Skip to main content

Intrinsic signalling factors associated with cancer cell-cell fusion

Abstract

Cellular fusion e.g. between cancer cells and normal cells represents a stepwise process that is tightly regulated. During a pre-hybrid preparation program somatic cells and/or cancer cells are promoted to a pro-fusogenic state as a prerequisite to prepare a fusion process. A pro-fusogenic state requires significant changes including restructure of the cytoskeleton, e.g., by the formation of F-actin. Moreover, distinct plasma membrane lipids such as phosphatidylserine play an important role during cell fusion. In addition, the expression of distinct fusogenic factors such as syncytins and corresponding receptors are of fundamental importance to enable cellular mergers. Subsequent hybrid formation and fusion are followed by a post-hybrid selection process. Fusion among normal cells is important and often required during organismal development. Cancer cells fusion appears more rarely and is associated with the generation of new cancer hybrid cell populations. These cancer hybrid cells contribute to an elevated tumour plasticity by altered metastatic behaviour, changes in therapeutic and apoptotic responses, and even in the formation of cancer stem/ initiating cells. While many parts within this multi-step cascade are still poorly understood, this review article predominantly focusses on the intracellular necessities for fusion among cancer cells or with other cell populations of the tumour microenvironment.

Video Abstract

Introduction

The hypothesis that cancer cells can fuse with other cells, such as macrophages, mesenchymal stroma-/stem-like cells (MSC), or further populations, with evolving cancer hybrids of an altered phenotype, was already postulated by the German physician Otto Aichel more than a century ago [1]. Such altered cancer hybrids can exhibit an enhanced metastatic capacity, an increased drug resistance and prospective cancer stem/initiating cells (CS/ICs) properties as described in a plethora of in vitro and in vivo studies (for review see: [2,3,4,5,6,7,8]). While these studies indicate that cancer cells and other cells could fuse, it not only remains unclear how this process is directed in detail, but also why cancer cells, or at least a certain subpopulation could fuse at all.

The fusion of cancer cells represents a pathophysiological process with increased malignancy that can ultimately be life-threatening to the organism. With respect to a variety of different tumour entities it is well known that development of metastases are the main cause of death in more than 90% of cases [9, 10]. Accordingly, cancer cell fusion, e.g. with MSC can develop cancer hybrid cells from a subsequent post-hybrid selection process (PHSP) with significantly increased metastatic capacity [11, 12].

Cancer hybrid cells may also contribute to immune escape [6, 13,14,15,16,17,18,19,20,21,22]. Gast et al. and Powell et al. suggested that leukocyte properties alone were sufficient to explain why (circulating) tumour × leukocyte hybrids may exhibit an immune-privileged phenotype [16, 17]. YKL-40 and immune checkpoint protein B7-2 (CD86) were elevated in glioblastoma × macrophage hybrids and independently suppressed anti-tumour immune factor levels of CD8+ cytotoxic T lymphocytes resulting in escape of immune surveillance [14]. Aguirre and colleagues observed that in vitro-derived cancer hybrid cells expressed high levels of the immune checkpoint ligand PD-L1, which was correlated to a reduced CD8+ lymphocyte proliferation in a PD-L1/PD-1 interaction-dependent manner [22]. Similarly, NK cell activity was significantly downregulated due to higher expression of NK cell inhibitors HLA class I members [22]. Likewise, HERV env proteins, such as syncytin-1 and syncytin-2, themselves exhibit immune suppressive properties, whereby it remains to be clarified how these proteins suppress host immunity [19, 20]. Syncytin-1 is commonly associated with (cancer) cell-cell fusion (Table 1) [23,24,25,26,27,28,29,30,31,32,33], but exhibits further tumour promoting characteristics, such as proliferation, invasion, metastasis [26, 34, 35], and possibly even immune escape. However, the precise role of cancer hybrids in immune escape still remains to be clarified. A possible focus could be on syncytins and checkpoint inhibitors.

Table 1 Syncytin-1 expression in cancer

In contrast to cancer cell fusion, physiological fusion processes, such as fertilization, placentation and myogenesis, are essential for an organism. Proper functioning of these fusion processes are ultimately required since dysregulation and/or dysfunction are associated with infertility and embryonic lethality [36,37,38,39,40,41,42,43]. Tissue regeneration represents another physiological cell-cell fusion process, which is facilitated by MSC and bone marrow-derived cells (BMDCs) [17, 44,45,46,47,48,49,50,51,52,53,54,55,56,57]. Even though the potency of BMDC-mediated tissue regeneration via cell-cell fusion was demonstrated in several in vivo studies [17, 44,45,46,47,48,49,50,51,52,53,54,55,56,57], the overall involvement of BMDCs in maintaining tissue homeostasis and repair remains less clear.

While all cell-cell fusion processes converge at a shared pathway of phospholipid bilayer fusion, the merger of cancer cells as compared to normal cells such as trophoblasts (placentation) and myoblasts (myogenesis) must be different processes. In addition to the fundamental difference on the physiology of the organism, this also includes the induction and regulation of cell-cell fusion. In particular, placentation and myogenesis include tightly regulated and very efficient fusion processes giving rise to a high number of viable multinucleated syncytial cells [42, 43, 58, 59]. In contrast, the fusion of cancer cells either with further cancer cells or with other cell populations like macrophages or MSC reveals a low fusion rate. Moreover, the overall survival of the resulting cancer hybrids is extremely low due to the reorganization of aneuploid chromosomal nuclei in a subsequent PHSP.

Accordingly, cancer cell fusion is influenced by both intrinsic signals and extracellular events. These questions will be explored and addressed in this review, with a particular focus on intrinsic signalling factors.

Similarities between physiological and patho-physiological cell-cell fusion events with a focus on fusogens, membrane phospholipids, and the actin cytoskeleton

The merger of bilayered phospholipid membranes of vesicles (e.g. endo-and exocytosis), organelles (e.g. mitochondria, exosomes), infection of host cells with enveloped viruses, or cell-cell merger represent fusion processes. These are complex, multi-factorial (including various proteins, phospholipids and biophysical conditions), energy depending, tightly and timely regulated but still scarcely understood actions (for review see: [3,4,5, 40, 60,61,62,63,64,65,66,67,68,69]). Figure 1 illustrates the apparently conserved pathway of lipid rearrangements in fusion of plasma membranes of cancer cells and normal cells, e.g. trophoblasts (placentation) and myoblasts (myogenesis) (Fig. 1).

Fig. 1
figure 1

Schematic scheme of decisive steps in membrane fusion. Fusogens ultimately catalyse the merger two phospholipid double membranes that should actually repel each other due to their negative charge

In addition to other proteins/ factors, a decisive role in cell-cell fusion is displayed by distinct proteins and phospholipids, which are indispensable for plasma membrane merger. These include so-called fusogens and their cognate receptors, as well as phosphatidylserine (PS), scramblase activity and PS-binding proteins [60,61,62, 64,65,66,67].

Fusogens and cognate receptors during physiological and non-physiological cell-cell fusion events

Although fusogens share the similar functionality to promote membrane merger, they differ in tissue-specific structure and compartmentalization and thus, display a marked heterogeneity. Syncytin-1 (human endogenous retrovirus-type W (HERV-W)) and syncytin-2 (human endogenous retrovirus-type FRD (HERV-FRD)) represent fusogens, which belong to the large family of HERV elements that account for about 8% of the human genome. Syncytin-1 and -2 are structurally related to class I viral fusogens [60, 61, 66, 70,71,72]. In a physiological environment these fusogens facilitate the fusion of villous and extravillous cytotrophoblasts to form syncytiotrophoblasts during placentation [73,74,75,76,77,78]. In the course of placenta formation Sugimoto and colleagues demonstrated that the human endogenous retroviral (HERV) element suppressyn impaired syncytin-1-mediated cytotrophoblast fusion by binding to the syncytin-1 receptor alanine, serine, cysteine transporter 2 (ASCT2) [79]. Moreover, increased suppressyn serum levels were found in women with placental defects in Down syndrome pregnancies [80]. In fact, a disturbed or dysregulated trophoblast syncytialization during placentation is commonly associated with an impaired maintenance and failed integrity of the blood-placental barrier. These effects could lead to pregnancy disorders with subsequent development of pre-eclampsia, HELLP syndrome, intrauterine growth retardation and even miscarriage [41, 79,80,81,82,83,84]. These unwanted complications and potential outcomes underline the importance of syncytin-1 functionality and proper cell-cell fusion during placental development.

In addition to placentation, syncytin-1 might also be involved in osteoclastogenesis [85, 86]. Here, data of Moller et al. revealed that syncytin-1 rather promoted cell fusions of two multi-nucleated osteoclasts, whereas the fusion of mono-nucleated pre-osteoclasts is directed by CD47 [85]. These findings suggest that different fusogens are involved in promoting and regulating either mono-nucleated or multi-nucleated cell fusion of the same cell type.

Other cell-type specific fusogens include the single pass transmembrane protein myomerger (myomixer—minion) and the seven transmembrane protein myomaker by promoting myoblast fusion [38,39,40]. A peak expression of both myomaker and myomerger is observed during skeletal muscle development [38]. Little if any multinucleated muscle cells were found in myomaker-myomerger knockout embryos [58, 87, 88]. This was further correlated with a lack of skeletal muscle wrapping, a transparent appearance and an overall early embryonic lethality [58, 87, 88].

At the level of gametes representing haploid sets of chromosomes, Juno and Izumo1 are important mediators of oocyte and sperm fusion [36, 37]. While Juno is a folate receptor (also known as folate receptor 4), Izumo1 represents a transmembrane protein in sperms. The ectodomain is composed of an immunoglobulin-like domain. The Izumo domain can be further divided into the N-terminal unstructured region and an α-helical core region important for sperm–egg binding [37]. Likewise, Juno and the tetraspanin-4 family protein CD9 may be important factors in oocytes. Accordingly, knockout of either Izumo1 (sperm), or Juno or CD9 (both oocyte) was associated with an impaired cell-cell fusion ability and, hence, correlated with infertility [37, 89, 90]. Recent work suggested that, in addition to its role in sperm to egg binding, Izumo1 exhibits fusogen activity [91]. Together, these data indicate the necessity of functioning fusogens and subsequent proper cell-cell fusion in various physiological processes and during development of an organism.

A different picture is observed in a patho-physiological environment such as the fusion of cancer cells. In contrast to the multiple fusogens that are involved in a variety of different physiological cell fusion events, only syncytin-1 has been identified with cancer cell-cell fusion and tumour progression so far [23,24,25,26,27,28,29,30,31,32,33, 35]. Interestingly, a potential association of syncytin-1 with cancer cell fusion was documented in various different tumour entities as summarised in Table 1.

It still remains unclear why syncytin-1 is expressed by cancer cells. Several studies have demonstrated an intrinsic basal syncytin-1 expression in various cancer cells and tumour types, which could be caused by changes in the promoter region and/ or other regulatory elements, such as 3’-long-term-repeat (LTR) and 5’-LTR regions [23,24,25,26,27,28,29,30,31,32,33]. Similarly, other data provided evidence that cytokines and an oestrogen responsive element, respectively, could induce, and thus increase intrinsic syncytin-1 expression levels in cancer cells and different tumour entities [23, 24, 26,27,28,29,30, 32]. Additionally, viral infections could also induce syncytin-1 expression in different cell types, such as Epstein-Barr virus (EBV) and human immunodeficiency virus (HIV) in astrocytes and monocytes [92, 93], and SARS-CoV-2 in Calu-3 cells and A459-ACE2 lung cancer cells [94]. Although these data indicate a possible link between certain viral infections and the induction of syncytin-1 expression, it is completely unclear if this actually plays a role in cancer cell fusion. Nevertheless, the tropism of viruses must also be taken into account, since viruses can only specifically infect their host cells.

Together, previous studies demonstrated that cancer cell-cell fusion is mediated primarily by syncytin-1, which appears similar to physiological syncytin-1-mediated cell-cell fusion events. However, no other fusogens have been identified so far in cancer cell fusion. In this context, studies by Uygur et al. showed that PC3 prostate cancer cells can fuse with muscle cells. In contrast to the muscle-specific fusogens myomaker and myomerger, however, this fusion process was also mediated by syncytin-1 [28].

Phosphatidyl serine (PS) in normal and cancer cell fusion

The membrane phospholipid component PS has been suggested as a uniquely conserved molecule in cell-cell and virus-cell fusion [62]. Under normal conditions PS is localised in the inner leaflet of the plasma membrane whereby translocation to the outer leaflet can be associated with apoptosis [95]. However, this PS shuttling is also essential for cell fusion. The two Ca2+-activated phospholipid scramblases (Ca2+-PLS) TMEM16E and TMEM16F, have been associated with cell-cell fusion [84, 96,97,98,99]. In particular, TMEM16F facilitates the translocation of PS from the inner to the outer leaflet of the plasma membrane which is essential for trophoblast fusion. Conversely, TMEM16F knockout mice exhibited a deficiency in trophoblast syncytialization and placental development, which was correlated to perinatal lethality [84].

Likewise, muscle progenitor cells from adult TMEM16E−/− knockout mice exhibited defective cell-cell fusion in culture and produced muscle fibres with significantly fewer nuclei as compared to controls [96]. This markedly reduced fusogenic capacity was associated with a decreased Ca2+-dependent PS exposure on the surface of TMEM16E−/− muscle progenitor cells and a decreased Ca2+ amplitude [96]. Re-expression of TMEM16E fully restored the fusogenic capacity of muscle progenitor cells concomitant with PS translocation and normalised Ca2+-dependent currents [96]. In opposite to these findings, however, studies by Gyobu and colleagues reported no apparent skeletal muscle abnormalities in TMEM16E−/− mice [97]. These discrepancies were hypothesised to be related to genetic differences in the murine animal models, to different approaches for down-modulation of TMEM16E, or to other experimental factors [96].

Fertilization represents another PS-dependent mechanism. Viable sperms containing membrane-associated PS and corresponding PS recognition receptors on oocytes have been suggested as key players in sperm × oocyte fusion [99]. Indeed, fertilization was impaired by both, masking of PS or functional disruption of PS receptor-specific signal transduction pathways [99]. Similarly, fertility was impaired in TMEM16E−/− mice, which, in addition to impaired PS translocation, was also attributed to a reduced sperm motility [97].

While the above summarised studies indicate the importance of PS in plasma membrane merger of normal cells, the role of PS in cancer cell-cell fusion is less clear. Only two studies so far suggested the involvement of PS in cancer cell-cell fusion [28, 100]. Muscle cell-mediated increase in IL-4 and IL-13 levels induced syncytin-1 and annexin A5 expression in human PC3 prostate cancer cells and subsequent cancer cell-cell fusion [28]. By contrast, hybridization of these two cell types was impaired by annexin A5 knockdown [28], which supports the necessity of PS even in cancer cell-cell fusion.

Whereas PS translocation to the outer plasma membrane leaflet can be associated with either apoptosis or cell fusion Noubissi et al. reported that the fusion between MSC and T47D human breast cancer cells or between MSC and MCF7 human breast cancer cells was significantly enhanced by apoptosis [100]. These findings suggest that apoptosis-induced cancer cell-cell fusion could result in viable cancer hybrids with a mixed genotype. Moreover, early apoptotic cells with still intact genome may represent the preferred fusion partners. Simultaneously, engaged and running pro-apoptotic pathways must be terminated in these newly formed cancer hybrids. Hochreiter-Hufford et al. demonstrated that apoptosis together with induced signalling via the PS receptor BAI1 promoted myoblast fusion [101]. Notably, apoptotic cells were only in contact with viable fusing myoblasts/myotubes, but did not merge with them indicating that cell-cell fusion was attributed to BAI1-induced signalling [101]. It is quite conceivable that a similar mechanism may occur in apoptosis-mediated fusion of cancer cells.

In brief, there is growing evidence for the importance of PS in plasma membrane-membrane fusion of normal cells and cancer cells [62, 66]. Nonetheless, the regulation of Ca2+-PLS activation and subsequent PS shuttling within the plasma membrane remains unclear although different Ca2+-PLS regulation pathways/ mechanisms have been identified [62, 66]. Moreover, potential signalling cascades in PS-relayed apoptosis and/or cell fusion represent crucial mosaic pieces within the multifactorial cell-cell-fusion machinery.

Actin in cell fusion

In addition to PS translocation and the expression of certain fusogens, restructure of certain cellular compartments including the actin cytoskeletal system is further required for cell-cell fusion [102, 103]. Thus, actin polymerization and associated cytoskeletal proteins play a substantial role to provide a fusion-permissive structure for the fusogenic cellular partners.

Changes in intracellular contractile elements of cytoskeletal proteins and a switch between a more soft and a rigid trabecular system of actin/myosin components promotes firm adhesion, higher migratory capacity, and facilitates cellular interactions. This is supported by the activation of the small GTPases Rac1 and Cdc42, and the Arp2/3-WASP complex which are key effectors of the actin cytoskeleton protrusion machinery to reorganise the actin cytoskeleton and promote an actin-mediated cell motility [104,105,106,107]. Interestingly, many genes required for myoblast fusion have been identified in fly, zebrafish and mouse, which have in common that they are participating in Arp2/3-mediated actin polymerization and formation of podosome-like structures [64, 108]. In fact, before the discovery of myomerger and myomixer, the formations of podosomes and invadopodia were thought to be instrumental in myoblast fusion. Reorganization and accumulation of F-actin results in the formation of F-actin-based protrusions that penetrate the target cell, causing the formation of fusion pores with ultimate fusion of the cells [108].

It is well-known that the Arp2/3 complex is deregulated in various tumours. Overexpression of Arp2/3-WASP is tightly associated with markedly enhanced cancer cell invasion and disease progression of several tumour types, including breast, lung, colorectal, prostate, and pancreas (for review see: [109]). The impact of Arp2/3-WASP in cancer cell-cell fusion is unclear, but previous work suggested a substantial role of F-actin polymerization and associated cytoskeletal protein alignment to enable a permissive microenvironment for the fusion of breast cancer cells with MSC within the tumour microenvironment [110, 111]. This fusion process could be inhibited by cytochalasin D which blocks elongation of actin filaments but exhibited no detectable effects on the expression of integrins and various cell adhesion molecules. Likewise, latrunculin B which belongs to a family of macrolide-structured toxins prevented the complex formation of monomeric G-actin with ATP and thereby impaired the energy-dependent generation of F-actin structures [110]. These findings suggest that cytoskeletal components including F-actin play an important role to provide a pro-fusogenic intracellular structure by interacting with distinct lipids of the cell membrane.

Taken together, cells are non-fusogenic per se, but must first undergo a pre-hybrid preparation program (PHPP) to promote transition into a so-called hypothetical pro-fusogenic state [4, 5, 60, 61, 112]. Fertilization, placentation, and myogenesis represent physiological cell-cell fusion processes that rely on important intrinsic signalling mechanisms. Structural cytoskeletal requirements in concert with sequential and timely availability of fusogens, corresponding fusogen receptors, and further proteins/phospholipids involved in fusion mediate transition into a pro-fusogenic state for preparation of cell fusion. However, cancer cells actually represent a non-fusogenic cell population although some studies indicated that cancer cells intrinsically can expressed fusogens such as syncytin-1 [23,24,25,26,27,28,29,30,31,32,33, 35]. A summary of these different models is presented in Fig. 2.

Fig. 2
figure 2

Homotypic and heterotypic cell-cell fusion. Physiological homotypic cell-cell fusion is characterised by a highly efficient fusion frequency and the generation of multiple syncytia, which is most likely attributed to a high number of cells in a pro-fusogenic state (A). However, it is also possible that pro-fusogenic cells fuse with non-fusogenic cells in a homotypic manner. Heterotypic cell-cell fusion is different since it remains unclear, which cells provide fusogens and exhibit further fusion relevant properties (B). A pro-fusogenic cell fuses with a cell in a non-fusogenic state, which only expresses cognate receptors and phosphatidylserine (PS) (adapted from [166])

Thus, some basic components of cell-cell fusion in physiological and pathophysiological processes may not differ from each other with respect to the involved proteins/phospholipids. Nevertheless, there are significant differences at the regulatory levels.

Differences between physiological and non-physiological cell-cell fusion events

The fundamental differences between physiological and pathophysiological cell-cell fusion events are their impact on the organism. Physiological fusion processes are essential for an organismal development whereby dysregulation and/or dysfunctions are associated e.g. with infertility and embryonic lethality [36,37,38,39,40,41,42,43]. In contrast, the pathophysiological process of cancer cell fusion is associated with a significantly enhanced tumour plasticity [12, 113]. Usually, this leads to disease progression due to an increased malignancy of the tumour hybrids [2,3,4,5,6,7,8]. Physiological and pathophysiological cell-cell fusion events unravel further differences, including the general fusion frequency, the regulation of the fusion process itself, the fusion partner, and the survival rate of the hybrid cells after a PHSP.

Regulation of cell-cell fusion and general fusion frequency

Fertilization, placentation, and myogenesis represent examples for physiological cell-cell fusion events that are characterised by an inherent time-dependent expression of specific fusogens and cognate receptors [3,4,5, 40, 60,61,62,63,64,65,66,67,68,69]. All of these fusion processes are running in a controlled manner and are terminated at a certain time point to avoid additional unwanted cell-cell fusion events. In particular, placentation and myogenesis represent highly effective cell-cell fusion events, which give rise to numerous syncytia in vitro and in vivo. For example, immortalised murine C2C12 myoblasts fused at a similar rate as compared to human skeletal myoblasts [59]. A lot of fusion events were also observed between nuclei present in cultured syncytia and fibroblasts expressing both, myomaker and myomerger [58]. Likewise, marked levels of myotube formation were determined by Isobe and colleagues using a novel fusion quantification system [42]. In this line, primary human trophoblasts cultured for 72 h and BeWo cells treated with forskolin for 48 h demonstrated a significant amount of fusion event [43]. These high cell-cell fusion rates during placentation and myogenesis are achieved by a precisely timed expression/activation of the cell-cell fusion machinery. For instance, a continuous syncytin-1 expression is detectable in villous cytotrophoblasts until 37 weeks of gestation [41]. Conversely, a peak expression of both, myomerger and myomerger is observed during skeletal muscle development [38].

In contrast, the fusion of cancer cells is different since only a certain subpopulation of cancer cells exhibits fusogenic properties. Moreover, the fusion frequency can change markedly between distinct cancer cell lines and read-out systems. Accordingly, determination of fusogenecity in vitro varied between 0.0066 and 6.5% [114,115,116,117,118] and in vivo between 0.5 and 51% [16, 17, 57, 115, 118,119,120]. However, the in vivo data have to be regarded very carefully as they do not necessarily represent the actual fusion frequency of the (cancer) cells. This particularly applies to detection limitations during in vivo studies with different tumour models, different readout systems, and especially different time points applied.

These data demonstrate that cancer cells apparently have an inherent fusogenecity that appears to be different among various cancer cell lines and only applies to a certain subpopulation. Moreover, the overall fusogenecity of cancer cells/ cell lines is rather low when compared to the rates observed during physiological cell-cell fusion events.

Cancer cell fusion partners: homotypic versus heterotypic fusion

Cell-cell fusion can be divided into homotypic and heterotypic processes. In homotypic cell-cell fusion events identical cell types are merging. Conversely, during heterotypic fusion hybrids are formed with different cell populations. Homotypic cell fusion predominantly applies to physiological processes, e.g. placentation and myogenesis (Fig. 2A) [3,4,5, 40, 60,61,62,63,64,65,66,67,68,69]. As an exception, heterotypic physiological cell-cell fusion is represented by the sperm and oocyte fusion with the rearrangement of haploid sets of chromosomes during fertilization [3,4,5, 40, 60,61,62,63,64,65,66,67,68,69].

Cancer cells may fuse in a homotypic as well as in a heterotypic manner [2,3,4,5,6,7,8]. According to the hypothesis of a pro-fusogenic state as a prerequisite for cell-cell fusion, either one or both cancer cells should acquire this state (Fig. 2A, B). A similar picture would emerge for heterotypic cancer cell fusion events (Fig. 2B). Either both or one of the cancer cell or normal cell would be in a pro-fusogenic state (Fig. 2B). In addition, a direct fusion between a pro-fusogenic cancer cell and a pro-fusogenic normal cell is also possible. This may account for a higher fusion rate in heterotypic cancer cell fusion since two independent pro-fusogenic cell types are involved. Supportive evidence for this model was obtained from previous work which determined a significantly lower homofusion (autofusion) rate by more than tenfold in various breast cancer cell lines when compared to a corresponding heterofusion with MSC [121].

As described above, expression of syncytin-1 in cancer cells appears to be primarily involved in cell fusion [23,24,25,26,27,28,29,30,31,32,33, 35]. These findings fit well to the assumption that “cancer cells fuse with other cells” (for review see [3,4,5, 61, 63, 122, 123]), which implies that cancer cells have acquired a pro-fusogenic state, express all necessary components of the fusion machinery and then actively fuse with other non-fusogenic cells (Fig. 3A).

Fig. 3
figure 3

In this model it is assumed that cancer cells fuse with other cells suggesting that all cells have entered a pre-hybrid preparation program (PHPP) by acquisition of a pro-fusogenic state (A). Alternatively, it cannot be ruled out that other cells in a pro-fusogenic state, such as macrophages, MSC, or stem cells could fuse with cancer cells in a non-fusogenic state (B). Similarly, both a cancer cell and a normal cell must have acquired a pro-fusogenic state for fusion (C). The resulting cancer hybrid populations from all of these alternatives have to undergo a post-hybrid selection process (PHSP) for re-organization of the chromosomal ploidy and corresponding metabolic adaptation to ensure survival

In addition, normal cells such as macrophages and MSC also exhibit fusogenic properties by acquisition of a pro-fusogenic state to form hybrids with other cells including cancer cells (Fig. 3B) [121, 124,125,126,127,128,129,130]. This assumption resembles MSC- and BMDC-mediated tissue regeneration e.g. during tumour growth-mediated chronic inflammation [17, 44,45,46,47,48,49,50,51,52,53,54,55,56,57, 131,132,133,134,135]. Tumour invading MSC and macrophages do not distinguish between normal cells and cancer cells and exhibit their regenerative capacity according to the requirements of the tissue damage [136, 137].

Heterokaryon-to-synkaryon transition/ ploidy reduction and post hybrid-selection process

Cell-cell fusion in neoplastic tissues represents a rare process. But even rarer is the survival of the resulting hybrid cells. The fusion of two cells does not simply result in the duplication of the karyotype. Rather, in case of division-active cells, such as cancer hybrids, two complex processes follow, which are the heterokaryon-to-synkaryon transition/ploidy reduction [135, 138,139,140] and PHSP [5]. Both processes are associated with aneuploidy and genomic instability due to chromosome missegregation, DNA damage, micronuclei formation and chromothripsis [141,142,143,144,145,146]. Thus, not only the fusion process itself, but also the survival probability of tumour hybrids in general is rather rare. Nonetheless, previous work assumed that despite a relatively low fusion frequency, fusion-mediated recombination could have a profound impact on clonal diversity and an overall increased intratumoural heterogeneity [113, 115].

This raises the question if cancer hybrid cells following HST/PR and PHSP are generally associated with increased malignancy? Besides increased malignancy, different outcomes have been described including reduced tumourigenicity [126, 147, 148] and tumour dormancy [149]. It is conceivable that HST/PR- and PSHP-associated cellular and genotypic stresses are survived primarily by those cancer hybrids in which apoptosis pathways are turned off and survival signalling pathways are active. Such cancer hybrids could exhibit a selection advantage over chemotherapeutic agents. Nonetheless, the fact that survival of fused cancer hybrid cells represents a rare event reflects one of the major differences to physiological cell-cell fusion processes.

Conclusion

Beside the importance of physiological cell fusion, there is profound scientific consensus that cell-cell fusion events also occur in human cancers and that disease progression can be related to the formation of tumour hybrids [16, 124, 150,151,152,153,154,155,156,157,158]. However, precise regulatory mechanisms to control e.g. fusion of mononucleated versus multinucleated cells are still obscure.

Although some mechanistic principles of cell-cell fusion do not markedly differ between physiological and pathophysiological processes (Fig. 4), other fundamental differences can be observed. These include, for example, the high fusion frequency in physiological processes such as placentation and myogenesis, which is ensured by a time-controlled upregulation of the fusion machinery. In contrast, fusion of cancer cells represents a rare process that is predominantly relayed by a basal expression of the fusogen syncytin-1. The rarity of cancer cell fusion events is also based on the limitation of fusogens expressed only in certain cancer subpopulations. In this context, the significantly higher rates of cancer cell heterofusions (e.g. with MSC) as compared to homofusions [121] are explained by the availability of more fusogenic partner cells and support the hypothetical fusion models (Figs. 2 and 3). In addition to the low cancer cell fusion rates, the number of surviving tumour hybrids is even far lower as a consequence of restoring DNA stability during the PHSP [5]. This is also consistent with mathematical models which show that despite a relatively low fusion frequency, fusion-mediated recombination could have a profound impact on clonal diversity and an overall increased intratumoural heterogeneity [115].

Fig. 4
figure 4

Cancer cell-cell fusion model which is focused predominantly on the expression of the fusogen syncytin-1 and includes some basal similarities to physiological fusion events. In this model it is assumed that a cancer cell with a re-organised actin cytoskeleton expresses syncytin-1 and a PS-binding receptor, which recognises ASCT2 and PS, respectively. A basal syncytin-1 expression could be attributed to promotor mutation and/or hypomethylation (TF = transcription factor). The majority of expressed syncytin-1 will remain in the cytosol. In some cells, syncytin-1 may translocate to the plasma membrane, which may be facilitated by altered cytoskeletal structures. The corresponding cell exhibits Ca2+-PLS activity, which is engaged by Ca2+ to shuttle PS and subsequently enable plasma membrane merger and cell fusion (modified according to [166])

If the amount of PHSP-surviving aneuploid cancer hybrid cells is obviously negligibly low why care for these cells? Previous work has demonstrated in vitro and in vivo that certain cancer hybrid cells with aneuploid karyotype, e.g. after fusion of breast cancer cells with MSC acquire a proliferation advantage during the selection process beside other properties such as enhanced metastatic capacities. Consequently, the cancer hybrid cells rapidly overgrow the initial cancer cells by exhibition of new properties [11, 119] and thus, expanding the tumour heterogeneity and plasticity. Of interest, a summarising compilation revealed various tumour-derived aneuploid cell lines. These include among others different carcinoma types and tissues such as 12 × breast, 4 × ovarian, 4 × cervical, 4 × endometrial, 4 × brain, 4 × lung, 7 × colon, 4 × liver, 5 × kidney, 5 × pancreas, 4 × gastric, 3 × prostate, beside melanoma, osteosarcoma, retinoblastoma, nasal, pharyngeal soft palate cancer, and 4 × leukaemia [159]. Although no direct proof is available, most of these spontaneously tumourigenic patient-derived cancer cells may be associated with previous fusion events after surviving from a PHSP as aneuploid cell types. Nevertheless, other forms of cancer cell mergers such as cannibalism or entosis can also contribute to aneuploid outcomes [160, 161] and even a more metastatic phenotype [162]. However, these findings demonstrate that a negligibly low amount of cancer hybrid cells can have a significant impact on the corresponding tumour development after all.

It still remains unclear why syncytin-1 is expressed only in a specific fraction of cancer cells. Previous work has demonstrated that proliferation, invasion and potentially metastasis of cancer cells may be influenced by syncytin-1 [26, 34, 35]. Similar findings have already been described for other HERV env elements [25, 163,164,165] indicating also a possible non-fusogenic role of syncytin-1 in cancer progression.

The timely orchestration of intrinsic signalling pathways involves actin-mediated intracellular restructuring to enable cytosolic syncytin-1 transport to the plasma membrane for a pro-fusogenic state. Mechanisms that facilitate this syncytin-1 translocation are unclear. In addition, Ca2+-PLS, particularly TMEM16F facilitates PS shuttling from the inner to the outer leaflet of the plasma membrane as an essential prerequisite for membrane merger. A variety of further components such as e.g. extracellular events [166] conclude a PHPP for subsequent cell fusion.

Interestingly, the fusogenic properties of macrophages and MSC are rather underestimated in the context of cancer cell-cell fusion. This is surprising, since many studies have shown that these cells can regenerate tissue damage through cell-cell fusion [17, 44,45,46,47,48,49,50,51,52,53,54,55,56,57]. Tumour invading macrophages and MSC seem to ignore as to whether normal cells or cancer cells represent their interaction partner. Moreover, the tumour microenvironment resembles chronic inflammatory tissue [131,132,133] which attracts macrophages and MSC and is a known inducer of cell-cell fusion [51, 56, 167].

It is therefore important to unravel the detailed interplay of the fusogenic contributors and their regulation particularly at the molecular level of the entire cell fusion process. Further insights into the underlying mechanisms could help to develop selective anti-cancer cell fusion strategies in tumour therapeutic approaches.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

ASCT2:

Alanine, serine, cysteine transporter 2

EBV:

Epstein-Barr virus

BMDCs:

Bone marrow-derived cells

Ca2+-PLS:

Ca2+-activated phospholipid scramblases

CS/ICs:

Cancer stem/initiating cells

HERV:

Human endogenous retroviral

HERV-FRD:

Human endogenous retrovirus-type FRD

HER2-W:

Human endogenous retrovirus-type W

HUVEC:

Human umbilical vein endothelial cells

HIV:

Human immunodeficiency virus

LTR:

Long-term-repeat

MSC:

Mesenchymal stroma-/stem-like cells

NSCLC:

Non-small cell lung carcinoma

OSCC:

Oral squamous carcinoma cells

PGCC:

Polygiant cancer cells

PHPP:

Pre-hybrid preparation program

PHSP:

Post-hybrid selection process

PS:

Phosphatidylserine

SARS-CoV-2:

Severe acute respiratory syndrome corona virus-2

syHP:

Syncytin-1 homologous protein

TMEM16:

Transmembrane member 16

UCC:

Urothelial cell carcinoma

References

  1. Aichel O. Über Zellverschmelzung mit quantitativ abnormer Chromosomenverteilung als Ursache der Geschwulstbildung. In: Leipzig RW, editor. Vorträge und Aufsätze über Entwicklungsmechanik der Organismen. Germany: Wilhelm Engelmann; 1911. p. 1–115.

    Google Scholar 

  2. Demin S, Berdieva M, Goodkov A. Cell-cell fusions and cell-in-cell phenomena in healthy cells and cancer: lessons from protists and invertebrates. Semin Cancer Biol. 2021;81:96–105.

    Article  PubMed  Google Scholar 

  3. Dittmar T, Weiler J, Luo T, Hass R. Cell-cell fusion mediated by viruses and HERV-derived fusogens in cancer initiation and progression. Cancers (Basel). 2021;13:5363.

    Article  CAS  PubMed  Google Scholar 

  4. Hass R, von der Ohe J, Dittmar T. Hybrid formation and fusion of cancer cells in vitro and in vivo. Cancers (Basel). 2021;13:4496.

    Article  CAS  PubMed  Google Scholar 

  5. Hass R, von der Ohe J, Dittmar T. Cancer cell fusion and post-hybrid selection process (PHSP). Cancers (Basel). 2021;13:4636.

    Article  CAS  PubMed  Google Scholar 

  6. 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;21:1872.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wang HF, Xiang W, Xue BZ, Wang YH, Yi DY, Jiang XB, Zhao HY, Fu P. Cell fusion in cancer hallmarks: current research status and future indications. Oncol Lett. 2021;22:530.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Zhang H, Ma H, Yang X, Fan L, Tian S, Niu R, Yan M, Zheng M, Zhang S. Cell Fusion-related proteins and signaling pathways, and their roles in the development and progression of cancer. Front Cell Dev Biol. 2021;9:809668.

    Article  PubMed  Google Scholar 

  9. Gupta GP, Massague J. Cancer metastasis: building a framework. Cell. 2006;127:679–95.

    Article  CAS  PubMed  Google Scholar 

  10. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70.

    Article  CAS  PubMed  Google Scholar 

  11. Melzer C, von der Ohe J, Hass R. Enhanced metastatic capacity of breast cancer cells after interaction and hybrid formation with mesenchymal stroma/stem cells (MSC). Cell Commun Signal. 2018;16:2.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Melzer C, Ohe JV, Hass R. Altered tumor plasticity after different cancer cell fusions with MSC. Int J Mol Sci. 2020;21:8347.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kaifi JT, Li G, Clawson G, Kimchi ET, Staveley-O’Carroll KF. Perioperative circulating tumor cell detection: current perspectives. Cancer Biol Ther. 2016;17:859–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shao R, Han X, Zhu B, Luo J, Zhang X, Chen H, Brown C, Burnside A, Fu G, Zhao F, et al. Fusion of invasive tumor cells with infiltrating macrophages fuels epithelial-mesenchymal transition and adaptive immune evasion. Res Sq. 2022.

  15. Jiang E, Yan T, Xu Z, Shang Z. Tumor microenvironment and cell fusion. Biomed Res Int. 2019;2019:5013592.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Gast CE, Silk AD, Zarour L, Riegler L, Burkhart JG, Gustafson KT, Parappilly MS, Roh-Johnson M, Goodman JR, Olson B, et al. Cell fusion potentiates tumor heterogeneity and reveals circulating hybrid cells that correlate with stage and survival. Sci Adv. 2018;4:eaat7828.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Powell AE, Anderson EC, Davies PS, Silk AD, Pelz C, Impey S, Wong MH. Fusion between Intestinal epithelial cells and macrophages in a cancer context results in nuclear reprogramming. Cancer Res. 2011;71:1497–505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tolosa JM, Schjenken JE, Clifton VL, Vargas A, Barbeau B, Lowry P, Maiti K, Smith R. The endogenous retroviral envelope protein syncytin-1 inhibits LPS/PHA-stimulated cytokine responses in human blood and is sorted into placental exosomes. Placenta. 2012;33:933–41.

    Article  CAS  PubMed  Google Scholar 

  19. Gao Y, Yu XF, Chen T. Human endogenous retroviruses in cancer: expression, regulation and function. Oncol Lett. 2021;21:121.

    Article  CAS  PubMed  Google Scholar 

  20. Dervan E, Bhattacharyya DD, McAuliffe JD, Khan FH, Glynn SA. Ancient adversary—HERV-K (HML-2) in cancer. Front Oncol. 2021;11:658489.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Barreto SG, Gardi N, Dutt S. Birth of a solid organ cancer-the cell fusion hypothesis presented with pancreatic cancer as a model: a narrative review. Chin Clin Oncol. 2021;10:45.

    Article  PubMed  Google Scholar 

  22. Aguirre LA, Montalban-Hernandez K, Avendano-Ortiz J, Marin E, Lozano R, Toledano V, Sanchez-Maroto L, Terron V, Valentin J, Pulido E, et al. Tumor stem cells fuse with monocytes to form highly invasive tumor-hybrid cells. Oncoimmunology. 2020;9:1773204.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Bjerregaard B, Holck S, Christensen IJ, Larsson LI. Syncytin is involved in breast cancer-endothelial cell fusions. Cell Mol Life Sci. 2006;63:1906–11.

    Article  CAS  PubMed  Google Scholar 

  24. Larsson LI, Holck S, Christensen IJ. Prognostic role of syncytin expression in breast cancer. Hum Pathol. 2007;38:726–31.

    Article  CAS  PubMed  Google Scholar 

  25. Yu H, Liu T, Zhao Z, Chen Y, Zeng J, Liu S, Zhu F. Mutations in 3’-long terminal repeat of HERV-W family in chromosome 7 upregulate syncytin-1 expression in urothelial cell carcinoma of the bladder through interacting with c-Myb. Oncogene. 2014;33:3947–58.

    Article  CAS  PubMed  Google Scholar 

  26. Strick R, Ackermann S, Langbein M, Swiatek J, Schubert SW, Hashemolhosseini S, Koscheck T, Fasching PA, Schild RL, Beckmann MW, Strissel PL. Proliferation and cell-cell fusion of endometrial carcinoma are induced by the human endogenous retroviral Syncytin-1 and regulated by TGF-beta. J Mol Med. 2007;85:23–38.

    Article  CAS  PubMed  Google Scholar 

  27. Fei F, Li C, Wang X, Du J, Liu K, Li B, Yao P, Li Y, Zhang S. Syncytin 1, CD9, and CD47 regulating cell fusion to form PGCCs associated with cAMP/PKA and JNK signaling pathway. Cancer Med. 2019;8:3047–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Uygur B, Leikina E, Melikov K, Villasmil R, Verma SK, Vary CPH, Chernomordik LV. Interactions with muscle cells boost fusion, stemness, and drug resistance of prostate cancer cells. Mol Cancer Res. 2019;17:806–20.

    Article  CAS  PubMed  Google Scholar 

  29. Chignola R, Sega M, Molesini B, Baruzzi A, Stella S, Milotti E. Collective radioresistance of T47D breast carcinoma cells is mediated by a Syncytin-1 homologous protein. PLoS One. 2019;14:e0206713.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yan TL, Wang M, Xu Z, Huang CM, Zhou XC, Jiang EH, Zhao XP, Song Y, Song K, Shao Z, et al. Up-regulation of syncytin-1 contributes to TNF-alpha-enhanced fusion between OSCC and HUVECs partly via Wnt/beta-catenin-dependent pathway. Sci Rep. 2017;7:40983.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fu Y, Zhuang X, Xia X, Li X, Xiao K, Liu X. Correlation between promoter hypomethylation and increased expression of Syncytin-1 in non-small cell lung cancer. Int J Gen Med. 2021;14:957–65.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Larsen JM, Christensen IJ, Nielsen HJ, Hansen U, Bjerregaard B, Talts JF, Larsson LI. Syncytin immunoreactivity in colorectal cancer: potential prognostic impact. Cancer Lett. 2009;280:44–9.

    Article  CAS  PubMed  Google Scholar 

  33. Benesova M, Trejbalova K, Kovarova D, Vernerova Z, Hron T, Kucerova D, Hejnar J. DNA hypomethylation and aberrant expression of the human endogenous retrovirus ERVWE1/syncytin-1 in seminomas. Retrovirology. 2017;14:20.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Zhou Y, Liu L, Liu Y, Zhou P, Yan Q, Yu H, Chen X, Zhu F. Implication of human endogenous retrovirus W family envelope in hepatocellular carcinoma promotes MEK/ERK-mediated metastatic invasiveness and doxorubicin resistance. Cell Death Discov. 2021;7:177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu C, Xu J, Wen F, Yang F, Li X, Geng D, Li L, Chen J, Zheng J. Upregulation of syncytin-1 promotes invasion and metastasis by activating epithelial-mesenchymal transition-related pathway in endometrial carcinoma. Onco Targets Ther. 2019;12:31–40.

    Article  CAS  PubMed  Google Scholar 

  36. Nishimura K, Han L, Bianchi E, Wright GJ, de Sanctis D, Jovine L. The structure of sperm Izumo1 reveals unexpected similarities with Plasmodium invasion proteins. Curr Biol. 2016;26:R661-662.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kato K, Satouh Y, Nishimasu H, Kurabayashi A, Morita J, Fujihara Y, Oji A, Ishitani R, Ikawa M, Nureki O. Structural and functional insights into IZUMO1 recognition by JUNO in mammalian fertilization. Nat Commun. 2016;7:12198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chen B, You W, Wang Y, Shan T. The regulatory role of Myomaker and Myomixer-Myomerger-Minion in muscle development and regeneration. Cell Mol Life Sci. 2020;77:1551–69.

    Article  CAS  PubMed  Google Scholar 

  39. Leikina E, Gamage DG, Prasad V, Goykhberg J, Crowe M, Diao J, Kozlov MM, Chernomordik LV, Millay DP. Myomaker and Myomerger work independently to control distinct steps of membrane remodeling during myoblast fusion. Dev Cell. 2018;46(767–780):e767.

    Article  Google Scholar 

  40. Yang Y, Margam NN. Structural insights into membrane fusion mediated by convergent small Fusogens. Cells. 2021;10:160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chen CP, Wang KG, Chen CY, Yu C, Chuang HC, Chen H. Altered placental syncytin and its receptor ASCT2 expression in placental development and pre-eclampsia. BJOG. 2006;113:152–8.

    Article  PubMed  Google Scholar 

  42. Isobe M, Suzuki Y, Sugiura H, Shibata M, Ohsaki Y, Kametaka S. Novel cell-based system to assay cell-cell fusion during myotube formation. Biomed Res. 2022;43:107–14.

    Article  CAS  PubMed  Google Scholar 

  43. Johnson LJ, Azari S, Webb A, Zhang X, Gavrilin MA, Marshall JM, Rood K, Seveau S. Human placental trophoblasts infected by listeria monocytogenes undergo a pro-inflammatory switch associated with poor pregnancy outcomes. Front Immunol. 2021;12:709466.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ferrand J, Noel D, Lehours P, Prochazkova-Carlotti M, Chambonnier L, Menard A, Megraud F, Varon C. Human bone marrow-derived stem cells acquire epithelial characteristics through fusion with gastrointestinal epithelial cells. PLoS One. 2011;6:e19569.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Willenbring H, Bailey AS, Foster M, Akkari Y, Dorrell C, Olson S, Finegold M, Fleming WH, Grompe M. Myelomonocytic cells are sufficient for therapeutic cell fusion in liver. Nat Med. 2004;10:744–8.

    Article  CAS  PubMed  Google Scholar 

  46. LaBarge MA, Blau HM. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell. 2002;111:589–601.

    Article  CAS  PubMed  Google Scholar 

  47. Weimann JM, Charlton CA, Brazelton TR, Hackman RC, Blau HM. Contribution of transplanted bone marrow cells to Purkinje neurons in human adult brains. Proc Natl Acad Sci U S A. 2003;100:2088–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Weimann JM, Johansson CB, Trejo A, Blau HM. Stable reprogrammed heterokaryons form spontaneously in Purkinje neurons after bone marrow transplant. Nat Cell Biol. 2003;5:959–66.

    Article  CAS  PubMed  Google Scholar 

  49. Camargo FD, Green R, Capetenaki Y, Jackson KA, Goodell MA. Single hematopoietic stem cells generate skeletal muscle through myeloid intermediates. Nat Med. 2003;9:1520–7.

    Article  CAS  PubMed  Google Scholar 

  50. Camargo FD, Finegold M, Goodell MA. Hematopoietic myelomonocytic cells are the major source of hepatocyte fusion partners. J Clin Invest. 2004;113:1266–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Davies PS, Powell AE, Swain JR, Wong MH. Inflammation and proliferation act together to mediate intestinal cell fusion. PLoS One. 2009;4:e6530.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Silk AD, Gast CE, Davies PS, Fakhari FD, Vanderbeek GE, Mori M, Wong MH. Fusion between hematopoietic and epithelial cells in adult human intestine. PLoS One. 2013;8:e55572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Terada N, Hamazaki T, Oka M, Hoki M, Mastalerz DM, Nakano Y, Meyer EM, Morel L, Petersen BE, Scott EW. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature. 2002;416:542–5.

    Article  CAS  PubMed  Google Scholar 

  54. Vassilopoulos G, Wang PR, Russell DW. Transplanted bone marrow regenerates liver by cell fusion. Nature. 2003;422:901–4.

    Article  CAS  PubMed  Google Scholar 

  55. Wang X, Willenbring H, Akkari Y, Torimaru Y, Foster M, Al-Dhalimy M, Lagasse E, Finegold M, Olson S, Grompe M. Cell fusion is the principal source of bone-marrow-derived hepatocytes. Nature. 2003;422:897–901.

    Article  CAS  PubMed  Google Scholar 

  56. Johansson CB, Youssef S, Koleckar K, Holbrook C, Doyonnas R, Corbel SY, Steinman L, Rossi FM, Blau HM. Extensive fusion of haematopoietic cells with Purkinje neurons in response to chronic inflammation. Nat Cell Biol. 2008;10:575–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rizvi AZ, Swain JR, Davies PS, Bailey AS, Decker AD, Willenbring H, Grompe M, Fleming WH, Wong MH. Bone marrow-derived cells fuse with normal and transformed intestinal stem cells. Proc Natl Acad Sci U S A. 2006;103:6321–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Quinn ME, Goh Q, Kurosaka M, Gamage DG, Petrany MJ, Prasad V, Millay DP. Myomerger induces fusion of non-fusogenic cells and is required for skeletal muscle development. Nat Commun. 2017;8:15665.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Cheng CS, El-Abd Y, Bui K, Hyun YE, Hughes RH, Kraus WE, Truskey GA. Conditions that promote primary human skeletal myoblast culture and muscle differentiation in vitro. Am J Physiol Cell Physiol. 2014;306:C385-395.

    Article  CAS  PubMed  Google Scholar 

  60. Hernandez JM, Podbilewicz B. The hallmarks of cell-cell fusion. Development. 2017;144:4481–95.

    Article  CAS  PubMed  Google Scholar 

  61. Podbilewicz B. Virus and cell fusion mechanisms. Annu Rev Cell Dev Biol. 2014;30:111–39.

    Article  CAS  PubMed  Google Scholar 

  62. Whitlock JM, Chernomordik LV. Flagging fusion: phosphatidylserine signaling in cell-cell fusion. J Biol Chem. 2021;296:100411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Willkomm L, Bloch W. State of the art in cell-cell fusion. Methods Mol Biol. 2015;1313:1–19.

    Article  PubMed  Google Scholar 

  64. Aguilar PS, Baylies MK, Fleissner A, Helming L, Inoue N, Podbilewicz B, Wang H, Wong M. Genetic basis of cell-cell fusion mechanisms. Trends Genet. 2013;29:427–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Helming L, Gordon S. Molecular mediators of macrophage fusion. Trends Cell Biol. 2009;19:514–22.

    Article  CAS  PubMed  Google Scholar 

  66. Brukman NG, Uygur B, Podbilewicz B, Chernomordik LV. How cells fuse. J Cell Biol. 2019;218:1436–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Petrany MJ, Millay DP. Cell fusion: merging membranes and making muscle. Trends Cell Biol. 2019;29:964–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Gao S, Hu J. Mitochondrial fusion: the machineries in and out. Trends Cell Biol. 2021;31:62–74.

    Article  CAS  PubMed  Google Scholar 

  69. Martens S, McMahon HT. Mechanisms of membrane fusion: disparate players and common principles. Nat Rev Mol Cell Biol. 2008;9:543–56.

    Article  CAS  PubMed  Google Scholar 

  70. Vance TDR, Lee JE. Virus and eukaryote fusogen superfamilies. Curr Biol. 2020;30:R750–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Garcia-Montojo M, Doucet-O’Hare T, Henderson L, Nath A. Human endogenous retrovirus-K (HML-2): a comprehensive review. Crit Rev Microbiol. 2018;44:715–38.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Durnaoglu S, Lee SK, Ahnn J. Syncytin, envelope protein of human endogenous retrovirus (HERV): no longer “fossil” in human genome. Anim Cells Syst (Seoul). 2021;25:358–68.

    Article  CAS  PubMed  Google Scholar 

  73. Lavialle C, Cornelis G, Dupressoir A, Esnault C, Heidmann O, Vernochet C, Heidmann T. Paleovirology of “syncytins”, retroviral env genes exapted for a role in placentation. Philos Trans R Soc Lond B Biol Sci. 2013;368:20120507.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Malassine A, Handschuh K, Tsatsaris V, Gerbaud P, Cheynet V, Oriol G, Mallet F, Evain-Brion D. Expression of HERV-W Env glycoprotein (syncytin) in the extravillous trophoblast of first trimester human placenta. Placenta. 2005;26:556–62.

    Article  CAS  PubMed  Google Scholar 

  75. Muir A, Lever AM, Moffett A. Human endogenous retrovirus-W envelope (syncytin) is expressed in both villous and extravillous trophoblast populations. J Gen Virol. 2006;87:2067–71.

    Article  CAS  PubMed  Google Scholar 

  76. Blond JL, Lavillette D, Cheynet V, Bouton O, Oriol G, Chapel-Fernandes S, Mandrand B, Mallet F, Cosset FL. An envelope glycoprotein of the human endogenous retrovirus HERV-W is expressed in the human placenta and fuses cells expressing the type D mammalian retrovirus receptor. J Virol. 2000;74:3321–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Frendo JL, Olivier D, Cheynet V, Blond JL, Bouton O, Vidaud M, Rabreau M, Evain-Brion D, Mallet F. Direct involvement of HERV-W Env glycoprotein in human trophoblast cell fusion and differentiation. Mol Cell Biol. 2003;23:3566–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Vargas A, Moreau J, Landry S, LeBellego F, Toufaily C, Rassart E, Lafond J, Barbeau B. Syncytin-2 plays an important role in the fusion of human trophoblast cells. J Mol Biol. 2009;392:301–18.

    Article  CAS  PubMed  Google Scholar 

  79. Sugimoto J, Sugimoto M, Bernstein H, Jinno Y, Schust D. A novel human endogenous retroviral protein inhibits cell-cell fusion. Sci Rep. 2013;3:1462.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Sugimoto J, Schust DJ, Yamazaki T, Kudo Y. Involvement of the HERV-derived cell-fusion inhibitor, suppressyn, in the fusion defects characteristic of the trisomy 21 placenta. Sci Rep. 2022;12:10552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Malassine A, Frendo JL, Evain-Brion D. Trisomy 21- affected placentas highlight prerequisite factors for human trophoblast fusion and differentiation. Int J Dev Biol. 2010;54:475–82.

    Article  CAS  PubMed  Google Scholar 

  82. Gauster M, Moser G, Orendi K, Huppertz B. Factors involved in regulating trophoblast fusion: potential role in the development of preeclampsia. Placenta. 2009;30:S49-54.

    Article  PubMed  Google Scholar 

  83. Vargas A, Zhou S, Ethier-Chiasson M, Flipo D, Lafond J, Gilbert C, Barbeau B. Syncytin proteins incorporated in placenta exosomes are important for cell uptake and show variation in abundance in serum exosomes from patients with preeclampsia. FASEB J. 2014;28:3703–19.

    Article  CAS  PubMed  Google Scholar 

  84. Zhang Y, Le T, Grabau R, Mohseni Z, Kim H, Natale DR, Feng L, Pan H, Yang H. TMEM16F phospholipid scramblase mediates trophoblast fusion and placental development. Sci Adv. 2020;6:eaba0310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Moller AM, Delaisse JM, Soe K. Osteoclast fusion: time-lapse reveals involvement of CD47 and Syncytin-1 at different stages of nuclearity. J Cell Physiol. 2017;232:1396–403.

    Article  CAS  PubMed  Google Scholar 

  86. Soe K, Andersen TL, Hobolt-Pedersen AS, Bjerregaard B, Larsson LI, Delaisse JM. Involvement of human endogenous retroviral syncytin-1 in human osteoclast fusion. Bone. 2011;48:837–46.

    Article  CAS  PubMed  Google Scholar 

  87. Bi P, Ramirez-Martinez A, Li H, Cannavino J, McAnally JR, Shelton JM, Sanchez-Ortiz E, Bassel-Duby R, Olson EN. Control of muscle formation by the fusogenic micropeptide myomixer. Science. 2017;356:323–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Millay DP, O’Rourke JR, Sutherland LB, Bezprozvannaya S, Shelton JM, Bassel-Duby R, Olson EN. Myomaker is a membrane activator of myoblast fusion and muscle formation. Nature. 2013;499:301–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Miyado K, Yoshida K, Yamagata K, Sakakibara K, Okabe M, Wang X, Miyamoto K, Akutsu H, Kondo T, Takahashi Y, et al. The fusing ability of sperm is bestowed by CD9-containing vesicles released from eggs in mice. Proc Natl Acad Sci U S A. 2008;105:12921–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Inoue N, Ikawa M, Isotani A, Okabe M. The immunoglobulin superfamily protein Izumo is required for sperm to fuse with eggs. Nature. 2005;434:234–8.

    Article  CAS  PubMed  Google Scholar 

  91. Brukman NG, Nakajima KP, Valansi C, Flyak K, Li X, Higashiyama T, Podbilewicz B. A novel function for the sperm adhesion protein IZUMO1 in cell-cell fusion. J Cell Biol. 2023;222:e202207147.

    Article  CAS  PubMed  Google Scholar 

  92. Mameli G, Poddighe L, Mei A, Uleri E, Sotgiu S, Serra C, Manetti R, Dolei A. Expression and activation by Epstein Barr virus of human endogenous retroviruses-W in blood cells and astrocytes: inference for multiple sclerosis. PLoS One. 2012;7:e44991.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Uleri E, Mei A, Mameli G, Poddighe L, Serra C, Dolei A. HIV Tat acts on endogenous retroviruses of the W family and this occurs via Toll-like receptor 4: inference for neuroAIDS. AIDS. 2014;28:2659–70.

    Article  CAS  PubMed  Google Scholar 

  94. Marston JL, Greenig M, Singh M, Bendall ML, Duarte RRR, Feschotte C, Iniguez LP, Nixon DF. SARS-CoV-2 infection mediates differential expression of human endogenous retroviruses and long interspersed nuclear elements. JCI Insight. 2021;6:e147170.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Martin SJ, Reutelingsperger CP, McGahon AJ, Rader JA, van Schie RC, LaFace DM, Green DR. Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl. J Exp Med. 1995;182:1545–56.

    Article  CAS  PubMed  Google Scholar 

  96. Whitlock JM, Yu K, Cui YY, Hartzell HC. Anoctamin 5/TMEM16E facilitates muscle precursor cell fusion. J Gen Physiol. 2018;150:1498–509.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Gyobu S, Miyata H, Ikawa M, Yamazaki D, Takeshima H, Suzuki J, Nagata S. A role of TMEM16E carrying a scrambling domain in sperm motility. Mol Cell Biol. 2016;36:645–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Zhang Y, Liang P, Yang L, Shan KZ, Feng L, Chen Y, Liedtke W, Coyne CB, Yang H. Functional coupling between TRPV4 channel and TMEM16F modulates human trophoblast fusion. Elife. 2022;11:e78840.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Rival CM, Xu W, Shankman LS, Morioka S, Arandjelovic S, Lee CS, Wheeler KM, Smith RP, Haney LB, Isakson BE, et al. Phosphatidylserine on viable sperm and phagocytic machinery in oocytes regulate mammalian fertilization. Nat Commun. 2019;10:4456.

    Article  PubMed  PubMed Central  Google Scholar 

  100. 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.

    Article  CAS  PubMed  Google Scholar 

  101. Hochreiter-Hufford AE, Lee CS, Kinchen JM, Sokolowski JD, Arandjelovic S, Call JA, Klibanov AL, Yan Z, Mandell JW, Ravichandran KS. Phosphatidylserine receptor BAI1 and apoptotic cells as new promoters of myoblast fusion. Nature. 2013;497:263–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Martin SG. Role and organization of the actin cytoskeleton during cell-cell fusion. Semin Cell Dev Biol. 2016;60:121–6.

    Article  CAS  PubMed  Google Scholar 

  103. Eitzen G. Actin remodeling to facilitate membrane fusion. Biochim Biophys Acta. 2003;1641:175–81.

    Article  CAS  PubMed  Google Scholar 

  104. Shen Q, Rahn JJ, Zhang J, Gunasekera N, Sun X, Shaw AR, Hendzel MJ, Hoffman P, Bernier A, Hugh JC. MUC1 initiates Src-CrkL-Rac1/Cdc42-mediated actin cytoskeletal protrusive motility after ligating intercellular adhesion molecule-1. Mol Cancer Res. 2008;6:555–67.

    Article  CAS  PubMed  Google Scholar 

  105. Gruenbaum-Cohen Y, Harel I, Umansky KB, Tzahor E, Snapper SB, Shilo BZ, Schejter ED. The actin regulator N-WASp is required for muscle-cell fusion in mice. Proc Natl Acad Sci U S A. 2012;109:11211–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Massarwa R, Carmon S, Shilo BZ, Schejter ED. WIP/WASp-based actin-polymerization machinery is essential for myoblast fusion in Drosophila. Dev Cell. 2007;12:557–69.

    Article  CAS  PubMed  Google Scholar 

  107. Zhang Y, Yang Y, Zhu Z, Ou G. WASP-Arp2/3-dependent actin polymerization influences fusogen localization during cell-cell fusion in Caenorhabditiselegans embryos. Biol Open. 2017;6:1324–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Abmayr SM, Pavlath GK. Myoblast fusion: lessons from flies and mice. Development. 2012;139:641–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Molinie N, Gautreau A. The Arp2/3 regulatory system and its deregulation in cancer. Physiol Rev. 2018;98:215–38.

    Article  CAS  PubMed  Google Scholar 

  110. Melzer C, von der Ohe J, Hass R. Involvement of actin cytoskeletal components in breast cancer cell fusion with human mesenchymal stroma/stem-like cells. Int J Mol Sci. 2019;20:876.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Hass R. Role of MSC in the tumor microenvironment. Cancers (Basel). 2020;12:2107.

    Article  CAS  PubMed  Google Scholar 

  112. Zhou X, Platt JL. Molecular and cellular mechanisms of Mammalian cell fusion. Adv Exp Med Biol. 2011;713:33–64.

    Article  CAS  PubMed  Google Scholar 

  113. Hass R, von der Ohe J, Ungefroren H. Impact of the tumor microenvironment on tumor heterogeneity and consequences for cancer cell plasticity and stemness. Cancers (Basel). 2020;12:3716.

    Article  CAS  PubMed  Google Scholar 

  114. Wakeling WF, Greetham J, Bennett DC. Efficient spontaneous fusion between some co-cultured cells, especially murine melanoma cells. Cell Biol Int. 1994;18:207–10.

    Article  CAS  PubMed  Google Scholar 

  115. Miroshnychenko D, Baratchart E, Ferrall-Fairbanks MC, Velde RV, Laurie MA, Bui MM, Tan AC, Altrock PM, Basanta D, Marusyk A. Spontaneous cell fusions as a mechanism of parasexual recombination in tumour cell populations. Nat Ecol Evol. 2021;5:379–91.

    Article  PubMed  Google Scholar 

  116. Fortuna MB, Dewey MJ, Furmanski P. Cell fusion in tumor development and progression: occurrence of cell fusion in primary methylcholanthrene-induced tumorigenesis. Int J Cancer. 1989;44:731–7.

    Article  CAS  PubMed  Google Scholar 

  117. Lu X, Kang Y. Efficient acquisition of dual metastasis organotropism to bone and lung through stable spontaneous fusion between MDA-MB-231 variants. Proc Natl Acad Sci U S A. 2009;106:9385–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Yan B, Wang J, Liu L. Chemotherapy promotes tumour cell hybridization in vivo. Tumour Biol. 2015;37:5025–30.

    Article  PubMed  Google Scholar 

  119. Melzer C, von der Ohe J, Hass R. In vivo cell fusion between mesenchymal stroma/stem-like cells and breast cancer cells. Cancers (Basel). 2019;11:185.

    Article  CAS  PubMed  Google Scholar 

  120. Ramakrishnan M, Mathur SR, Mukhopadhyay A. Fusion derived epithelial cancer cells express hematopoietic markers and contribute to stem cell and migratory phenotype in ovarian carcinoma. Cancer Res. 2013;73:5360–70.

    Article  CAS  PubMed  Google Scholar 

  121. Melzer C, von der Ohe J, Hass R. In vitro fusion of normal and neoplastic breast epithelial cells with human mesenchymal stroma/stem cells (MSC) partially involves TNF receptor signaling. Stem Cells. 2018;36:977–89.

    Article  CAS  PubMed  Google Scholar 

  122. Pawelek JM, Chakraborty AK. Fusion of tumour cells with bone marrow-derived cells: a unifying explanation for metastasis. Nat Rev Cancer. 2008;8:377–86.

    Article  CAS  PubMed  Google Scholar 

  123. Duelli D, Lazebnik Y. Cell fusion: a hidden enemy? Cancer Cell. 2003;3:445–8.

    Article  CAS  PubMed  Google Scholar 

  124. Clawson GA, Matters GL, Xin P, Imamura-Kawasawa Y, Du Z, Thiboutot DM, Helm KF, Neves RI, Abraham T. Macrophage-tumor cell fusions from peripheral blood of melanoma patients. PLoS One. 2015;10:e0134320.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Kemeny LV, Kurgyis Z, Buknicz T, Groma G, Jakab A, Zanker K, Dittmar T, Kemeny L, Nemeth IB. Melanoma cells can adopt the phenotype of stromal fibroblasts and macrophages by spontaneous cell fusion in vitro. Int J Mol Sci. 2016;17:826.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Lizier M, Anselmo A, Mantero S, Ficara F, Paulis M, Vezzoni P, Lucchini F, Pacchiana G. Fusion between cancer cells and macrophages occurs in a murine model of spontaneous neu+ breast cancer without increasing its metastatic potential. Oncotarget. 2016;7:60793.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Shabo I, Midtbo K, Andersson H, Akerlund E, Olsson H, Wegman P, Gunnarsson C, Lindstrom A. Macrophage traits in cancer cells are induced by macrophage-cancer cell fusion and cannot be explained by cellular interaction. BMC Cancer. 2015;15:922.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Hass R, von der Ohe J, Ungefroren H. Potential role of MSC/cancer cell fusion and EMT for breast cancer stem cell formation. Cancers (Basel). 2019;11:1432.

    Article  CAS  PubMed  Google Scholar 

  129. Dornen J, Myklebost O, Dittmar T. 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;21:9636.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Sottile F, Aulicino F, Theka I, Cosma MP. Mesenchymal stem cells generate distinct functional hybrids in vitro via cell fusion or entosis. Sci Rep. 2016;6:36863.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–9.

    Article  CAS  PubMed  Google Scholar 

  132. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539–45.

    Article  CAS  PubMed  Google Scholar 

  133. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44.

    Article  CAS  PubMed  Google Scholar 

  134. Dittmar T, Zanker KS. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Dornen J, Sieler M, Weiler J, Keil S, Dittmar T. Cell fusion-mediated tissue regeneration as an inducer of polyploidy and aneuploidy. Int J Mol Sci. 2020;21:1811.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Hass R, Otte A. Mesenchymal stem cells as all-round supporters in a normal and neoplastic microenvironment. Cell Commun Signal. 2012;10:26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Melzer C, Yang Y, Hass R. Interaction of MSC with tumor cells. Cell Commun Signal. 2016;14:20.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Duncan AW, Hickey RD, Paulk NK, Culberson AJ, Olson SB, Finegold MJ, Grompe M. Ploidy reductions in murine fusion-derived hepatocytes. PLoS Genet. 2009;5:e1000385.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Duncan AW, Taylor MH, Hickey RD, Hanlon Newell AE, Lenzi ML, Olson SB, Finegold MJ, Grompe M. The ploidy conveyor of mature hepatocytes as a source of genetic variation. Nature. 2010;467:707–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Bjerkvig R, Tysnes BB, Aboody KS, Najbauer J, Terzis AJ. Opinion: the origin of the cancer stem cell: current controversies and new insights. Nat Rev Cancer. 2005;5:899–904.

    Article  CAS  PubMed  Google Scholar 

  141. Ly P, Cleveland DW. Rebuilding chromosomes after catastrophe: emerging mechanisms of chromothripsis. Trends Cell Biol. 2017;27:917–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Rode A, Maass KK, Willmund KV, Lichter P, Ernst A. Chromothripsis in cancer cells: an update. Int J Cancer. 2016;138:2322–33.

    Article  CAS  PubMed  Google Scholar 

  143. Bakhoum SF, Kabeche L, Compton DA, Powell SN, Bastians H. Mitotic DNA damage response: at the crossroads of structural and numerical cancer chromosome instabilities. Trends Cancer. 2017;3:225–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. He B, Gnawali N, Hinman AW, Mattingly AJ, Osimani A, Cimini D. Chromosomes missegregated into micronuclei contribute to chromosomal instability by missegregating at the next division. Oncotarget. 2019;10:2660–74.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Janssen A, van der Burg M, Szuhai K, Kops GJ, Medema RH. Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science. 2011;333:1895–8.

    Article  CAS  PubMed  Google Scholar 

  146. Passerini V, Ozeri-Galai E, de Pagter MS, Donnelly N, Schmalbrock S, Kloosterman WP, Kerem B, Storchova Z. The presence of extra chromosomes leads to genomic instability. Nat Commun. 2016;7:10754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Rachkovsky M, Sodi S, Chakraborty A, Avissar Y, Bolognia J, McNiff JM, Platt J, Bermudes D, Pawelek J. Melanoma x macrophage hybrids with enhanced metastatic potential. Clin Exp Metastasis. 1998;16:299–312.

    Article  CAS  PubMed  Google Scholar 

  148. Melzer C, von der Ohe J, Hass R. MSC stimulate ovarian tumor growth during intercellular communication but reduce tumorigenicity after fusion with ovarian cancer cells. Cell Commun Signal. 2018;16:67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Melzer C, Ohe JV, Luo T, Hass R. Spontaneous fusion of MSC with breast cancer cells can generate tumor dormancy. Int J Mol Sci. 2021;22:5930.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Chakraborty A, Lazova R, Davies S, Backvall H, Ponten F, Brash D, Pawelek J. Donor DNA in a renal cell carcinoma metastasis from a bone marrow transplant recipient. Bone Marrow Transpl. 2004;34:183–6.

    Article  CAS  Google Scholar 

  151. LaBerge G, Duvall E, Grasmick Z, Haedicke K, Galan A, Pawelek J. A melanoma patient with macrophage-cancer cell hybrids in the primary tumor, a lymph node metastasis and a brain metastasis. Cancer Genet. 2021;256–257:162–4.

    Article  PubMed  Google Scholar 

  152. LaBerge GS, Duvall E, Grasmick Z, Haedicke K, Pawelek J. A melanoma lymph node metastasis with a donor-patient hybrid genome following bone marrow transplantation: a second case of leucocyte-tumor cell hybridization in cancer metastasis. PLoS One. 2017;12:e0168581.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Lazova R, Laberge GS, Duvall E, Spoelstra N, Klump V, Sznol M, Cooper D, Spritz RA, Chang JT, 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. 2013;8:e66731.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Yilmaz Y, Lazova R, Qumsiyeh M, Cooper D, Pawelek J. Donor Y chromosome in renal carcinoma cells of a female BMT recipient: visualization of putative BMT-tumor hybrids by FISH. Bone Marrow Transpl. 2005;35:1021–4.

    Article  CAS  Google Scholar 

  155. Manjunath Y, Mitchem JB, Suvilesh KN, Avella DM, Kimchi ET, Staveley-O’Carroll KF, Deroche CB, Pantel K, Li G, Kaifi JT. Circulating giant tumor-macrophage fusion cells are independent prognosticators in non-small cell lung cancer patients. J Thorac Oncol. 2020;15:1460–71.

    Article  CAS  PubMed  Google Scholar 

  156. Dietz MS, Sutton TL, Walker BS, Gast CE, Zarour L, Sengupta SK, Swain JR, Eng J, Parappilly M, Limbach K, et al. Relevance of circulating hybrid cells as a non-invasive biomarker for myriad solid tumors. Sci Rep. 2021;11:13630.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Clawson GA, Kimchi E, Patrick SD, Xin P, Harouaka R, Zheng S, Berg A, Schell T, Staveley-O’Carroll KF, Neves RI, et al. Circulating tumor cells in melanoma patients. PLoS One. 2012;7:e41052.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Ruano APC, Gadelha Guimaraes AP, Braun AC, Flores B, Tariki MS, Abdallah EA, Torres JA, Nunes DN, Tirapelli B, de Lima VCC, et al. Fusion cell markers in circulating tumor cells from patients with high-grade ovarian serous carcinoma. Int J Mol Sci. 2022;23:14687.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Melzer C, von der Ohe J, Hass R. Concise review: crosstalk of mesenchymal stroma/stem-like cells with cancer cells provides therapeutic potential. Stem Cells. 2018;36:951–68.

    Article  CAS  PubMed  Google Scholar 

  160. Janssen A, Medema RH. Entosis: aneuploidy by invasion. Nat Cell Biol. 2011;13:199–201.

    Article  CAS  PubMed  Google Scholar 

  161. Xia P, Wang S, Guo Z, Yao X. Emperipolesis, entosis and beyond: dance with fate. Cell Res. 2008;18:705–7.

    Article  CAS  PubMed  Google Scholar 

  162. Chen YC, Gonzalez ME, Burman B, Zhao X, Anwar T, Tran M, Medhora N, Hiziroglu AB, Lee W, Cheng YH, et al. Mesenchymal stem/stromal cell engulfment reveals metastatic advantage in breast cancer. Cell Rep. 2019;27(3916–3926):e3915.

    Google Scholar 

  163. Zhou F, Li M, Wei Y, Lin K, Lu Y, Shen J, Johanning GL, Wang-Johanning F. Activation of HERV-K Env protein is essential for tumorigenesis and metastasis of breast cancer cells. Oncotarget. 2016;7:84093–117.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Li M, Radvanyi L, Yin B, Rycaj K, Li J, Chivukula R, Lin K, Lu Y, Shen J, Chang DZ, et al. Downregulation of human endogenous retrovirus type K (HERV-K) viral env RNA in pancreatic cancer cells decreases cell proliferation and tumor growth. Clin Cancer Res. 2017;23:5892–911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Lemaitre C, Tsang J, Bireau C, Heidmann T, Dewannieux M. A human endogenous retrovirus-derived gene that can contribute to oncogenesis by activating the ERK pathway and inducing migration and invasion. PLoS Pathog. 2017;13:e1006451.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Dittmar T, Hass R. Extracellular events involved in cancer cell-cell fusion. Int J Mol Sci. 2022;23:16071.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. McNally AK, Anderson JM. Macrophage fusion and multinucleated giant cells of inflammation. Adv Exp Med Biol. 2011;713:97–111.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

not applicable

Funding

Open Access funding enabled and organized by Projekt DEAL. This work was supported by a grant from the Niedersächsische Krebsgesellschaft e.V. with respect to the NDR charity campaign ‘Hand in Hand für Norddeutschland 2019’ to Ralf Hass.

Author information

Authors and Affiliations

Authors

Contributions

TD and RH designed the study, analysed and interpreted the data and wrote the manuscript. TD and RH prepared all figures. Both authors have read and agreed to the published version of the manuscript. TD and RH are both corresponding authors.

Corresponding authors

Correspondence to Thomas Dittmar or Ralf Hass.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dittmar, T., Hass, R. Intrinsic signalling factors associated with cancer cell-cell fusion. Cell Commun Signal 21, 68 (2023). https://doi.org/10.1186/s12964-023-01085-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-023-01085-5

Keywords