation, especially for those tumors with p53 inactivation.
Collapse
Affiliation(s)
- Chunfang Liu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| | - Asad Moten
- Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Zhan Ma
- Department of Laboratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
67
|
Voelkel-Johnson C. Sphingolipids in embryonic development, cell cycle regulation, and stemness - Implications for polyploidy in tumors. Semin Cancer Biol 2022; 81:206-219. [PMID: 33429049 PMCID: PMC8263803 DOI: 10.1016/j.semcancer.2020.12.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/26/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022]
Abstract
The aberrant biology of polyploid giant cancer cells (PGCC) includes dysregulation of the cell cycle, induction of stress responses, and dedifferentiation, all of which are likely accompanied by adaptations in biophysical properties and metabolic activity. Sphingolipids are the second largest class of membrane lipids and play important roles in many aspects of cell biology that are potentially relevant to polyploidy. We have recently shown that the function of the sphingolipid enzyme acid ceramidase (ASAH1) is critical for the ability of PGCC to generate progeny by depolyploidization but mechanisms by which sphingolipids contribute to polyploidy and generation of offspring with stem-like properties remain elusive. This review discusses the role of sphingolipids during embryonic development, cell cycle regulation, and stem cells in an effort to highlight parallels to polyploidy.
Collapse
Affiliation(s)
- Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
68
|
Targeting tumor cell senescence and polyploidy as potential therapeutic strategies. Semin Cancer Biol 2022; 81:37-47. [PMID: 33358748 PMCID: PMC8214633 DOI: 10.1016/j.semcancer.2020.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 01/14/2023]
Abstract
Senescence is a unique state of growth arrest that develops in response to a plethora of cellular stresses, including replicative exhaustion, oxidative injury, and genotoxic insults. Senescence has been implicated in the pathogenesis of multiple aging-related pathologies, including cancer. In cancer, senescence plays a dual role, initially acting as a barrier against tumor progression by enforcing a durable growth arrest in premalignant cells, but potentially promoting malignant transformation in neighboring cells through the secretion of pro-tumorigenic drivers. Moreover, senescence is induced in tumor cells upon exposure to a wide variety of conventional and targeted anticancer drugs (termed Therapy-Induced Senescence-TIS), representing a critical contributing factor to therapeutic outcomes. As with replicative or oxidative senescence, TIS manifests as a complex phenotype of macromolecular damage, energetic dysregulation, and altered gene expression. Senescent cells are also frequently polyploid. In vitro studies have suggested that polyploidy may confer upon senescent tumor cells the ability to escape from growth arrest, thereby providing an additional avenue whereby tumor cells escape the lethality of anticancer treatment. Polyploidy in tumor cells is also associated with persistent energy production, chromatin remodeling, self-renewal, stemness and drug resistance - features that are also associated with escape from senescence and conversion to a more malignant phenotype. However, senescent cells are highly heterogenous and can present with variable phenotypes, where polyploidy is one component of a complex reversion process. Lastly, emerging efforts to pharmacologically target polyploid tumor cells might pave the way towards the identification of novel targets for the elimination of senescent tumor cells by the incorporation of senolytic agents into cancer therapeutic strategies.
Collapse
|
69
|
Saini G, Joshi S, Garlapati C, Li H, Kong J, Krishnamurthy J, Reid MD, Aneja R. Polyploid giant cancer cell characterization: New frontiers in predicting response to chemotherapy in breast cancer. Semin Cancer Biol 2022; 81:220-231. [PMID: 33766651 PMCID: PMC8672208 DOI: 10.1016/j.semcancer.2021.03.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023]
Abstract
Although polyploid cells were first described nearly two centuries ago, their ability to proliferate has only recently been demonstrated. It also becomes increasingly evident that a subset of tumor cells, polyploid giant cancer cells (PGCCs), play a critical role in the pathophysiology of breast cancer (BC), among other cancer types. In BC, PGCCs can arise in response to therapy-induced stress. Their progeny possess cancer stem cell (CSC) properties and can repopulate the tumor. By modulating the tumor microenvironment (TME), PGCCs promote BC progression, chemoresistance, metastasis, and relapse and ultimately impact the survival of BC patients. Given their pro- tumorigenic roles, PGCCs have been proposed to possess the ability to predict treatment response and patient prognosis in BC. Traditionally, DNA cytometry has been used to detect PGCCs.. The field will further derive benefit from the development of approaches to accurately detect PGCCs and their progeny using robust PGCC biomarkers. In this review, we present the current state of knowledge about the clinical relevance of PGCCs in BC. We also propose to use an artificial intelligence-assisted image analysis pipeline to identify PGCC and map their interactions with other TME components, thereby facilitating the clinical implementation of PGCCs as biomarkers to predict treatment response and survival outcomes in BC patients. Finally, we summarize efforts to therapeutically target PGCCs to prevent chemoresistance and improve clinical outcomes in patients with BC.
Collapse
Affiliation(s)
- Geetanjali Saini
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Shriya Joshi
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | | | - Hongxiao Li
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA; Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA; Department of Computer Science, Georgia State University, Atlanta, GA, USA; Department of Computer Science, Emory University, Atlanta, GA, USA
| | | | - Michelle D Reid
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
70
|
White-Gilbertson S, Lu P, Esobi I, Echesabal-Chen J, Mulholland PJ, Gooz M, Ogretmen B, Stamatikos A, Voelkel-Johnson C. Polyploid giant cancer cells are dependent on cholesterol for progeny formation through amitotic division. Sci Rep 2022; 12:8971. [PMID: 35624221 PMCID: PMC9142539 DOI: 10.1038/s41598-022-12705-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/06/2022] [Indexed: 12/16/2022] Open
Abstract
Polyploid Giant Cancer Cells (PGCC) are increasingly being recognized as drivers of cancer recurrence. Therapy stress promotes the formation of these cells, which upon stress cessation often successfully generate more aggressive progeny that repopulate the tumor. Therefore, identification of potential PGCC vulnerabilities is key to preventing therapy failure. We have previously demonstrated that PGCC progeny formation depends on the lysosomal enzyme acid ceramidase (ASAH1). In this study, we compared transcriptomes of parental cancer cells and PGCC in the absence or presence of the ASAH1 inhibitor LCL521. Results show that PGCC express less INSIG1, which downregulates cholesterol metabolism and that inhibition of ASAH1 increased HMGCR which is the rate limiting enzyme in cholesterol synthesis. Confocal microscopy revealed that ceramide and cholesterol do not colocalize. Treatment with LCL521 or simvastatin to inhibit ASAH1 or HMGCR, respectively, resulted in accumulation of ceramide at the cell surface of PGCC and prevented PGCC progeny formation. Our results suggest that similarly to inhibition of ASAH1, disruption of cholesterol signaling is a potential strategy to interfere with PGCC progeny formation.
Collapse
Affiliation(s)
- Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Basic Science Building, MSC250504, 173 Ashley Ave., Charleston, SC, USA
| | - Ping Lu
- Department of Microbiology and Immunology, Medical University of South Carolina, Basic Science Building, MSC250504, 173 Ashley Ave., Charleston, SC, USA
| | - Ikechukwu Esobi
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, USA
| | - Jing Echesabal-Chen
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston Alcohol Research Center, Charleston, USA
| | - Monika Gooz
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, USA
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, USA
| | - Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Basic Science Building, MSC250504, 173 Ashley Ave., Charleston, SC, USA.
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, USA.
| |
Collapse
|
71
|
Amaya C, Smith ER, Xu XX. Low Intensity Ultrasound as an Antidote to Taxane/Paclitaxel-induced Cytotoxicity. J Cancer 2022; 13:2362-2373. [PMID: 35517405 PMCID: PMC9066212 DOI: 10.7150/jca.71263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
The taxane family of compounds, including Taxol/paclitaxel and Taxotere/docetaxel, are surprisingly successful drugs used in combination or alone for the treatment of most major solid tumors, especially metastatic cancer. The drugs are commonly used in regimen with other agents (often platinum drugs) as frontline treatment, or used as a single agent in a dose dense regimen for recurrent cancer. The major side effects of taxanes are peripheral neuropathy, alopecia, and neutropenia, which are grave burden for patients and limit the full potential of the taxane drugs. Especially in the current treatment protocol for peripheral neuropathy, taxane dosage is reduced once the symptoms present, resulting in the loss of full or optimal cancer killing activity. Substantial efforts have been made to address the problem of cytotoxic side effects of taxanes, though strategies remain very limited. Following administration of the taxane compound by infusion, taxane binds to cellular microtubules and is sequestered within the cells for several days. Taxane stabilizes and interferes with microtubule function, leading to ultimate death of cancer cells, but also damages hair follicles, peripheral neurons, and hemopoietic stem cells. Currently, cryo-treatment is practiced to limit exposure and side effects of the drug during infusion, though the effectiveness is uncertain or limited. A recent laboratory finding may provide a new strategy to counter taxane cytotoxicity, that a brief exposure to low density ultrasound waves was sufficient to eliminate paclitaxel cytotoxicity cells in culture by transiently breaking microtubule filaments, which were then relocated to lysosomes for disposal. Thus, ultrasonic force to break rigid microtubules is an effective solution to counter taxane cytotoxicity. The discovery and concept of low intensity ultrasound as an antidote may have the potential to provide a practical strategy to counter paclitaxel-induced peripheral neuropathy and alopecia that resulted from chemotherapy. Taxanes are a class of important drugs used in chemotherapy to treat several major cancers. This article reviews a new laboratory discovery that ultrasound can be used as an antidote for the peripheral cytotoxicity of taxane drugs and discusses the potential development and application of low intensity ultrasound to prevent side effects in chemotherapeutic treatment of cancer patients.
Collapse
Affiliation(s)
- Celina Amaya
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Elizabeth R Smith
- Department of Obstetrics, Gynecology and Reproductive Science, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Xiang-Xi Xu
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, 1120 NW 14th Street, Miami, FL, USA
| |
Collapse
|
72
|
Pustovalova M, Blokhina T, Alhaddad L, Chigasova A, Chuprov-Netochin R, Veviorskiy A, Filkov G, Osipov AN, Leonov S. CD44+ and CD133+ Non-Small Cell Lung Cancer Cells Exhibit DNA Damage Response Pathways and Dormant Polyploid Giant Cancer Cell Enrichment Relating to Their p53 Status. Int J Mol Sci 2022; 23:ijms23094922. [PMID: 35563313 PMCID: PMC9101266 DOI: 10.3390/ijms23094922] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 01/10/2023] Open
Abstract
Cancer stem cells (CSCs) play a critical role in the initiation, progression and therapy relapse of many cancers including non-small cell lung cancer (NSCLC). Here, we aimed to address the question of whether the FACS-sorted CSC-like (CD44 + &CD133 +) vs. non-CSC (CD44-/CD133- isogenic subpopulations of p53wt A549 and p53null H1299 cells differ in terms of DNA-damage signaling and the appearance of "dormant" features, including polyploidy, which are early markers (predictors) of their sensitivity to genotoxic stress. X-ray irradiation (IR) at 5 Gy provoked significantly higher levels of the ATR-Chk1/Chk2-pathway activity in CD44-/CD133- and CD133+ subpopulations of H1299 cells compared to the respective subpopulations of A549 cells, which only excited ATR-Chk2 activation as demonstrated by the Multiplex DNA-Damage/Genotoxicity profiling. The CD44+ subpopulations did not demonstrate IR-induced activation of ATR, while significantly augmenting only Chk2 and Chk1/2 in the A549- and H1299-derived cells, respectively. Compared to the A549 cells, all the subpopulations of H1299 cells established an increased IR-induced expression of the γH2AX DNA-repair protein. The CD44-/CD133- and CD133+ subpopulations of the A549 cells revealed IR-induced activation of ATR-p53-p21 cell dormancy signaling-mediated pathway, while none of the CD44+ subpopulations of either cell line possessed any signs of such activity. Our data indicated, for the first time, the transcription factor MITF-FAM3C axis operative in p53-deficient H1299 cells, specifically their CD44+ and CD133+ populations, in response to IR, which warrants further investigation. The p21-mediated quiescence is likely the predominant surviving pathway in CD44-/CD133- and CD133+ populations of A549 cells as indicated by single-cell high-content imaging and analysis of Ki67- and EdU-coupled fluorescence after IR stress. SA-beta-galhistology revealed that cellular-stress-induced premature senescence (SIPS) likely has a significant influence on the temporary dormant state of H1299 cells. For the first time, we demonstrated polyploid giant and/or multinucleated cancer-cell (PGCC/MGCC) fractions mainly featuring the progressively augmenting Ki67low phenotype in CD44+ and CD133+ A549 cells at 24-48 h after IR. In contrast, the Ki67high phenotype enrichment in the same fractions of all the sorted H1299 cells suggested an increase in their cycling/heterochromatin reorganization activity after IR stress. Our results proposed that entering the "quiescence" state rather than p21-mediated SIPS may play a significant role in the survival of p53wt CSC-like NSCLC cells after IR. The results obtained are important for the selection of therapeutic schemes for the treatment of patients with NSCLC, depending on the functioning of the p53 system in tumor cells.
Collapse
Affiliation(s)
- Margarita Pustovalova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
- Correspondence: (M.P.); (S.L.)
| | - Taisia Blokhina
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Lina Alhaddad
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
| | - Anna Chigasova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Roman Chuprov-Netochin
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
| | - Alexander Veviorskiy
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Gleb Filkov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- Laboratory of Medical Informatics, Novgorod Technical School, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia
| | - Andreyan N. Osipov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
- Correspondence: (M.P.); (S.L.)
| |
Collapse
|
73
|
Was H, Borkowska A, Bagues A, Tu L, Liu JYH, Lu Z, Rudd JA, Nurgali K, Abalo R. Mechanisms of Chemotherapy-Induced Neurotoxicity. Front Pharmacol 2022; 13:750507. [PMID: 35418856 PMCID: PMC8996259 DOI: 10.3389/fphar.2022.750507] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Since the first clinical trials conducted after World War II, chemotherapeutic drugs have been extensively used in the clinic as the main cancer treatment either alone or as an adjuvant therapy before and after surgery. Although the use of chemotherapeutic drugs improved the survival of cancer patients, these drugs are notorious for causing many severe side effects that significantly reduce the efficacy of anti-cancer treatment and patients’ quality of life. Many widely used chemotherapy drugs including platinum-based agents, taxanes, vinca alkaloids, proteasome inhibitors, and thalidomide analogs may cause direct and indirect neurotoxicity. In this review we discuss the main effects of chemotherapy on the peripheral and central nervous systems, including neuropathic pain, chemobrain, enteric neuropathy, as well as nausea and emesis. Understanding mechanisms involved in chemotherapy-induced neurotoxicity is crucial for the development of drugs that can protect the nervous system, reduce symptoms experienced by millions of patients, and improve the outcome of the treatment and patients’ quality of life.
Collapse
Affiliation(s)
- Halina Was
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Agata Borkowska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Ana Bagues
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain.,High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC), URJC, Alcorcón, Spain.,Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Longlong Tu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Julia Y H Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Zengbing Lu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - John A Rudd
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.,The Laboratory Animal Services Centre, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia.,Department of Medicine Western Health, University of Melbourne, Melbourne, VIC, Australia.,Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Raquel Abalo
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón, Spain.,Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), URJC, Alcorcón, Spain.,Grupo de Trabajo de Ciencias Básicas en Dolor y Analgesia de la Sociedad Española del Dolor, Madrid, Spain
| |
Collapse
|
74
|
Bowers RR, Andrade MF, Jones CM, White-Gilbertson S, Voelkel-Johnson C, Delaney JR. Autophagy modulating therapeutics inhibit ovarian cancer colony generation by polyploid giant cancer cells (PGCCs). BMC Cancer 2022; 22:410. [PMID: 35421971 PMCID: PMC9012005 DOI: 10.1186/s12885-022-09503-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Genomic instability and chemoresistance can arise in cancer due to a unique form of plasticity: that of polyploid giant cancer cells (PGCCs). These cells form under the stress of chemotherapy and have higher than diploid chromosome content. PGCCs are able to then repopulate tumors through an asymmetric daughter cell budding process. PGCCs have been observed in ovarian cancer histology, including the deadly and common form high-grade serous ovarian carcinoma (HGSC). We previously discovered that drugs which disrupt the cellular recycling process of autophagy are uniquely efficacious in pre-clinical HGSC models. While autophagy induction has been associated with PGCCs, it has never been previously investigated if autophagy modulation interacts with the PGCC life cycle and this form of tumor cell plasticity. METHODS CAOV3 and OVCAR3 ovarian cancer cell lines were treated with carboplatin or docetaxel to induce PGCC formation. Microscopy was used to characterize and quantify PGCCs formed by chemotherapy. Two clinically available drugs that inhibit autophagy, hydroxychloroquine and nelfinavir, and a clinically available activator of autophagy, rapamycin, were employed to test the effect of these autophagy modulators on PGCC induction and subsequent colony formation from PGCCs. Crystal violet-stained colony formation assays were used to quantify the tumor-repopulating stage of the PGCC life cycle. RESULTS Autophagy inhibitors did not prevent PGCC formation in OVCAR3 or CAOV3 cells. Rapamycin did not induce PGCC formation on its own nor did it exacerbate PGCC formation by chemotherapy. However, hydroxychloroquine prevented efficient colony formation in CAOV3 PGCCs induced by carboplatin (27% inhibition) or docetaxel (41% inhibition), as well as in OVCAR3 cells (95% and 77%, respectively). Nelfinavir similarly prevented colony formation in CAOV3 PGCCs induced by carboplatin (64% inhibition) or docetaxel (94% inhibition) as well as in OVCAR3 cells (89% and 80%, respectively). Rapamycin surprisingly also prevented PGCC colony outgrowth (52-84% inhibition). CONCLUSIONS While the autophagy previously observed to correlate with PGCC formation is unlikely necessary for PGCCs to form, autophagy modulating drugs severely impair the ability of HGSC PGCCs to form colonies. Clinical trials which utilize hydroxychloroquine, nelfinavir, and/or rapamycin after chemotherapy may be of future interest.
Collapse
Affiliation(s)
- Robert R Bowers
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Maya F Andrade
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Christian M Jones
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Joe R Delaney
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
75
|
Mesenchymal Stem Cells and their Derived Exosomes Promote Malignant Phenotype of Polyploid Non-Small-Cell Lung Cancer Cells through AMPK Signaling Pathway. Anal Cell Pathol 2022; 2022:8708202. [PMID: 35419253 PMCID: PMC9001126 DOI: 10.1155/2022/8708202] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy is an important method for the treatment of non-small-cell lung cancer (NSCLC), but it can lead to side effects and polyploid cancer cells. The polyploid cancer cells can live and generate daughter cancer cells via budding. Mesenchymal stem cells (MSCs) are pluripotent stem cells with repair and regeneration functions and can resist tissue damage caused by tumor therapy. This study is aimed at investigating the effects of MSCs and their derived exosomes on the biological characteristics of polyploid NSCLC cells and the potential mechanisms. We found that MSC conditioned medium (CM), MSCs, and MSC-exosomes had no effect on cell proliferation of the polyploid A549 and H1299 cells. Compared with the control group, MSCs and MSC-exosomes significantly promoted epithelial mesenchymal transformation, cell migration, antiapoptosis, and autophagy in the polyploid A549 and H1299 by activating AMPK signaling pathway, but no significant changes were observed in MSC-CM treatment. These results revealed that MSCs and MSC-exosomes promoted malignant phenotype of polyploid NSCLC cells through the AMPK signaling pathway.
Collapse
|
76
|
Vainshelbaum NM, Salmina K, Gerashchenko BI, Lazovska M, Zayakin P, Cragg MS, Pjanova D, Erenpreisa J. Role of the Circadian Clock "Death-Loop" in the DNA Damage Response Underpinning Cancer Treatment Resistance. Cells 2022; 11:880. [PMID: 35269502 PMCID: PMC8909334 DOI: 10.3390/cells11050880] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/14/2022] [Accepted: 03/01/2022] [Indexed: 12/11/2022] Open
Abstract
Here, we review the role of the circadian clock (CC) in the resistance of cancer cells to genotoxic treatments in relation to whole-genome duplication (WGD) and telomere-length regulation. The CC drives the normal cell cycle, tissue differentiation, and reciprocally regulates telomere elongation. However, it is deregulated in embryonic stem cells (ESCs), the early embryo, and cancer. Here, we review the DNA damage response of cancer cells and a similar impact on the cell cycle to that found in ESCs—overcoming G1/S, adapting DNA damage checkpoints, tolerating DNA damage, coupling telomere erosion to accelerated cell senescence, and favouring transition by mitotic slippage into the ploidy cycle (reversible polyploidy). Polyploidy decelerates the CC. We report an intriguing positive correlation between cancer WGD and the deregulation of the CC assessed by bioinformatics on 11 primary cancer datasets (rho = 0.83; p < 0.01). As previously shown, the cancer cells undergoing mitotic slippage cast off telomere fragments with TERT, restore the telomeres by ALT-recombination, and return their depolyploidised offspring to telomerase-dependent regulation. By reversing this polyploidy and the CC “death loop”, the mitotic cycle and Hayflick limit count are thus again renewed. Our review and proposed mechanism support a life-cycle concept of cancer and highlight the perspective of cancer treatment by differentiation.
Collapse
Affiliation(s)
- Ninel Miriam Vainshelbaum
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
- Faculty of Biology, University of Latvia, LV-1050 Riga, Latvia
| | - Kristine Salmina
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Bogdan I. Gerashchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, 03022 Kyiv, Ukraine;
| | - Marija Lazovska
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Pawel Zayakin
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Mark Steven Cragg
- Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
| | - Dace Pjanova
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Jekaterina Erenpreisa
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| |
Collapse
|
77
|
You B, Xia T, Gu M, Zhang Z, Zhang Q, Shen J, Fan Y, Yao H, Pan S, Lu Y, Cheng T, Yang Z, He X, Zhang H, Shi M, Liu D, You Y. AMPK-mTOR-Mediated Activation of Autophagy Promotes Formation of Dormant Polyploid Giant Cancer Cells. Cancer Res 2022; 82:846-858. [PMID: 34965934 PMCID: PMC9359740 DOI: 10.1158/0008-5472.can-21-2342] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 11/20/2021] [Accepted: 12/27/2021] [Indexed: 01/07/2023]
Abstract
Dormant cancer cells that survive anticancer therapy can lead to cancer recurrence and disseminated metastases that prove fatal in most cases. Recently, specific dormant polyploid giant cancer cells (PGCC) have drawn our attention because of their association with the clinical risk of nasopharyngeal carcinoma (NPC) recurrence, as demonstrated by previous clinical data. In this study, we report the biological properties of PGCC, including mitochondrial alterations, and reveal that autophagy is a critical mechanism of PGCC induction. Moreover, pharmacologic or genetic inhibition of autophagy greatly impaired PGCC formation, significantly suppressing metastasis and improving survival in a mouse model. Mechanistically, chemotherapeutic drugs partly damaged mitochondria, which then produced low ATP levels and activated autophagy via the AMPK-mTOR pathway to promote PGCC formation. Analysis of the transcriptional and epigenetic landscape of PGCC revealed overexpression of RIPK1, and the scaffolding function of RIPK1 was required for AMPK-mTOR pathway-induced PGCC survival. High numbers of PGCCs correlated with shorter recurrence time and worse survival outcomes in patients with NPC. Collectively, these findings suggest a therapeutic approach of targeting dormant PGCCs in cancer. SIGNIFICANCE Pretreatment with an autophagy inhibitor before chemotherapy could prevent formation of therapy-induced dormant polyploid giant cancer cells, thereby reducing recurrence and metastasis of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Bo You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Tian Xia
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Miao Gu
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Zhenxin Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Qicheng Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Jianhong Shen
- Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yue Fan
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Hui Yao
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Si Pan
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Yingna Lu
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Tianyi Cheng
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Zhiyuan Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Xin He
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hao Zhang
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Muqi Shi
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and MOE, Nantong University, Nantong, Jiangsu Province, China.,Corresponding Authors: Yiwen You, Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, 40 Qing Nian Dong Lu, Chongchuan District, Nantong, Jiangsu Province, China, 226007, China. Phone: 135-8522-9333; E-mail: ; and Dong Liu, Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and MOE, Nantong University, 9 Siyuan Road, Chongchuan District, Nantong, Jiangsu Province, China, 226019, China. Phone: 8618-6051-33927; E-mail:
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,College of Medicine, Nantong, Jiangsu Province, China.,Corresponding Authors: Yiwen You, Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, 40 Qing Nian Dong Lu, Chongchuan District, Nantong, Jiangsu Province, China, 226007, China. Phone: 135-8522-9333; E-mail: ; and Dong Liu, Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and MOE, Nantong University, 9 Siyuan Road, Chongchuan District, Nantong, Jiangsu Province, China, 226019, China. Phone: 8618-6051-33927; E-mail:
| |
Collapse
|
78
|
Olszewska A, Borkowska A, Granica M, Karolczak J, Zglinicki B, Kieda C, Was H. Escape From Cisplatin-Induced Senescence of Hypoxic Lung Cancer Cells Can Be Overcome by Hydroxychloroquine. Front Oncol 2022; 11:738385. [PMID: 35127467 PMCID: PMC8813758 DOI: 10.3389/fonc.2021.738385] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/23/2021] [Indexed: 12/23/2022] Open
Abstract
Chemotherapy is the commonly used treatment for advanced lung cancer. However, it produces side effects such as the development of chemoresistance. A possible responsible mechanism may be therapy-induced senescence (TIS). TIS cells display increased senescence-associated β-galactosidase (SA-β-gal) activity and irreversible growth arrest. However, recent data suggest that TIS cells can reactivate their proliferative potential and lead to cancer recurrence. Our previous study indicated that reactivation of proliferation by TIS cells might be related with autophagy modulation. However, exact relationship between both processes required further studies. Therefore, the aim of our study was to investigate the role of autophagy in the senescence-related chemoresistance of lung cancer cells. For this purpose, human and murine lung cancer cells were treated with two commonly used chemotherapeutics: cisplatin (CIS), which forms DNA adducts or docetaxel (DOC), a microtubule poison. Hypoxia, often overlooked in experimental settings, has been implicated as a mechanism responsible for a significant change in the response to treatment. Thus, cells were cultured under normoxic (~19% O2) or hypoxic (1% O2) conditions. Herein, we show that hypoxia increases resistance to CIS. Lung cancer cells cultured under hypoxic conditions escaped from CIS-induced senescence, displayed reduced SA-β-gal activity and a decreased percentage of cells in the G2/M phase of the cell cycle. In turn, hypoxia increased the proliferation of lung cancer cells and the proportion of cells proceeding to the G0/G1 phase. Further molecular analyses demonstrated that hypoxia inhibited the prosenescent p53/p21 signaling pathway and induced epithelial to mesenchymal transition in CIS-treated cancer cells. In cells treated with DOC, such effects were not observed. Of importance, pharmacological autophagy inhibitor, hydroxychloroquine (HCQ) was capable of overcoming short-term CIS-induced resistance of lung cancer cells in hypoxic conditions. Altogether, our data demonstrated that hypoxia favors cancer cell escape from CIS-induced senescence, what could be overcome by inhibition of autophagy with HCQ. Therefore, we propose that HCQ might be used to interfere with the ability of senescent cancer cells to repopulate following exposure to DNA-damaging agents. This effect, however, needs to be tested in a long-term perspective for preclinical and clinical applications.
Collapse
Affiliation(s)
- Aleksandra Olszewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Agata Borkowska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Monika Granica
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Doctoral School of Translational Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Justyna Karolczak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Bartosz Zglinicki
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Halina Was
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- *Correspondence: Halina Was,
| |
Collapse
|
79
|
Heng J, Heng HH. Genome Chaos, Information Creation, and Cancer Emergence: Searching for New Frameworks on the 50th Anniversary of the "War on Cancer". Genes (Basel) 2021; 13:genes13010101. [PMID: 35052441 PMCID: PMC8774498 DOI: 10.3390/genes13010101] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 12/26/2022] Open
Abstract
The year 2021 marks the 50th anniversary of the National Cancer Act, signed by President Nixon, which declared a national “war on cancer.” Powered by enormous financial support, this past half-century has witnessed remarkable progress in understanding the individual molecular mechanisms of cancer, primarily through the characterization of cancer genes and the phenotypes associated with their pathways. Despite millions of publications and the overwhelming volume data generated from the Cancer Genome Project, clinical benefits are still lacking. In fact, the massive, diverse data also unexpectedly challenge the current somatic gene mutation theory of cancer, as well as the initial rationales behind sequencing so many cancer samples. Therefore, what should we do next? Should we continue to sequence more samples and push for further molecular characterizations, or should we take a moment to pause and think about the biological meaning of the data we have, integrating new ideas in cancer biology? On this special anniversary, we implore that it is time for the latter. We review the Genome Architecture Theory, an alternative conceptual framework that departs from gene-based theories. Specifically, we discuss the relationship between genes, genomes, and information-based platforms for future cancer research. This discussion will reinforce some newly proposed concepts that are essential for advancing cancer research, including two-phased cancer evolution (which reconciles evolutionary contributions from karyotypes and genes), stress-induced genome chaos (which creates new system information essential for macroevolution), the evolutionary mechanism of cancer (which unifies diverse molecular mechanisms to create new karyotype coding during evolution), and cellular adaptation and cancer emergence (which explains why cancer exists in the first place). We hope that these ideas will usher in new genomic and evolutionary conceptual frameworks and strategies for the next 50 years of cancer research.
Collapse
Affiliation(s)
- Julie Heng
- Harvard College, 16 Divinity Ave, Cambridge, MA 02138, USA;
| | - Henry H. Heng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Correspondence:
| |
Collapse
|
80
|
Peng J, Shi S, Yu J, Liu J, Wei H, Song H, Wang S, Li Z, He S, Li L, Zhang H, Yan Z, Zhao R, Liu Y, Liu Y, Li J, Zhang R, Wang W. miR-378d suppresses malignant phenotype of ESCC cells through AKT signaling. Cancer Cell Int 2021; 21:702. [PMID: 34937563 PMCID: PMC8697470 DOI: 10.1186/s12935-021-02403-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/08/2021] [Indexed: 01/12/2023] Open
Abstract
Background Post-resistance progress in paclitaxel (PTX) treatment remains a major challenge in tumor treatment. A high dose of PTX was used for cell lines to analyze the changes in molecular expression. The miR-378d was sharply reduced in surviving cells, but the role of miR-378d in Esophageal squamous cell carcinoma (ESCC) remained unclear. Methods We analyzed the relationship between miR-378d expression and the clinicopathological features of ESCC. We constructed miR-378d silent expression cell lines to study phenotypes and molecular mechanisms. Results The miR-378d expression was associated with good prognosis in patients with ESCC. miR-378d inhibition promoted chemo-resistance, monoclonal formation, EMT, migration, invasion, stemness, and metastasis of ESCC cells. miR-378d can target downregulated AKT1. Conclusions Therefore, miR-378d expression is a good prognostic factor of patients with ESCC and regulates the malignant phenotype of tumor cells through AKT. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02403-y.
Collapse
|
81
|
Anatskaya OV, Vinogradov AE. Whole-Genome Duplications in Evolution, Ontogeny, and Pathology: Complexity and Emergency Reserves. Mol Biol 2021. [DOI: 10.1134/s0026893321050022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
82
|
Arman T, Baron JA, Lynch KD, White LA, Aldan J, Clarke JD. MCLR-elicited hepatic fibrosis and carcinogenic gene expression changes persist in rats with diet-induced nonalcoholic steatohepatitis through a 4-week recovery period. Toxicology 2021; 464:153021. [PMID: 34740672 PMCID: PMC8629135 DOI: 10.1016/j.tox.2021.153021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/08/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) causes liver extracellular matrix (ECM) remodeling and is a risk factor for fibrosis and hepatocellular carcinoma (HCC). Microcystin-LR (MCLR) is a hepatotoxin produced by fresh-water cyanobacteria that causes a NASH-like phenotype, liver fibrosis, and is also a risk factor for HCC. The focus of the current study was to investigate and compare hepatic recovery after cessation of MCLR exposure in healthy versus NASH animals. Male Sprague-Dawley rats were fed either a control or a high fat/high cholesterol (HFHC) diet for eight weeks. Animals received either vehicle or 30 μg/kg MCLR (i.p: 2 weeks, alternate days). Animals were euthanized at one of three time points: at the completion of the MCLR exposure period and after 2 and 4 weeks of recovery. Histological staining suggested that after four weeks of recovery the MCLR-exposed HFHC group had less steatosis and more fibrosis compared to the vehicle-exposed HFHC group and MCLR-exposed control group. RNA-Seq analysis revealed dysregulation of ECM genes after MCLR exposure in both control and HFHC groups that persisted only in the HFHC groups during recovery. After 4 weeks of recovery, MCLR hepatotoxicity in pre-existing NASH persistently dysregulated genes related to cellular differentiation and HCC. These data demonstrate impaired hepatic recovery and persistent carcinogenic changes after MCLR toxicity in pre-existing NASH.
Collapse
Affiliation(s)
- Tarana Arman
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, United States
| | - J Allen Baron
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, United States
| | - Katherine D Lynch
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, United States
| | - Laura A White
- Washington Animal Disease Diagnostic Laboratory, Washington State University, Pullman, WA, 99164, United States
| | - Johnny Aldan
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, United States
| | - John D Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, United States.
| |
Collapse
|
83
|
Neural is Fundamental: Neural Stemness as the Ground State of Cell Tumorigenicity and Differentiation Potential. Stem Cell Rev Rep 2021; 18:37-55. [PMID: 34714532 DOI: 10.1007/s12015-021-10275-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 01/07/2023]
Abstract
Tumorigenic cells are similar to neural stem cells or embryonic neural cells in regulatory networks, tumorigenicity and pluripotent differentiation potential. By integrating the evidence from developmental biology, tumor biology and evolution, I will make a detailed discussion on the observations and propose that neural stemness underlies two coupled cell properties, tumorigenicity and pluripotent differentiation potential. Neural stemness property of tumorigenic cells can hopefully integrate different observations/concepts underlying tumorigenesis. Neural stem cells and tumorigenic cells share regulatory networks; both exhibit neural stemness, tumorigenicity and pluripotent differentiation potential; both depend on expression or activation of ancestral genes; both rely primarily on aerobic glycolytic metabolism; both can differentiate into various cells/tissues that are derived from three germ layers, leading to tumor formation resembling severely disorganized or more degenerated process of embryonic tissue differentiation; both are enriched in long genes with more splice variants that provide more plastic scaffolds for cell differentiation, etc. Neural regulatory networks, which include higher levels of basic machineries of cell physiological functions and developmental programs, work concertedly to define a basic state with fast cell cycle and proliferation. This is predestined by the evolutionary advantage of neural state, the ground or initial state for multicellularity with adaptation to an ancient environment. Tumorigenesis might represent a process of restoration of neural ground state, thereby restoring a state with fast proliferation and pluripotent differentiation potential in somatic cells. Tumorigenesis and pluripotent differentiation potential might be better understood from understanding neural stemness, and cancer therapy should benefit more from targeting neural stemness.
Collapse
|
84
|
Li Z, Zheng M, Zhang H, Yang X, Fan L, Fu F, Fu J, Niu R, Yan M, Zhang S. Arsenic Trioxide Promotes Tumor Progression by Inducing the Formation of PGCCs and Embryonic Hemoglobin in Colon Cancer Cells. Front Oncol 2021; 11:720814. [PMID: 34676163 PMCID: PMC8523995 DOI: 10.3389/fonc.2021.720814] [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: 06/16/2021] [Accepted: 09/17/2021] [Indexed: 12/24/2022] Open
Abstract
Arsenic trioxide (ATO) has been used to treat acute promyelocytic leukemia. However, it is not effective in treating solid tumors such as colorectal cancer. We have previously reported that polyploid giant cancer cells (PGCCs) exhibiting the characteristics of cancer stem cells can be generated by various inducers. In this study, ATO was used to induce the formation of PGCCs in LoVo and Hct116 colon cancer cell lines. The migration, invasion, and proliferation abilities of colon cancer cells with and without ATO treatment were assessed by wound-healing, transwell, and plate colony formation assays. The expression of epithelial to mesenchymal transition-related proteins and erythroid differentiation-related proteins in colon cancer cells was further evaluated by western blot and immunocytochemical assays. LoVo and Hct116 cells were transfected with a eukaryotic expression vector for green fluorescent protein (GFP), red fluorescent protein (RFP), H2B-GFP, and H2B-mCherry to study PGCCs formation via cell fusion. WB and ICC assays were performed to assess the expression of cell fusion-related proteins. MG132, small interfering RNA-glial cell missing 1 (GCM1), and chromatin immunoprecipitation-polymerase chain reaction assays were performed to study the role of GCM1/syncytin-1-mediated cell fusion. Clinically, the significance of cell fusion-related proteins and erythroid differentiation-related proteins expression in human colorectal cancer tissues was evaluated. Results of our study showed that ATO induced the formation of PGCCs, and the daughter cells derived from PGCCs gained a mesenchymal phenotype and exhibited strong migration, invasion, and proliferation abilities. PGCCs also produced embryonic hemoglobin-delta and -zeta with strong oxygen-binding ability and erythroid differentiation-related proteins after ATO treatment. In addition, cell fusion was observed during the formation of PGCCs, indicated by the presence of yellow fluorescence via the GCM1/syncytin-1 signaling pathway. Clinically, the expression of cell fusion-related and erythroid differentiation-related proteins gradually increased with the progression of human colorectal cancer tissues. In conclusion, ATO can promote tumor progression by inducing the formation of PGCCs via GCM1/syncytin-1-mediated cell fusion. PGCCs can produce daughter cells with high invasion and migration abilities and embryonic hemoglobin with strong oxygen binding ability, promoting survival of tumor cells in a hypoxic microenvironment.
Collapse
Affiliation(s)
- Zugui Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Hao Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Linlin Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fangmei Fu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China.,Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junjie Fu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China.,Graduate School, Tianjin Medical University, Tianjin, China
| | - Rui Niu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Man Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
85
|
Liu J, Erenpreisa J, Sikora E. Polyploid giant cancer cells: An emerging new field of cancer biology. Semin Cancer Biol 2021; 81:1-4. [PMID: 34695579 DOI: 10.1016/j.semcancer.2021.10.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jinsong Liu
- Department of Anatomical Pathology and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | - Ewa Sikora
- Laboratory of Moleuclar Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
86
|
Liu J, Niu N, Li X, Zhang X, Sood AK. The life cycle of polyploid giant cancer cells and dormancy in cancer: Opportunities for novel therapeutic interventions. Semin Cancer Biol 2021; 81:132-144. [PMID: 34670140 DOI: 10.1016/j.semcancer.2021.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023]
Abstract
Recent data suggest that most genotoxic agents in cancer therapy can lead to shock of genome and increase in cell size, which leads whole genome duplication or multiplication, formation of polyploid giant cancer cells, activation of an early embryonic program, and dedifferentiation of somatic cells. This process is achieved via the giant cell life cycle, a recently proposed mechanism for malignant transformation of somatic cells. Increase in both cell size and ploidy allows cells to completely or partially restructures the genome and develop into a blastocyst-like structure, similar to that observed in blastomere-stage embryogenesis. Although blastocyst-like structures with reprogrammed genome can generate resistant or metastatic daughter cells or benign cells of different lineages, they also acquired ability to undergo embryonic diapause, a reversible state of suspended embryonic development in which cells enter dormancy for survival in response to environmental stress. Therapeutic agents can activate this evolutionarily conserved developmental program, and when cells awaken from embryonic diapause, this leads to recurrence or metastasis. Understanding of the key mechanisms that regulate the different stages of the giant cell life cycle offers new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jinsong Liu
- Departments of Anatomic Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Departments of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Na Niu
- Departments of Anatomic Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaoran Li
- Departments of Anatomic Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xudong Zhang
- Departments of Anatomic Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Anil K Sood
- Departments of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
87
|
Liu H, Xia T, You Y, Zhang Q, Ni H, Liu Y, Liu Y, Xu Y, You B, Zhang Z. Characteristics and clinical significance of polyploid giant cancer cells in laryngeal carcinoma. Laryngoscope Investig Otolaryngol 2021; 6:1228-1234. [PMID: 34667869 PMCID: PMC8513447 DOI: 10.1002/lio2.667] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/16/2021] [Accepted: 09/12/2021] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVES We aimed to construct an induction system for polyploid giant cancer cells (PGCCs), as well as to investigate PGCC features and clinical significance. METHODS A laryngeal neoplasm-PGCC induction system was constructed using paclitaxel liposomes (PTX). We used western blots to compare expression of epithelial-mesenchymal transition-related proteins, stem cell interrelated proteins, and cyclin-associated proteins. We then measured PGCC count in tissue samples of patients with laryngeal neoplasms and analyzed its relationship with prognosis. Statistical significance was determined using t-tests. RESULTS PTX successfully induced PGCCs. Western blotting showed that CyclinB1, CDC25C, CDK1, E-cadherin, and EIF-4A expression decreased in PGCCs compared with normal cancer cells, whereas vimentin and CD133 expression increased. Number of PGCCs in laryngeal cancer tissues and overall survival time were inversely correlated (P < .05). CONCLUSIONS PTX successfully induces PGCC formation in laryngeal carcinoma, which may be the cause of poor prognosis in patients with laryngeal cancer.Level of Evidence: 4.
Collapse
Affiliation(s)
- Hui‐Ting Liu
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Tian Xia
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Yi‐Wen You
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Qi‐Cheng Zhang
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Hao‐sheng Ni
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Yi‐Fei Liu
- Department of PathologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Yuan‐Ru Liu
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Yu‐Qing Xu
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
| | - Bo You
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Zhen‐Xin Zhang
- Otorhinolaryngology Head and Neck Surgery DepartmentAffiliated Hospital of Nantong UniversityNantongChina
- Otolaryngology Head and Neck Surgery InstituteAffiliated Hospital of Nantong UniversityNantongChina
| |
Collapse
|
88
|
IL-6 promotes drug resistance through formation of polyploid giant cancer cells and stromal fibroblast reprogramming. Oncogenesis 2021; 10:65. [PMID: 34588424 PMCID: PMC8481288 DOI: 10.1038/s41389-021-00349-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/15/2021] [Accepted: 08/23/2021] [Indexed: 12/23/2022] Open
Abstract
To understand the role of polyploid giant cancer cells (PGCCs) in drug resistance and disease relapse, we examined the mRNA expression profile of PGCCs following treatment with paclitaxel in ovarian cancer cells. An acute activation of IL-6 dominated senescence-associated secretory phenotype lasted 2–3 weeks and declined during the termination phase of polyploidy. IL-6 activates embryonic stemness during the initiation of PGCCs and can reprogram normal fibroblasts into cancer-associated fibroblasts (CAFs) via increased collagen synthesis, activation of VEGF expression, and enrichment of CAFs and the GPR77 + /CD10 + fibroblast subpopulation. Blocking the IL-6 feedback loop with tocilizumab or apigenin prevented PGCC formation, attenuated embryonic stemness and the CAF phenotype, and inhibited tumor growth in a patient-derived xenograft high-grade serous ovarian carcinoma model. Thus, IL-6 derived by PGCCs is capable of reprogramming both cancer and stromal cells and contributes to the evolution and remodeling of cancer. Targeting IL-6 in PGCCs may represent a novel approach to combating drug resistance.
Collapse
|
89
|
Kostecka LG, Pienta KJ, Amend SR. Lipid droplet evolution gives insight into polyaneuploid cancer cell lipid droplet functions. Med Oncol 2021; 38:133. [PMID: 34581907 PMCID: PMC8478749 DOI: 10.1007/s12032-021-01584-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/12/2021] [Indexed: 12/16/2022]
Abstract
Lipid droplets (LDs) are found throughout all phyla across the tree of life. Originating as pure energy stores in the most basic organisms, LDs have evolved to fill various roles as regulators of lipid metabolism, signaling, and trafficking. LDs have been noted in cancer cells and have shown to increase tumor aggressiveness and chemotherapy resistance. A certain transitory state of cancer cell, the polyaneuploid cancer cell (PACC), appears to have higher LD levels than the cancer cell from which they are derived. PACCs are postulated to be the mediators of metastasis and resistance in many different cancers. Utilizing the evolutionarily conserved roles of LDs to protect from cellular lipotoxicity allows PACCs to survive otherwise lethal stressors. By better understanding how LDs have evolved throughout different phyla we will identify opportunities to target LDs in PACCs to increase therapeutic efficiency in cancer cells.
Collapse
Affiliation(s)
- Laurie G Kostecka
- The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, MD, 21287, USA. .,Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, MD, 21287, USA.,Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Sarah R Amend
- The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, MD, 21287, USA.,Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
90
|
Song Y, Zhao Y, Deng Z, Zhao R, Huang Q. Stress-Induced Polyploid Giant Cancer Cells: Unique Way of Formation and Non-Negligible Characteristics. Front Oncol 2021; 11:724781. [PMID: 34527590 PMCID: PMC8435787 DOI: 10.3389/fonc.2021.724781] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Polyploidy is a conserved mechanism in cell development and stress responses. Multiple stresses of treatment, including radiation and chemotherapy drugs, can induce the polyploidization of tumor cells. Through endoreplication or cell fusion, diploid tumor cells convert into giant tumor cells with single large nuclei or multiple small nucleuses. Some of the stress-induced colossal cells, which were previously thought to be senescent and have no ability to proliferate, can escape the fate of death by a special way. They can remain alive at least before producing progeny cells through asymmetric cell division, a depolyploidization way named neosis. Those large and danger cells are recognized as polyploid giant cancer cells (PGCCs). Such cells are under suspicion of being highly related to tumor recurrence and metastasis after treatment and can bring new targets for cancer therapy. However, differences in formation mechanisms between PGCCs and well-accepted polyploid cancer cells are largely unknown. In this review, the methods used in different studies to induce polyploid cells are summarized, and several mechanisms of polyploidization are demonstrated. Besides, we discuss some characteristics related to the poor prognosis caused by PGCCs in order to provide readers with a more comprehensive understanding of these huge cells.
Collapse
Affiliation(s)
- Yanwei Song
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yucui Zhao
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Deng
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruyi Zhao
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Huang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
91
|
De Blander H, Morel AP, Senaratne AP, Ouzounova M, Puisieux A. Cellular Plasticity: A Route to Senescence Exit and Tumorigenesis. Cancers (Basel) 2021; 13:4561. [PMID: 34572787 PMCID: PMC8468602 DOI: 10.3390/cancers13184561] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 01/10/2023] Open
Abstract
Senescence is a dynamic, multistep program that results in permanent cell cycle arrest and is triggered by developmental or environmental, oncogenic or therapy-induced stress signals. Senescence is considered as a tumor suppressor mechanism that prevents the risk of neoplastic transformation by restricting the proliferation of damaged cells. Cells undergoing senescence sustain important morphological changes, chromatin remodeling and metabolic reprogramming, and secrete pro-inflammatory factors termed senescence-associated secretory phenotype (SASP). SASP activation is required for the clearance of senescent cells by innate immunity. Therefore, escape from senescence and the associated immune editing would be a prerequisite for tumor initiation and progression as well as therapeutic resistance. One of the possible mechanisms for overcoming senescence could be the acquisition of cellular plasticity resulting from the accumulation of genomic alterations and genetic and epigenetic reprogramming. The modified composition of the SASP produced by these reprogrammed cancer cells would create a permissive environment, allowing their immune evasion. Additionally, the SASP produced by cancer cells could enhance the cellular plasticity of neighboring cells, thus hindering their recognition by the immune system. Here, we propose a comprehensive review of the literature, highlighting the role of cellular plasticity in the pro-tumoral activity of senescence in normal cells and in the cancer context.
Collapse
Affiliation(s)
- Hadrien De Blander
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
| | - Anne-Pierre Morel
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
- Institut Curie “EMT and Cancer Cell Plasticity”, Consortium Centre Léon Bérard, 69008 Lyon, France
| | - Aruni P. Senaratne
- UMR3664—Nuclear Dynamics, Development, Biology, Cancer, Genetics and Epigenetics, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Maria Ouzounova
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
- Institut Curie “EMT and Cancer Cell Plasticity”, Consortium Centre Léon Bérard, 69008 Lyon, France
- CNRS UMR3666, Inserm U1143, Cellular and Chemical Biology, Institut Curie, PSL Research University, 75005 Paris, France
| | - Alain Puisieux
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
- Institut Curie “EMT and Cancer Cell Plasticity”, Consortium Centre Léon Bérard, 69008 Lyon, France
- CNRS UMR3666, Inserm U1143, Cellular and Chemical Biology, Institut Curie, PSL Research University, 75005 Paris, France
| |
Collapse
|
92
|
Zhang DY, Monteiro MJ, Liu JP, Gu WY. Mechanisms of cancer stem cell senescence: Current understanding and future perspectives. Clin Exp Pharmacol Physiol 2021; 48:1185-1202. [PMID: 34046925 DOI: 10.1111/1440-1681.13528] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 05/24/2021] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are a small population of heterogeneous tumor cells with the capacity of self-renewal and aberrant differentiation for immortality and divergent lineages of cancer cells. In contrast to bulky tumor cells, CSCs remain less differentiated and resistant to therapy even when targeted with tissue-specific antigenic markers. This makes CSCs responsible for not only tumor initiation, development, but also tumor recurrence. Emerging evidence suggests that CSCs can undergo cell senescence, a non-proliferative state of cells in response to stress. While cell senescence attenuates tumor cell proliferation, it is commonly regarded as a tumor suppressive mechanism. However, mounting research indicates that CSC senescence also provides these cells with the capacity to evade cytotoxic effects from cancer therapy, exacerbating cancer relapse and metastasis. Recent studies demonstrate that senescence drives reprogramming of cancer cell toward stemness and promotes CSC generation. In this review, we highlight the origin, heterogeneity and senescence regulatory mechanisms of CSCs, the complex relationship between CSC senescence and tumor therapy, and the recent beneficial effects of senotherapy on eliminating senescent tumor cells.
Collapse
Affiliation(s)
- Da-Yong Zhang
- Department of Clinical Medicine, Zhejiang University City College, Hangzhou, China
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, Australia
| | - Michael J Monteiro
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, Australia
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, Hangzhou, China
- Department of Immunology, Monash University Faculty of Medicine, Prahran, Vic, Australia
- Hudson Institute of Medical Research, and Department of Molecular and Translational Science, Monash University Faculty of Medicine, Clayton, Vic, Australia
| | - Wen-Yi Gu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
93
|
Heng E, Moy A, Liu G, Heng HH, Zhang K. ER Stress and Micronuclei Cluster: Stress Response Contributes to Genome Chaos in Cancer. Front Cell Dev Biol 2021; 9:673188. [PMID: 34422803 PMCID: PMC8371933 DOI: 10.3389/fcell.2021.673188] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/12/2021] [Indexed: 12/31/2022] Open
Affiliation(s)
- Eric Heng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Amanda Moy
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Guo Liu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Henry H Heng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
94
|
Thura M, Ye Z, Al-Aidaroos AQ, Xiong Q, Ong JY, Gupta A, Li J, Guo K, Ang KH, Zeng Q. PRL3 induces polypoid giant cancer cells eliminated by PRL3-zumab to reduce tumor relapse. Commun Biol 2021; 4:923. [PMID: 34326464 PMCID: PMC8322210 DOI: 10.1038/s42003-021-02449-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 07/13/2021] [Indexed: 12/18/2022] Open
Abstract
PRL3, a unique oncotarget, is specifically overexpressed in 80.6% of cancers. In 2003, we reported that PRL3 promotes cell migration, invasion, and metastasis. Herein, firstly, we show that PRL3 induces Polyploid Giant Cancer Cells (PGCCs) formation. PGCCs constitute stem cell-like pools to facilitate cell survival, chemo-resistance, and tumor relapse. The correlations between PRL3 overexpression and PGCCs attributes raised possibilities that PRL3 could be involved in PGCCs formation. Secondly, we show that PRL3+ PGCCs co-express the embryonic stem cell markers SOX2 and OCT4 and arise mainly due to incomplete cytokinesis despite extensive DNA damage. Thirdly, we reveal that PRL3+ PGCCs tolerate prolonged chemotherapy-induced genotoxic stress via suppression of the pro-apoptotic ATM DNA damage-signaling pathway. Fourthly, we demonstrated PRL3-zumab, a First-in-Class humanized antibody drug against PRL3 oncotarget, could reduce tumor relapse in 'tumor removal' animal model. Finally, we confirmed that PGCCs were enriched in relapse tumors versus primary tumors. PRL3-zumab has been approved for Phase 2 clinical trials in Singapore, US, and China to block all solid tumors. This study further showed PRL3-zumab could potentially serve an 'Adjuvant Immunotherapy' after tumor removal surgery to eliminate PRL3+ PGCC stem-like cells, preventing metastasis and relapse.
Collapse
Affiliation(s)
- Min Thura
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Zu Ye
- MD Anderson Cancer Centre, The University of Texas, Houston, TX, USA
| | - Abdul Qader Al-Aidaroos
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Qiancheng Xiong
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Jun Yi Ong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Abhishek Gupta
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Jie Li
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Ke Guo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Koon Hwee Ang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Qi Zeng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore. .,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
95
|
Farina AR, Cappabianca LA, Zelli V, Sebastiano M, Mackay AR. Mechanisms involved in selecting and maintaining neuroblastoma cancer stem cell populations, and perspectives for therapeutic targeting. World J Stem Cells 2021; 13:685-736. [PMID: 34367474 PMCID: PMC8316860 DOI: 10.4252/wjsc.v13.i7.685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric neuroblastomas (NBs) are heterogeneous, aggressive, therapy-resistant embryonal tumours that originate from cells of neural crest (NC) origin and in particular neuroblasts committed to the sympathoadrenal progenitor cell lineage. Therapeutic resistance, post-therapeutic relapse and subsequent metastatic NB progression are driven primarily by cancer stem cell (CSC)-like subpopulations, which through their self-renewing capacity, intermittent and slow cell cycles, drug-resistant and reversibly adaptive plastic phenotypes, represent the most important obstacle to improving therapeutic outcomes in unfavourable NBs. In this review, dedicated to NB CSCs and the prospects for their therapeutic eradication, we initiate with brief descriptions of the unique transient vertebrate embryonic NC structure and salient molecular protagonists involved NC induction, specification, epithelial to mesenchymal transition and migratory behaviour, in order to familiarise the reader with the embryonic cellular and molecular origins and background to NB. We follow this by introducing NB and the potential NC-derived stem/progenitor cell origins of NBs, before providing a comprehensive review of the salient molecules, signalling pathways, mechanisms, tumour microenvironmental and therapeutic conditions involved in promoting, selecting and maintaining NB CSC subpopulations, and that underpin their therapy-resistant, self-renewing metastatic behaviour. Finally, we review potential therapeutic strategies and future prospects for targeting and eradication of these bastions of NB therapeutic resistance, post-therapeutic relapse and metastatic progression.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Lucia Annamaria Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy.
| |
Collapse
|
96
|
Cancer recurrence and lethality are enabled by enhanced survival and reversible cell cycle arrest of polyaneuploid cells. Proc Natl Acad Sci U S A 2021; 118:2020838118. [PMID: 33504594 PMCID: PMC7896294 DOI: 10.1073/pnas.2020838118] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We present a unifying theory to explain cancer recurrence, therapeutic resistance, and lethality. The basis of this theory is the formation of simultaneously polyploid and aneuploid cancer cells, polyaneuploid cancer cells (PACCs), that avoid the toxic effects of systemic therapy by entering a state of cell cycle arrest. The theory is independent of which of the classically associated oncogenic mutations have already occurred. PACCs have been generally disregarded as senescent or dying cells. Our theory states that therapeutic resistance is driven by PACC formation that is enabled by accessing a polyploid program that allows an aneuploid cancer cell to double its genomic content, followed by entry into a nondividing cell state to protect DNA integrity and ensure cell survival. Upon removal of stress, e.g., chemotherapy, PACCs undergo depolyploidization and generate resistant progeny that make up the bulk of cancer cells within a tumor.
Collapse
|
97
|
Zhao Q, Zhang K, Li Z, Zhang H, Fu F, Fu J, Zheng M, Zhang S. High Migration and Invasion Ability of PGCCs and Their Daughter Cells Associated With the Nuclear Localization of S100A10 Modified by SUMOylation. Front Cell Dev Biol 2021; 9:696871. [PMID: 34336846 PMCID: PMC8322665 DOI: 10.3389/fcell.2021.696871] [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: 04/18/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Our previous studies have confirmed that cobalt chloride (CoCl2) or chemoradiotherapy could induce the formation of polyploid tumor giant cells (PGCCs). Polyploid giant cancer cells are a special subpopulation of cancer cells that contribute to solid tumor heterogeneity. The size of PGCC was at least three times larger than regular diploid cancer cells. PGCCs have the properties of cancer stem cells (CSCs) and can express CSC markers CD44 and CD133. Daughter cells derived from PGCCs have strong proliferation, infiltration and migration abilities. However, the detailed molecular mechanism of daughter cells expressing mesenchymal phenotype and displaying strong abilities of proliferation and migration is unclear. As a plasminogen receptor, S100A10 which is closely associated with the invasion and metastasis of malignant tumors, was highly expressed in PGCCs with their daughter cells. In this study, CoCl2 was used to induce the formation of PGCCs in LoVo and HCT116 CRC cells. Cell functional experiments, co-immunoprecipitation, MG132 and ginkgolic acid treatment, western blot, and ChIP-Seq were used to identify the mechanism of S100A10 nuclear location. The proliferation and migration abilities of PGCCs and their daughter cells decreased significantly after S100A10 knockdown. In the control cells, S100A10 was mainly ubiquitinated, while in PGCCs and daughter cells, S100A10 was mainly SUMOylated, which was associated with S100A10 nuclear location. After SUMO1 was inhibited, the nuclear S100A10 in PGCCs and daughter cells decreased, and their proliferation and migration abilities significantly decreased. ChIP-Seq combined with real-time fluorescent quantitative PCR showed that S100A10 regulated the expression of neutrophil defensin 3 (DEFA3), receptor-type tyrosine-protein phosphatase N2 (PTPRN2), and rho guanine nucleotide exchange factor 18 (ARHGEF18), which were associated with actin dynamics and cytoskeleton remodeling. The expression of S100A10 in the nuclei and cytoplasm of rectal cancer after neoadjuvant chemoradiation (nCRT) and liver metastases increased compared with that in rectal cancer without nCRT. Taken together, the expression and nuclear localization of S100A10 modified by SUMOylation were associated with the high proliferation and migration of PGCCs and their daughter cells, and the differentiation, metastases, and relapse of CRCs by regulating the expression of ARHGEF18, PTPRN2, and DEFA3.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, China.,Tianjin Medical University, Tianjin, China
| | - Kexin Zhang
- Graduate School, School of Medicine, Nankai University, Tianjin, China
| | - Zugui Li
- 3Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hao Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, China.,3Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fangmei Fu
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, China.,3Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junjie Fu
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, China.,Tianjin Medical University, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| |
Collapse
|
98
|
Kostecka LG, Pienta KJ, Amend SR. Polyaneuploid Cancer Cell Dormancy: Lessons From Evolutionary Phyla. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.660755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Dormancy is a key survival strategy in many organisms across the tree of life. Organisms that utilize some type of dormancy (hibernation, aestivation, brumation, diapause, and quiescence) are able to survive in habitats that would otherwise be uninhabitable. Induction into dormant states is typically caused by environmental stress. While organisms are dormant, their physical activity is minimal, and their metabolic rates are severely depressed (hypometabolism). These metabolic reductions allow for the conservation and distribution of energy while conditions in the environment are poor. When conditions are more favorable, the organisms are then able to come out of dormancy and reengage in their environment. Polyaneuploid cancer cells (PACCs), proposed mediators of cancer metastasis and resistance, access evolutionary programs and employ dormancy as a survival mechanism in response to stress. Quiescence, the type of dormancy observed in PACCs, allows these cells the ability to survive stressful conditions (e.g., hypoxia in the microenvironment, transiting the bloodstream during metastasis, and exposure to chemotherapy) by downregulating and altering metabolic function, but then increasing metabolic activities again once stress has passed. We can gain insights regarding the mechanisms underlying PACC dormancy by looking to the evolution of dormancy in different organisms.
Collapse
|
99
|
Acikgoz E, Soner BC, Ozdil B, Guven M. CD133+/CD44+ prostate cancer stem cells exhibit embryo-like behavior patterns. Acta Histochem 2021; 123:151743. [PMID: 34157581 DOI: 10.1016/j.acthis.2021.151743] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs), which act as an important bridge between cancer formation and embryonic development, represent a small population associated with tumor initiation, drug resistance, metastasis and recurrence. CSCs have the ability to form spheroids in three-dimensional culture systems. Tumor spheroids derived from CSCs with symmetric and asymmetric division patterns were found to contain highly heterogeneous cell groups. The biological behavior patterns which some CSCs display serve as an important bridge between cancer formation and embryonic development. The cell population in the DU-145 prostate cancer cell line with surface markers CD133+/CD44+ was isolated by FACS. Prostate spheroids were formed by using agarose-coated plates. The morphological characteristics of the cell population within spheroid structure and the expression of Ki-67 and Caspase-3 were investigated by histochemical methods. In this study, we observed that CD133+/CD44+ prostate CSCs form different spheroid structures as well as normal spheroid structures: i) some spheroid structures formed with a highly transparent zone on the outer part of the spheroid, in addition to the normal spheroidal zones and ii) spheroidal structures obtained from prostate CD1334+/CD44+ CSCs that share the same microenvironment are hollow spheres similar to the blastula-like structure in the embryo. These spheroidal structures exhibiting embryo-like properties indicate that the expression of embryonic factors might be reiterated in CSCs. Further investigation of the formation mechanism of the transparent zone and the hollow sphere will shed light on the embryonic origin of prostate cancer and the design of new therapeutic strategies.
Collapse
Affiliation(s)
- Eda Acikgoz
- Van Yuzuncu Yil University, Faculty of Medicine, Department of Histology and Embryology, Van, 65080, Turkey.
| | - Burak Cem Soner
- Izmir Demokrasi University, Faculty of Medicine, Department of Pharmacology, Izmir, Turkey
| | - Berrin Ozdil
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey; Ege University, Faculty of Medicine, Department of Histology and Embryology, Izmir, Turkey
| | - Mustafa Guven
- Van Yuzuncu Yil University, Faculty of Medicine, Van, Turkey
| |
Collapse
|
100
|
Wang XF, Yang SA, Gong S, Chang CH, Portilla JM, Chatterjee D, Irianto J, Bao H, Huang YC, Deng WM. Polyploid mitosis and depolyploidization promote chromosomal instability and tumor progression in a Notch-induced tumor model. Dev Cell 2021; 56:1976-1988.e4. [PMID: 34146466 DOI: 10.1016/j.devcel.2021.05.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/18/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Ploidy variation is a cancer hallmark and is frequently associated with poor prognosis in high-grade cancers. Using a Drosophila solid-tumor model where oncogenic Notch drives tumorigenesis in a transition-zone microenvironment in the salivary gland imaginal ring, we find that the tumor-initiating cells normally undergo endoreplication to become polyploid. Upregulation of Notch signaling, however, induces these polyploid transition-zone cells to re-enter mitosis and undergo tumorigenesis. Growth and progression of the transition-zone tumor are fueled by a combination of polyploid mitosis, endoreplication, and depolyploidization. Both polyploid mitosis and depolyploidization are error prone, resulting in chromosomal copy-number variation and polyaneuploidy. Comparative RNA-seq and epistasis analysis reveal that the DNA-damage response genes, also active during meiosis, are upregulated in these tumors and are required for the ploidy-reduction division. Together, these findings suggest that polyploidy and associated cell-cycle variants are critical for increased tumor-cell heterogeneity and genome instability during cancer progression.
Collapse
Affiliation(s)
- Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Sheng-An Yang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Shangyu Gong
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Chih-Hsuan Chang
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Juan Martin Portilla
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Deeptiman Chatterjee
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Hongcun Bao
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University Louisiana Center Research Center, New Orleans, LA 70112, USA; Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|