1
|
Merckens A, Sieler M, Keil S, Dittmar T. Altered Phenotypes of Breast Epithelial × Breast Cancer Hybrids after ZEB1 Knock-Out. Int J Mol Sci 2023; 24:17310. [PMID: 38139138 PMCID: PMC10744253 DOI: 10.3390/ijms242417310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
ZEB1 plays a pivotal role in epithelial-to-mesenchymal transition (EMT), (cancer) cell stemness and cancer therapy resistance. The M13HS tumor hybrids, which were derived from spontaneous fusion events between the M13SV1-EGFP-Neo breast epithelial cells and HS578T-Hyg breast cancer cells, express ZEB1 and exhibit prospective cancer stem cell properties. To explore a possible correlation between the ZEB1 and stemness/ EMT-related properties in M13HS tumor hybrids, ZEB1 was knocked-out by CRISPR/Cas9. Colony formation, mammosphere formation, cell migration, invasion assays, flow cytometry and Western blot analyses were performed for the characterization of ZEB1 knock-out cells. The ZEB1 knock-out in M13HS tumor cells was not correlated with the down-regulation of the EMT-related markers N-CADHERIN (CDH2) and VIMENTIN and up-regulation of miR-200c-3p. Nonetheless, both the colony formation and mammosphere formation capacities of the M13HS ZEB1 knock-out cells were markedly reduced. Interestingly, the M13HS-2 ZEB1-KO cells harbored a markedly higher fraction of ALDH1-positive cells. The Transwell/ Boyden chamber migration assay data indicated a reduced migratory activity of the M13HS ZEB1-knock-out tumor hybrids, whereas in scratch/ wound-healing assays only the M13SH-8 ZEB1-knock-out cells possessed a reduced locomotory activity. Similarly, only the M13HS-8 ZEB1-knock-out tumor hybrids showed a reduced invasion capacity. Although the ZEB1 knock-out resulted in only moderate phenotypic changes, our data support the role of ZEB1 in EMT and stemness.
Collapse
Affiliation(s)
| | | | | | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58448 Witten, Germany; (A.M.); (M.S.); (S.K.)
| |
Collapse
|
2
|
Alrumaihi F. The Multi-Functional Roles of CCR7 in Human Immunology and as a Promising Therapeutic Target for Cancer Therapeutics. Front Mol Biosci 2022; 9:834149. [PMID: 35874608 PMCID: PMC9298655 DOI: 10.3389/fmolb.2022.834149] [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: 12/13/2021] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
An important hallmark of the human immune system is to provide adaptive immunity against pathogens but tolerance toward self-antigens. The CC-chemokine receptor 7 (CCR7) provides a significant contribution in guiding cells to and within lymphoid organs and is important for acquiring immunity and tolerance. The CCR7 holds great importance in establishing thymic architecture and function and naïve and regulatory T-cell homing in the lymph nodes. Similarly, the receptor is a key regulator in cancer cell migration and the movement of dendritic cells. This makes the CCR7 an important receptor as a drug and prognostic marker. In this review, we discussed several biological roles of the CCR7 and its importance as a drug and prognostic marker.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
3
|
Generation of Cancer Stem/Initiating Cells by Cell-Cell Fusion. Int J Mol Sci 2022; 23:ijms23094514. [PMID: 35562905 PMCID: PMC9101717 DOI: 10.3390/ijms23094514] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
CS/ICs have raised great expectations in cancer research and therapy, as eradication of this key cancer cell type is expected to lead to a complete cure. Unfortunately, the biology of CS/ICs is rather complex, since no common CS/IC marker has yet been identified. Certain surface markers or ALDH1 expression can be used for detection, but some studies indicated that cancer cells exhibit a certain plasticity, so CS/ICs can also arise from non-CS/ICs. Another problem is intratumoral heterogeneity, from which it can be inferred that different CS/IC subclones must be present in the tumor. Cell–cell fusion between cancer cells and normal cells, such as macrophages and stem cells, has been associated with the generation of tumor hybrids that can exhibit novel properties, such as an enhanced metastatic capacity and even CS/IC properties. Moreover, cell–cell fusion is a complex process in which parental chromosomes are mixed and randomly distributed among daughter cells, resulting in multiple, unique tumor hybrids. These, if they have CS/IC properties, may contribute to the heterogeneity of the CS/IC pool. In this review, we will discuss whether cell–cell fusion could also lead to the origin of different CS/ICs that may expand the overall CS/IC pool in a primary tumor.
Collapse
|
4
|
Kaigorodova EV, Kozik AV, Zavaruev IS, Grishchenko MY. Hybrid/Atypical Forms of Circulating Tumor Cells: Current State of the Art. BIOCHEMISTRY (MOSCOW) 2022; 87:380-390. [PMID: 35527376 PMCID: PMC8993035 DOI: 10.1134/s0006297922040071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cancer is one of the most common diseases worldwide, and its treatment is associated with many challenges such as drug and radioresistance and formation of metastases. These difficulties are due to tumor heterogeneity, which has many causes. One may be the cell fusion, a process that is relevant to both physiological (e.g., wound healing) and pathophysiological (cancer and viral infection) processes. This literature review aimed to summarize the existing data on the hybrid/atypical forms of circulating cancer cells and their role in tumor progression. For that, the bioinformatics search in universal databases, such as PubMed, NCBI, and Google Scholar was conducted by using the keywords “hybrid cancer cells”, “cancer cell fusion”, etc. In this review the latest information related to the hybrid tumor cells, theories of their genesis, characteristics of different variants with data from our own researches are presented. Many aspects of the hybrid cell research are still in their infancy. However, with the level of knowledge already accumulated, circulating hybrids such as CAML and CHC could be considered as promising biomarkers of cancerous tumors, and even more as a new approach to cancer treatment.
Collapse
Affiliation(s)
- Evgeniya V Kaigorodova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
- Siberian State Medical University, Tomsk, 634050, Russia
| | - Alexey V Kozik
- Siberian State Medical University, Tomsk, 634050, Russia
| | | | | |
Collapse
|
5
|
Manjunath Y, Suvilesh KN, Mitchem JB, Avella Patino DM, Kimchi ET, Staveley-O'Carroll KF, Pantel K, Yi H, Li G, Harris PK, Chaudhuri AA, Kaifi JT. Circulating Tumor-Macrophage Fusion Cells and Circulating Tumor Cells Complement Non-Small-Cell Lung Cancer Screening in Patients With Suspicious Lung-RADS 4 Nodules. JCO Precis Oncol 2022; 6:e2100378. [PMID: 35417204 PMCID: PMC9012602 DOI: 10.1200/po.21.00378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/30/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Low-dose computed tomography (LDCT) screening of high-risk patients decreases lung cancer-related mortality. However, high false-positive rates associated with LDCT result in unnecessary interventions. To distinguish non-small-cell lung cancer (NSCLC) from benign nodules, in the present study, we integrated cellular liquid biomarkers in patients with suspicious lung nodules (lung cancer screening reporting and data system [Lung-RADS] 4). METHODS Prospectively, 7.5 mL of blood was collected from 221 individuals (training set: 90 nonscreened NSCLC patients, 74 high-risk screening patients with no/benign nodules [Lung-RADS 1-3], and 20 never smokers; validation set: 37 patients with suspicious nodules [Lung-RADS 4]). Circulating tumor cells (CTCs), CTC clusters, and tumor-macrophage fusion (TMF) cells were identified by blinded analyses. Screening patients underwent a median of two LDCTs (range, 1-4) with a median surveillance time of 30 (range, 11-50) months. RESULTS In the validation set of 37 Lung-RADS 4 patients, all circulating cellular biomarker counts (P < .005; Wilcoxon test) and positivity rates were significantly higher in 23 biopsy-proven NSCLC patients (CTCs: 23 of 23 [100%], CTC clusters: 6 of 23 [26.1%], and TMF cells: 15 of 23 [65.2%]) than in 14 patients with biopsy-proven benign nodules (6 of 14 [42.9%], 0 of 14 [0%], and 2 of 14 [14.3%]). On the basis of cutoff values from the training set, logistic regression with receiver operating characteristic and area under the curve analyses demonstrated that CTCs (sensitivity: 0.870, specificity: 1.0, and area under the curve: 0.989) and TMF cells (0.652; 0.880; 0.790) complement LDCT in diagnosing NSCLC in Lung-RADS 4 patients. CONCLUSION Cellular liquid biomarkers have a potential to complement LDCT interpretation of suspicious Lung-RADS 4 nodules to distinguish NSCLC from benign lung nodules. A future prospective, large-scale, multicenter clinical trial should validate the role of cellular liquid biomarkers in improving diagnostic accuracy in high-risk patients with Lung-RADS 4 nodules.
Collapse
Affiliation(s)
- Yariswamy Manjunath
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| | - Kanve Nagaraj Suvilesh
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
| | - Jonathan B. Mitchem
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Diego M. Avella Patino
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| | - Eric T. Kimchi
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Kevin F. Staveley-O'Carroll
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Klaus Pantel
- Institute for Tumor Biology, University of Hamburg, Hamburg, Germany
| | - Huang Yi
- Departments of Radiation Oncology, Genetics, and Computer Science and Engineering, Washington University School of Medicine, St Louis, MO
| | - Guangfu Li
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| | - Peter K. Harris
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
- Departments of Radiation Oncology, Genetics, and Computer Science and Engineering, Washington University School of Medicine, St Louis, MO
| | - Aadel A. Chaudhuri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
- Departments of Radiation Oncology, Genetics, and Computer Science and Engineering, Washington University School of Medicine, St Louis, MO
| | - Jussuf T. Kaifi
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
6
|
Hybrid Formation and Fusion of Cancer Cells In Vitro and In Vivo. Cancers (Basel) 2021; 13:cancers13174496. [PMID: 34503305 PMCID: PMC8431460 DOI: 10.3390/cancers13174496] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cell fusion as a fundamental biological process is required for various physiological processes, including fertilization, placentation, myogenesis, osteoclastogenesis, and wound healing/tissue regeneration. However, cell fusion is also observed during pathophysiological processes like tumor development. Mesenchymal stroma/stem-like cells (MSC) which play an important role within the tumor microenvironment like other cell types such as macrophages can closely interact and hybridize with cancer cells. The formation of cancer hybrid cells can involve various different mechanisms whereby the genomic parts of the hybrid cells require rearrangement to form a new functional hybrid cell. The fusion of cancer cells with neighboring cell types may represent an important mechanism during tumor development since cancer hybrid cells are detectable in various tumor tissues. During this rare event with resulting genomic instability the cancer hybrid cells undergo a post-hybrid selection process (PHSP) to reorganize chromosomes of the two parental nuclei whereby the majority of the hybrid population undergoes cell death. The remaining cancer hybrid cells survive by displaying altered properties within the tumor tissue. Abstract The generation of cancer hybrid cells by intra-tumoral cell fusion opens new avenues for tumor plasticity to develop cancer stem cells with altered properties, to escape from immune surveillance, to change metastatic behavior, and to broaden drug responsiveness/resistance. Genomic instability and chromosomal rearrangements in bi- or multinucleated aneuploid cancer hybrid cells contribute to these new functions. However, the significance of cell fusion in tumorigenesis is controversial with respect to the low frequency of cancer cell fusion events and a clonal advantage of surviving cancer hybrid cells following a post-hybrid selection process. This review highlights alternative processes of cancer hybrid cell development such as entosis, emperipolesis, cannibalism, therapy-induced polyploidization/endoreduplication, horizontal or lateral gene transfer, and focusses on the predominant mechanisms of cell fusion. Based upon new properties of cancer hybrid cells the arising clinical consequences of the subsequent tumor heterogeneity after cancer cell fusion represent a major therapeutic challenge.
Collapse
|
7
|
Cell Fusion of Mesenchymal Stem/Stromal Cells and Breast Cancer Cells Leads to the Formation of Hybrid Cells Exhibiting Diverse and Individual (Stem Cell) Characteristics. Int J Mol Sci 2020; 21:ijms21249636. [PMID: 33348862 PMCID: PMC7765946 DOI: 10.3390/ijms21249636] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the most common diseases worldwide, and treatment bears many challenges such as drug and radioresistance and formation of metastases. These difficulties are due to tumor heterogeneity, which has many origins. One may be cell fusion, a process that is relevant in both physiological (e.g., wound healing) and pathophysiological (cancer and viral infection) processes. In this study, we examined if cell fusion between mesenchymal stem/stromal cells (MSCs) and breast cancer (BC) cells occurs and if newly generated hybrid cells may exhibit cancer stem/initiating cell (CS/IC) characteristics. Therefore, several methods such as mammosphere assay, AldeRed assay, flow cytometry (CD24, CD44, CD104) and Western blot analysis (of epithelial to mesenchymal transition markers such as SNAIL, SLUG and Twist) were applied. In short, four different hybrid clones, verified by short tandem repeat (STR) analysis, were analyzed; each expressed an individual phenotype that seemed not to be explicitly related to either a more stem cell or cancer cell phenotype. These results show that cancer cells and MSCs are able to fuse spontaneously in vitro, thereby giving rise to hybrid cells with new properties, which likely indicate that cell fusion may be a trigger for tumor heterogeneity.
Collapse
|
8
|
Circulating Giant Tumor-Macrophage Fusion Cells Are Independent Prognosticators in Patients With NSCLC. J Thorac Oncol 2020; 15:1460-1471. [DOI: 10.1016/j.jtho.2020.04.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/09/2020] [Accepted: 04/28/2020] [Indexed: 12/24/2022]
|
9
|
Zhang LN, Zhang DD, Yang L, Gu YX, Zuo QP, Wang HY, Xu J, Liu DX. Roles of cell fusion between mesenchymal stromal/stem cells and malignant cells in tumor growth and metastasis. FEBS J 2020; 288:1447-1456. [PMID: 33070450 DOI: 10.1111/febs.15483] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/21/2020] [Accepted: 07/08/2020] [Indexed: 01/02/2023]
Abstract
Invasion and metastasis are the basic characteristics and important markers of malignant tumors, which are also the main cause of death in cancer patients. Epithelial-mesenchymal transition (EMT) is recognized as the first step of tumor invasion and metastasis. Many studies have demonstrated that cell fusion is a common phenomenon and plays a critical role in cancer development and progression. At present, cancer stem cell fusion has been considered as a new mechanism of cancer metastasis. Mesenchymal stromal/stem cell (MSC) is a kind of adult stem cells with high self-renewal ability and multidifferentiation potential, which is used as a very promising fusogenic candidate in the tumor microenvironment and has a crucial role in cancer progression. Many research results have shown that MSCs are involved in the regulation of tumor growth and metastasis through cell fusion. However, the role of cell fusion between MSCs and malignant cells in tumor growth and metastasis is still controversial. Several studies have demonstrated that MSCs can enhance malignant characteristics, promoting tumor growth and metastasis by fusing with malignant cells, while other conflicting reports believe that MSCs can reduce tumorigenicity upon fusion with malignant cells. In this review, we summarize the recent research on cell fusion events between MSCs and malignant cells in tumor growth and metastasis. The elucidation of the molecular mechanisms between MSC fusion and tumor metastasis may provide an effective strategy for tumor biotherapy.
Collapse
Affiliation(s)
- Li-Na Zhang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Di-Di Zhang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Lei Yang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Yu-Xuan Gu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Qiu-Ping Zuo
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Hao-Yi Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Jia Xu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Dian-Xin Liu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| |
Collapse
|
10
|
Human Endogenous Retrovirus K in Cancer: A Potential Biomarker and Immunotherapeutic Target. Viruses 2020; 12:v12070726. [PMID: 32640516 PMCID: PMC7412025 DOI: 10.3390/v12070726] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022] Open
Abstract
In diseases where epigenetic mechanisms are changed, such as cancer, many genes show altered gene expression and inhibited genes become activated. Human endogenous retrovirus type K (HERV-K) expression is usually inhibited in normal cells from healthy adults. In tumor cells, however, HERV-K mRNA expression has been frequently documented to increase. Importantly, HERV-K-derived proteins can act as tumor-specific antigens, a class of neoantigens, and induce immune responses in different types of cancer. In this review, we describe the function of the HERV-K HML-2 subtype in carcinogenesis as biomarkers, and their potential as targets for cancer immunotherapy.
Collapse
|
11
|
Manjunath Y, Porciani D, Mitchem JB, Suvilesh KN, Avella DM, Kimchi ET, Staveley-O’Carroll KF, Burke DH, Li G, Kaifi JT. Tumor-Cell-Macrophage Fusion Cells as Liquid Biomarkers and Tumor Enhancers in Cancer. Int J Mol Sci 2020; 21:E1872. [PMID: 32182935 PMCID: PMC7084898 DOI: 10.3390/ijms21051872] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 02/06/2023] Open
Abstract
Although molecular mechanisms driving tumor progression have been extensively studied, the biological nature of the various populations of circulating tumor cells (CTCs) within the blood is still not well understood. Tumor cell fusion with immune cells is a longstanding hypothesis that has caught more attention in recent times. Specifically, fusion of tumor cells with macrophages might lead to the development of metastasis by acquiring features such as genetic and epigenetic heterogeneity, chemotherapeutic resistance, and immune tolerance. In addition to the traditional FDA-approved definition of a CTC (CD45-, EpCAM+, cytokeratins 8+, 18+ or 19+, with a DAPI+ nucleus), an additional circulating cell population has been identified as being potential fusions cells, characterized by distinct, large, polymorphonuclear cancer-associated cells with a dual epithelial and macrophage/myeloid phenotype. Artificial fusion of tumor cells with macrophages leads to migratory, invasive, and metastatic phenotypes. Further studies might investigate whether these have a potential impact on the immune response towards the cancer. In this review, the background, evidence, and potential relevance of tumor cell fusions with macrophages is discussed, along with the potential role of intercellular connections in their formation. Such fusion cells could be a key component in cancer metastasis, and therefore, evolve as a diagnostic and therapeutic target in cancer precision medicine.
Collapse
Affiliation(s)
- Yariswamy Manjunath
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - David Porciani
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA; (D.P.); (D.H.B.)
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, USA
| | - Jonathan B. Mitchem
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Kanve N. Suvilesh
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
| | - Diego M. Avella
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Eric T. Kimchi
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Donald H. Burke
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA; (D.P.); (D.H.B.)
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, USA
- Department of Biochemistry, University of Missouri, Columbia, MO 65212, USA
| | - Guangfu Li
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA; (D.P.); (D.H.B.)
| | - Jussuf T. Kaifi
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| |
Collapse
|
12
|
Tumor Microenvironment and Cell Fusion. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5013592. [PMID: 31380426 PMCID: PMC6657644 DOI: 10.1155/2019/5013592] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 12/14/2022]
Abstract
Cell fusion is a highly regulated biological process that occurs under both physiological and pathological conditions. The cellular and extracellular environment is critical for the induction of the cell-cell fusion. Aberrant cell fusion is initiated during tumor progression. Tumor microenvironment is a complex dynamic system formed by the interaction between tumor cells and their surrounding cells. Cell-cell fusion mediates direct interaction between tumor cells and their surrounding cells and is associated with tumor initiation and progression. Various microenvironmental factors affect cell fusion in tumor microenvironment and generate hybrids that acquire genomes of both parental cells and exhibit novel characteristics, such as tumor stem cell-like properties, radioresistance, drug resistance, immune evasion, and enhanced migration and invasion abilities, which are closely related to the initiation, invasion, and metastasis of tumor. The phenotypic characteristics of hybrids are based on the phenotypes of parental cells, and the fusion of tumor cells with diverse types of microenvironmental fusogenic cells is concomitant with phenotypic heterogeneity. This review highlights the types of fusogenic cells in tumor microenvironment that can fuse with tumor cells and their specific significance and summarizes the various microenvironmental factors affecting tumor cell fusion. This review may be used as a reference to develop strategies for future research on tumor cell fusion and the exploration of cell fusion-based antitumor therapies.
Collapse
|
13
|
Zhang LN, Kong CF, Zhao D, Cong XL, Wang SS, Ma L, Huang YH. Fusion with mesenchymal stem cells differentially affects tumorigenic and metastatic abilities of lung cancer cells. J Cell Physiol 2018; 234:3570-3582. [PMID: 30417342 DOI: 10.1002/jcp.27011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Cell fusion plays a crucial role in cancer progression and leads to massive aberrant changes in chromosome and gene expression involved in tumor metastasis. Cancer cells can fuse with many cell types, including stromal cells, epithelial cells, macrophages, and endothelial cells. Mesenchymal stem cells (MSCs) have been reported to migrate and incorporate into tumor sites during cancer progression. However, the underlying mechanism of stem cell fusion in tumor metastasis has not been fully deciphered. In this research, we established a cell fusion model between lung cancer cells and MSCs in vitro. We found that the hybrid cells showed enhanced metastatic capacity with increased expression of MMP-2 and MMP-9, whereas the proliferation ability was inhibited and cell cycle was blocked in the G0 /G1 phase with elevated expression of p21, p27, and p53. Moreover, the hybrid cells lost epithelial morphology and exhibited an epithelial-mesenchymal transition (EMT) change with downregulation of E-cadherin and upregulation of N-cadherin, Vimentin, α-SMA and Fibronectin1. Meanwhile, the expressions of EMT transcription factors, including Snail1, Slug, Twist1, Zeb1, and Zeb2, were also increased in hybrid cells. More important, the fusion hybrids acquired stem cell-like properties, which exhibited increased expression stem cell transcription factors Oct4, Sox2, Nanog, Kif4 as well as Bmi1. Taken together, our results suggested that cell fusion between lung cancer cells and MSCs offered enhanced metastatic capacity and characteristics of cancer stem cell by undergoing EMT. This study will contribute to explaning the origin of lung cancer stem cells and to elucidate the role of cell fusion in cancer metastasis.
Collapse
Affiliation(s)
- Li-Na Zhang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Chen-Fei Kong
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Dan Zhao
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xian-Ling Cong
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Shen-Sen Wang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Ling Ma
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Ying-Hui Huang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| |
Collapse
|
14
|
Grandi N, Tramontano E. HERV Envelope Proteins: Physiological Role and Pathogenic Potential in Cancer and Autoimmunity. Front Microbiol 2018; 9:462. [PMID: 29593697 PMCID: PMC5861771 DOI: 10.3389/fmicb.2018.00462] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
Human endogenous retroviruses (HERVs) are relics of ancient infections accounting for about the 8% of our genome. Despite their persistence in human DNA led to the accumulation of mutations, HERVs are still contributing to the human transcriptome, and a growing number of findings suggests that their expression products may have a role in various diseases. Among HERV products, the envelope proteins (Env) are currently highly investigated for their pathogenic properties, which could likely be participating to several disorders with complex etiology, particularly in the contexts of autoimmunity and cancer. In fact, HERV Env proteins have been shown, on the one side, to trigger both innate and adaptive immunity, prompting inflammatory, cytotoxic and apoptotic reactions; and, on the other side, to prevent the immune response activation, presenting immunosuppressive properties and acting as immune downregulators. In addition, HERV Env proteins have been shown to induce abnormal cell-cell fusion, possibly contributing to tumor development and metastasizing processes. Remarkably, even highly defective HERV env genes and alternative env splicing variants can provide further mechanisms of pathogenesis. A well-known example is the HERV-K(HML2) env gene that, depending on the presence or the absence of a 292-bp deletion, can originate two proteins of different length (Np9 and Rec) proposed to have oncogenic properties. The understanding of their involvement in complex pathological disorders made HERV Env proteins potential targets for therapeutic interventions. Of note, a monoclonal antibody directed against a HERV-W Env is currently under clinical trial as therapeutic approach for multiple sclerosis, representing the first HERV-based treatment. The present review will focus on the current knowledge of the HERV Env expression, summarizing its role in human physiology and its possible pathogenic effects in various cancer and autoimmune disorders. It moreover analyzes HERV Env possible exploitation for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Nicole Grandi
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Enzo Tramontano
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| |
Collapse
|
15
|
Noubissi FK, Ogle BM. Cancer Cell Fusion: Mechanisms Slowly Unravel. Int J Mol Sci 2016; 17:ijms17091587. [PMID: 27657058 PMCID: PMC5037852 DOI: 10.3390/ijms17091587] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/26/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023] Open
Abstract
Although molecular mechanisms and signaling pathways driving invasion and metastasis have been studied for many years, the origin of the population of metastatic cells within the primary tumor is still not well understood. About a century ago, Aichel proposed that cancer cell fusion was a mechanism of cancer metastasis. This hypothesis gained some support over the years, and recently became the focus of many studies that revealed increasing evidence pointing to the possibility that cancer cell fusion probably gives rise to the metastatic phenotype by generating widespread genetic and epigenetic diversity, leading to the emergence of critical populations needed to evolve resistance to the treatment and development of metastasis. In this review, we will discuss the clinical relevance of cancer cell fusion, describe emerging mechanisms of cancer cell fusion, address why inhibiting cancer cell fusion could represent a critical line of attack to limit drug resistance and to prevent metastasis, and suggest one new modality for doing so.
Collapse
Affiliation(s)
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Lillehei Heart Institute, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Institute for Engineering and Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
16
|
Kuźnar-Kamińska B, Mikuła-Pietrasik J, Sosińska P, Książek K, Batura-Gabryel H. COPD promotes migration of A549 lung cancer cells: the role of chemokine CCL21. Int J Chron Obstruct Pulmon Dis 2016; 11:1061-6. [PMID: 27307721 PMCID: PMC4888725 DOI: 10.2147/copd.s96490] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Patients with COPD develop lung cancer more frequently than healthy smokers. At the same time, molecular mediators promoting various aspects of cancer cell progression are still elusive. In this report, we examined whether COPD can be coupled with increased migration of non-small-cell lung cancer cells A549 and, if so, whether this effect may be related to altered production and activity of chemokines CCL21, CXCL5, and CXCL12. The study showed that the migration of A549 cells through the polycarbonate membrane and basement membrane extract toward a chemotactic gradient elicited by serum from patients with COPD was markedly higher as compared with serum from healthy donors. The concentration of CCL21 and CXCL12, but not CXCL5, in serum from patients with COPD was also increased. Experiments in which CCL21- and CXCL12-dependent signaling was blocked revealed that increased migration of the cancer cells upon treatment with serum from patients with COPD was mediated exclusively by CCL21. Collectively, our results indicate that COPD may contribute to the progression of lung cancer via CCL21-dependent intensification of cancer cell migration.
Collapse
Affiliation(s)
| | | | - Patrycja Sosińska
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Krzysztof Książek
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | | |
Collapse
|
17
|
Tosun S, Fried S, Niggemann B, Zänker KS, Dittmar T. Hybrid Cells Derived from Human Breast Cancer Cells and Human Breast Epithelial Cells Exhibit Differential TLR4 and TLR9 Signaling. Int J Mol Sci 2016; 17:ijms17050726. [PMID: 27187369 PMCID: PMC4881548 DOI: 10.3390/ijms17050726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/15/2016] [Accepted: 05/03/2016] [Indexed: 02/07/2023] Open
Abstract
TLRs are important receptors of cells of the innate immune system since they recognize various structurally conserved molecular patterns of different pathogens as well as endogenous ligands. In cancer, the role of TLRs is still controversial due to findings that both regression and progression of tumors could depend on TLR signaling. In the present study, M13SV1-EGFP-Neo human breast epithelial cells, MDA-MB-435-Hyg human breast cancer cells and two hybrids M13MDA435-1 and -3 were investigated for TLR4 and TLR9 expression and signaling. RT-PCR data revealed that LPS and CpG-ODN induced the expression of pro-inflammatory cytokines, like IFN-β, TNF-α, IL-1β and IL-6 in hybrid cells, but not parental cells. Interestingly, validation of RT-PCR data by Western blot showed detectable protein levels solely after LPS stimulation, suggesting that regulatory mechanisms are also controlled by TLR signaling. Analysis of pAKT and pERK1/2 levels upon LPS and CpG-ODN stimulation revealed a differential phosphorylation pattern in all cells. Finally, the migratory behavior of the cells was investigated showing that both LPS and CpG-ODN potently blocked the locomotory activity of the hybrid cells in a dose-dependent manner. In summary, hybrid cells exhibit differential TLR4 and TLR9 signaling.
Collapse
Affiliation(s)
- Songül Tosun
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 10, 58448 Witten, Germany.
| | - Sabrina Fried
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 10, 58448 Witten, Germany.
- Faculty of Medicine, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany.
| | - Bernd Niggemann
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 10, 58448 Witten, Germany.
| | - Kurt S Zänker
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 10, 58448 Witten, Germany.
| | - Thomas Dittmar
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 10, 58448 Witten, Germany.
| |
Collapse
|
18
|
Single-cell RNA-seq reveals activation of unique gene groups as a consequence of stem cell-parenchymal cell fusion. Sci Rep 2016; 6:23270. [PMID: 26997336 PMCID: PMC4800419 DOI: 10.1038/srep23270] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 03/01/2016] [Indexed: 12/26/2022] Open
Abstract
Fusion of donor mesenchymal stem cells with parenchymal cells of the recipient can occur in the brain, liver, intestine and heart following transplantation. The therapeutic benefit or detriment of resultant hybrids is unknown. Here we sought a global view of phenotypic diversification of mesenchymal stem cell-cardiomyocyte hybrids and associated time course. Using single-cell RNA-seq, we found hybrids consistently increase ribosome components and decrease genes associated with the cell cycle suggesting an increase in protein production and decrease in proliferation to accommodate the fused state. But in the case of most other gene groups, hybrids were individually distinct. In fact, though hybrids can express a transcriptome similar to individual fusion partners, approximately one-third acquired distinct expression profiles in a single day. Some hybrids underwent reprogramming, expressing pluripotency and cardiac precursor genes latent in parental cells and associated with developmental and morphogenic gene groups. Other hybrids expressed genes associated with ontologic cancer sets and two hybrids of separate experimental replicates clustered with breast cancer cells, expressing critical oncogenes and lacking tumor suppressor genes. Rapid transcriptional diversification of this type garners consideration in the context of cellular transplantation to damaged tissues, those with viral infection or other microenvironmental conditions that might promote fusion.
Collapse
|
19
|
Fried S, Tosun S, Troost G, Keil S, Zaenker KS, Dittmar T. Lipopolysaccharide (LPS) Promotes Apoptosis in Human Breast Epithelial × Breast Cancer Hybrids, but Not in Parental Cells. PLoS One 2016; 11:e0148438. [PMID: 26863029 PMCID: PMC4749126 DOI: 10.1371/journal.pone.0148438] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 01/18/2016] [Indexed: 01/03/2023] Open
Abstract
Toll-like receptors (TLRs) belong to the group of pathogen recognition receptors known to play a crucial role in the innate immune system. In cancer, TLR expression is still debated controversially due to contradictory results reporting that both induction of apoptosis as well as tumor progression could depend on TLR signaling, whereby recent data rather indicate a pro-tumorigenic effect. The biological phenomenon of cell fusion has been associated with cancer progression due to findings revealing that fusion-derived hybrid cells could exhibit properties like an increased metastatogenic capacity and an increased drug resistance. Thus, M13MDA435 hybrid cell lines, which derived from spontaneous fusion events between human M13SV1-EGFP-Neo breast epithelial cells and human MDA-MB-435-Hyg breast cancer cells, were investigated. Cultivation of cells in the presence of the TLR4 ligand LPS potently induced apoptosis in all hybrid clones, but not in parental cells, which was most likely attributed to differential kinetics of the TLR4 signal transduction cascade. Activation of this pathway concomitant with NF-κB nuclear translocation and TNF-α expression was solely observed in hybrid cells. However, induction of LPS mediated apoptosis was not TNF-α dependent since TNF-α neutralization was not correlated to a decreased amount of dead cells. In addition to TNF-α, LPS also caused IFN-β expression in hybrid clones 1 and 3. Interestingly, hybrid clones differ in the mode of LPS induced apoptosis. While neutralization of IFN-β was sufficient to impair the LPS induced apoptosis in M13MDA435-1 and -3 hybrids, the amount of apoptotic M13MDA435-2 and -4 hybrid cells remained unchanged in the presence of neutralizing IFN-β antibodies. In summary, the fusion of non-LPS susceptible parental human breast epithelial cells and human breast cancer cells gave rise to LPS susceptible hybrid cells, which is in view with the cell fusion hypothesis that hybrid cells could exhibit novel properties.
Collapse
Affiliation(s)
- Sabrina Fried
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research, Witten/Herdecke University, Stockumer Str. 10, Witten, Germany
| | - Songuel Tosun
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research, Witten/Herdecke University, Stockumer Str. 10, Witten, Germany
| | - Gabriele Troost
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research, Witten/Herdecke University, Stockumer Str. 10, Witten, Germany
| | - Silvia Keil
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research, Witten/Herdecke University, Stockumer Str. 10, Witten, Germany
| | - Kurt S. Zaenker
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research, Witten/Herdecke University, Stockumer Str. 10, Witten, Germany
| | - Thomas Dittmar
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research, Witten/Herdecke University, Stockumer Str. 10, Witten, Germany
- * E-mail:
| |
Collapse
|
20
|
Tissue Regeneration in the Chronically Inflamed Tumor Environment: Implications for Cell Fusion Driven Tumor Progression and Therapy Resistant Tumor Hybrid Cells. Int J Mol Sci 2015; 16:30362-81. [PMID: 26703575 PMCID: PMC4691180 DOI: 10.3390/ijms161226240] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 12/19/2022] Open
Abstract
The biological phenomenon of cell fusion in a cancer context is still a matter of controversial debates. Even though a plethora of in vitro and in vivo data have been published in the past decades the ultimate proof that tumor hybrid cells could originate in (human) cancers and could contribute to the progression of the disease is still missing, suggesting that the cell fusion hypothesis is rather fiction than fact. However, is the lack of this ultimate proof a valid argument against this hypothesis, particularly if one has to consider that appropriate markers do not (yet) exist, thus making it virtually impossible to identify a human tumor cell clearly as a tumor hybrid cell. In the present review, we will summarize the evidence supporting the cell fusion in cancer concept. Moreover, we will refine the cell fusion hypothesis by providing evidence that cell fusion is a potent inducer of aneuploidy, genomic instability and, most likely, even chromothripsis, suggesting that cell fusion, like mutations and aneuploidy, might be an inducer of a mutator phenotype. Finally, we will show that "accidental" tissue repair processes during cancer therapy could lead to the origin of therapy resistant cancer hybrid stem cells.
Collapse
|
21
|
Cancer (stem) cell differentiation: An inherent or acquired property? Med Hypotheses 2015; 85:1012-8. [PMID: 26347071 DOI: 10.1016/j.mehy.2015.08.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 08/23/2015] [Indexed: 02/08/2023]
Abstract
There is a growing list of data indicating that cancer (stem) cells could functionally adapt foreign tissue features, such as endothelial-like cells or neuroendocrine cells, express lineage markers or could differentiate into various lineages in response to appropriate differentiation criteria. The finding that cancer (stem) cells may possess some kind of differentiation capacity poses the question whether this might be an inherent or acquired property. Cancer stem cells share stem cell characteristics and may thus possess an inherent differentiation capacity enabling the cells to respond to various differentiation stimuli. Considering the plasticity of cancer (stem) cells, even non-tumorigenic (and putatively non-differentiable) tumor cells could give rise to tumorigenic tumor stem cells, exhibiting stem cell characteristics including an inherent differentiation capacity. On the contrary, cancer (stem) cells may have acquired differentiation capacity as a consequence of a previous cell fusion event with cell types exhibiting differentiation potential and being fusogenic, such as macrophages or stem cells. Of pivotal interest in a tumor context are macrophages, which chiefly foster the chronically inflamed tumor microenvironment. Because chronically inflamed tissue is a well-known trigger for cell fusion and both macrophages and stem cells are highly fusogenic we conclude that cell fusion events between these cell types and cancer (stem) cells should frequently occur, thereby giving rise to hybrid cells exhibiting not only novel properties, like an enhanced metastatogenic phenotype, but also parental characteristics, such as differentiation capacity. Conceivably, the combination of both properties might be advantageous for metastasizing cancer (stem) cells to adapt better and faster to a foreign organ tissue environment.
Collapse
|
22
|
Wei HJ, Nickoloff JA, Chen WH, Liu HY, Lo WC, Chang YT, Yang PC, Wu CW, Williams DF, Gelovani JG, Deng WP. FOXF1 mediates mesenchymal stem cell fusion-induced reprogramming of lung cancer cells. Oncotarget 2015; 5:9514-29. [PMID: 25237908 PMCID: PMC4253450 DOI: 10.18632/oncotarget.2413] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Several reports suggest that malignant cells generate phenotypic diversity through fusion with various types of stromal cells within the tumor microenvironment. Mesenchymal stem cell (MSC) is one of the critical components in the tumor microenvironment and a promising fusogenic candidate, but the underlying functions of MSC fusion with malignant cell have not been fully examined. Here, we demonstrate that MSCs fuse spontaneously with lung cancer cells, and the latter is reprogrammed to slow growth and stem-like state. Transcriptome profiles reveal that lung cancer cells are reprogrammed to a more benign state upon MSC fusion. We further identified FOXF1 as a reprogramming mediator that contributes not only to the reprogramming toward stemness but also to the p21-regulated growth suppression in fusion progeny. Collectively, MSC fusion does not enhance the intrinsic malignancy of lung cancer cells. The anti-malignant effects of MSC fusion-induced reprogramming on lung cancer cells were accomplished by complementation of tumorigenic defects, including restoration of p21 function and normal terminal differentiation pathways as well as up-regulation of FOXF1, a putative tumor suppressor. Such fusion process raises the therapeutic potential that MSC fusion can be utilized to reverse cellular phenotypes in cancer.
Collapse
Affiliation(s)
- Hong-Jian Wei
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan. Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Wei-Hong Chen
- Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Hen-Yu Liu
- Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan. School of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ya-Ting Chang
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Pan-Chyr Yang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Cheng-Wen Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei 112, Taiwan
| | - David F Williams
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC 27157, USA
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI 48201, USA Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Win-Ping Deng
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan. Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
23
|
Noubissi FK, Harkness T, Alexander CM, Ogle BM. Apoptosis-induced cancer cell fusion: a mechanism of breast cancer metastasis. FASEB J 2015; 29:4036-45. [PMID: 26085132 DOI: 10.1096/fj.15-271098] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 06/02/2015] [Indexed: 02/06/2023]
Abstract
Although cancer cell fusion has been suggested as a mechanism of cancer metastasis, the underlying mechanisms defining this process are poorly understood. In a recent study, apoptotic cells were newly identified as a type of cue that induces signaling via phosphatidylserine receptors to promote fusion of myoblasts. The microenvironment of breast tumors is often hypoxic, and because apoptosis is greatly increased in hypoxic conditions, we decided to investigate whether the mechanism of breast cancer cell fusion with mesenchymal stem/multipotent stromal cells (MSCs) involves apoptosis. We used a powerful tool for identification and tracking of hybrids based on bimolecular fluorescence complementation (BiFC) and found that breast cancer cells fused spontaneously with MSCs. This fusion was significantly enhanced with hypoxia and signaling associated with apoptotic cells, especially between nonmetastatic breast cancer cells and MSCs. In addition, the hybrids showed a significantly higher migratory capacity than did the parent cells. Taken together, these findings describe a mechanism by which hypoxia-induced apoptosis stimulates fusion between MSCs and breast tumor cells resulting in hybrids with an enhanced migratory capacity that may enable their dissemination to distant sites or metastases. In the long run, this study may provide new strategies for developing novel drugs for preventing cancer metastasis.
Collapse
Affiliation(s)
- Felicite K Noubissi
- *Department of Biomedical Engineering, Stem Cell Institute, Lillehei Heart Institute, Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA; and Department of Biomedical Engineering and Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ty Harkness
- *Department of Biomedical Engineering, Stem Cell Institute, Lillehei Heart Institute, Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA; and Department of Biomedical Engineering and Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Caroline M Alexander
- *Department of Biomedical Engineering, Stem Cell Institute, Lillehei Heart Institute, Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA; and Department of Biomedical Engineering and Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Brenda M Ogle
- *Department of Biomedical Engineering, Stem Cell Institute, Lillehei Heart Institute, Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA; and Department of Biomedical Engineering and Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
24
|
El-Ghonaimy EA, El-Shinawi M, Ibrahim SA, El-Ghazaly H, Abd-El-Tawab R, Nouh MA, El-Mamlouk T, Mohamed MM. Positive lymph-node breast cancer patients - activation of NF-κB in tumor-associated leukocytes stimulates cytokine secretion that promotes metastasis via C-C chemokine receptor CCR7. FEBS J 2014; 282:271-82. [DOI: 10.1111/febs.13124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 08/23/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022]
Affiliation(s)
| | | | | | | | | | - Mohamed A. Nouh
- Department of Pathology; National Cancer institute; Cairo University; Giza Egypt
| | | | | |
Collapse
|
25
|
Ma S, Shi Y, Pang Y, Dong F, Cheng H, Hao S, Xu J, Zhu X, Yuan W, Cheng T, Zheng G. Notch1-induced T cell leukemia can be potentiated by microenvironmental cues in the spleen. J Hematol Oncol 2014; 7:71. [PMID: 25366136 PMCID: PMC4229605 DOI: 10.1186/s13045-014-0071-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 01/10/2023] Open
Abstract
Background Leukemia is a systemic malignancy originated from hematopoietic cells. The extracellular environment has great impacts on the survival, proliferation and dissemination of leukemia cells. The spleen is an important organ for extramedullary hematopoiesis and a common infiltration site in lymphoid malignancies. Splenomegaly, frequently observed in T cell acute lymphoblastic leukemia (T-ALL), is associated with poor prognosis. However, how the spleen microenvironment distinctly affects T-ALL cells as opposed to bone marrow (BM) microenvironment has not been addressed. Methods A Notch1-induced mouse T-ALL model was applied in this study. Flow cytometry and two-photon fluorescence microscopy were used to analyze early distribution of T-ALL cells. MILLIPLEX® MAP Multiplex Immunoassay was performed to measure cytokine/chemokine levels in different microenvironments. Transwell and co-culture experiments were used to test the effects of splenic microenvironment in vitro. Splenectomy was performed to assess the organ specific impact on the survival of T-ALL-bearing mice. Results More leukemia cells were detected in the spleen than in the BM after injection of T-ALL cells by flow cytometry and two-photon fluorescence microscopy analysis. By screening a panel of cytokines/chemokines, a higher level of MIP-3β was found in the splenic microenvironment than BM microenvironment. In vitro transwell experiment further confirmed that MIP-3β recruits T-ALL cells which express a high level of MIP-3β receptor, CCR7. Furthermore, the splenic microenvironment stimulates T-ALL cells to express a higher level of MIP-3β, which further recruits T-ALL cells to the spleen. Co-culture experiment found that the splenic microenvironment more potently stimulated the proliferation and migration of T-ALL cells than BM. Moreover, the mice transplanted with T-ALL cells from the spleen had a shorter life span than those transplanted from BM, suggesting increased potency of the T-ALL cells induced by the splenic microenvironment. In addition, splenectomy prolonged the survival of leukemic mice. Conclusions Our study demonstrates an organ specific effect on leukemia development. Specifically, T-ALL cells can be potentiated by splenic microenvironment and thus spleen may serve as a target organ for the treatment of some types of leukemia. Electronic supplementary material The online version of this article (doi:10.1186/s13045-014-0071-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shihui Ma
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| | - Yingxu Shi
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China. .,Current address of Yingxu Shi: Affiliated Hospital Clinical Laboratory, Inner Mongolian Medical University, Hohhot, China.
| | - Yakun Pang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| | - Sha Hao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| | - Jing Xu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China. .,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China. .,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China. .,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Guoguang Zheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China. .,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
26
|
Rommerswinkel N, Niggemann B, Keil S, Zänker KS, Dittmar T. Analysis of cell migration within a three-dimensional collagen matrix. J Vis Exp 2014:e51963. [PMID: 25350138 DOI: 10.3791/51963] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The ability to migrate is a hallmark of various cell types and plays a crucial role in several physiological processes, including embryonic development, wound healing, and immune responses. However, cell migration is also a key mechanism in cancer enabling these cancer cells to detach from the primary tumor to start metastatic spreading. Within the past years various cell migration assays have been developed to analyze the migratory behavior of different cell types. Because the locomotory behavior of cells markedly differs between a two-dimensional (2D) and three-dimensional (3D) environment it can be assumed that the analysis of the migration of cells that are embedded within a 3D environment would yield in more significant cell migration data. The advantage of the described 3D collagen matrix migration assay is that cells are embedded within a physiological 3D network of collagen fibers representing the major component of the extracellular matrix. Due to time-lapse video microscopy real cell migration is measured allowing the determination of several migration parameters as well as their alterations in response to pro-migratory factors or inhibitors. Various cell types could be analyzed using this technique, including lymphocytes/leukocytes, stem cells, and tumor cells. Likewise, also cell clusters or spheroids could be embedded within the collagen matrix concomitant with analysis of the emigration of single cells from the cell cluster/ spheroid into the collagen lattice. We conclude that the 3D collagen matrix migration assay is a versatile method to analyze the migration of cells within a physiological-like 3D environment.
Collapse
Affiliation(s)
- Nadine Rommerswinkel
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University
| | - Bernd Niggemann
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University
| | - Silvia Keil
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University
| | - Kurt S Zänker
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University
| | - Thomas Dittmar
- Institute of Immunology & Experimental Oncology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University;
| |
Collapse
|
27
|
Goldenberg DM, Rooney RJ, Loo M, Liu D, Chang CH. In-vivo fusion of human cancer and hamster stromal cells permanently transduces and transcribes human DNA. PLoS One 2014; 9:e107927. [PMID: 25259521 PMCID: PMC4178054 DOI: 10.1371/journal.pone.0107927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/19/2014] [Indexed: 12/16/2022] Open
Abstract
After demonstrating, with karyotyping, polymerase chain reaction (PCR) and fluorescence in-situ hybridization, the retention of certain human chromosomes and genes following the spontaneous fusion of human tumor and hamster cells in-vivo, it was postulated that cell fusion causes the horizontal transmission of malignancy and donor genes. Here, we analyzed gene expression profiles of 3 different hybrid tumors first generated in the hamster cheek pouch after human tumor grafting, and then propagated in hamsters and in cell cultures for years: two Hodgkin lymphomas (GW-532, GW-584) and a glioblastoma multiforme (GB-749). Based on the criteria of MAS 5.0 detection P-values ≤0.065 and at least a 2-fold greater signal expression value than a hamster melanoma control, we identified 3,759 probe sets (ranging from 1,040 to 1,303 in each transplant) from formalin-fixed, paraffin-embedded sections of the 3 hybrid tumors, which unambiguously mapped to 3,107 unique Entrez Gene IDs, representative of all human chromosomes; however, by karyology, one of the hybrid tumors (GB-749) had a total of 15 human chromosomes in its cells. Among the genes mapped, 39 probe sets, representing 33 unique Entrez Gene IDs, complied with the detection criteria in all hybrid tumor samples. Five of these 33 genes encode transcription factors that are known to regulate cell growth and differentiation; five encode cell adhesion- and transmigration-associated proteins that participate in oncogenesis and/or metastasis and invasion; and additional genes encode proteins involved in signaling pathways, regulation of apoptosis, DNA repair, and multidrug resistance. These findings were corroborated by PCR and reverse transcription PCR, showing the presence of human alphoid (α)-satellite DNA and the F11R transcripts in additional tumor transplant generations. We posit that in-vivo fusion discloses genes implicated in tumor progression, and gene families coding for the organoid phenotype. Thus, cancer cells can transduce adjacent stromal cells, with the resulting progeny having permanently transcribed genes with malignant and other gene functions of the donor DNA. Using heterospecific in-vivo cell fusion, genes encoding oncogenic and organogenic traits may be identified.
Collapse
Affiliation(s)
- David M. Goldenberg
- Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, New Jersey, United States of America
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
- * E-mail:
| | - Robert J. Rooney
- Genome Explorations, Inc., Memphis, Tennessee, United States of America
| | - Meiyu Loo
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
| | - Donglin Liu
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
| | - Chien-Hsing Chang
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
| |
Collapse
|