Cell‑cell fusion as an important mechanism of tumor metastasis (Review)

  • Authors:
    • Xiao-Chun Peng
    • Min Zhang
    • Ying-Ying Meng
    • Yan-Fang Liang
    • Ying-Ying Wang
    • Xiao-Qin Liu
    • Wen-Qi Cai
    • Yang Zhou
    • Xian-Wang Wang
    • Zhao-Wu Ma
    • Ying Xiang
    • Li-Si Zeng
    • Shu-Zhong Cui
    • Liu-Ming Yang
    • Hong-Wu Xin
  • View Affiliations

  • Published online on: May 31, 2021     https://doi.org/10.3892/or.2021.8096
  • Article Number: 145
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cell‑cell fusion is a dynamic biological phenomenon, which plays an important role in various physiological processes, such as tissue regeneration. Similarly, normal cells, particularly bone marrow‑derived cells (BMDCs), may attempt to fuse with cancer cells to rescue them. The rescue may fail, but the fused cells end up gaining the motility traits of BMDCs and become metastatic due to the resulting genomic instability. In fact, cell‑cell fusion was demonstrated to occur in vivo in cancer and was revealed to promote tumor metastasis. However, its existence and role may be underestimated, and has not been widely acknowledged. In the present review, the milestones in cell fusion research were highlighted, the evidence for cell‑cell fusion in vitro and in vivo in cancer was evaluated, and the current understanding of the molecular mechanisms by which cell‑cell fusion occurs was summarized, to emphasize their important role in tumor metastasis. The summary provided in the present review may promote further study into this process and result in novel discoveries of strategies for future treatment of tumor metastasis.

Introduction

Tumors are hypothesized to originate as a result of and progress due to sequential genetic and epigenetic mutations of cells. Tumors originate gradually from the tumor stem cells (TSCs) that accrue several mutations (16). In recent years, it has been indicated that the origin of tumors or TSCs involves cell-cell fusion (16). It was originally hypothesized that tumor cells possessed characteristics indicative of aneuploidy and chromosomal disorders. Therefore, it was reasonable to hypothesize that these features of tumors may be associated with cell-cell fusion. It would be interesting to determine if the fusion of a cancerous cell with a normal healthy cell, such as a migrating bone marrow-derived cell (BMDCs), may give rise to unique features in the resultant cell, such as increased tumor initiation, tumor metastasis and/or drug resistance capacity.

Milestones in cell-cell fusion research

Cell-cell fusion, also termed cell hybridization, refers to the process of the fusion of two or more cells into a single hybrid cell, with the formation of a single nucleus possessing genetic information from two or more lineages (7). At the beginning of the process, the membranes begin to fuse, followed by fusion of the cytoplasm and the nuclei, ultimately resulting in the formation of a single cell (8). In multicellular organisms, cell fusion is a basic developmental and physiological process. The fusion of a sperm and egg cell is one of the most classical examples of cell fusion. In 2002, Mohler et al (9) first identified that the eff-1 gene was essential for developmental cell fusion. In 2004, Shemer et al successfully demonstrated that the expression of EFF-1 protein leads to cell fusion, and that it could cause independent cell fusion in the absence of other proteins (10).

Cell-cell fusion can occur in vivo in an organism and in vitro in cell cultures, both spontaneously and artificially. In a laboratory, researchers can use an external agent, such as viral fusion agent (Sendai virus), chemical fusion agent (polyethylene glycol) or electric shock, to induce cell-cell fusion in vitro between the same or different cell types.

The major milestones in the study of cell-cell fusion are summarized in (Tables I and II; Fig. 1). In the 1930′s, scientists observed the presence of multinucleated cells in smallpox, chickenpox, measles and other infectious diseases, and rabbit homotypic cell fusion in vivo in the formation of foreign body giant cells (11). In 1954, Enders and Peebles (12) reported that human multinucleated giant cells or syncytia were formed in vitro as a result of measles viral infection (12). In 1961, Barski (13) observed the somatic cell fusion phenomenon in tissue cultures. In 1962, Furusawa and Cutting (14) discovered that the hemagglutinating virus caused fusion of mouse Ehrlich ascites tumor cells in vitro. In 1965, Cascardo and Carzon (15) found and confirmed that the inactivated measles virus under the appropriate conditions could also induce human cell fusion in vitro. Harris and Watkins (16) reported the fusion of human HeLa cells and mouse Ehrlich ascites tumor cells in vitro. In 1968, Goldenberg (17) reported the fusion of human tumor and normal animal host cells in vivo. In 1970, a polykaryocyte was discovered (18) and Goldenberg et al (19) reported evidence of the fusion of transplanted human cancer cells with normal hamster cells in vivo. In 1984, Klein et al (20) reported the spontaneous fusion of mouse melanoma cells in vitro. In 1994, Lapidot et al (21) reported the generation of cancer stem cells from mouse cell fusion in vivo. In 1995, Gibson et al (22) observed spontaneous mouse heterotypic cell fusion in vivo, and in 2013, Goldenberg et al (23) reported cell-cell fusion of human lymphoma and rodent host cells in vivo.

Table I.

Milestones in cell-cell fusion.

Table I.

Milestones in cell-cell fusion.

Author(s), yearCell-cell fusion(Refs.)
Forkner, 1930Pulmonary tuberculosis, smallpox, varicella, measles, and rabbit homotypic cell fusion in vivo(11)
Enders and Peebles, 1954Human multinucleated giant cells or syncytia were formed in vitro(12)
Barski, 1961Somatic cell fusion was observed in tissue culture(13)
Furusawa and Cutting, 1962A hemagglutinating virus induced mouse Ehrlick ascites tumor cell fusion in vitro(14)
Cascardo and Karzon, 1965Inactivated virus induced human cell fusion in vitro(15)
Harris and Watkins, 1965Cell fusion between human and mouse cells in vitro(16)
Goldenberg, 1968Human tumor and normal animal cell fusion in vivo(17)
Poste, 1970Polykaryocyte was found(18)
Goldenberg et al, 1974Fusion between transplanted human cancer cells and normal hamster cells in vivo(19)
Klein et al, 1984Spontaneous fusion between mouse melanoma cells in vitro(20)
Lapidot et al, 1994Cancer stem cells generated by mouse cell fusion in vivo(21)
Gibson et al, 1995Spontaneous mouse heterotypic cell fusion in vivo(22)
Mohler et al, 2002The gene eff-1 was essential for developmental cell fusion(9)
Goldenberg et al, 2013Cell fusion between human lymphoma and rodent cells in vivo(23)

Table II.

More recent important discoveries in cell-cell fusion.

Table II.

More recent important discoveries in cell-cell fusion.

Author(s), yearSpeciesCell 1Cell 2EvidenceFunctionMechanism(Refs.)
Kato et al, 2016MouseEggSpermSperm-egg fusion assayFertilization sperm-egg fusion 1 and egg IZUMO1 receptor, JUNOSperm Izumo(24)
Smith et al, 2017YeastYeastYeastYeast mating assaysFertilizationCell division cycle 42 pseudogene 1-Fus2p interaction(25)
Yang et al, 2017C. elegansSeam cellHyp7 cellLive cell imaging in C. elegans embryo and larvae Spectraplakin links EFF-1 to the actin cytoskeleton(26)
Di Gioia et al, 2017Human and mouseHuman myoblastMouse C2C12 cellsCell fusion assay in vitro and allelic comple mentation in vivoCarey-Fineman-Ziter syndromeMyomaker, myoblast fusion factor(27)
Lee et al, 2017Human and hamsterHuman embryo kidney 293T cellsHamster ovary K1 cellsWestern blot, immunofluores cence staining, flow cytometry Lipid raft-associated stomatin(28)

In recent years, increasing evidence of cell-cell fusion and their underlying mechanisms have been reported (Table II; Fig. 1). In 2016, the interaction between the sperm protein Izumo sperm-egg fusion 1 and egg the protein IZUMO1 receptor, JUNO was revealed to mediate mouse fertilization (24). In 2017, cell-cell fusion was demonstrated to be mediated by cell division cycle 42 pseudogene 1-Fus2p and spectraplakin-EFF-1 interactions in yeast and C. elegans, respectively (25,26). In 2017, the myomaker, a myoblast fusion actor gene, was reported to be involved in cell-cell fusion, leading to Carey-Fineman-Ziter syndrome (27), and in 2017, lipid raft-associated stomatin was reported to form a molecular assembly that promoted membrane fusion (28).

Cell-cell fusion in vitro in cancer

A tumor is formed by the continuous proliferation of transformed cells, and may progress to become more carcinogenic through continuous evolution. Abnormal proliferation of non-physiological fusion cells in multicellular organisms may be one of the causes of tumor formation and progression. There is a considerable body of knowledge supporting the occurrence of spontaneous cell-cell fusion in vitro in cell cultures between cancerous and other cell types as demonstrated in Table III and Fig. 2. These studies have investigated the fusion of cancer cells with endothelial cells, BMDCs and epithelial cells.

Table III.

Cell-cell fusion between cancer and other cells in vitro.

Table III.

Cell-cell fusion between cancer and other cells in vitro.

Author(s), yearSpeciesCancerOther cell involvedMethodMechanism(Refs.)
Mortensen et al, 2004HumanBreast cancerEndothelial cellsCell culture, ICC, FISH.None(29)
Song et al, 2014HumanOral cancerEndothelial cellsCell fusion assays, block assay, IHC, ICC, FCTNF-α, VCAM-1/VLA-4(30)
Rappa et al, 2012HumanBreast cancerStromal cellsViral vectors, wound-healing assay, invasion assays, implantation, gene expression, FM.None(33)
Dittmar et al, 2011HumanBreast cancerEpithelial cellsCell co-culture, short-tandem-repeat analysis, RT-qPCR, FC, cytotoxicity assayNone(65)
Ozel et al, 2012HumanBreast cancerEpithelial cellsFC, cell migration, WBAKT, RAF-1-MAPKp42/44(66)
Mortensen et al, 2004HumanBreast cancerEpithelial cellsA fluorescence double reporter vector, Cre transduction, blocking experimentsTNF-α, hypoxia(29)
Bhatia et al, 2008HumanProstate cancerEpithelial cellsRetroviral vector, prospective cell-fusion, tumorigenicity assay, WT, IF, RT-qPCRp16, p53, hTERT(68)
He et al, 2016HumanOvarian cancerEmbryonic stem cellFusion experiment, RT-PCR, WT, mouse model, cell growthSuppressing p53 and PTEN(51)
Wang et al, 2016HumanHepatocellular carcinomaStem cellsSingle-cell fusion technique, RT-qPCR, FC, tumorigenicity assayUnknown(34)
Noubissi et al, 2015HumanBreast cancerMSCsBiFC, coculture experiments, apoptotic and hypoxic treatment, annexin V apoptosis assayHypoxia-induced apoptosis stimulates fusion(35)
Melzer et al, 2019HumanBreast cancerMSCsCell culture, FC, cell cycle analysis, RT-qPCR, mass spectrometryActin cytoskeletal components(37)
Wang et al, 2012HumanEsophageal carcinomaUmbilical cord, MSCsXenograft assays, transfection, WTDUSP6/MKP3 increased MAPK(52)
Fan and Lu, 2014HumanEsophageal carcinomaBone hemopoietic stem cellsCell fusion experimentNone(53)
Xu et al, 2014HumanLung cancer (HCC827)MSCsCo-culture, migration and invasion assays, FC and cell sorting of heterotypic hybrids, IF, RT-qPCREMT increased stemness of tumorigenic hybrids.(38)
Sun et al, 2015HumanGliomaBMSCsTube formation assay of the fused cells, ICC, IHCNone(40)
Li et al, 2014HumanLiver cancerBMSCsCell culture, chromosome analysis, cell invasion and migration assays, WTNone(42)
Yin et al, 2020HumanProstate cancerNeural stem cellNeural differentiation, Cell proliferation in 3-D, species-specific PCR, WB, IFTumor cell heterogeneity(43)
Wang et al, 2017HumanMelanomaMacrophagesPolyethylene glycol induced fusionNone(44)
Ding et al, 2012HumanBreast cancerMacrophagesIHC, cell tracker dye staining, PEG-mediated cell fusion, mammosphere formation assay, FCNone(46)
Chakraborty et al, 2001HumanMelanomaMacrophages GnT-V and β1,6-branching enhanced in glycoproteins(47)
Kemény et al, 2016; Kurgyis et al, 2016HumanMelanomaMacrophagesCell culture, detection of spontaneous cell fusion, fluorescent live cell imaging, IFNone(48,49)
Xue et al, 2015HumanGastric cancerMSCsThe hybrids were generated by using PEG1500, RT-PCR, IFNone(39)
Lindström et al, 2017HumanBreast cancerMacrophagesCell culture, cell fusion, radiation, clonogenic assayNone(50)
Gauck et al, 2017HumanBreast cancerBreast epithelial cell Mammosphere-formation, karyotype analysis, cell morphology, cell migrationNone(67)
Liu et al, 2018HumanCervical cancerT effector cellsCell fusion assay and RT-qPCRmicroRNA-181 enhances HeV F-/G-mediated cell fusion(94)
Song et al, 2012HumanOral squamousEndothelial cellsCell co-culture, RT-qPCR, IHC, block and carcinomaWnt/β-catenin- syncytin-1 enhance assay contributed to TNF-α-enhanced fusion(32)
Yoo et al, 2010HumanMel-DSP2 cellCHO-DSP1 cellStable reporter fusion assay, syncytium morphology assay, confocal microscopy, immunoprecipitation, WTVZV gB/gH-gL mediated cell-cell fusion(55)
Kawada et al, 2003HumanCervical cancer293T cellsCell-cell fusion assays, pseudotyped virus entry assay, crystallization and structure determinationHexamer-of-trimer interfaces enhance cell-cell fusion(56)
Chakraborty et al, 2000HamsterMelanomaCHO-K1 Cre cellsCell-cell fusion assay, quantitative cre reporter assaygB modulated cell fusion via an ITIM-mediated Y881 phosphorylation(70)
Melzer et al, 2018MouseBreast cancerMSCCell-cell fusion assayNone(36)
Zhang et al, 2019MouseLung cancerMSCsKaryotyping, RT-qPCR, WB, cell proliferation, colony formation, DNA ploidy, wound healing, Transwell migration and invasion, xenograft, ICCCancer metastasis and cancer stem cell features(41)
Hu et al, 2017MouseHepatocellular carcinomaDendritic cellsIF, ELISA, FC, histology, IHCMIP-10 alleviated immunosuppressive tumor environment(64)
Zhang et al, 2019MurineBreast cancerMacrophageCell fusion, RT-qPCR, WB, CCK-8 assayCancer proliferation, migration and invasion(45)

[i] FC, flow cytometry; FISH, fluorescent in situ hybridization; IF, immunofluorescence; FM, fluorescence microscopy; ICC, immunocytochemistry; TNF-α, tumor necrosis factor-α; WB, western blotting; CCK-8, Cell Counting Kit-8; RT-qPCR, reverse transcription-quantitative PCR; MSC, mesenchymal stem cell; BMSC, bone marrow-derived stem cell.

Endothelial cells line the inner side of blood and lymphatic vessels, and cancer cells must cross this barrier to gain access to the circulation, and cross again to exit and metastasize. Fusions between cancerous and endothelial cells were revealed to occur in vitro in co-cultures of human breast cancer cells and endothelial cells (29). These observations demonstrated a novel type of cancer-endothelial cell interaction, which may be of fundamental importance in the process of metastasis (29). Song et al (30) demonstrated that the oral cancer cell line SCC9 could spontaneously fuse with co-cultured endothelial cells, and the resultant hybrid cells exhibited continuous division and proliferation following re-plating and thawing. Such hybrids express markers of both of the parental cells, and undergo nuclear fusion, resulting in the acquisition of novel properties, enhanced drug resistance and improved survival potential. The hybrid cells comprised a significant portion of the tumor composition as demonstrated by immunostaining and FISH analysis, even though the hybrid cells and SCC9 cells were inoculated with a ratio of 1:10,000 cells (30). These experimental findings provided further evidence supporting the hypothesis that cell fusion may be involved in cancer progression (31,32).

Human BMDCs, including embryonic stem cells (ESCs), hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), macrophages and dendritic cells (DCs) have been reported to fuse in vitro with various types of cancer cells spontaneously (Table III; Fig. 2). Spontaneous in vitro formation of heterotypic hybrids was revealed to occur between human bone marrow-derived multipotent stromal cells and two different breast cancer cell lines, MDA-MB-231 and MA11 cells (33). The resultant fused cells formed of hepatocellular carcinoma cells and hESCs, expressed both cancer and stemness markers and exhibited increased drug resistance and enhanced tumorigenesis (34). MSCs and breast cancer cell fusion resulted in hybrids with enhanced migratory capacity, which promoted breast cancer metastasis (35,36). In 2019, it was demonstrated that actin cytoskeletal components served an important role in the cell fusion between breast cancer cells and MSCs (37). Cell fusion between lung cancer cells and MSCs provided a non-mutation-dependent mechanism that contributed to the aberrant gene expression patterns, and gave rise to highly malignant subpopulations with epithelial-mesenchymal transition (EMT) and TSC-like properties (38). Cell fusion between hMSCs and gastric cancer cells may contribute to the generation of tumorigenic hybrids, with EMT and TSC-like properties (39). The spontaneous in vitro fusion of mouse hMSCs and human SU3 glioma stem/progenitor cells is one of the driving factors for glioma neovascularization (40). Cell fusion between MSCs and lung cancer cells enhanced the metastatic capacity and characteristics of cancer stem cells by undergoing EMT (41). The hybrid cells that were formed of human liver cancer cells and mouse MSCs exhibited increased expression of E-cadherin, vimentin, twist, snail, and matrix metalloproteinase 2 and 9, were aneuploid, possessed enhanced invasive and migratory capacities and generated an increased number of metastatic liver and lung lesions (42). In 2020, prostate cancer cells were revealed to exhibit characteristics associated with neuroendocrine function and heterogeneity following fusion with bystander neural stem cells in the tumor microenvironment (43). Macrophages serve an important role during the development of cancer, such as in breast cancer and melanoma (44). Macrophage-breast cancer cell hybrids become more proliferative and invasive as they undergo EMT and following increased activity of the Wnt/β-catenin signaling pathway (45), and may also acquire TSC properties (46). Fusion between cancer cells and macrophages generates metastatic hybrids with genetic and phenotypic characteristics of both maternal cells. Fusion hybrids of macrophages and melanoma cells exhibited upregulated expression of N-acetylglucosaminyltransferase V, β1-6 branching and were metastatic (47). Melanoma-peritumoral stromal cell fusion may assist in explaining the high rate of recurrence of melanomas in patients following removal of the primary tumors (48,49). Macrophage-cancer cell fusion was reported to generate a subpopulation of radiotherapy-resistant cells with enhanced DNA-repair capacity (50).

However, it is not always the case that fusion cells will exhibit increased tumorigenicity or TSC-like properties. He et al (51) reported in 2017 that hESCs and ovarian cancer cells can fuse in vitro spontaneously, and the fused cells interestingly exhibited epigenetic changes that led to inhibition of growth, which may provide a novel direction for the treatment of ovarian cancer. Although cell fusion between BMDCs and somatic cells may be the origin of TSCs, the hybrid cells that form as a result of the fusion of human HSCs and esophageal carcinoma cells did not generate esophageal TSCs (52,53). DC-cancer cell fusion vaccines are an attractive modality for the treatment of several types of cancers, such as prostate, liver, gastric, colorectal, lung and breast cancer (5463). The cytotoxic T chemokine interferon-induced protein-10 was demonstrated to enhance the antitumor effects of DC/tumor cell fusion vaccines by alleviating the immunosuppressive tumor environment (64).

In addition, cancer cells can fuse with normal epithelial cells. The hybrid cells derived from the spontaneous fusion between the breast epithelial cell line M13SV1-EGFP-Neo and two breast cancer cell lines, HS578T-Hyg and MDA-MB-435-Hyg, both exhibited increased migratory capacity and increased drug resistance towards chemotherapeutic drugs, such as doxorubicin and paclitaxel. This finding further supported the hypothesis that cell fusion may give rise to drug resistant and metastatic cells (65). Human breast cancer cells and breast epithelial cell fusion was observed and verified using a Cre-loxP-based double fluorescence reporter system (35,66). The fusion between human breast epithelial cells and breast cancer cells gave rise to hybrid cells that possessed certain TSC or tumor initiating cell-like properties, indicating that cell fusion may be a mechanism underlying how tumor cells come to acquire a TSC phenotype (67). Additionally, the fusion of senescent human prostate epithelial cells and cancer cells was reported to promote tumor development in prostate cancer (68).

Cell-cell fusion in vivo in cancer

Tumor cells may fuse with several different types of cells, including stromal cells, epithelial cells and endothelial cells in vivo. Cell-cell fusion in vivo provides more convincing evidence of the involvement of this process in cancer development and progression than cell-cell fusion in vitro. However, providing direct evidence of cell-cell fusion at the DNA level is considerably more difficult, particularly for human cell-human cell fusions in vivo. There are >30 reports of cell-cell fusions in vivo between tumor cells and normal cells, in most of which, macrophages or other BMDCs are a component cell of the fusion (Table IV; Fig. 2) (50). These reports primarily revealed cell fusion between mouse-mouse cells or human-mouse cells, with only a few reports demonstrating fusion between human-human cells.

Table IV.

Cell-cell fusion between cancerous cells and other cells in vivo.

Table IV.

Cell-cell fusion between cancerous cells and other cells in vivo.

Author(s), yearSpeciesPartner cellCancer cellMethodMechanisms(Refs.)
Goldenberg et al, 2013HumanStromaLymphomaBMT, FISH, PCR, IHCTumor heterogeneity and progression(23)
Pawelek and Chakraborty, 2008; Harkness et al, 2013HumanBMDCsRenal cell carcinomaBMTTumor metastasis and recurrence(76,77)
Lazova et al, 2013; LaBerge et al, 2017HumanBMDCsMelanomaBMT, PCR, forensic genetic analyses of STR loci, allelic stutterTumor metastasis(80,81)
Andersen et al, 2007HumanOsteoclastsMyelomaCell culture, histology, TUNEL assay, microscopy, BrdU labellingNone(82)
Chakraborty et al, 2004HumanHematopoietic stem cellsRenal cell carcinomaAllogeneic liver and BMTCancer progression(78)
Clawson et al, 2015HumanMacrophagesMelanomaXenograft, IF, 3D confocal microscopy, live cell microscopyFusion cells at the periphery of primary tumors became metastasis initiating cells.(83)
Kurgyis et al, 2016HumanStromal cellsMelanomaLaser-capture microdissection and DNA mutationCXCR4, CD44(49)
Melzer et al, 2019HumanMSCsBreast cancerCell culture, mouse experiments, FC, RT-PCRTumor heterogeneity(84)
Martin-Padura et al, 2012Human and mouseMacrophages (mouse)Acute myeloid leukemia (Human)Mouse and human leukemia transplants, IF, PCR, IHC, FACSNone(74)
Goldenberg et al, 2012Human and hamsterStromal cells (hamster)Glioblastoma (Human)Transplantation, FISH, PCR, IHCNone(91)
Mortensen et al, 2004Human and mouseMouse endothelial cellsHuman breast cancerCell culture, FISH, IHCNone(29)
Chitwood et al, 2018Human and mouseMouse MSCsHuman breast cancerCell culture, RNA-Seq, Flox-luc mice, hematoxylin and eosin staining, IF, qPCRCancer proliferation and metastases(75)
Luo et al, 2016MouseBMSCsProstate cancerBMT, IHC, IF, RT-qPCR, FCNone(71)
Chakraborty et al, 2000MouseLungMelanomaMigration assay, FC of DNA content, histology, DNA sequencing, WTMetastasis(70)
Kerbel et al, 1983MouseMSCsSarcomasCell culture, chromosome analysis, serology, cytotoxic T-cell H-2 antigens typingMetastasis(69)
Jacobsen et al, 2006MouseStromaBreast cancerCell culture, IHC karyotyping, xenograftsNone(73)
Sun et al, 2019MouseBMSCGliomaA dual-color fluorescent protein tracer model, RT-qPCR, WB, ICC, IHC, tube formation, tumorigenicityAngiogenic effect(72)

[i] BMT, bone-marrow transplantation; FC, Flow cytometry; FISH, fluorescent in situ hybridization; IF, immunofluorescence; FM, Fluorescence microscopy; ICC, Immunocytochemistry; WB, western blotting; RT-qPCR, reverse transcription-quantitative PCR; MSC, mesenchymal stem cell; BMSC, bone marrow-derived stem cell; BMDC, bone marrow derived cell.

Mouse malignant cells were reported to fuse in vivo spontaneously with normal mouse cells. For example, spontaneous cell fusion in vivo was demonstrated between the mouse sarcoma cell line, MDW4, and normal mouse host cells, through the co-expression of their different major histocompatibility complex antigens in the fusion cells (69). In another example, mouse melanoma cells were revealed to fuse spontaneously in vivo with mouse host cells, and the fusion cells were indicated to serve an initiating mechanism for melanoma lung metastasis (70). A BALB/c nude mouse is an albino mouse with a tyrosine protein kinase homozygous mutation (c/c), which is a rate-limiting enzyme in the formation of melanin. Although the malignant melanoma cells transplanted into the mice were able to produce wild-type tyrosine kinase (C/C), the resulting tumors produced little or no melanin and became pigment-free. Although metastases occurred frequently in these mice, the tumors were small, had no pigment in the lungs and could be tolerated by the mice. In one mouse, however, a tumor that produced melanin was generated near the site of the implant, in the tail dermis. The tail of the mouse was cut off and was observed to ascertain if there were any distant metastases. After 5 weeks, the mice began to die, and there was considerable pigment transfer in the lungs. DNA analysis revealed that the metastatic cells had a C/c phenotype, indicating that they were fused and derived from the fusion of the transplanted tumor and host cells. The DNA content of the cells derived from the metastatic foci increased by 30–40%, chemotaxis was enhanced in vitro, and the activity and expression of tyrosinase was increased. Additionally, it also produced large melanin granules and exhibited autophagy, which included the formation of melanosomes (70). Histopathological analysis of the site of origin indicated that the mice exhibited macrophage infiltration, which may support the possibility of fusion between melanoma tumor cells and macrophages. Recently, mouse bone marrow MSC and mouse prostate cancer cell fusion in vivo was reported, which may serve a role in promoting cancer progression (71). In 2019, mouse MSCs were revealed to fuse with glioma stem cells, and the hybrids exhibited enhanced angiogenic effects compared with the parental glioma cells both in vivo and in vitro (72).

Human lymphoma cells were reported to fuse in vivo with hamster stromal cells, and this was one of the first reports of in vivo cell-cell fusion of human tumor cells with a rodent host cell, indicating that the horizontal transfer of tumor DNA to adjacent stromal cells may be implicated in tumor heterogeneity and progression. The hybrid xenografts had a gene signature of B-cell malignancy (23). Synkaryons were formed in the solid tumor by spontaneous fusion between the malignant human breast epithelium and the surrounding normal mouse stroma. The transformed hybrid cells were tumorigenic with histopathological features of malignancy, indicating a novel mechanism for tumor progression (73), and the breast cancer progressed with cancer cell heterogeneity and generated invasive and metastatic breast cancer cells within the populations of non-metastatic cells in the primary tumor. In addition, the fusion of human acute leukemia cells with rodent macrophages may be a mechanism of gene transfer for cancer dissemination, and the fused cells may be used to identify, as of yet, unrecognized leukemogenic genes that are conserved in the hybrid cells and are able to perpetuate leukemia in vivo (74). Human breast cancer cells spontaneously fuse with mouse endothelial cells resulting in viable and actively dividing hybrid cells, which exhibit an enhanced capability to traverse the endothelial barrier and metastasize (29). Human breast cancer cells were also revealed to fuse with mouse MSCs spontaneously in vivo, and a significantly higher number of hybrids resided in the metastatic tumors compared with the primary tumors, supporting the possibility that hybrids can emerge from the primary tumors and become metastatic (75).

However, due to the lack of specific DNA markers of both fusion partner cells, the direct evidence of human-human cell fusion in vivo remains lacking. Human-human cell fusion in vivo was reported between human cancer cells and human BMDCs (76,77). Studies have demonstrated the presence of donor cell genes in recipient malignant cells after bone marrow transplantation (BMT), supporting the possibility of donor-recipient cell fusion in vivo (77). In a previous study, donor DNA was detected in the recipient tumors by continued genetic analysis of renal cell carcinoma specimens from allogeneic BMT patients who developed secondary malignancies (78). Donor DNA was analyzed by laser capture microdissection of the tumor cells followed by PCR. In another study, patients receiving radiotherapy and immunosuppression prior to transplantation increased the likelihood of recurrence of the tumors and the donor BMDCs were found in the tumors of the patients (76). Other researchers discovered that early papillary renal cell carcinoma originated from male to female HSC transplantation, and showed trisomy 17 characteristics, which is common in early stage renal cell carcinoma and other types of tumors; ~1% of trisomy 17 of the tumor cells also contained a Y chromosome. It is worth noting that Y chromosome-containing and chromosome 17 paired tumor cells clustered in the tumor during mitotic anaphase. In addition, tumor cells containing the Y chromosome appeared in ~10% of the tumor cells, indicating clonal growth of these cells. As aforementioned, HSCs were associated with tumor cells. However, it is possible that the tumor cells originated from the donor HSCs alone, that no fusion had occurred, and the Y chromosome was lost during tumor growth and proliferation (79). In another similar study, the tumor cells containing the Y chromosome were revealed in two patients with intestinal cancer and one patient with lung cancer who had previously received a male HSC transplant. The presence of XXY or XXXY chromosome phenotypes detected by XY fluorescence in situ hybridization analysis supported the notion that the tumors originated from a cancer cell-BMDC fusion (77). The first and second pieces of convincing evidence of human cell-human cell fusions in vivo came from the detection of a short tandem repeat of parental cell alleles (80,81). Both donor and recipient DNA were detected in single cells of melanoma lymph-node and brain metastases from sex mismatched BMT female cancer patients.

Potential human-human cell fusion was reported in vivo between malignant cells and macrophages. Potential fusion cells may originate through spontaneous fusion in vivo between human myeloma cells and human osteoclasts, as supported by the presence of chromosomal translocations specific for the myeloma cells in the osteoclast nuclei of patients with myeloma (82). Osteoclast-myeloma hybrids reflect a previously unrecognized mechanism of bone destruction. Transcriptional activation of both malignant and normal nuclei was observed in the tumor-associated osteoclasts derived from the patients with melanoma. In these osteoclasts, 30% of the nuclei were derived from the malignant cells. In a previous study, potential fusion cells of human melanoma cells and human macrophages were reported in the peripheral blood of patients with cutaneous melanomas, and they possessed the ability to form metastatic lesions when transplanted into mice (83). The researchers isolated and cultured the circulating tumor cells from the patients with melanoma and termed them fusions of macrophages with tumor cells (MTFs). They discovered that MTFs exhibited a macrophage-like appearance, but contained melanosomes. MTFs also expressed pan-macrophage and M2-macrophage markers (such as CD14 and CD68, as well as CD163, CD204 and CD206, respectively), melanocyte-specific markers (activated leukocyte cell adhesion molecule and melan-A), epithelial biomarkers (keratin 1 and epithelial cell adhesion molecule), the pro-carcinogenic cytokine macrophage migration inhibitory factor, and cancer stem cell markers [C-X-C motif chemokine receptor 4 (CXCR4) and CD44]. They also demonstrated that 5×105 cultured human melanoma MTFs could induce the formation of metastatic tumors when subcutaneously injected into nude mice. The melanoma-derived BRAF (V600E) mutation was also detected in the micro-dissected peritumoral stromal cells of patients, indicating the occurrence of a potential in vivo fusion between human melanoma cells and human stromal cells (49). These potential hybrid cells display the phenotype of stromal cells and are therefore undetectable during routine histological assessments. In 2019, the in vivo fusion between human breast cancer cells and human MSCs was also found when co-injected in mice, and this fusion increased tumor heterogeneity (84).

Mechanisms of cell-cell fusion in cancer

Cell-cell fusion in cancer may involve several steps (Fig. 3). Here, MSCs are used as an example to illustrate the different steps of fusion with a cancer cell. The first step of cell-cell fusion includes the recruitment of an MSC to the tumor microenvironment through tumor-secreted cytokines, such as C-C motif chemokine ligand 2 and VEGF-C (8587). The MSC then undergoes polarization and acquires a competent phenotype, which is followed by the binding of the fusion partners, cell membrane and cytoplasm fusion and then nuclei fusion. During these processes, cytokines, such as β1,6-branched polylactosamines, CXCR4, CD44 and TNF-α serve important roles (8891). The fused cells may promote tumorigenesis, metastasis and drug resistance by releasing cytokines, such as IFN-γ, angiotensin, COX-2, IL-1β and S100A4 (92,93).

Importantly, in vitro studies on virus-cell fusion and cell-cell fusion between cancer and other cells have provided a tool to understand the mechanisms of cell-cell fusion. Glycoprotein B (gB) of VZV was reported to serve a role in cell-cell fusion (94). Strict regulation of VZV gB/gH-gL-mediated cell-cell fusion between Mel-DSP2 cells and CHO-DSP1 cells through gBcyt and gHcyt was revealed to be required for effective viral propagation (55). The identification of the role of the gB lysine cluster in cell-cell fusion regulation revealed the molecular mechanisms that govern VZV syncytium formation during infection (55). Hexamer-of-trimers assembly of gB was important during fusion pore formation in both cell-cell fusion and virus-cell fusion systems (56). gB-modulated melanoma and CHO-K1 cell fusion was mediated via a T-cell immunoreceptor with Ig and ITIM domain-mediated Y881 phosphorylation-dependent mechanism, supporting a unique concept that intracellular signaling through this gBcyt motif regulates VZV syncytia formation and is essential for skin cancer pathogenesis (57). MicroRNA-181 was demonstrated to suppress ephrin receptors that negatively regulate henipavirus glycoprotein-mediated cell-cell fusion (95). CXCR4 was identified as a key fusion gene involved in cell-cell fusion. Hu et al (96) reported that urine-derived stem cells could fuse with different types of liver cells by upregulating CXCR4 expression during liver tissue recovery, following injury. Fusions of human melanoma cells and human macrophages were reported in the peripheral blood of patients with cutaneous melanomas, and they possessed the ability to form metastatic lesions when transplanted into mice, as cultured human melanoma fusion cells could induce metastatic tumors when subcutaneously injected into nude mice (83). In addition, other signaling pathways, such as the Wnt/β-catenin pathway may serve a role in cell-cell fusion in cancer. The Wnt/β-catenin pathway activation-dependent upregulation of syncytin-1 was found to be involved in TNF-α-induced cell-cell fusion between oral cancer cells and endothelial cells (54). However, additional in vivo studies are required to determine the roles and mechanisms of cell fusion in tumor progression.

Conclusions

Cell-cell fusion in vitro is a recognized biological process, which occurs not only under physiological conditions, but also during tumorigenesis and tumor metastasis. In the present review, the important pro-tumorigenic and pro-metastatic roles of cell-cell fusion were discussed. It is hypothesized that cell-cell fusion is an important mechanism that enables tumor metastasis, and may be one of the primary causes of tumor metastasis in the majority of different types of cancer. In fact, cell-cell fusion has been targeted for cancer therapy; VSV-G-mediated neural stem cell-glioma cell fusion was induced in vivo as a form of glioma therapy (97). However, further probing the molecular and cellular mechanisms of cell fusion in the context of tumor progression may pave the way for the development of novel techniques for the treatment of cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (81872412 to XHW, 81602303 to XY, 31700736 to WXW), the National innovation and entrepreneurship training program for College Students (202010489017 to PXC), the Hubei Province Health and Family Planning Scientific Research Project (WJ2016-Y-10 to PXC), the Jingzhou Science and Technology Development Planning Project (JZKJ15063 to WXW), the Hubei Province Scientific and Technological Research Project (Q20171306 to XWW), the Hubei Province Natural Science Foundation of China (2017CFB786 to PXC, 2016CFB180 to WXW), and the Guangzhou Key Medical Discipline Construction Project (CSZ).

Availability of data and materials

Not applicable.

Authors' contributions

HWX designed and supervised the study. YFL, YYW, YYM and XQL reviewed the references. XCP and MZ wrote the manuscript. WQC, YZ, XWW and ZWM contributed to tables and figures. YYM, YX, LSZ, LMY and SZC revised the manuscript. HWX and XCP acquired funding. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Fonseca NA, Cruz AF, Moura V, Simões S and Moreira JN: The cancer stem cell phenotype as a determinant factor of the heterotypic nature of breast tumors. Crit Rev Oncol Hematol. 113:111–121. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Xin HW, Hari DM, Mullinax JE, Ambe CW, Koizumi T, Ray S, Anderson AJ, Wiegand GW, Garfield SH, Thorgeirsson SS and Avital I: Tumor-initiating label-retaining cancer cells in human gastrointestinal cancers undergo asymmetric cell division. Stem Cells. 30:591–598. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Xin HW, Ambe CM, Ray S, Kim BK, Koizumi T, Wiegand GW, Hari D, Mullinax JE, Jaiswal KR, Garfield SH, et al: Wnt and the cancer niche: Paracrine interactions with gastrointestinal cancer cells undergoing asymmetric cell division. J Cancer. 4:447–457. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Xin HW, Ambe CM, Miller TC, Chen JQ, Wiegand GW, Anderson AJ, Ray S, Mullinax JE, Hari DM, Koizumi T, et al: Liver label retaining cancer cells are relatively resistant to the reported anti-cancer stem cell drug metformin. J Cancer. 7:1142–11451. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Xin HW, Ambe CM, Hari DM, Wiegand GW, Miller TC, Chen JQ, Anderson AJ, Ray S, Mullinax JE, Koizumi T, et al: Label-retaining liver cancer cells are relatively resistant to sorafenib. Gut. 62:1777–1786. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Hari D, Xin HW, Jaiswal K, Wiegand G, Kim BK, Ambe C, Burka D, Koizumi T, Ray S, Garfield S, et al: Isolation of live label-retaining cells and cells undergoing asymmetric cell division via nonrandom chromosomal cosegregation from human cancers. Stem Cells Dev. 20:1649–1658. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Kiberstis PA: Micromanaging muscle cell fusion. Science. 356:280–281. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Wang J, Liu Q, Luo K, Chen X, Xiao J, Zhang C, Tao M, Zhao R and Liu S: Cell fusion as the formation mechanism of unreduced gametes in the gynogenetic diploid hybrid fish. Sci Rep. 6:316582016. View Article : Google Scholar : PubMed/NCBI

9 

Mohler WA, Shemer G, del Campo JJ, Valansi C, Opoku-Serebuoh E, Scranton V, Assaf N, White JG and Podbilewicz B: The type I membrane protein EFF-1 is essential for developmental cell fusion. Dev Cell. 2:355–362. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Shemer G, Suissa M, Kolotuev I, Nguyen KC, Hall DH and Podbilewicz B: EFF-1 is sufficient to initiate and execute tissue-specific cell fusion in C. elegans. Curr Biol. 14:1587–1591. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Forkner CE: The origin and fate of two types of multi-nucleated giant cells in the circulating blood. J Exp Med. 52:279–297. 1930. View Article : Google Scholar : PubMed/NCBI

12 

Enders JF and Peebles TC: Propagation in tissue cultures of cytopathogenic agents from patients with measles. Proc Soc Exp Biol Med. 86:277–286. 1954. View Article : Google Scholar : PubMed/NCBI

13 

Barski G: ‘Hybrid’ cell clones isolated from mixed cell cultures. C R Hebd Seances Acad Sci. 253:1186–1188. 1961.(In French). PubMed/NCBI

14 

Furusawa E and Cutting W: Loss of neurotropic pathogenicity and hemagglutinating property of Columbia SK virus by in vitro cultivation in sarcoma 180 ascites cells. Proc Soc Exp Biol Med. 109:417–421. 1962. View Article : Google Scholar : PubMed/NCBI

15 

Cascardo MR and Karzon DT: Measles virus giant cell induction factor (fusion factor). Virology. 26:311–325. 1965. View Article : Google Scholar : PubMed/NCBI

16 

Harris H and Watkins JF: Hybrid cells derived from mouse and man: Artificial heterokaryons of mammalian cells from different species. Nature. 205:640–646. 1965. View Article : Google Scholar : PubMed/NCBI

17 

Goldenberg DM: On the progression of malignancy: A hypothesis. Klin Wochenschr. 46:898–899. 1968.(In German). View Article : Google Scholar : PubMed/NCBI

18 

Poste G: Virus-induced polykaryocytosis and the mechanism of cell fusion. Adv Virus Res. 16:303–356. 1970. View Article : Google Scholar : PubMed/NCBI

19 

Goldenberg DM, Pavia RA and Tsao MC: In vivo hybridisation of human tumour and normal hamster cells. Nature. 250:649–651. 1974. View Article : Google Scholar : PubMed/NCBI

20 

Klein PA, Xiang JH and Kimura AK: Melanoma cells growing in aggregates on a non-adhesive poly(HEMA) substrate exhibit polykaryocytosis but do not develop an increased metastatic capability. Clin Exp Metastasis. 2:287–295. 1984. View Article : Google Scholar : PubMed/NCBI

21 

Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA and Dick JE: A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 367:645–648. 1994. View Article : Google Scholar : PubMed/NCBI

22 

Gibson AJ, Karasinski J, Relvas J, Moss J, Sherratt TG, Strong PN and Watt DJ: Dermal fibroblasts convert to a myogenic lineage in mdx mouse muscle. J Cell Sci. 108:207–214. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Goldenberg DM, Gold DV, Loo M, Liu D, Chang CH and Jaffe ES: Horizontal transmission of malignancy: In-vivo fusion of human lymphomas with hamster stroma produces tumors retaining human genes and lymphoid pathology. PLoS One. 8:e553242013. View Article : Google Scholar : PubMed/NCBI

24 

Kato K, Satouh Y, Nishimasu H, Kurabayashi A, Morita J, Fujihara Y, Oji A, Ishitani R, Ikawa M and Nureki O: Structural and functional insights into IZUMO1 recognition by JUNO in mammalian fertilization. Nat Commun. 7:121982016. View Article : Google Scholar : PubMed/NCBI

25 

Smith JA, Hall AE and Rose MD: Membrane curvature directs the localization of Cdc42p to novel foci required for cell-cell fusion. J Cell Biol. 216:3971–3980. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Yang Y, Zhang Y, Li WJ, Jiang Y, Zhu Z, Hu H, Li W, Wu JW, Wang ZX, Dong MQ, et al: Spectraplakin induces positive feedback between fusogens and the actin cytoskeleton to promote cell-cell fusion. Dev Cell. 41:107–120. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Di Gioia SA, Connors S, Matsunami N, Cannavino J, Rose MF, Gilette NM, Artoni P, de Macena Sobreira NL, Chan WM, Webb BD, et al: A defect in myoblast fusion underlies Carey-Fineman-Ziter syndrome. Nat Commun. 8:160772017. View Article : Google Scholar : PubMed/NCBI

28 

Lee JH, Hsieh CF, Liu HW, Chen CY, Wu SC, Chen TW, Hsu CS, Liao YH, Yang CY, Shyu JF, et al: Lipid raft-associated stomatin enhances cell fusion. FASEB J. 31:47–59. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Mortensen K, Lichtenberg J, Thomsen PD and Larsson LI: Spontaneous fusion between cancer cells and endothelial cells. Cell Mol Life Sci. 61:2125–2131. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Song K, Song Y, Zhao XP, Shen H, Wang M, Yan TL, Liu K and Shang ZJ: Oral cancer/endothelial cell fusion experiences nuclear fusion and acquisition of enhanced survival potential. Exp Cell Res. 328:156–163. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Raj AT, Kheur S, Patil VR and Gupta AA: Assessing the role of cell fusion in cancer metastasis. Oral Oncol. 90:124–125. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Song K, Zhu F, Zhang Hz and Shang Zj: Tumor necrosis factor-α enhanced fusions between oral squamous cell carcinoma cells andendothelial cells via VCAM-1/VLA-4 pathway. Exp Cell Res. 318:1707–1715. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Rappa G, Mercapide J and Lorico A: Spontaneous formation of tumorigenic hybrids between breast cancer and multipotent stromal cells is a source of tumor heterogeneity. Am J Pathol. 180:2504–2515. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Wang R, Chen S, Li C, Ng KTP, Kong Cw, Cheng J, Cheng SH, Li RA, Lo CM, Man K and Sun D: Fusion with stem cell makes the hepatocellular carcinoma cells similar to liver tumor--initiating cells. BMC Cancer. 16:562016. View Article : Google Scholar : PubMed/NCBI

35 

Noubissi FK, Harkness T, Alexander CM and Ogle BM: Apoptosis-induced cancer cell fusion: A mechanism of breast cancer metastasis. FASEB J. 29:4036–4045. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Melzer C, von der Ohe J and Hass R: Enhanced metastatic capacity of breast cancer cells after interaction and hybrid formation with mesenchymal stroma/stem cells (MSC). Cell Commun Signal. 16:22018. View Article : Google Scholar : PubMed/NCBI

37 

Melzer C, von der Ohe J and Hass R: Involvement of actin cytoskeletal components in breast cancer cell fusion with human mesenchymal stroma/stem-like cells. Int J Mol Sci. 20:8762019. View Article : Google Scholar : PubMed/NCBI

38 

Xu MH, Gao X, Luo D, Zhou XD, Xiong W and Liu GX: EMT and acquisition of stem cell-like properties are involved in spontaneous formation of tumorigenic hybrids between lung cancer and bone marrow-derived mesenchymal stem cells. PLoS One. 9:e878932014. View Article : Google Scholar : PubMed/NCBI

39 

Xue J, Zhu Y, Sun Z, Ji R, Zhang X, Xu W, Yuan X, Zhang B, Yan Y, Yin L, et al: Tumorigenic hybrids between mesenchymal stem cells and gastric cancer cells enhanced cancer proliferation, migration and stemness. BMC Cancer. 15:7932015. View Article : Google Scholar : PubMed/NCBI

40 

Sun C, Zhao D, Dai X, Chen J, Rong X, Wang H, Wang A, Li M, Dong J, Huang Q and Lan Q: Fusion of cancer stem cells and mesenchymal stem cells contributes to glioma neovascularization. Oncol Rep. 34:2022–2030. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Zhang LN, Kong CF, Zhao D, Cong XL, Wang SS, Ma L and Huang YH: Fusion with mesenchymal stem cells differentially affects tumorigenic and metastatic abilities of lung cancer cells. J Cell Physiol. 234:3570–3582. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Li H, Feng Z, Tsang TC, Tang T, Jia X, He X, Pennington ME, Badowski MS, Liu AKM, Chen D, et al: Fusion of HepG2 cells with mesenchymal stem cells increases cancer associated and malignant properties: An in vivo metastasis model. Oncol Rep. 32:539–547. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Yin L, Hu P, Shi X, Qian W, Zhau HE, Pandol SJ, Lewis MS, Chung LWK and Wang R: Cancer cell's neuroendocrine feature can be acquired through cell-cell fusion during cancer-neural stem cell interaction. Sci Rep. 10:12162020. View Article : Google Scholar : PubMed/NCBI

44 

Wang H, Yang L, Wang D, Zhang Q and Zhang L: Pro-tumor activities of macrohpages in the progression of melanoma. Hum Vaccin Immunother. 13:1556–1562. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Zhang LN, Huang YH and Zhao L: Fusion of macrophages promotes breast cancer cell proliferation, migration and invasion through activating epithelial-mesenchymal transition and Wnt/β-catenin signaling pathway. Arch Biochem Biophys. 676:1081372019. View Article : Google Scholar : PubMed/NCBI

46 

Ding J, Jin W, Chen C, Shao Z and Wu J: Tumor associated macrophage × cancer cell hybrids may acquire cancer stem cell properties in breast cancer. PLoS One. 7:e419422012. View Article : Google Scholar : PubMed/NCBI

47 

Chakraborty AK, Pawelek J, Ikeda Y, Miyoshi E, Kolesnikova N, Funasaka Y, Ichihashi M and Taniguchi N: Fusion hybrids with macrophage and melanoma cells up-regulate N-acetylglucosaminyltransferase V, beta1-6 branching, and metastasis. Cell Growth Differ. 12:623–630. 2001.PubMed/NCBI

48 

Kemény LV, Kurgyis Z, Buknicz T, Groma G, Jakab A, Zänker K, Dittmar T, Kemény L and Németh IB: Melanoma cells can adopt the phenotype of stromal fibroblasts and macrophages by spontaneous cell fusion in vitro. Int J Mol Sci. 17:8262016. View Article : Google Scholar

49 

Kurgyis Z, Kemény LV, Buknicz T, Groma G, Oláh J, Jakab A, Polyánka H, Zänker K, Dittmar T, Kemény L and Németh IB: Melanoma-Derived BRAF (V600E) mutation in peritumoral stromal cells: Implications for in vivo cell fusion. Int J Mol Sci. 17:9802016. View Article : Google Scholar : PubMed/NCBI

50 

Lindström A, Midtbö K, Arnesson LG, Garvin S and Shabo I: Fusion between M2-macrophages and cancer cells results in a subpopulation of radioresistant cells with enhanced DNA-repair capacity. Oncotarget. 8:51370–51386. 2017. View Article : Google Scholar

51 

He K, Qu H, Xu LN, Gao J, Cheng FY, Xiang P and Zhou CQ: Epigenetics changes caused by the fusion of human embryonic stem cell and ovarian cancer cells. Biosci Rep. 36:e003782016. View Article : Google Scholar : PubMed/NCBI

52 

Wang Y, Fan H, Zhou B, Ju Z, Yu L, Guo L, Han J and Lu S: Fusion of human umbilical cord mesenchymal stem cells with esophageal carcinoma cells inhibits the tumorigenicity of esophageal carcinoma cells. Int J Oncol. 40:370–377. 2012.PubMed/NCBI

53 

Fan H and Lu S: Fusion of human bone hemopoietic stem cell with esophageal carcinoma cells didn't generate esophageal cancer stem cell. Neoplasma. 61:540–545. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Kim TB, Park HK, Chang JH, Choi IH, Kim KH, Yoon SJ, Lee MS, Jung H and Kim CS: The establishment of dendritic cell-tumor fusion vaccines for hormone refractory prostate cancer cell. Korean J Urol. 51:139–144. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Yoo C, Do HA, Jeong IG, Park H, Hwang JJ, Hong JH, Cho JS, Choo MS, Ahn H and Kim CS: Efficacy of dendritic cells matured early with OK-432 (Picibanil), prostaglandin E2, and interferon-alpha as a vaccine for a hormone refractory prostate cancer cell line. J Korean Med Sci. 25:1284–1290. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Kawada M, Ikeda H, Takahashi T, AIshizu A, Ishikura H, Katoh H and Yoshiki T: Vaccination of fusion cells of rat dendritic and carcinoma cells prevents tumor growth in vivo. Int J Cancer. 105:520–526. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Matsumoto S, Saito H, Tsujitani S and Ikeguchi M: Allogeneic gastric cancer cell-dendritic cell hybrids induce tumor antigen (carcinoembryonic antigen) specific CD8(+) T cells. Cancer Immunol Immunother. 55:131–139. 2006. View Article : Google Scholar : PubMed/NCBI

58 

Koido S, Hara E, Homma S, Torii A, Toyama Y, Kawahara H, Watanabe M, Yanaga K, Fujise K, Tajiri H, et al: Dendritic cells fused with allogeneic colorectal cancer cell line present multiple colorectal cancer-specific antigens and induce antitumor immunity against autologous tumor cells. Clin Cancer Res. 11:7891–7900. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Zhang K, Gao PF, Yu PW, Rao Y and Zhou LX: Study on biological characters of SGC7901 gastric cancer cell-dendritic cell fusion vaccines. World J Gastroenterol. 12:3438–3441. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Imura K, Ueda Y, Hayashi T, Itoh T, Shimizu K, Tamai H, Yano Y, Naito K, Kohara J, Nakane K, et al: Induction of cytotoxic T lymphocytes against human cancer cell lines using dendritic cell-tumor cell hybrids generated by a newly developed electrofusion technique. Int J Oncol. 29:531–539. 2006.PubMed/NCBI

61 

Zhang Y, Ma B, Zhou Y, Zhang M, Qiu X, Sui Y, Zhang X, Ma B and Fan Q: Dendritic cells fused with allogeneic breast cancer cell line induce tumor antigen-specific CTL responses against autologous breast cancer cells. Breast Cancer Res Treat. 105:277–286. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Koido S, Tanaka Y, Tajiri H and Gong J: Generation and functional assessment of antigen-specific T cells stimulated by fusions of dendritic cells and allogeneic breast cancer cells. Vaccine. 25:2610–2619. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Serhal K, Baillou C, Ghinea N, Fontanges P, Dupuy FP, Lemoine FM and Lacave R: Characteristics of hybrid cells obtained by dendritic cell/tumour cell fusion in a T-47D breast cancer cell line model indicate their potential as anti-tumour vaccines. Int J Oncol. 31:1357–1365. 2007.PubMed/NCBI

64 

Hu Z, Chen J, Zhou S, Yang N, Duan S, Zhang Z, Su J, He J, Zhang Z, Lu X and Zhao Y: Mouse IP-10 gene delivered by folate-modified chitosan nanoparticles and dendritic/tumor cells fusion vaccine effectively inhibit the growth of hepatocellular carcinoma in mice. Theranostics. 7:1942–1952. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Dittmar T, Schwitalla S, Seidel J, Haverkampf S, Reith G, Meyer-Staeckling S, Brandt BH, Niggemann B and Zänker KS: Characterization of hybrid cells derived from spontaneous fusion events between breast epithelial cells exhibiting stem-like characteristics and breast cancer cells. Clin Exp Metastasis. 28:75–90. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Ozel C, Seidel J, Meyer-Staeckling S, Brandt BH, Niggemann B, Zänker KS and Dittmar T: Hybrid cells derived from breast epithelial cell/breast cancer cell fusion events show a differential RAF-AKT crosstalk. Cell Commun Signal. 10:102012. View Article : Google Scholar : PubMed/NCBI

67 

Gauck D, Keil S, Niggemann B, Zänker KS and Dittmar T: Hybrid clone cells derived from human breast epithelial cells and human breast cancer cells exhibit properties of cancer stem/initiating cells. BMC Cancer. 17:5152017. View Article : Google Scholar : PubMed/NCBI

68 

Bhatia B, Multani AS, Patrawala L, Chen X, Calhoun-Davis T, Zhou J, Schroeder L, Schneider-Broussard R, Shen J, Pathak S, et al: Evidence that senescent human prostate epithelial cells enhance tumorigenicity: Cell fusion as a potential mechanism and inhibition by p16INK4a and hTERT. Int J Cancer. 122:1483–1495. 2008. View Article : Google Scholar : PubMed/NCBI

69 

Kerbel RS, Lagarde AE, Dennis JW and Donaghue TP: Spontaneous fusion in vivo between normal host and tumor cells: Possible contribution to tumor progression and metastasis studied with a lectin-resistant mutant tumor. Mol Cell Biol. 3:523–538. 1983. View Article : Google Scholar : PubMed/NCBI

70 

Chakraborty AK, Sodi S, Rachkovsky M, Kolesnikova N, Platt JT, Bolognia JL and Pawelek JM: A spontaneous murine melanoma lung metastasis comprised of host × tumor hybrids. Cancer Res. 60:2512–2519. 2000.PubMed/NCBI

71 

Luo F, Liu T, Wang J, Li J, Ma P, Ding H, Feng G, Lin D, Xu Y and Yang K: Bone marrow mesenchymal stem cells participate in prostate carcinogenesis and promote growth of prostate cancer by cell fusion in vivo. Oncotarget. 7:30924–30934. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Sun C, Dai X, Zhao D, Wang H, Rong X, Huang Q and Lan Q: Mesenchymal stem cells promote glioma neovascularization in vivo by fusing with cancer stem cells. BMC Cancer. 19:12402019. View Article : Google Scholar : PubMed/NCBI

73 

Jacobsen BM, Harrell JC, Jedlicka P, Borges VF, Varella-Garcia M and Horwitz KB: Spontaneous fusion with, and transformation of mouse stroma by, malignant human breast cancer epithelium. Cancer Res. 66:8274–8279. 2006. View Article : Google Scholar : PubMed/NCBI

74 

Martin-Padura I, Marighetti P, Gregato G, Agliano A, Malazzi O, Mancuso P, Pruneri G, Viale A and Bertolini F: Spontaneous cell fusion of acute leukemia cells and macrophages observed in cells with leukemic potential. Neoplasia. 14:1057–1066. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Chitwood CA, Dietzsch C, Jacobs G, McArdle T, Freeman BT, Banga A, Noubissi FK and Ogle BM: Breast tumor cell hybrids form spontaneously in vivo and contribute to breast tumor metastases. APL Bioeng. 2:0319072018. View Article : Google Scholar : PubMed/NCBI

76 

Pawelek JM and Chakraborty AK: The cancer cell-leukocyte fusion theory of metastasis. Adv Cancer Res. 101:397–444. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Harkness T, Weaver BA, Alexander CM and Ogle BM: Cell fusion in tumor development: Accelerated genetic evolution. Crit Rev Oncog. 18:19–42. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Chakraborty A, Lazova R, Davies S, Bäckvall H, Ponten F, Brash D and Pawelek J: Donor DNA in a renal cell carcinoma metastasis from a bone marrow transplant recipient. Bone Marrow Transplant. 34:183–186. 2004. View Article : Google Scholar : PubMed/NCBI

79 

Yilmaz Y, Lazova R, Qumsiyeh M, Cooper D, Pawelek J and Donor Y: Chromosome in renal carcinoma cells of a female BMT recipient: Visualization of putative BMT-tumor hybrids by FISH. Bone Marrow Transplant. 35:1021–1024. 2005. View Article : Google Scholar : PubMed/NCBI

80 

Lazova R, Laberge GS, Duvall E, Spoelstra N, Klump V, Sznol M, Cooper D, Spritz RA, Chang JT and Pawelek JM: A melanoma brain metastasis with a donor-patient hybrid genome following bone marrow transplantation: First evidence for fusion in human cancer. PLoS One. 8:e667312013. View Article : Google Scholar : PubMed/NCBI

81 

LaBerge GS, Duvall E, Grasmick Z, Haedicke K and Pawelek J: A melanoma lymphnode metastasis with a donor-patient hybrid genome following bone marrow transplantation: A second case of leucocyte-tumor cell hybridization in cancer metastasis. PLoS One. 12:e01685812017. View Article : Google Scholar : PubMed/NCBI

82 

Andersen TL, Boissy P, Sondergaard TE, Kupisiewicz K, Plesner T, Rasmussen T, Haaber J, Kølvraa S and Delaissé JM: Osteoclast nuclei of myeloma patients show chromosome translocations specific for the myeloma cell clone: A new type of cancer-host partnership? J Pathol. 211:10–17. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Clawson GA, Matters GL, Xin P, Imamura-Kawasawa Y, Du Z, Thiboutot DM, Helm KF, Neves RI and Abraham T: Macrophage-tumor cell fusions from peripheral blood of melanoma patients. PLoS One. 10:e01343202015. View Article : Google Scholar : PubMed/NCBI

84 

Melzer C, von der Ohe J and Hass R: In vivo cell fusion between mesenchymal stroma/stem-like cells and breast cancer cells. Cancers (Basel). 110:1852019. View Article : Google Scholar

85 

Hong S, Zhang P, Zhang H, Jia L, Qu X, Yang Q, Rong F and Kong B: Enforced effect of tk-MCP-1 fusion gene in ovarian cancer. J Exp Clin Cancer Res. 31:742012. View Article : Google Scholar : PubMed/NCBI

86 

Zhang D, Li B, Shi J, Zhao L, Zhang X, Wang C, Hou S, Qian W, Kou G, Wang H and Guo Y: Suppression of tumor growth and metastasis by simultaneously blocking vascular endothelial growth factor (VEGF)-A and VEGF-C with a receptor-immunoglobulin fusion protein. Cancer Res. 70:2495–2503. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Tammela T, Zarkada G, Nurmi H, Jakobsson L, Heinolainen K, Tvorogov D, Zheng W, FrancoC A, Murtomäki A, Aranda E, et al: VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing Notch signalling. Nat Cell Biol. 13:1202–1213. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Liang AL, Qian HL, Zhang TT, Zhou N, Wang HJ, Men XT, Qi W, Zhang PP, Fu M, Liang X, et al: Bifunctional fused polypeptide inhibits the growth and metastasis of breast cancer. Drug Des Devel Ther. 9:5671–5686. 2015.PubMed/NCBI

89 

Beha N, Harder M, Ring S, Kontermann RE and Müller D: IL15-based trifunctional antibody-fusion proteins with costimulatory TNF-superfamily ligands in the single-chain format for cancer immunotherapy. Mol Cancer Ther. 18:1278–1288. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Weiler J and Dittmar T: Minocycline impairs TNF-α-induced cell fusion of M13SV1-Cre cells with MDA-MB-435-pFDR1 cells by suppressing NF-κB transcriptional activity and its induction of target-gene expression of fusion-relevant factors. Cell Commun Signal. 17:712019. View Article : Google Scholar : PubMed/NCBI

91 

Goldenberg DM, Zagzag D, Heselmeyer-Haddad KM, Garcia LYB, Ried T, Loo M, Chang CH and Gold DV: Horizontal transmission and retention of malignancy, as well as functional human genes, after spontaneous fusion of human glioblastoma and hamster host cells in vivo. Int J Cancer. 131:49–58. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Lee SH, Lee YP, Kim SY, Jeong MS, Lee MJ, Kang HW, Jeong HJ, Kim DW, Sohn EJ, Jang SH, et al: Inhibition of LPS-induced cyclooxygenase 2 and nitric oxide production by transduced PEP-1-PTEN fusion protein in raw 264.7 macrophage cells. Exp Mol Med. 40:629–638. 2008. View Article : Google Scholar : PubMed/NCBI

93 

Wolf S, Haase-Kohn C, Lenk J, Hoppmann S, Bergmann R, Steinbach J and Pietzsch J: Expression, purification and fluorine-18 radiolabeling of recombinant S100A4: A potential probe for molecular imaging of receptor for advanced glycation endproducts in vivo? Amino Acids. 41:809–820. 2011. View Article : Google Scholar : PubMed/NCBI

94 

Liu XQ, Xin HY, Lyu YN, Ma ZW, Peng XC, Xiang Y, Wang YY, Wu ZJ, Cheng JT, Ji JF, et al: Oncolytic herpes simplex virus tumor targeting and neutralization escape by engineering viral envelope glycoproteins. Drug Deliv. 25:1950–1962. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Foo CH, Rootes CL, Cowley K, Marsh GA, Gould CM, Deffrasnes C, Cowled CJ, Klein R, Riddell SJ and Middleton D: Dual microRNA screens reveal that the immune-responsive miR-181 promotes henipavirus entry and cell-cell fusion. PLoS Pathog. 12:e10059742016. View Article : Google Scholar : PubMed/NCBI

96 

Hu C, He Y, Liu D, Zhao L, Fang S, Tan B, Dong S, Wang Y, He T and Bi Y: Hypoxia preconditioning promotes the proliferation and migration of urine-derived stem cells in chronically injured liver of mice by upregulating CXCR4. Stem Cells Dev. 15:526–536. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Luo Y, Zhu D, Lam DH, Huang J, Tang Y, Luo X and Wang S: A double-switch cell fusion-inducible transgene expression system for neural stem cell-based antiglioma gene therapy. Stem Cells Int. 2015:6490802015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2021
Volume 46 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Peng X, Zhang M, Meng Y, Liang Y, Wang Y, Liu X, Cai W, Zhou Y, Wang X, Ma Z, Ma Z, et al: Cell‑cell fusion as an important mechanism of tumor metastasis (Review). Oncol Rep 46: 145, 2021
APA
Peng, X., Zhang, M., Meng, Y., Liang, Y., Wang, Y., Liu, X. ... Xin, H. (2021). Cell‑cell fusion as an important mechanism of tumor metastasis (Review). Oncology Reports, 46, 145. https://doi.org/10.3892/or.2021.8096
MLA
Peng, X., Zhang, M., Meng, Y., Liang, Y., Wang, Y., Liu, X., Cai, W., Zhou, Y., Wang, X., Ma, Z., Xiang, Y., Zeng, L., Cui, S., Yang, L., Xin, H."Cell‑cell fusion as an important mechanism of tumor metastasis (Review)". Oncology Reports 46.1 (2021): 145.
Chicago
Peng, X., Zhang, M., Meng, Y., Liang, Y., Wang, Y., Liu, X., Cai, W., Zhou, Y., Wang, X., Ma, Z., Xiang, Y., Zeng, L., Cui, S., Yang, L., Xin, H."Cell‑cell fusion as an important mechanism of tumor metastasis (Review)". Oncology Reports 46, no. 1 (2021): 145. https://doi.org/10.3892/or.2021.8096