Next Article in Journal
Recent Advances in Ovarian Cancer: Therapeutic Strategies, Potential Biomarkers, and Technological Improvements
Next Article in Special Issue
The E1a Adenoviral Gene Upregulates the Yamanaka Factors to Induce Partial Cellular Reprogramming
Previous Article in Journal
Hypothesis: Emerging Roles for Aryl Hydrocarbon Receptor in Orchestrating CoV-2-Related Inflammation
Previous Article in Special Issue
Roles of Podoplanin in Malignant Progression of Tumor
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epithelial Mesenchymal Transition (EMT) and Associated Invasive Adhesions in Solid and Haematological Tumours

School of Life Sciences and Health, University of Roehampton, London SW15 4JD, UK
*
Author to whom correspondence should be addressed.
Cells 2022, 11(4), 649; https://doi.org/10.3390/cells11040649
Submission received: 13 January 2022 / Revised: 7 February 2022 / Accepted: 8 February 2022 / Published: 13 February 2022

Abstract

:
In solid tumours, cancer cells that undergo epithelial mesenchymal transition (EMT) express characteristic gene expression signatures that promote invasive migration as well as the development of stemness, immunosuppression and drug/radiotherapy resistance, contributing to the formation of currently untreatable metastatic tumours. The cancer traits associated with EMT can be controlled by the signalling nodes at characteristic adhesion sites (focal contacts, invadopodia and microtentacles) where the regulation of cell migration, cell cycle progression and pro-survival signalling converge. In haematological tumours, ample evidence accumulated during the last decade indicates that the development of an EMT-like phenotype is indicative of poor disease prognosis. However, this EMT phenotype has not been directly linked to the assembly of specific forms of adhesions. In the current review we discuss the role of EMT in haematological malignancies and examine its possible link with the progression towards more invasive and aggressive forms of these tumours. We also review the known types of adhesions formed by haematological malignancies and speculate on their possible connection with the EMT phenotype. We postulate that understanding the architecture and regulation of EMT-related adhesions will lead to the discovery of new therapeutic interventions to overcome disease progression and resistance to therapies.

1. Introduction

Epithelium integrity is maintained by the apical-basal polarity of epithelial cells generated by adhesions at cell-cell junctions and with the basal lamina. However, under specific physiological conditions, epithelial cells lose the contacts with neighbouring cells and the subjacent matrix, adopting a highly motile mesenchymal phenotype. This cell behaviour is called epithelial-mesenchymal transition (EMT) and it is critical for tissue morphogenesis during embryonic development and in adulthood for wound healing [1,2]. At the molecular level, EMT is characterised by the downregulation of the cell adhesion protein E-cadherin, leading to the disassembly of intercellular junctions and the initial dissociation of epithelia. In parallel, cells undergo major cytoskeletal rearrangements involving the downregulation of keratins and the upregulation of vimentin, which facilitates the highly migratory behaviour in the dissociated cells [2]. Several well characterised transcription factors (TFs) are upregulated during EMT such as SNAIL/SNAI1, SLUG/SNAI2, TWIST and the ZEB family of TFs, which orchestrate the downregulation of E-cadherin [3] and the upregulation and activation of N-cadherin and the intermediate filament vimentin [4]. Reciprocally, upregulated vimentin can promote the expression or the activity of EMT TFs and other signalling pathways favouring an invasive phenotype [5,6].
EMT is also thought to play a central role in cancer progression by facilitating the invasive behaviour of tumour cells, enabling tissue infiltration and the formation of metastatic foci [7,8]. Consequently, expression of EMT markers is associated with an aggressive malignant phenotype and correlates with the poor prognosis of cancer patients [8,9,10]. Over the last decade, development of an EMT phenotype in tumour cells in cancers of epithelial origin (carcinomas) has also been associated with acquisition of further malignant traits including cancer stem cell properties [11,12], immunosuppression [13,14], as well as resistance to therapeutic agents [15,16,17,18,19] and to radiotherapy [20,21].
The induction and maintenance of EMT is highly regulated by extracellular signals during embryonic development, tissue repair and tumour progression. These include chemical cues such as cytokines, chemokines, the presence of reactive oxygen species (ROS) [22] and hypoxia [23,24], as well as mechanochemical signals provided by changes in the rigidity of the surrounding extracellular matrix [25,26]. Specifically in tumours, autocrine and paracrine stimulation of cancer cells by cytokines such as TGFβ1 induce EMT while promoting an immunosuppressive microenvironment [27,28]. Additionally, macrophages and cancer associated fibroblasts present in the tumour microenvironment can secrete classical EMT-inducing cytokines/chemokines including HGF, SDF1α, EGF, PDGF as well as TGFβ1 [24,28]. Cytokines also secreted by the tumour stroma and firstly identified as pro-survival, immunosuppressive or as chemotactic signals to recruit immune cells to the tumour microenvironment are now known to also induce EMT in cancer cells. Some examples are IL-6, IL-8, IL-10 and TNFα [27,29]. The hypoxic microenvironment generated in growing tumours prior to vascularisation is another extensively studied factor that promotes EMT in cancer cells [23,30,31]. Hypoxia drives EMT by triggering various signalling pathways [32], the most commonly known being the stabilisation of Hypoxia Inducible Factor-1 α (HIF-1α) [33], that results in increased activity of the EMT TFs SLUG [33], SNAIL [32] and TWIST [34].
EMT can be reversed by a process known as mesenchymal-epithelial transition (MET). The coordination of EMT and MET is critical in both physiological and pathological conditions [2]. During the formation of metastatic foci, EMT facilitates the dissemination of cancer cells from the primary tumour to the blood and lymph circulation systems. After extravasation of circulating tumour cells to a secondary organ, they undergo MET to convert from a non- or low proliferative, migratory phenotype to a proliferative type that will generate a secondary tumour. However, the current scientific consensus in the field establishes that only under some very specific circumstances during development do epithelial cells switch completely from an epithelial to a fully mesenchymal phenotype [2]. It is currently agreed that there is a spectrum of possible intermediate stages characteristic of specific EMT programmes that encompass the loss of characteristic features of epithelial cells while acquiring the traits of mesenchymal cells, resulting in a more invasive/motile phenotype [2]. It remains undetermined whether the molecular and cellular EMT features in these programs comprise discrete stages of a linear spectrum of EMT- MET conversion or whether, instead, they represent a continuum of possible non-epithelial states expressed depending on the biological context [2].
In cancer cells, gene mutations and/or the influence of the tumour stroma may induce patterns of expression of epithelial and mesenchymal markers resulting in particular EMT phenotypes [2,10,35]. This epithelial/mesenchymal plasticity spectrum provides tumours with a landscape of possible phenotypes with multiple associated functions [36] and capacity for adaptation to various microenvironments [37,38]. For example, circulating tumour cells express characteristic intermediate epithelial-mesenchymal traits [37,38] whereas once the cells colonise a secondary site, they undergo a further MET conversion [39,40]. Additionally, certain EMT profiles have been associated with the subsequent expression of gene signatures of tumour subpopulations in skin and breast carcinomas that may allow these cells to colonise specific organs [36]. This possible control of gene expression by distinct EMT profiles may infer cancer-stem cell [41,42] and/or drug resistant traits to subpopulations of tumour cells [13,15,16,17,18,19], leading to refractory disease and relapse in cancer patients.
A substantial body of evidence supports the central role of EMT in the invasive behaviour and the metastatic disease of carcinomas such as lung, prostate, colon, liver, pancreatic and breast cancers [24]. However, in the last decade it has become evident that an EMT behaviour is also present in non-epithelial solid tumours of mesenchymal origin and in haematological malignancies [9,10,23,43]. This EMT-like behaviour in mesenchymal tumours may echo the loss of polarity and increased migratory capacity of endothelial cells during tissue repair and angiogenesis in an EMT-related process named endothelial-mesenchymal transition [1,44].
This review will emphasize the development of EMT-phenotypes during tumour progression of haematological malignancies. We will discuss the current understanding of the specific molecular EMT features described in haematological cancers and propose possible associated adhesomes formed by haematological tumour cells that may facilitate this invasive and aggressive phenotype.

2. EMT in Haematological Malignancies

Haematopoietic cells derive from the mesoderm and, therefore, have a mesenchymal developmental origin, however, the expression of EMT-like signatures with upregulation of canonical mesenchymal markers has been described in all types of haematological malignancies (lymphomas, multiple myeloma and lymphoid and myeloid leukaemia) [10]. The current literature strongly indicates a clear correlation between the expression of these EMT signatures, or genetic abnormalities in EMT TFs, and poor prognosis of patients [9,45,46,47,48,49,50,51]. One of the most common characteristics is the expression of high levels of vimentin in various forms of aggressive haematological tumours [9,52,53]. However, the exact biological role of the EMT markers in haematopoietic cancers remains largely unstudied.
In particular, the actual correlation between the higher levels of EMT markers with a more motile phenotype, which is a key EMT feature, has not yet been studied in great depth in haematological malignancies in comparison to solid tumours. The majority of the studies have concentrated on various other tumour traits that may explain the correlation between expression of EMT signatures and the poor prognosis of patients. For example, the expression of EMT TFs has been associated with the repression of differentiation markers, correlating with more aggressive forms of haematological malignancies. Increased expression of ZEB1 promotes the methylation and downregulation of B-Cell Lymphoma protein 6 (BCL6), a key transcription factor that promotes differentiation of B cells and whose expression is associated with a benign profile [45]. A correlation has also been established between EMT and resistance to therapies. ZEB1 has been shown to promote drug resistance in mantle cell lymphoma by activating proliferation-associated genes while repressing pro-apoptotic ones and regulating the expression levels of membrane transporters involved in drug influx and efflux [46]. The upregulation of TWIST1 levels in chronic myeloid leukaemia (CML) promotes resistance to the therapeutic drug imatinib [50]. However, the cellular and molecular TWIST1-dependent mechanisms that may explain the lack of response to this drug have not yet been elucidated. Similarly, expression of EMT TFs in multiple myeloma cells in response to cues from the tumour microenvironment correlates with resistance to the clinical drugs dexamethasone and Velcade (bortezomib) [54].
Only a few of these studies have addressed the connection between the expression of EMT markers and increased cell migration. In paediatric anaplastic large cell lymphoma cells, expression of the oncogenic protein anaplastic lymphoma kinase (ALK) correlated with expression of higher levels of TWIST1. Downregulation of TWIST1 in these cells inhibited invasive migration and reinstated the efficacy of therapeutic drugs [49]. In MLL-AF9 driven acute myeloid leukaemia (AML), poor prognosis in patients correlated with expression of EMT markers and experimental downregulation of ZEB1 in AML cells inhibited the invasive capacity of this aggressive cancer [9].
Multiple myeloma is the haematological tumour where the EMT phenotype has been studied in more depth, and there is substantial evidence of the acquisition of a migratory phenotype orchestrated by the EMT program [23,43,51,54]. Multiple myeloma results from the accumulation of malignant plasma cells in disseminated tumour foci within the bone marrow. In 2012, the Ghobrial lab demonstrated that during tumour progression, hypoxia induced an EMT program in multiple myeloma cells that resulted in increased mobilisation and formation of further bone marrow foci [23]. This program was the result of the upregulation of HIF-1α leading to expression of SNAIL and downregulation of E-cadherin [23]. It was then proposed that multiple myeloma may be envisaged as a metastatic disease where progression results from continuing trafficking of myeloma cells from initial proliferating tumours in the bone marrow that acquire a hypoxic-driven EMT phenotype as they expand, leading to mobilisation of myeloma cells undergoing EMT [43]. Mobilised myeloma cells into the circulation express cell adhesion molecules such as β7 integrins [55] that would facilitate the interaction of circulating cells with the bone marrow endothelium and the extravasation into a new site. The re-entry into the bone marrow in this vascularised normoxic environment would repress the expression of HIF-1 α, silencing the EMT program and promoting a transition similar to MET in epithelial tumours, leading to the proliferation of myeloma cells and development of a new tumour [23]. Additional factors that may lead to the mobilisation of myeloma cells through the acquisition of an EMT-phenotype include the increased concentration of cytokines such as IL-6, TNFα, VEGF, HGF and IGF-1 in the tumour microenvironment during the symptomatic phase of the disease [43]. More recently, further evidence shows that cytogenetic abnormalities associated with a poor prognosis in multiple myeloma [51] as well as the composition of the extracellular matrix of the myeloma bone marrow microenvironment [54] promote an EMT invasive phenotype in these cancer cells.
Taken together, the studies in haematological malignancies indicate a compelling connection between expression of EMT markers and poor prognosis (Table 1). In addition, the acquisition of an invasive migratory phenotype has been corroborated in the limited number of in depth studies so far available (Table 1). However, the exact coordination between the levels of expression of EMT TFs and markers, and the migratory features of haematological cancer cells is practically unknown with exception of a few studies in multiple myeloma. More significantly, the exact adhesions and cytoskeletal configurations that may be assembled by haematological cancer cells undergoing EMT, have not been yet elucidated. In the next section, we review the structure, regulation and known roles of EMT-related adhesions in solid tumours and discuss the invasive adhesions so far described in haematological malignancies and their possible, although not yet proved, association with EMT.

3. Invasive Adhesions Formed by Cells Undergoing EMT in Solid Tumours: Are They Related to Adhesions Described in Haematological Malignancies?

Cell adhesion molecules and associated proteins, in addition to anchoring cells to the surrounding microenvironment and facilitating migration, are known to regulate gene expression and cell cycle progression [56,57,58,59]. For instance, integrin mediated signalling at adhesions modulates the activity of cell cycle check points such as the downregulation of CDK inhibitors and the induction of cyclin D during progression of the G1 phase [56,57,58,59]. Initiation of DNA synthesis requires sustained adhesion signalling [58,60] and transition from G2 to mitosis is also dependent on the level of formation of cell adhesions [61,62]. The recent finding of the direct binding between the cell cycle regulator CDK1 and talin, a key component of integrin-mediated adhesions, further reinforces a critical link between the organisation of cell adhesions and cell cycle progression [63]. Thus, during EMT the rearrangement of cell adhesions is likely to influence fundamental cell functions that determine the proliferative and invasive capacity of cancer cells and, therefore, tumour progression. Consequently, therapeutically targeting the signalling complexes at adhesion sites associated with EMT may not only repress the invasive capacity of cancer cells but it may also block survival and cell cycle progression, preventing tumour development and/or resistance to drug treatments [11,12,13,14,15,16,17,18,19,20,21].
We propose that to identify effective therapeutic targets for aggressive tumours, it is imperative to investigate the signalosomes at cell adhesions formed in cancer cells that have developed an invasive EMT phenotype. It is then essential to understand the architecture and regulation of EMT-related adhesions.

3.1. Adhesions Formed in Solid Tumours Undergoing EMT

The onset of the EMT process requires the reorganisation of filamentous actin (F-actin) microfilaments and microtubules in coordination with the reticular assembly of intermediate filaments of upregulated vimentin, while cells acquire migration polarity. Polarised cells display a distinctive protrusive leading edge and a contractile rear end that facilitates motility. Polymerisation of F-actin is the driving force that pushes the cell membrane at the leading edge, forming finger like protrusions containing parallel bundles of linear actin filaments called filopodia, or sheet like extensions shaped by the formation of a branched network of F-actin, called lamellipodia. Filopodia are generally thought to detect and integrate chemotactic signals that will determine the directionality of the cell movement [64] and they can mechanically facilitate cell displacement [65,66]. Lamellipodia are responsible for the substantial protrusive activity at the leading edge of mesenchymal migrating cells. Interestingly, filopodia can transform into lamellipodia by initiating dendritic actin nucleation on originally linear actin filaments, showing that these structures cooperate and are inter-convertible [67].
Nascent actin filaments polymerising at the cell membrane are shaped by various families of actin binding proteins. Formins are responsible for the linear polymerisation of F-actin in filopodia while fascin cross-links the formed filaments in parallel bundles [68,69]. Formation of filopodia [70,71] downstream of activation of TWIST1 and SNAIL1 [72] and the expression of formins [73,74] regulate the migration of cancer cells undergoing EMT. Additionally, the upregulation of formins contributes to metastasis formation [75], and high levels of fascin correlate with poor prognosis in a number of cancers including breast, lung, gastrointestinal and oral squamous cell carcinoma [64,76,77], suggesting that their role in EMT contributes to cell invasion and the formation of secondary tumour foci. Increased expression of fascin is also associated with the development of EMT-mediated drug resistance in hepatocellular carcinoma [78].
Equally relevant for the development of an effective EMT phenotype in cancer cells is the formation of lamellipodia. F-actin assembly at lamellipodia is controlled by the Arp 2/3 complex, a multimeric protein that promotes lateral branching of polymerising actin filaments. Activation of the actin nucleation-promoting factors N-WASP/WASP and Scar/WAVE, downstream of the Rho-GTPases Cdc42 and Rac, respectively [79,80], regulates the activity of the Arp2/3 complex. The formation of lamellipodia in cancer cells undergoing EMT [81], together with the expression of high levels of the Arp2/3 complex and WAVE2 correlates with poor prognosis in several types of tumours [82,83] and strongly suggests a significant role of lamellipodia in the invasive migration of metastatic cells.
Both filopodia and lamellipodia need to be stabilised by clusters of cell adhesion molecules called focal contacts that dynamically tether the membrane on the surface of neighbouring cells or on the surrounding extracellular matrix. In cells undergoing EMT, the cell adhesion molecules identified at focal contacts include proteoglycans (characteristically CD44) and integrins [84]. These initial cytoskeletal and/or adhesion rearrangements may further develop into additional adhesive and protrusive structures such as invadopodia or microtentacles.

3.1.1. Invasive Focal Contacts

Focal contacts that sustain filopodia and lamellipodia during EMT also show the capacity for local degradation of the extracellular matrix. CD44 clustered at focal contacts can form a complex with the metalloprotease (MMP) MT1-MMP at lamellipodia [85] suggesting a possible role of this receptor in organising and directing the dynamics of degradation sites. Additionally, CD44 creates a significant signalling node involved in EMT by regulating the activity of Src kinase [86]. Integrins have also been implicated in regulating the degradative function of focal contacts through the intracellular interaction with ILK, a kinase that facilitates the localised secretion of MMP9 [87].
Invasive focal contacts are also critical for the development of other cancer traits such as drug resistance. Targeting the cholesterol-rich domains (also called lipid rafts) that contribute to integrin clustering inhibits the signalling at adhesion sites, blocking migration while re-sensitizing drug-resistant cancer cells to therapeutic drugs [88]. These data reinforce the idea that targeting adhesions formed during EMT not only will prevent cancer cell migration but will also block the activation of signalling pathways involved in cancer cell survival, stemness and drug resistance.

3.1.2. Invadopodia

Invadopodia are F-actin dependent adhesions that protrude at the ventral surface of a cell, belonging to a larger family of adhesions called invadosomes, which also include podosomes [89,90,91]. Whilst similar, podosomes and invadopodia are structurally distinct, and form in different cell types. Podosomes are typically found in cells of monocytic lineage, endothelial cells [91,92,93], smooth muscle cells [94], and neuronal growth cones [95]; invadopodia are only observed in tumour cells and Src-transformed fibroblasts [91,96]. Invadopodia attach cells to the extracellular matrix while focally secreting MMPs. This process facilitates penetration of the extracellular matrix leading to subsequent cell migration and tissue invasion [97,98,99]. Formation of invadopodia is closely associated with the development of an EMT phenotype, and their function is to facilitate the infiltration of cancer cells across tissue barriers such as the endothelial basal lamina in blood vessels [100,101]. The structure of invadopodia comprises a protrusive F-actin-rich core assembled by cortactin, N-WASP, and the Arp2/3 complex, which is linked to the plasma membrane by cell adhesion proteins including integrins and CD44 [96,97]. Unlike podosomes, targeted microtubules and vimentin polymerisation can drive further extension of mature invadopodia into the extracellular matrix [102].
There is recent evidence indicating that the formation of invadopodia is coupled to cell cycle progression and EMT. Expression of invadopodia constituents is increased in G1, correlating with the enhanced capacity for degradation of the extracellular matrix of cancer cells [103]. Additionally, induction of the cell cycle checkpoint kinase 2 (Chk2) leads to cell cycle arrest in G2/M and suppression of TWIST1, SNAIL1, ZEB1 and vimentin expression, resulting in inhibition of invadopodia formation and cancer cell invasion in p53-mutated cells [104]. It is tempting to speculate that, reciprocally, invadopodia formation may have an impact in the regulation of the cell cycle during EMT, since vimentin is a key constituent of invadopodia and it has been shown to regulate cell cycle progression through its interaction with 14-3-3 proteins [105,106]. Vimentin availability and its post-translational modifications at invadopodia may affect its capacity for sequestration of free 14-3-3 and the formation of complexes mediated by these proteins, which in turn would affect the transition in G1/S and G2/M phases of the cell cycle [107].

3.1.3. Microtentacles

Cancer cells form microtubule-based cell long tubular membrane protrusions used for cell migration that receive myriad names within the established literature [108,109]. As these protrusions are all structurally and functionally related, in this review they will all be referred to as microtentacles (McTNs). McTNs are CD44 dependent membrane protrusions containing actin and vimentin in addition to microtubules [110,111]. McTNs are typically around 1 µm in diameter and allow for invasive migration within tissues [111]. They can also be assembled by detached circulating tumour cells [109], and they facilitate integrin-dependent adherence to the endothelium to penetrate endothelial cell-cell junctions [112,113]. Expression of MT1-MMP at McTNs facilitates degradation of endothelial cell-cell connections and the basement membrane allowing extravasation of the circulating tumour cells [114,115]. Although in some instances McTNs may be morphologically quite similar to canonical filopodia, they clearly differ structurally, as they contain vimentin, microtubules, and their actin content is cortical rather than central [116]. Additionally, actin polymerisation drives filopodia protrusion whereas the formation of McTNs requires a fine balance between microtubule and actin polymerisation, which promote McTNs extension and retraction, respectively [111]. Formation of McTNs has been linked directly to the development of EMT. Upregulation of TWIST1 downregulates the expression of the tubulin tyrosine ligase enzyme, resulting in an accumulation of detyrosinated α-tubulin. This leads to microtubule polymerisation, the extension of McTNs, and reattachment and infiltration of cancer cells across endothelial layers [113,117].
Assembly of McTNs and invadopodia are interchangeable in breast cancer cells. The activation of Src kinase promotes the diversion towards the formation of invadopodia, whereas the repression of Src activity leads to the assembly of McTNs [118]. This interconversion provides breast cancer cells with a plasticity to form metastatic foci that may result in drug resistance to Src inhibitors.

3.2. Adhesions Formed in Haematological Cancer Cells

In contrast to solid tumours, in haematological malignancies there is a paucity of published studies that clearly connect the development of the EMT phenotype with the formation of specific types of adhesions. However, there is strong evidence that haematological tumours can form adhesions that are structurally and/or functionally related to those formed in solid tumours undergoing EMT. The following adhesions below have been observed in haematological tumour cells and display close similarities to the EMT-related adhesions formed in solid tumours. We propose that these adhesions should be investigated in the future in the context of EMT-like processes in haematological malignancies.

3.2.1. Invasive Focal Contacts

In general, haematological cancer cells in in vitro culture do not readily attach to plastic, and only a small proportion of cells form small and labile focal contacts on extracellular matrix proteins such as fibronectin by clustering integrins and associated proteins. These adhesions contain many components present in focal contacts assembled by cancer cells undergoing EMT in solid tumours including Src kinases, FAK, p130Cas, Cbl, Abi1 and CrkL [119,120,121], but they lack some of the integrin-binding proteins that confer adhesion stability such as paxillin and vinculin [119,120]. However, in response to activation with 12-O-Tetradecanoylphorbol-13-acetate (TPA), phorbol-12-myristate-13-acetate (PMA) or certain cytokines, paxillin and vinculin are incorporated into these initial focal contacts (Figure 1), correlating with the increased attachment of cell cultures [120]. These mature focal contacts have also been shown to be sites of secretion of metalloproteases such as MMP2 and MMP9 [121], suggesting a role for remodelling and/or degradation of the extracellular matrix. Focal contacts have been proposed to be involved in cell adhesion mediated drug resistance to clinical drugs by mediating the interaction of leukaemia and multiple myeloma cells with the surrounding tissue stroma [55,122]. They have also been shown to modulate cell division by facilitating the ILK-dependent orientation of the centrosomes and the organisation of the mitotic spindle [123], indicating a role in cell proliferation. Targeting the disassembly of focal contacts in combination with cytotoxic drugs could improve therapies for relapse and refractory leukaemia patients [124,125,126].

3.2.2. Podosomes

Similarly to invadopodia, podosomes are sites of attachment and localised secretion of metalloproteinases that under normal physiological conditions mediate the migration of normal myeloid cells across tissue boundaries. In osteoclasts, podosomes facilitate migration on the bone surface and they can mature into an actin structure called the sealing zone that creates a confined area for secretion of acidic enzymes and proteases to dissolve the bone matrix. In vitro, TPA/PMA activated myeloid leukaemia cells can also form podosomes [127], which suggests a possible role of these structures in facilitating leukaemia cell motility. The factors that may determine the choice of formation of podosomes or focal contacts in similarly activated myeloid leukaemia cells remain to be investigated, as does whether these cell adhesions are interchangeable. Podosomes have been shown to facilitate the invasive motility of B-CLL cells across endothelial barriers [128]. In contrast to normal B-cells, which are completely devoid of podosomes, B-cell leukaemic cells can form degradative podosomes without any additional in vitro activation [128,129]. We have also observed the formation of podosomes with typical F-actin cores and a ring of integrin associated proteins such as talin in primary multiple myeloma cells seeded on fibronectin (Figure 2). Other authors have shown that myeloma cell lines and primary cells from myeloma patients can also be differentiated into osteoclastic cells and degrade the bone matrix, a feature that is not associated with normal B-plasma cells and that requires podosome formation [130,131].
The modulation of the signalosome in podosomes may not only affect the migratory capacity of haematological tumour cells but also the expression of genes involved in cell survival, proliferation and secretion of cytokines, critical mechanisms involved in cancer progression, immunosuppression and drug resistance. Several of the proteins involved in organising the F-actin architecture of podosomes, such as WASP [132] and the Arp2/3 complex [133,134], have also been shown to translocate to the nucleus where they regulate transcription of various cytokines [132] and the activity of RNA polymerase II [133], respectively. Interestingly, the regulation of podosomal components may be particular to haematological tumours in comparison to normal myeloid cells. For example, the phosphorylated form of the transcription factor STAT5 accumulates in podosomes formed by activated CML cells, whereas it localises to the nucleus in normal myeloid cells [135]. The exact role of the recruitment of STAT5 to podosomes is presently unknown, but it may regulate the expression of genes controlled by this TF. Taken together, the current studies suggest that in haematological malignancies, podosomes may facilitate invasive migration and they may be involved in the regulation of gene expression.

3.2.3. Podia

Podia are tubular membrane projections formed by protrusive activity and sustained by integrins and CD44 mediated adhesions that are observed in haematopoietic stem cells [136,137] as well as in leukaemia [137] and multiple myeloma cells [138] (Figure 3). These structures may be reminiscent of the McTNs assembled by cells undergoing EMT in solid tumours. They may also be related to protrusive structures named magnupodia recently described in polarised migrating haematopoietic stem cells [136].
Chemotactic and EMT promoting signals such as SDF1-α induce podia formation in leukaemia [140] and haematopoietic stem cells [136]. Podia assembly requires the activation of the degradative enzyme elastase in leukaemia cells [140] but the possible extracellular matrix degradation function of podia has not been demonstrated. It is thought that podia may contribute to the egress and exit of leukaemia cells from the circulation, leading to the formation of distant foci [140]. Interestingly, inhibition of elastase activity in leukaemia cells inhibited cell migration and promoted proliferation [140], a phenotype similar to that of cells undergoing MET. However, no clear link between podia formation and any stages of the EMT process has been demonstrated. The detailed molecular structure, exact function and the regulation of podia in haematological malignancies is largely undefined.
In summary, the possible connection between the formation of podia, invasive focal contacts or podosomes and the EMT phenotype in haematological malignancies remains to be elucidated. This is an emerging field of research in haematological tumours with significant potential to identify critical signalling nodes involved in invasive migration, metastatic behaviour, and drug resistance.

4. Final Remarks

In epithelial tumours, the upregulation of the core EMT TFs (ZEB family, SNAI1/2 and TWIST) and the intermediate filament vimentin are related to the development of different types of invasive adhesions such as degradative focal contacts, invadopodia and microtentacles. In haematological tumours, it remains largely unknown whether the expression of EMT TFs and the development of cytoskeletal and adhesive structures are coordinated similarly to epithelial tumours; or whether the expression changes are constrained in distinctive combinations according to the microenvironment and/or the type of haematological cancer. Notably, the formation of particular invasive adhesions associated with EMT remain largely unstudied in haematological cancers. Given the substantial evidence indicating that the development of EMT signatures correlates with poor prognosis in leukaemia, lymphoma and multiple myeloma, it will be critical in the coming years to determine the adhesions and cytoskeletal organisation associated with this phenotype. Several drugs that may interfere with the signalosomes at adhesion sites and/or cytoskeletal -remodelling are already available and could be used as migrastatics: compounds interfering with the migration of invasive cancers [141,142,143] to improve therapeutic interventions and avoid metastasis and relapse.
Determining the composition and regulation of EMT-associated adhesions in haematological malignancies will improve our understanding of the development not only of invasive migration but also the mechanisms leading to drug resistance and immunosuppression. It is anticipated that this research will result in the identification of new therapeutic targets that alone or in combination with cytotoxic therapies will allow for the treatment of patients refractory to current therapies.

Author Contributions

Both authors contributed in the literature review, discussion and manuscript writing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received funding from Cancer Research UK (Project numbers C34579/A14248 and C34579/A20784) and from the funds for research-led teaching activities from the University of Roehampton.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Ines M. Anton, Gareth E. Jones and Claire M. Wells for their very valuable critical revision of the manuscript.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Kovacic, J.C.; Mercader, N.; Torres, M.; Boehm, M.; Fuster, V. Epithelial-to-mesenchymal and endothelial-to-mesenchymal transition: From cardiovascular development to disease. Circulation 2012, 125, 1795–1808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Yang, J.; Antin, P.; Berx, G.; Blanpain, C.; Brabletz, T.; Bronner, M.; Campbell, K.; Cano, A.; Casanova, J.; Christofori, G.; et al. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2020, 21, 341–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Thiery, J.P.; Sleeman, J.P. Complex networks orchestrate epithelial-mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 2006, 7, 131–142. [Google Scholar] [CrossRef] [PubMed]
  4. Meng, J.; Chen, S.; Han, J.X.; Qian, B.; Wang, X.R.; Zhong, W.L.; Qin, Y.; Zhang, H.; Gao, W.F.; Lei, Y.Y.; et al. Twist1 Regulates Vimentin through Cul2 Circular RNA to Promote EMT in Hepatocellular Carcinoma. Cancer Res. 2018, 78, 4150–4162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Virtakoivu, R.; Mai, A.; Mattila, E.; De Franceschi, N.; Imanishi, S.Y.; Corthals, G.; Kaukonen, R.; Saari, M.; Cheng, F.; Torvaldson, E.; et al. Vimentin-ERK Signaling Uncouples Slug Gene Regulatory Function. Cancer Res. 2015, 75, 2349–2362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Satelli, A.; Li, S. Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell. Mol. Life Sci. CMLS 2011, 68, 3033–3046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Thiery, J.P. EMT: An Update. Methods Mol. Biol. 2021, 2179, 35–39. [Google Scholar] [CrossRef] [PubMed]
  8. Sato, M.; Shames, D.S.; Hasegawa, Y. Emerging evidence of epithelial-to-mesenchymal transition in lung carcinogenesis. Respirology 2012, 17, 1048–1059. [Google Scholar] [CrossRef]
  9. Stavropoulou, V.; Kaspar, S.; Brault, L.; Sanders, M.A.; Juge, S.; Morettini, S.; Tzankov, A.; Iacovino, M.; Lau, I.J.; Milne, T.A.; et al. MLL-AF9 Expression in Hematopoietic Stem Cells Drives a Highly Invasive AML Expressing EMT-Related Genes Linked to Poor Outcome. Cancer Cell 2016, 30, 43–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kahlert, U.D.; Joseph, J.V.; Kruyt, F.A.E. EMT- and MET-related processes in nonepithelial tumors: Importance for disease progression, prognosis, and therapeutic opportunities. Mol. Oncol. 2017, 11, 860–877. [Google Scholar] [CrossRef]
  11. Guo, W.; Keckesova, Z.; Donaher, J.L.; Shibue, T.; Tischler, V.; Reinhardt, F.; Itzkovitz, S.; Noske, A.; Zurrer-Hardi, U.; Bell, G.; et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell 2012, 148, 1015–1028. [Google Scholar] [CrossRef] [Green Version]
  12. Scheel, C.; Weinberg, R.A. Phenotypic plasticity and epithelial-mesenchymal transitions in cancer and normal stem cells? Int. J. Cancer 2011, 129, 2310–2314. [Google Scholar] [CrossRef] [Green Version]
  13. Terry, S.; Savagner, P.; Ortiz-Cuaran, S.; Mahjoubi, L.; Saintigny, P.; Thiery, J.P.; Chouaib, S. New insights into the role of EMT in tumor immune escape. Mol. Oncol. 2017, 11, 824–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Noman, M.Z.; Janji, B.; Abdou, A.; Hasmim, M.; Terry, S.; Tan, T.Z.; Mami-Chouaib, F.; Thiery, J.P.; Chouaib, S. The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology 2017, 6, e1263412. [Google Scholar] [CrossRef] [PubMed]
  15. Recondo, G.; Mezquita, L.; Facchinetti, F.; Planchard, D.; Gazzah, A.; Bigot, L.; Rizvi, A.Z.; Frias, R.L.; Thiery, J.P.; Scoazec, J.Y.; et al. Diverse Resistance Mechanisms to the Third-Generation ALK Inhibitor Lorlatinib in ALK-Rearranged Lung Cancer. Clin. Cancer Res. 2020, 26, 242–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sha, J.; Bai, Y.; Ngo, H.X.; Okui, T.; Kanno, T. Overview of Evidence-Based Chemotherapy for Oral Cancer: Focus on Drug Resistance Related to the Epithelial-Mesenchymal Transition. Biomolecules 2021, 11, 893. [Google Scholar] [CrossRef]
  17. Mirzaei, S.; Abadi, A.J.; Gholami, M.H.; Hashemi, F.; Zabolian, A.; Hushmandi, K.; Zarrabi, A.; Entezari, M.; Aref, A.R.; Khan, H.; et al. The involvement of epithelial-to-mesenchymal transition in doxorubicin resistance: Possible molecular targets. Eur. J. Pharm. 2021, 908, 174344. [Google Scholar] [CrossRef]
  18. Zhang, N.; Ng, A.S.; Cai, S.; Li, Q.; Yang, L.; Kerr, D. Novel therapeutic strategies: Targeting epithelial-mesenchymal transition in colorectal cancer. Lancet. Oncol. 2021, 22, e358–e368. [Google Scholar] [CrossRef]
  19. Zhang, B.; Li, Y.; Wu, Q.; Xie, L.; Barwick, B.; Fu, C.; Li, X.; Wu, D.; Xia, S.; Chen, J.; et al. Acetylation of KLF5 maintains EMT and tumorigenicity to cause chemoresistant bone metastasis in prostate cancer. Nat. Commun. 2021, 12, 1714. [Google Scholar] [CrossRef]
  20. Yu, X.; Wang, Q.; Liu, B.; Zhang, N.; Cheng, G. Vitamin D Enhances Radiosensitivity of Colorectal Cancer by Reversing Epithelial-Mesenchymal Transition. Front. Cell Dev. Biol. 2021, 9, 684855. [Google Scholar] [CrossRef]
  21. Galeaz, C.; Totis, C.; Bisio, A. Radiation Resistance: A Matter of Transcription Factors. Front. Oncol. 2021, 11, 662840. [Google Scholar] [CrossRef] [PubMed]
  22. Ramundo, V.; Giribaldi, G.; Aldieri, E. Transforming Growth Factor-beta and Oxidative Stress in Cancer: A Crosstalk in Driving Tumor Transformation. Cancers 2021, 13, 3093. [Google Scholar] [CrossRef] [PubMed]
  23. Azab, A.K.; Hu, J.; Quang, P.; Azab, F.; Pitsillides, C.; Awwad, R.; Thompson, B.; Maiso, P.; Sun, J.D.; Hart, C.P.; et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood 2012, 119, 5782–5794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ribatti, D.; Tamma, R.; Annese, T. Epithelial-Mesenchymal Transition in Cancer: A Historical Overview. Transl. Oncol. 2020, 13, 100773. [Google Scholar] [CrossRef] [PubMed]
  25. Dai, J.; Qin, L.; Chen, Y.; Wang, H.; Lin, G.; Li, X.; Liao, H.; Fang, H. Matrix stiffness regulates epithelial-mesenchymal transition via cytoskeletal remodeling and MRTF-A translocation in osteosarcoma cells. J. Mech. Behav. Biomed. Mater. 2019, 90, 226–238. [Google Scholar] [CrossRef] [PubMed]
  26. Fattet, L.; Jung, H.Y.; Matsumoto, M.W.; Aubol, B.E.; Kumar, A.; Adams, J.A.; Chen, A.C.; Sah, R.L.; Engler, A.J.; Pasquale, E.B.; et al. Matrix Rigidity Controls Epithelial-Mesenchymal Plasticity and Tumor Metastasis via a Mechanoresponsive EPHA2/LYN Complex. Dev. Cell 2020, 54, 302–316. [Google Scholar] [CrossRef]
  27. Chattopadhyay, I.; Ambati, R.; Gundamaraju, R. Exploring the Crosstalk between Inflammation and Epithelial-Mesenchymal Transition in Cancer. Mediat. Inflamm. 2021, 2021, 9918379. [Google Scholar] [CrossRef]
  28. Scheel, C.; Eaton, E.N.; Li, S.H.; Chaffer, C.L.; Reinhardt, F.; Kah, K.J.; Bell, G.; Guo, W.; Rubin, J.; Richardson, A.L.; et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell 2011, 145, 926–940. [Google Scholar] [CrossRef] [Green Version]
  29. Baram, T.; Rubinstein-Achiasaf, L.; Ben-Yaakov, H.; Ben-Baruch, A. Inflammation-Driven Breast Tumor Cell Plasticity: Stemness/EMT, Therapy Resistance and Dormancy. Front. Oncol. 2020, 10, 614468. [Google Scholar] [CrossRef]
  30. Tao, J.; Yang, G.; Zhou, W.; Qiu, J.; Chen, G.; Luo, W.; Zhao, F.; You, L.; Zheng, L.; Zhang, T.; et al. Targeting hypoxic tumor microenvironment in pancreatic cancer. J. Hematol. Oncol. 2021, 14, 14. [Google Scholar] [CrossRef]
  31. Gonzalez-Gonzalez, R.; Ortiz-Sarabia, G.; Molina-Frechero, N.; Salas-Pacheco, J.M.; Salas-Pacheco, S.M.; Lavalle-Carrasco, J.; Lopez-Verdin, S.; Tremillo-Maldonado, O.; Bologna-Molina, R. Epithelial-Mesenchymal Transition Associated with Head and Neck Squamous Cell Carcinomas: A Review. Cancers 2021, 13, 3027. [Google Scholar] [CrossRef]
  32. Choi, B.J.; Park, S.A.; Lee, S.Y.; Cha, Y.N.; Surh, Y.J. Hypoxia induces epithelial-mesenchymal transition in colorectal cancer cells through ubiquitin-specific protease 47-mediated stabilization of Snail: A potential role of Sox9. Sci. Rep. 2017, 7, 15918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lin, C.W.; Wang, L.K.; Wang, S.P.; Chang, Y.L.; Wu, Y.Y.; Chen, H.Y.; Hsiao, T.H.; Lai, W.Y.; Lu, H.H.; Chang, Y.H.; et al. Daxx inhibits hypoxia-induced lung cancer cell metastasis by suppressing the HIF-1alpha/HDAC1/Slug axis. Nat. Commun. 2016, 7, 13867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Yang, M.H.; Wu, M.Z.; Chiou, S.H.; Chen, P.M.; Chang, S.Y.; Liu, C.J.; Teng, S.C.; Wu, K.J. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat. Cell Biol. 2008, 10, 295–305. [Google Scholar] [CrossRef] [PubMed]
  35. Chaffer, C.L.; San Juan, B.P.; Lim, E.; Weinberg, R.A. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016, 35, 645–654. [Google Scholar] [CrossRef] [PubMed]
  36. Pastushenko, I.; Brisebarre, A.; Sifrim, A.; Fioramonti, M.; Revenco, T.; Boumahdi, S.; Van Keymeulen, A.; Brown, D.; Moers, V.; Lemaire, S.; et al. Identification of the tumour transition states occurring during EMT. Nature 2018, 556, 463–468. [Google Scholar] [CrossRef] [PubMed]
  37. Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Schliekelman, M.J.; Taguchi, A.; Zhu, J.; Dai, X.; Rodriguez, J.; Celiktas, M.; Zhang, Q.; Chin, A.; Wong, C.H.; Wang, H.; et al. Molecular portraits of epithelial, mesenchymal, and hybrid States in lung adenocarcinoma and their relevance to survival. Cancer Res. 2015, 75, 1789–1800. [Google Scholar] [CrossRef] [Green Version]
  39. Ocana, O.H.; Corcoles, R.; Fabra, A.; Moreno-Bueno, G.; Acloque, H.; Vega, S.; Barrallo-Gimeno, A.; Cano, A.; Nieto, M.A. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 2012, 22, 709–724. [Google Scholar] [CrossRef] [Green Version]
  40. Tsai, J.H.; Donaher, J.L.; Murphy, D.A.; Chau, S.; Yang, J. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell 2012, 22, 725–736. [Google Scholar] [CrossRef] [Green Version]
  41. Ye, X.; Tam, W.L.; Shibue, T.; Kaygusuz, Y.; Reinhardt, F.; Ng Eaton, E.; Weinberg, R.A. Distinct EMT programs control normal mammary stem cells and tumour-initiating cells. Nature 2015, 525, 256–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. May, C.D.; Sphyris, N.; Evans, K.W.; Werden, S.J.; Guo, W.; Mani, S.A. Epithelial-mesenchymal transition and cancer stem cells: A dangerously dynamic duo in breast cancer progression. Breast Cancer Res. 2011, 13, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ghobrial, I.M. Myeloma as a model for the process of metastasis: Implications for therapy. Blood 2012, 120, 20–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Alvandi, Z.; Bischoff, J. Endothelial-Mesenchymal Transition in Cardiovascular Disease. Arter. Thromb. Vasc. Biol. 2021, 41, 2357–2369. [Google Scholar] [CrossRef]
  45. Lemma, S.; Karihtala, P.; Haapasaari, K.M.; Jantunen, E.; Soini, Y.; Bloigu, R.; Pasanen, A.K.; Turpeenniemi-Hujanen, T.; Kuittinen, O. Biological roles and prognostic values of the epithelial-mesenchymal transition-mediating transcription factors Twist, ZEB1 and Slug in diffuse large B-cell lymphoma. Histopathology 2013, 62, 326–333. [Google Scholar] [CrossRef] [PubMed]
  46. Sanchez-Tillo, E.; Fanlo, L.; Siles, L.; Montes-Moreno, S.; Moros, A.; Chiva-Blanch, G.; Estruch, R.; Martinez, A.; Colomer, D.; Gyorffy, B.; et al. The EMT activator ZEB1 promotes tumor growth and determines differential response to chemotherapy in mantle cell lymphoma. Cell Death Differ. 2014, 21, 247–257. [Google Scholar] [CrossRef] [PubMed]
  47. Wong, H.K.; Gibson, H.; Hake, T.; Geyer, S.; Frederickson, J.; Marcucci, G.; Caligiuri, M.A.; Porcu, P.; Mishra, A. Promoter-Specific Hypomethylation Is Associated with Overexpression of PLS3, GATA6, and TWIST1 in the Sezary Syndrome. J. Investig. Derm. 2015, 135, 2084–2092. [Google Scholar] [CrossRef] [Green Version]
  48. Mishra, A.; La Perle, K.; Kwiatkowski, S.; Sullivan, L.A.; Sams, G.H.; Johns, J.; Curphey, D.P.; Wen, J.; McConnell, K.; Qi, J.; et al. Mechanism, Consequences, and Therapeutic Targeting of Abnormal IL15 Signaling in Cutaneous T-cell Lymphoma. Cancer Discov. 2016, 6, 986–1005. [Google Scholar] [CrossRef] [Green Version]
  49. Zhang, J.; Wang, P.; Wu, F.; Li, M.; Sharon, D.; Ingham, R.J.; Hitt, M.; McMullen, T.P.; Lai, R. Aberrant expression of the transcriptional factor Twist1 promotes invasiveness in ALK-positive anaplastic large cell lymphoma. Cell Signal. 2012, 24, 852–858. [Google Scholar] [CrossRef]
  50. Cosset, E.; Hamdan, G.; Jeanpierre, S.; Voeltzel, T.; Sagorny, K.; Hayette, S.; Mahon, F.X.; Dumontet, C.; Puisieux, A.; Nicolini, F.E.; et al. Deregulation of TWIST-1 in the CD34+ compartment represents a novel prognostic factor in chronic myeloid leukemia. Blood 2011, 117, 1673–1676. [Google Scholar] [CrossRef] [Green Version]
  51. Cheong, C.M.; Mrozik, K.M.; Hewett, D.R.; Bell, E.; Panagopoulos, V.; Noll, J.E.; Licht, J.D.; Gronthos, S.; Zannettino, A.C.W.; Vandyke, K. Twist-1 is upregulated by NSD2 and contributes to tumour dissemination and an epithelial-mesenchymal transition-like gene expression signature in t(4;14)-positive multiple myeloma. Cancer Lett. 2020, 475, 99–108. [Google Scholar] [CrossRef] [PubMed]
  52. Peng, Y.; Li, F.; Zhang, P.; Wang, X.; Shen, Y.; Feng, Y.; Jia, Y.; Zhang, R.; Hu, J.; He, A. IGF-1 promotes multiple myeloma progression through PI3K/Akt-mediated epithelial-mesenchymal transition. Life Sci. 2020, 249, 117503. [Google Scholar] [CrossRef] [PubMed]
  53. Wu, S.; Du, Y.; Beckford, J.; Alachkar, H. Upregulation of the EMT marker vimentin is associated with poor clinical outcome in acute myeloid leukemia. J. Transl. Med. 2018, 16, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ibraheem, A.; Attar-Schneider, O.; Dabbah, M.; Dolberg Jarchowsky, O.; Tartakover Matalon, S.; Lishner, M.; Drucker, L. BM-MSCs-derived ECM modifies multiple myeloma phenotype and drug response in a source-dependent manner. Transl. Res. 2019, 207, 83–95. [Google Scholar] [CrossRef] [PubMed]
  55. Neri, P.; Ren, L.; Azab, A.K.; Brentnall, M.; Gratton, K.; Klimowicz, A.C.; Lin, C.; Duggan, P.; Tassone, P.; Mansoor, A.; et al. Integrin beta7-mediated regulation of multiple myeloma cell adhesion, migration, and invasion. Blood 2011, 117, 6202–6213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Klein, E.A.; Yin, L.; Kothapalli, D.; Castagnino, P.; Byfield, F.J.; Xu, T.; Levental, I.; Hawthorne, E.; Janmey, P.A.; Assoian, R.K. Cell-cycle control by physiological matrix elasticity and in vivo tissue stiffening. Curr. Biol. CB 2009, 19, 1511–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Assoian, R.K.; Schwartz, M.A. Coordinate signaling by integrins and receptor tyrosine kinases in the regulation of G1 phase cell-cycle progression. Curr. Opin. Genet. Dev. 2001, 11, 48–53. [Google Scholar] [CrossRef]
  58. Schwartz, M.A.; Assoian, R.K. Integrins and cell proliferation: Regulation of cyclin-dependent kinases via cytoplasmic signaling pathways. J. Cell Sci. 2001, 114, 2553–2560. [Google Scholar] [CrossRef]
  59. Welsh, C.F.; Roovers, K.; Villanueva, J.; Liu, Y.; Schwartz, M.A.; Assoian, R.K. Timing of cyclin D1 expression within G1 phase is controlled by Rho. Nat. Cell Biol. 2001, 3, 950–957. [Google Scholar] [CrossRef]
  60. Assoian, R.K. Anchorage-dependent cell cycle progression. J. Cell Biol. 1997, 136, 1–4. [Google Scholar] [CrossRef] [Green Version]
  61. Jones, M.C.; Askari, J.A.; Humphries, J.D.; Humphries, M.J. Cell adhesion is regulated by CDK1 during the cell cycle. J. Cell Biol. 2018, 217, 3203–3218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Marchesi, S.; Montani, F.; Deflorian, G.; D'Antuono, R.; Cuomo, A.; Bologna, S.; Mazzoccoli, C.; Bonaldi, T.; Di Fiore, P.P.; Nicassio, F. DEPDC1B coordinates de-adhesion events and cell-cycle progression at mitosis. Dev. Cell 2014, 31, 420–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gough, R.E.; Jones, M.C.; Zacharchenko, T.; Le, S.; Yu, M.; Jacquemet, G.; Muench, S.P.; Yan, J.; Humphries, J.D.; Jorgensen, C.; et al. Talin mechanosensitivity is modulated by a direct interaction with cyclin-dependent kinase-1. J. Biol. Chem. 2021, 297, 100837. [Google Scholar] [CrossRef] [PubMed]
  64. Yilmaz, M.; Christofori, G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009, 28, 15–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Adebowale, K.; Gong, Z.; Hou, J.C.; Wisdom, K.M.; Garbett, D.; Lee, H.P.; Nam, S.; Meyer, T.; Odde, D.J.; Shenoy, V.B.; et al. Enhanced substrate stress relaxation promotes filopodia-mediated cell migration. Nat. Mater. 2021, 20, 1290–1299. [Google Scholar] [CrossRef] [PubMed]
  66. Li, X.; Zhang, Q.; Yan, S.; Li, M.; You, R. Topographic cues reveal filopodia-mediated cell locomotion in 3D microenvironment. Biointerphases 2020, 15, 031001. [Google Scholar] [CrossRef] [PubMed]
  67. Mongiu, A.K.; Weitzke, E.L.; Chaga, O.Y.; Borisy, G.G. Kinetic-structural analysis of neuronal growth cone veil motility. J. Cell Sci. 2007, 120, 1113–1125. [Google Scholar] [CrossRef] [Green Version]
  68. Le Clainche, C.; Carlier, M.F. Regulation of actin assembly associated with protrusion and adhesion in cell migration. Physiol. Rev. 2008, 88, 489–513. [Google Scholar] [CrossRef] [Green Version]
  69. Svitkina, T.M.; Bulanova, E.A.; Chaga, O.Y.; Vignjevic, D.M.; Kojima, S.; Vasiliev, J.M.; Borisy, G.G. Mechanism of filopodia initiation by reorganization of a dendritic network. J. Cell Biol. 2003, 160, 409–421. [Google Scholar] [CrossRef]
  70. Nonpanya, N.; Sanookpan, K.; Joyjamras, K.; Wichadakul, D.; Sritularak, B.; Chaotham, C.; Chanvorachote, P. Norcycloartocarpin targets Akt and suppresses Akt-dependent survival and epithelial-mesenchymal transition in lung cancer cells. PLoS ONE 2021, 16, e0254929. [Google Scholar] [CrossRef]
  71. Sanookpan, K.; Nonpanya, N.; Sritularak, B.; Chanvorachote, P. Ovalitenone Inhibits the Migration of Lung Cancer Cells via the Suppression of AKT/mTOR and Epithelial-to-Mesenchymal Transition. Molecules 2021, 26, 638. [Google Scholar] [CrossRef] [PubMed]
  72. Sibony-Benyamini, H.; Gil-Henn, H. Invadopodia: The leading force. Eur. J. Cell Biol. 2012, 91, 896–901. [Google Scholar] [CrossRef] [PubMed]
  73. Li, Y.Y.; Jiang, G.T.; Chen, L.J.; Jiang, Y.H.; Jiao, J.D. Formin mDia1 contributes to migration and epithelial-mesenchymal transition of tubular epithelial cells exposed to TGF-beta1. J. Cell. Biochem. 2019, 3861–3870. [Google Scholar] [CrossRef]
  74. Rana, M.K.; Aloisio, F.M.; Choi, C.; Barber, D.L. Formin-dependent TGF-beta signaling for epithelial to mesenchymal transition. Mol. Biol. Cell 2018, 29, 1465–1475. [Google Scholar] [CrossRef] [PubMed]
  75. Yang, X.Y.; Liao, J.J.; Xue, W.R. FMNL1 down-regulation suppresses bone metastasis through reducing TGF-beta1 expression in non-small cell lung cancer (NSCLC). Biomed. Pharmacother. 2019, 117, 109126. [Google Scholar] [CrossRef] [PubMed]
  76. Rodrigues, P.C.; Sawazaki-Calone, I.; Ervolino de Oliveira, C.; Soares Macedo, C.C.; Dourado, M.R.; Cervigne, N.K.; Miguel, M.C.; Ferreira do Carmo, A.; Lambert, D.W.; Graner, E.; et al. Fascin promotes migration and invasion and is a prognostic marker for oral squamous cell carcinoma. Oncotarget 2017, 8, 74736–74754. [Google Scholar] [CrossRef] [Green Version]
  77. Ristic, B.; Kopel, J.; Sherazi, S.A.A.; Gupta, S.; Sachdeva, S.; Bansal, P.; Ali, A.; Perisetti, A.; Goyal, H. Emerging Role of Fascin-1 in the Pathogenesis, Diagnosis, and Treatment of the Gastrointestinal Cancers. Cancers 2021, 13, 2536. [Google Scholar] [CrossRef]
  78. Zhang, Y.; Lu, Y.; Zhang, C.; Huang, D.; Wu, W.; Zhang, Y.; Shen, J.; Cai, Y.; Chen, W.; Yao, W. FSCN1 increases doxorubicin resistance in hepatocellular carcinoma through promotion of epithelial-mesenchymal transition. Int. J. Oncol. 2018, 52, 1455–1464. [Google Scholar] [CrossRef]
  79. Ibarra, N.; Pollitt, A.; Insall, R.H. Regulation of actin assembly by SCAR/WAVE proteins. Biochem. Soc. Trans. 2005, 33, 1243–1246. [Google Scholar] [CrossRef]
  80. Stradal, T.E.; Rottner, K.; Disanza, A.; Confalonieri, S.; Innocenti, M.; Scita, G. Regulation of actin dynamics by WASP and WAVE family proteins. Trends Cell Biol. 2004, 14, 303–311. [Google Scholar] [CrossRef]
  81. Khoury, H.; Dankort, D.L.; Sadekova, S.; Naujokas, M.A.; Muller, W.J.; Park, M. Distinct tyrosine autophosphorylation sites mediate induction of epithelial mesenchymal like transition by an activated ErbB-2/Neu receptor. Oncogene 2001, 20, 788–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Iwaya, K.; Norio, K.; Mukai, K. Coexpression of Arp2 and WAVE2 predicts poor outcome in invasive breast carcinoma. Mod. Pathol. 2007, 20, 339–343. [Google Scholar] [CrossRef] [PubMed]
  83. Iwaya, K.; Oikawa, K.; Semba, S.; Tsuchiya, B.; Mukai, Y.; Otsubo, T.; Nagao, T.; Izumi, M.; Kuroda, M.; Domoto, H.; et al. Correlation between liver metastasis of the colocalization of actin-related protein 2 and 3 complex and WAVE2 in colorectal carcinoma. Cancer Sci. 2007, 98, 992–999. [Google Scholar] [CrossRef] [PubMed]
  84. Datta, A.; Deng, S.; Gopal, V.; Yap, K.C.; Halim, C.E.; Lye, M.L.; Ong, M.S.; Tan, T.Z.; Sethi, G.; Hooi, S.C.; et al. Cytoskeletal Dynamics in Epithelial-Mesenchymal Transition: Insights into Therapeutic Targets for Cancer Metastasis. Cancers 2021, 13, 1882. [Google Scholar] [CrossRef]
  85. Mori, H.; Tomari, T.; Koshikawa, N.; Kajita, M.; Itoh, Y.; Sato, H.; Tojo, H.; Yana, I.; Seiki, M. CD44 directs membrane-type 1 matrix metalloproteinase to lamellipodia by associating with its hemopexin-like domain. Embo J. 2002, 21, 3949–3959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Pastushenko, I.; Mauri, F.; Song, Y.; de Cock, F.; Meeusen, B.; Swedlund, B.; Impens, F.; Van Haver, D.; Opitz, M.; Thery, M.; et al. Fat1 deletion promotes hybrid EMT state, tumour stemness and metastasis. Nature 2021, 589, 448–455. [Google Scholar] [CrossRef]
  87. Tsirtsaki, K.; Gkretsi, V. The focal adhesion protein Integrin-Linked Kinase (ILK) as an important player in breast cancer pathogenesis. Cell Adh. Migr. 2020, 14, 204–213. [Google Scholar] [CrossRef]
  88. Jin, H.; He, Y.; Zhao, P.; Hu, Y.; Tao, J.; Chen, J.; Huang, Y. Targeting lipid metabolism to overcome EMT-associated drug resistance via integrin beta3/FAK pathway and tumor-associated macrophage repolarization using legumain-activatable delivery. Theranostics 2019, 9, 265–278. [Google Scholar] [CrossRef]
  89. van den Dries, K.; Linder, S.; Maridonneau-Parini, I.; Poincloux, R. Probing the mechanical landscape—New insights into podosome architecture and mechanics. J. Cell Sci. 2019, 132. [Google Scholar] [CrossRef]
  90. van den Dries, K.; Nahidiazar, L.; Slotman, J.A.; Meddens, M.B.M.; Pandzic, E.; Joosten, B.; Ansems, M.; Schouwstra, J.; Meijer, A.; Steen, R.; et al. Modular actin nano-architecture enables podosome protrusion and mechanosensing. Nat. Commun. 2019, 10, 5171. [Google Scholar] [CrossRef] [Green Version]
  91. Linder, S. Invadosomes at a glance. J.Cell Sci. 2009, 122, 3009–3013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Alonso, F.; Spuul, P.; Genot, E. Podosomes in endothelial cell--microenvironment interactions. Curr. Opin. Hematol. 2020, 27, 197–205. [Google Scholar] [CrossRef] [PubMed]
  93. Chen, Y.W.; Lai, C.S.; Chen, Y.F.; Chiu, W.T.; Chen, H.C.; Shen, M.R. STIM1-dependent Ca(2+) signaling regulates podosome formation to facilitate cancer cell invasion. Sci. Rep. 2017, 7, 11523. [Google Scholar] [CrossRef] [Green Version]
  94. Lener, T.; Burgstaller, G.; Crimaldi, L.; Lach, S.; Gimona, M. Matrix-degrading podosomes in smooth muscle cells. Eur. J. Cell Biol. 2006, 85, 183–189. [Google Scholar] [CrossRef] [PubMed]
  95. Santiago-Medina, M.; Gregus, K.A.; Nichol, R.H.; O'Toole, S.M.; Gomez, T.M. Regulation of ECM degradation and axon guidance by growth cone invadosomes. Development 2015, 142, 486–496. [Google Scholar] [CrossRef] [Green Version]
  96. Murphy, D.A.; Courtneidge, S.A. The ‘ins’ and ‘outs’ of podosomes and invadopodia: Characteristics, formation and function. Nat. Rev. Mol. Cell Biol. 2011, 12, 413–426. [Google Scholar] [CrossRef] [Green Version]
  97. Branch, K.M.; Hoshino, D.; Weaver, A.M. Adhesion rings surround invadopodia and promote maturation. Biol. Open 2012, 1, 711–722. [Google Scholar] [CrossRef] [Green Version]
  98. Poincloux, R.; Lizarraga, F.; Chavrier, P. Matrix invasion by tumour cells: A focus on MT1-MMP trafficking to invadopodia. J. Cell Sci. 2009, 122, 3015–3024. [Google Scholar] [CrossRef] [Green Version]
  99. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef]
  100. Zhang, D.; Bi, J.; Liang, Q.; Wang, S.; Zhang, L.; Han, F.; Li, S.; Qiu, B.; Fan, X.; Chen, W.; et al. VCAM1 Promotes Tumor Cell Invasion and Metastasis by Inducing EMT and Transendothelial Migration in Colorectal Cancer. Front. Oncol. 2020, 10, 1066. [Google Scholar] [CrossRef]
  101. Borriello, L.; Karagiannis, G.S.; Duran, C.L.; Coste, A.; Oktay, M.H.; Entenberg, D.; Condeelis, J.S. The role of the tumor microenvironment in tumor cell intravasation and dissemination. Eur. J. Cell Biol. 2020, 99, 151098. [Google Scholar] [CrossRef]
  102. Schoumacher, M.; Goldman, R.D.; Louvard, D.; Vignjevic, D.M. Actin, microtubules, and vimentin intermediate filaments cooperate for elongation of invadopodia. J. Cell Biol. 2010, 189, 541–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Bayarmagnai, B.; Perrin, L.; Esmaeili Pourfarhangi, K.; Grana, X.; Tuzel, E.; Gligorijevic, B. Invadopodia-mediated ECM degradation is enhanced in the G1 phase of the cell cycle. J. Cell Sci. 2019, 132. [Google Scholar] [CrossRef] [Green Version]
  104. Nayak, D.; Kumar, A.; Chakraborty, S.; Rasool, R.U.; Amin, H.; Katoch, A.; Gopinath, V.; Mahajan, V.; Zilla, M.K.; Rah, B.; et al. Inhibition of Twist1-mediated invasion by Chk2 promotes premature senescence in p53-defective cancer cells. Cell Death Differ. 2017, 24, 1275–1287. [Google Scholar] [CrossRef] [Green Version]
  105. Tzivion, G.; Luo, Z.J.; Avruch, J. Calyculin A-induced vimentin phosphorylation sequesters 14-3-3 and displaces other 14-3-3 partners in vivo. J. Biol. Chem. 2000, 275, 29772–29778. [Google Scholar] [CrossRef] [Green Version]
  106. Kidd, M.E.; Shumaker, D.K.; Ridge, K.M. The role of vimentin intermediate filaments in the progression of lung cancer. Am. J. Respir. Cell Mol. Biol. 2014, 50, 1–6. [Google Scholar] [CrossRef] [PubMed]
  107. Hermeking, H.; Benzinger, A. 14-3-3 proteins in cell cycle regulation. Semin. Cancer Biol. 2006, 16, 183–192. [Google Scholar] [CrossRef]
  108. Franchi, M.; Piperigkou, Z.; Riti, E.; Masola, V.; Onisto, M.; Karamanos, N.K. Long filopodia and tunneling nanotubes define new phenotypes of breast cancer cells in 3D cultures. Matrix Biol. Plus 2020, 6–7, 100026. [Google Scholar] [CrossRef]
  109. Li, P.; Chen, Y.; Peng, Y.; Zhang, Y.; Zhou, H.; Chen, X.; Li, T.; Li, S.; Yang, H.; Wu, C.; et al. Notch-1 signaling promotes reattachment of suspended cancer cells by cdc42-dependent microtentacles formation. Cancer Sci. 2021, 112, 4894. [Google Scholar] [CrossRef] [PubMed]
  110. Killilea, A.N.; Csencsits, R.; Le, E.; Patel, A.M.; Kenny, S.J.; Xu, K.; Downing, K.H. Cytoskeletal organization in microtentacles. Exp. Cell Res. 2017, 357, 291–298. [Google Scholar] [CrossRef]
  111. Wolf, K.J.; Shukla, P.; Springer, K.; Lee, S.; Coombes, J.D.; Choy, C.J.; Kenny, S.J.; Xu, K.; Kumar, S. A mode of cell adhesion and migration facilitated by CD44-dependent microtentacles. Proc. Natl. Acad. Sci. USA 2020, 117, 11432–11443. [Google Scholar] [CrossRef] [PubMed]
  112. Whipple, R.A.; Cheung, A.M.; Martin, S.S. Detyrosinated microtubule protrusions in suspended mammary epithelial cells promote reattachment. Exp. Cell Res. 2007, 313, 1326–1336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Charpentier, M.; Martin, S. Interplay of Stem Cell Characteristics, EMT, and Microtentacles in Circulating Breast Tumor Cells. Cancers 2013, 5, 1545–1565. [Google Scholar] [CrossRef] [Green Version]
  114. Cepeda, M.A.; Pelling, J.J.; Evered, C.L.; Williams, K.C.; Freedman, Z.; Stan, I.; Willson, J.A.; Leong, H.S.; Damjanovski, S. Less is more: Low expression of MT1-MMP is optimal to promote migration and tumourigenesis of breast cancer cells. Mol. Cancer 2016, 15, 65. [Google Scholar] [CrossRef] [Green Version]
  115. Perentes, J.Y.; Kirkpatrick, N.D.; Nagano, S.; Smith, E.Y.; Shaver, C.M.; Sgroi, D.; Garkavtsev, I.; Munn, L.L.; Jain, R.K.; Boucher, Y. Cancer cell-associated MT1-MMP promotes blood vessel invasion and distant metastasis in triple-negative mammary tumors. Cancer Res. 2011, 71, 4527–4538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Latario, C.J.; Schoenfeld, L.W.; Howarth, C.L.; Pickrell, L.E.; Begum, F.; Fischer, D.A.; Grbovic-Huezo, O.; Leach, S.D.; Sanchez, Y.; Smith, K.D.; et al. Tumor microtubes connect pancreatic cancer cells in an Arp2/3 complex-dependent manner. Mol. Biol. Cell 2020, 31, 1259–1272. [Google Scholar] [CrossRef] [PubMed]
  117. Whipple, R.A.; Matrone, M.A.; Cho, E.H.; Balzer, E.M.; Vitolo, M.I.; Yoon, J.R.; Ioffe, O.B.; Tuttle, K.C.; Yang, J.; Martin, S.S. Epithelial-to-mesenchymal transition promotes tubulin detyrosination and microtentacles that enhance endothelial engagement. Cancer Res. 2010, 70, 8127–8137. [Google Scholar] [CrossRef] [Green Version]
  118. Balzer, E.M.; Whipple, R.A.; Thompson, K.; Boggs, A.E.; Slovic, J.; Cho, E.H.; Matrone, M.A.; Yoneda, T.; Mueller, S.C.; Martin, S.S. c-Src differentially regulates the functions of microtentacles and invadopodia. Oncogene 2010, 29, 6402–6408. [Google Scholar] [CrossRef] [Green Version]
  119. Roselova, P.; Obr, A.; Holoubek, A.; Grebenova, D.; Kuzelova, K. Adhesion structures in leukemia cells and their regulation by Src family kinases. Cell Adh. Migr. 2018, 12, 286–298. [Google Scholar] [CrossRef] [Green Version]
  120. Maia, V.; Ortiz-Rivero, S.; Sanz, M.; Gutierrez-Berzal, J.; Alvarez-Fernandez, I.; Gutierrez-Herrero, S.; de Pereda, J.M.; Porras, A.; Guerrero, C. C3G forms complexes with Bcr-Abl and p38alpha MAPK at the focal adhesions in chronic myeloid leukemia cells: Implication in the regulation of leukemic cell adhesion. Cell Commun. Signal. 2013, 11, 9. [Google Scholar] [CrossRef] [Green Version]
  121. Vacca, A.; Ria, R.; Presta, M.; Ribatti, D.; Iurlaro, M.; Merchionne, F.; Tanghetti, E.; Dammacco, F. alpha(v)beta(3) integrin engagement modulates cell adhesion, proliferation, and protease secretion in human lymphoid tumor cells. Exp. Hematol. 2001, 29, 993–1003. [Google Scholar] [CrossRef]
  122. Chen, W.C.; Hu, G.; Hazlehurst, L.A. Contribution of the bone marrow stromal cells in mediating drug resistance in hematopoietic tumors. Curr. Opin. Pharm. 2020, 54, 36–43. [Google Scholar] [CrossRef] [PubMed]
  123. Krenn, P.W.; Hofbauer, S.W.; Pucher, S.; Hutterer, E.; Hinterseer, E.; Denk, U.; Asslaber, D.; Ganghammer, S.; Sternberg, C.; Neureiter, D.; et al. ILK Induction in Lymphoid Organs by a TNFalpha-NF-kappaB-Regulated Pathway Promotes the Development of Chronic Lymphocytic Leukemia. Cancer Res. 2016, 76, 2186–2196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Ouchani, F.; Devy, J.; Rusciani, A.; Helesbeux, J.J.; Salesse, S.; Letinois, I.; Gras-Billart, D.; Duca, L.; Duval, O.; Martiny, L.; et al. Targeting focal adhesion assembly by ethoxyfagaronine prevents lymphoblastic cell adhesion to fibronectin. Anal. Cell Pathol. (Amst.) 2012, 35, 267–284. [Google Scholar] [CrossRef] [PubMed]
  125. Bosch, R.; Dieguez-Gonzalez, R.; Cespedes, M.V.; Parreno, M.; Pavon, M.A.; Granena, A.; Sierra, J.; Mangues, R.; Casanova, I. A novel inhibitor of focal adhesion signaling induces caspase-independent cell death in diffuse large B-cell lymphoma. Blood 2011, 118, 4411–4420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Rothe, K.; Babaian, A.; Nakamichi, N.; Chen, M.; Chafe, S.C.; Watanabe, A.; Forrest, D.L.; Mager, D.L.; Eaves, C.J.; Dedhar, S.; et al. Integrin-Linked Kinase Mediates Therapeutic Resistance of Quiescent CML Stem Cells to Tyrosine Kinase Inhibitors. Cell Stem Cell 2020, 27, 110–124. [Google Scholar] [CrossRef] [PubMed]
  127. Gaidano, G.; Bergui, L.; Schena, M.; Gaboli, M.; Cremona, O.; Marchisio, P.C.; Caligaris-Cappio, F. Integrin distribution and cytoskeleton organization in normal and malignant monocytes. Leukemia 1990, 4, 682–687. [Google Scholar]
  128. Redondo-Munoz, J.; Escobar-Diaz, E.; Samaniego, R.; Terol, M.J.; Garcia-Marco, J.A.; Garcia-Pardo, A. MMP-9 in B-cell chronic lymphocytic leukemia is up-regulated by alpha4beta1 integrin or CXCR4 engagement via distinct signaling pathways, localizes to podosomes, and is involved in cell invasion and migration. Blood 2006, 108, 3143–3151. [Google Scholar] [CrossRef] [Green Version]
  129. Caligaris-Cappio, F.; Bergui, L.; Tesio, L.; Corbascio, G.; Tousco, F.; Marchisio, P.C. Cytoskeleton organization is aberrantly rearranged in the cells of B chronic lymphocytic leukemia and hairy cell leukemia. Blood 1986, 67, 233–239. [Google Scholar] [CrossRef] [Green Version]
  130. Calvani, N.; Cafforio, P.; Silvestris, F.; Dammacco, F. Functional osteoclast-like transformation of cultured human myeloma cell lines. Br. J. Haematol. 2005, 130, 926–938. [Google Scholar] [CrossRef]
  131. Calvani, N.; Silvestris, F.; Cafforio, P.; Dammacco, F. Osteoclast-like cell formation by circulating myeloma B lymphocytes: Role of RANK-L. Leuk. Lymphoma 2004, 45, 377–380. [Google Scholar] [CrossRef] [PubMed]
  132. Taylor, M.D.; Sadhukhan, S.; Kottangada, P.; Ramgopal, A.; Sarkar, K.; D'Silva, S.; Selvakumar, A.; Candotti, F.; Vyas, Y.M. Nuclear role of WASp in the pathogenesis of dysregulated TH1 immunity in human Wiskott-Aldrich syndrome. Sci. Transl. Med. 2010, 2, 37ra44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Yoo, Y.; Wu, X.; Guan, J.L. A novel role of the actin-nucleating Arp2/3 complex in the regulation of RNA polymerase II-dependent transcription. J.Biol. Chem. 2007, 282, 7616–7623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Yoshida, T.; Shimada, K.; Oma, Y.; Kalck, V.; Akimura, K.; Taddei, A.; Iwahashi, H.; Kugou, K.; Ohta, K.; Gasser, S.M.; et al. Actin-related protein Arp6 influences H2A.Z-dependent and -independent gene expression and links ribosomal protein genes to nuclear pores. PLoS Genet. 2010, 6, e1000910. [Google Scholar] [CrossRef]
  135. Poincloux, R.; Cougoule, C.; Daubon, T.; Maridonneau-Parini, I.; Le Cabec, V. Tyrosine-phosphorylated STAT5 accumulates on podosomes in Hck-transformed fibroblasts and chronic myeloid leukemia cells. J. Cell Physiol. 2007, 213, 212–220. [Google Scholar] [CrossRef] [PubMed]
  136. Bessy, T.; Candelas, A.; Souquet, B.; Saadallah, K.; Schaeffer, A.; Vianay, B.; Cuvelier, D.; Gobaa, S.; Nakid-Cordero, C.; Lion, J.; et al. Hematopoietic progenitors polarize in contact with bone marrow stromal cells in response to SDF1. J. Cell Biol. 2021, 220, e202005085. [Google Scholar] [CrossRef]
  137. Holloway, W.; Martinez, A.R.; Oh, D.J.; Francis, K.; Ramakrishna, R.; Palsson, B.O. Key adhesion molecules are present on long podia extended by hematopoietic cells. Cytometry 1999, 37, 171–177. [Google Scholar] [CrossRef]
  138. Ramasamy, K.; Anwar, A.; Kazmi, M.A.; Y, C. Podia in Multiple Myeloma (MM) Cells Promote Adhesion with Bone Marrow (BM) Fibroblastic Stromal Cells. In Proceedings of the International Conference on The Tumour Microenvironment in the Haematological Malignancies and its Therapeutic Targeting, Lisbon, Portugal, 7–9 May 2015. [Google Scholar]
  139. Ramasamy, K.; Khatun, H.; Macpherson, L.; Caley, M.P.; Sturge, J.; Mufti, G.J.; Schey, S.A.; Calle, Y. Fluorescence-based experimental model to evaluate the concomitant effect of drugs on the tumour microenvironment and cancer cells. Br. J. Haematol. 2012, 157, 564–579. [Google Scholar] [CrossRef]
  140. Tavor, S.; Petit, I.; Porozov, S.; Goichberg, P.; Avigdor, A.; Sagiv, S.; Nagler, A.; Naparstek, E.; Lapidot, T. Motility, proliferation, and egress to the circulation of human AML cells are elastase dependent in NOD/SCID chimeric mice. Blood 2005, 106, 2120–2127. [Google Scholar] [CrossRef] [Green Version]
  141. Gandalovicova, A.; Rosel, D.; Fernandes, M.; Vesely, P.; Heneberg, P.; Cermak, V.; Petruzelka, L.; Kumar, S.; Sanz-Moreno, V.; Brabek, J. Migrastatics-Anti-metastatic and Anti-invasion Drugs: Promises and Challenges. Trends Cancer 2017, 3, 391–406. [Google Scholar] [CrossRef] [Green Version]
  142. Orgaz, J.L.; Crosas-Molist, E.; Sadok, A.; Perdrix-Rosell, A.; Maiques, O.; Rodriguez-Hernandez, I.; Monger, J.; Mele, S.; Georgouli, M.; Bridgeman, V.; et al. Myosin II Reactivation and Cytoskeletal Remodeling as a Hallmark and a Vulnerability in Melanoma Therapy Resistance. Cancer Cell 2020, 37, 85–103. [Google Scholar] [CrossRef] [PubMed]
  143. Hegde, S.; Gasilina, A.; Wunderlich, M.; Lin, Y.; Buchholzer, M.; Krumbach, O.H.F.; Akbarzadeh, M.; Ahmadian, M.R.; Seibel, W.; Zheng, Y.; et al. Inhibition of the RacGEF VAV3 by the small molecule IODVA1 impedes RAC signaling and overcomes resistance to tyrosine kinase inhibition in acute lymphoblastic leukemia. Leukemia 2021, 1–11. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Formation of mature focal contacts in AML cells activated with TPA. THP-1 cells were seeded on fibronectin coated coverslips (10 μg/mL) n and left untreated (AC) or were treated with TPA (30 nM for 48 h). Cells were fixed with 4% paraformaldehyde and permeabilised with 0.05% Triton X-100 and immuno-stained to detect the distribution of vinculin (A,D) and F-actin (B,E). Merged images are shown in (C,F). Vinculin was quite homogenously distributed on the surface of untreated THP-1 cells, whereas TPA activation induced the clustering of vinculin into focal contacts (arrows in D). Bar 10 μm.
Figure 1. Formation of mature focal contacts in AML cells activated with TPA. THP-1 cells were seeded on fibronectin coated coverslips (10 μg/mL) n and left untreated (AC) or were treated with TPA (30 nM for 48 h). Cells were fixed with 4% paraformaldehyde and permeabilised with 0.05% Triton X-100 and immuno-stained to detect the distribution of vinculin (A,D) and F-actin (B,E). Merged images are shown in (C,F). Vinculin was quite homogenously distributed on the surface of untreated THP-1 cells, whereas TPA activation induced the clustering of vinculin into focal contacts (arrows in D). Bar 10 μm.
Cells 11 00649 g001
Figure 2. Formation of podosomes in multiple myeloma cells. CD138 positive cells were isolated from bone marrow aspirates from multiple myeloma patients. Isolated cells were seeded on fibronectin-coated coverslips and incubated for 24 h. Cultures were fixed with 4% paraformaldehyde and permeabilised with 0.05% Triton X-100 and immuno-stained to detect the distribution of talin (A) and F-actin (B). Merged images are shown in (C). Magnification of the boxed areas with talin, vinculin and merged staining are shown at the bottom. Bar 10 μm.
Figure 2. Formation of podosomes in multiple myeloma cells. CD138 positive cells were isolated from bone marrow aspirates from multiple myeloma patients. Isolated cells were seeded on fibronectin-coated coverslips and incubated for 24 h. Cultures were fixed with 4% paraformaldehyde and permeabilised with 0.05% Triton X-100 and immuno-stained to detect the distribution of talin (A) and F-actin (B). Merged images are shown in (C). Magnification of the boxed areas with talin, vinculin and merged staining are shown at the bottom. Bar 10 μm.
Cells 11 00649 g002
Figure 3. Formation of podia in multiple myeloma cells. The enhanced-Green Fluorescent Protein (eGFP)-expressing multiple myeloma cell line RPMI-8226 (eGFP_RPMI 28226) (A) and the monomeric Cherry (m-Cherry)-expressing bone marrow stromal cell line HS5 (mCherry-HS5) (B) were generated as previously described [139]. eGFP-RPMI 8226 were co-cultured for three days with mCherry-HS5 cells, fixed with 4% paraformaldehyde and permeabilised with 0.05% Triton X-100 and stained with Alexa 647-Phalloidin to detect the distribution of F-actin (C). Merged images are shown in (D). Magnification of the boxed areas with eGFP and F-actin staining are shown at the bottom. eGFP-RPMI 8226 cells formed podia (white arrows) elongating along the surface of mCherry-HS5 cells. Bar 10 μm.
Figure 3. Formation of podia in multiple myeloma cells. The enhanced-Green Fluorescent Protein (eGFP)-expressing multiple myeloma cell line RPMI-8226 (eGFP_RPMI 28226) (A) and the monomeric Cherry (m-Cherry)-expressing bone marrow stromal cell line HS5 (mCherry-HS5) (B) were generated as previously described [139]. eGFP-RPMI 8226 were co-cultured for three days with mCherry-HS5 cells, fixed with 4% paraformaldehyde and permeabilised with 0.05% Triton X-100 and stained with Alexa 647-Phalloidin to detect the distribution of F-actin (C). Merged images are shown in (D). Magnification of the boxed areas with eGFP and F-actin staining are shown at the bottom. eGFP-RPMI 8226 cells formed podia (white arrows) elongating along the surface of mCherry-HS5 cells. Bar 10 μm.
Cells 11 00649 g003
Table 1. Upregulation of canonical EMT markers in haematological malignancies correlates with poor prognosis and/or the enhanced migration of cancer cells.
Table 1. Upregulation of canonical EMT markers in haematological malignancies correlates with poor prognosis and/or the enhanced migration of cancer cells.
Mechanism
Regulated
EMT TFHaematological
Malignancy Type
References
Resistance to therapy and poor prognosisZEB1B-Cell Lymphoma[45]
Mantle cell lymphoma[46]
TWIST1CML[50]
SNAIL, SLUGMultiple myeloma[54]
ZEB1, HGFMLL-AF9 AML[9]
Enhanced capacity for cell migrationTWIST1Paediatric anaplastic[49]
large cell lymphoma/
ZEB1MLL-AF9 AML[9]
TWIST1, SNAIL, SLUGMultiple myeloma[23,51,54]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Greaves, D.; Calle, Y. Epithelial Mesenchymal Transition (EMT) and Associated Invasive Adhesions in Solid and Haematological Tumours. Cells 2022, 11, 649. https://doi.org/10.3390/cells11040649

AMA Style

Greaves D, Calle Y. Epithelial Mesenchymal Transition (EMT) and Associated Invasive Adhesions in Solid and Haematological Tumours. Cells. 2022; 11(4):649. https://doi.org/10.3390/cells11040649

Chicago/Turabian Style

Greaves, David, and Yolanda Calle. 2022. "Epithelial Mesenchymal Transition (EMT) and Associated Invasive Adhesions in Solid and Haematological Tumours" Cells 11, no. 4: 649. https://doi.org/10.3390/cells11040649

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop