Abstract

Epithelial–mesenchymal plasticity (EMP) is a key driver of cancer metastasis and therapeutic resistance, through which cancer cells can reversibly and dynamically alter their molecular and functional traits along the epithelial–mesenchymal spectrum. While cells in the epithelial phenotype are usually tightly adherent, less metastatic, and drug-sensitive, those in the hybrid epithelial/mesenchymal and/or mesenchymal state are more invasive, migratory, drug-resistant, and immune-evasive. Single-cell studies have emerged as a powerful tool in gaining new insights into the dynamics of EMP across various cancer types. Here, we review many recent studies that employ single-cell analysis techniques to better understand the dynamics of EMP in cancer both in vitro and in vivo. These single-cell studies have underlined the plurality of trajectories cells can traverse during EMP and the consequent heterogeneity of hybrid epithelial/mesenchymal phenotypes seen at both preclinical and clinical levels. They also demonstrate how diverse EMP trajectories may exhibit hysteretic behavior and how the rate of such cell-state transitions depends on the genetic/epigenetic background of recipient cells, as well as the dose and/or duration of EMP-inducing growth factors. Finally, we discuss the relationship between EMP and patient survival across many cancer types. We also present a next set of questions related to EMP that could benefit much from single-cell observations and pave the way to better tackle phenotypic switching and heterogeneity in clinic.
Introduction
Phenotypic plasticity—the ability of a genotype to enable multiple phenotypes in response to varying environmental conditions—is crucial for survival. A ubiquitous phenomenon seen across biological organisms and contexts, phenotypic plasticity can be considered as an evolutionary strategy for adapting to variable environments.1 One popular graphical representation of phenotypic plasticity is Waddington’s “epigenetic landscape” which conceptualizes embryonic development of multicellular organisms as a sequence of successive cell-fate decision-making bifurcations, where “master regulators” of specific cellular lineages at these branch points shape the dynamical trajectory of phenotypic differentiation.2 Plasticity is not, however, restricted to development; it comes into play as cells take their reprogramming excursions during injury repair or also in pathophysiological conditions such as cancer and fibrosis. It is now considered as a key enabling hallmark of cancer aggressiveness.3
Phenotypic plasticity comes in various manifestations; one of the well-studied canonical instances is epithelial–mesenchymal plasticity (EMP). EMP involves reversible cell-state changes along the epithelial–hybrid–mesenchymal spectrum with varying molecular, functional, and cell morphological attributes.4 It is observed in multiple stages of embryonic development, such as gastrulation, mammary gland development,5 and neural crest formation,6 and is implicated in cancer metastasis and somatic cell reprogramming7,8 to induced pluripotent stem cells (iPSCs)9 as well. Recent progress in single-cell investigations has unravelled novel insights into underlying molecular programs controlling the directionality and scope of phenotypic plasticity trajectories being traversed in a high-dimensional space. Here, we highlight the salient features of EMP dynamics in carcinomas that have been elucidated by single-cell investigations.
EMP is comprised of epithelial-to-mesenchymal transition (EMT) and its reverse, mesenchymal-to-epithelial transition (MET). During EMT, epithelial cells partially or fully lose their typical features such as tight cell–cell adhesion and apicobasal polarity and gain migratory and invasive features. These changes can, in principle, be reversed during MET. The dynamics of EMT and MET are regulated by a complex network of transcription factors (TFs), microRNAs, epigenetic modulators, long noncoding RNAs, and external microenvironment signals such as hypoxia and matrix stiffness.10 Most of the experimental investigations in EMP dynamics have been at a transcriptomic level,11,12 but other axes such as morphology, proteomics, and epigenetics are becoming increasingly prevalent.13−15
Such multidimensional analysis of EMP has enabled visualizing it from a dynamical systems perspective, where a cell state can be specified using a set of state variables (expression levels, cell shapes, etc.). A stable cell state can then be viewed as an “attractor” in multidimensional cell-state space and can be specified by its state variables.16 Cell-state transitions such as EMT/MET can be considered as a switch from one attractor to another in a multistable landscape (Figure 1). Single-cell investigations have begun to answer many salient features about cell-state transition dynamics: the existence of hybrid epithelial/mesenchymal (E/M) phenotypes, plurality of cell-state transition paths, reversibility, and symmetry of these transitions, etc.
Figure 1.

Schematic of a possible landscape showing cell states during EMP. Contour map showing different possible cell states and trajectories in the EMP landscape. Dotted arrows show reversible transitions while solid arrows show irreversible transitions.
Molecular and Functional Characterization of Hybrid E/M States
Earlier thought of as a binary process, EMP is now understood as incorporating one or more hybrid E/M states, as demonstrated via multiple experimental and computational analyses.17−20 One critical open question in the growing field of hybrid E/M cell states is identifying necessary and sufficient molecular or functional attributes of cancer cells to characterize them as a hybrid E/M phenotype. Traditionally, a hybrid cell type is indicated by coexpression of specific epithelial and mesenchymal marker genes in the same cell.21 However, this set of markers to characterize the hybrid E/M states are often quite context-specific across multiple cancer types.22−25
Single-cell investigations have provided unprecedented insights into phenotypic heterogeneity along the EMP spectrum, revealing multiple hybrid E/M states with varied molecular and functional traits. For instance, multiple hybrid E/M subpopulations were identified in skin and mammary primary tumors by screening cell surface markers. These subpopulations displayed varying invasiveness, cellular plasticity, and metastatic potentials, which were distinct from those of the extreme epithelial or mesenchymal states.26 The loss of function of cadherin Fat1 via mutation or deletion in mouse and human squamous cell carcinoma was found to promote the hybrid E/M state, enabling tumor initiation, progression, and metastasis.27 Strikingly, despite different surface marker patterns, hybrid E/M cells were found to be more plastic relative to the epithelial and mesenchymal ones.26 A possible explanation behind this observation was offered by recent computational analysis of multiple regulatory networks underlying EMP. These networks contained “teams” of nodes that stabilized epithelial and mesenchymal phenotypes, but no such team was observed for hybrid E/M states, thus enabling their higher propensity to switch.28 Another single-cell analysis tracked cancer cells undergoing full and partial EMT using live imaging, tamoxifen-inducible lineage-tracing systems, and 5-cell RNA sequencing in the MMTV–PyMT mouse model of metastatic breast cancer. While cells undergoing full EMT failed to colonize the lungs, partial EMT (or hybrid E/M) ones contributed majorly to lung metastasis and chemoresistance29 (Figure 2). These trends are reminiscent of in vitro observations that hybrid E/M cells had almost 10 times higher mammosphere-forming ability than epithelial or mesenchymal cells.20 A recent study establishing single-cell clones from the heterogeneous SUM149PT breast cancer line showed that clones exhibiting a hybrid E/M phenotype had both higher plasticity and tumor-initiating ability than the clones representing either end of the epithelial–hybrid–mesenchymal spectrum.19 Together, these observations underscore that despite plurality and marker-specific identification of hybrid E/M phenotypes, higher plasticity and metastatic potential are their functional hallmarks.
Figure 2.
Summary of experimental methodologies for single-cell analysis of EMP. Single-cell RNA-Seq, single-cell lineage tracing, mass cytometry-based experiments, and real-time single-cell imaging are the most common laboratory methods to study EMP at a single-cell resolution.
One reason for multiple hybrid E/M phenotypes may be semi-independent changes along the epithelial and mesenchymal axes. For instance, single-cell RNA-seq (scRNA-seq) time-course transcriptomic profiles of TGF-β induction of MCF10A cells up to 10 days reported a delayed and gradual decrease of the canonical epithelial marker CDH1 but an immediate loss of S100 calcium-binding protein A9 (S100A9).30 This observation raises an important question as to whether heterogeneity can be seen as a function of time or also as a function of dose of the EMT-inducing signal. To address this question, in a recent scRNA-seq experiment, MCF10A cells were treated with TGF-β at various doses and time points up to 14 days.31 At a 14-day time point, with increasing dose, the cells progressed from (epithelial high, mesenchymal low) to (epithelial low, mesenchymal high) state and showed a saturating behavior at higher doses. A similar observation was made for characteristic epithelial (e.g., CDH1) and mesenchymal (e.g., VIM, FN1) genes. Intriguingly, time-dependent EMT demonstrated more significant separability in the continuum of cell states noted, as compared to dose-dependent behavior. When temporal resolution is missing explicitly from the experimental design, pseudotime analysis is often implemented to identify the cell-state transition trajectory. Such analysis for in vivo single-cell RNA-seq data from squamous cell carcinoma identified that the most likely EMP trajectory was dependent more on an increase in mesenchymal than on a concomitant decrease in epithelial.32 These observations showcase that a decrease in epithelial and an increase in mesenchymal gene expression programs may be asynchronous, thus enabling a multitude of hybrid E/M states.
Another possible reason enabling multiple hybrid E/M phenotypes is varying levels of coordination among the various axes of EMT—molecular EMT, morphological EMT, and functional EMT.33 For instance, single-cell mechanical investigation and spatial EMT-related gene expression profiling illustrated that leader cells in collective cell migration of A549 lung cancer cells were more elongated and softer (lower Young’s modulus) than the follower ones. These cells expressed mesenchymal markers (SNAI1, VIM) while still retaining epithelial (CDH1) ones, exhibiting a partial EMT phenotype.34 Another study using image quantification to track EMT progression revealed a partial EMT phenotype with a unique cytoskeletal signature, consistent with decreased stiffness compared to mesenchymal cells.35 Tracking the morphological changes upon treatment with EMT-inducing growth factors has helped infer transition rates among different morphological cell states and has been used to develop classifiers to segregate epithelial vs mesenchymal states with 92% accuracy.36−38 Such characterization can be coupled with scRNA-seq data to extract information about E–M heterogeneity. An example would be the development of a functional single-cell selection pipeline that allowed screening based on time-varying cellular dynamics or morphology and selective labeling of cells among a heterogeneous population.39 Monitoring the migration trajectories of individual cells in the MCF10 cell line, fast-moving cells were identified and phototagged. These cells were spindle-shaped, exhibiting a mesenchymal-like phenotype. They were later segregated for single-cell transcriptomic analysis, which revealed an upregulation of EMT-driving pathways TGF-β and NF-κB in these cells, compared to the slow-moving ones. This study thus demonstrated how cells identified to be mesenchymal using image analysis (Figure 2) exhibited concomitant gene expression profiles too, i.e., well-coordinated molecular and morphological EMT.
How many phenotypes exist along the epithelial–hybrid–mesenchymal spectrum is an open question. While scRNA-seq data can help identify prevalent heterogeneity, pinpointing an optimal number of meaningful clusters in such high-dimensional data is a challenge from both the methodical and conceptual standpoints. A scRNA-seq study in genetic mouse models of pancreatic ductal adenocarcinoma (PDAC) revealed ∼50 cancer cell clusters spanning the epithelial–mesenchymal continuum; they observed similar trends in human PDAC samples.40 Simulations for large regulatory networks underlying EMP support the idea of a spectrum of hybrid E/M states.41,42 However, it remains to be understood whether these states are functionally very distinct from one another or that at least some of them can be considered as “microstates” within a “macrostate”.43,44
Plurality of Trajectories Enabling Varying Levels of EMP
Plasticity refers to the change of phenotype either spontaneously (i.e., without an obvious external trigger) due to biological noise or driven by specific microenvironmental cues. In cell biology, intrinsic factors such as epigenetic variations or stochastic fluctuations in gene expression or levels of signaling molecules can lead to changes in the phenotype. For instance, microRNA–mRNA interaction-driven oscillations seen without any explicit feedback loops can explain observed cell-state transitions on a slower time scale. Such scenarios may explain intrinsic and reversible EMP.45 As cells change phenotypes, they can be conceptualized as traveling along diverse paths in a high-dimensional gene expression space. Thus, characterizing cellular trajectories in this landscape is crucial in determining the allowed and forbidden cell states/phenotypes and may point toward a more rational design of differentiation therapy in cancer.46
In the quest for finding a common EMT gene expression program, Cook and Vanderhyden performed scRNA-seq across 12 EMT time-course experiments to measure the gene expression profiles of 103 999 cells from 960 samples.47 They assessed four different cancer cell lines (A549, lung; DU145, prostate; MCF7, breast; and OVCA420, ovarian) and exposed them to three EMT-inducing factors (TGFB1, EGF, and TNF). Among the three EMT inducers, TGFB1 was found to be the most potent. The study highlighted the context specificity of transcriptional dynamics of EMT, where only 22% of response genes, on average, were shared between any two of the 12 EMT time courses. Moreover, the single-cell transcriptomic profiles were grouped based on cell lines rather than by the inducing growth factor, revealing that the genetic background of cells can govern the manifestation of EMP. In another study, they analyzed scRNA-seq data spanning eight different cancer types from 266 tumors.48 They reported a high degree of intratumoral heterogeneity, which could be influenced by the tumor microenvironment and signaling associated with combinations of common regulatory pathways. Using a general EMP signature to query samples from TCGA (The Cancer Genome Atlas), they found EMP to be associated with reduced progression-free intervals and changes in immune cell proportions within the tumor microenvironment. scRNA-seq data can also be used to deconvolute the extent of the EMP signature coming from cancer cells vs that from stromal cells,49 thus possibly reconciling conflicting reports of clinicopathological association of EMP.50 Clarifying the contribution of cancer cells vs stromal cells in bulk transcriptomic readouts of EMP is crucial because recent lineage-tracing experiments in mouse models of metastatic breast cancer have suggested that a complete transition to a mesenchymal state is rare; rather, cells often underwent a partial EMT.29 Such preclinical observations reinforce the trends witnessed in three-dimensional reconstruction of the primary tumor samples that single-cell migration is an extremely rare phenomenon; instead, cells migrated mostly as “tumor buds” with simultaneous expression of both E-cadherin (epithelial cell–cell adhesion molecule) and Zeb1 (EMT-inducing transcription factor), thus displaying hybrid E/M state(s).51
The prevalence and plurality of hybrid E/M phenotypes raise the question of how many paths can cells take en route to EMT in a high-dimensional space. To address this question, Wang et al. recorded single-cell trajectories of A549 lung cancer cells labeled endogenously with Vimentin-RFP that underwent EMT for varying concentrations of TGF-β.52 They identified two parallel trajectories in a multidimensional cell-feature space, thus indicating that the transition dynamics proceeds via parallel trajectories. They conjectured that with increasing TGF-β concentrations, the initial epithelial attractor collides with two saddle node points in a sequential manner, and a new mesenchymal attractor emerges. How similar or different the transcriptomic profiles of cells moving along these two paths are yet remains to be elucidated. Nonetheless, such temporal mapping allows investigating the high-dimensional EMT landscape from a dynamical systems theory lens, as applied earlier to reveal independent trajectories for evolution of drug-tolerant cells in individual melanoma cells treated with the BRAF inhibitor vemurafenib.53,54 Parallel activation of EMT signaling pathways was also reported in scRNA-seq analysis of MCF10A cells treated with TGFβ up to a duration of 8 days,30 and cells were found to progress through EMT at different speeds despite identical duration and dose of the inducing signal, illustrating dynamic nongenetic heterogeneity. Computational modeling has predicted that parallel progression through alternative transition paths can accelerate EMT,43 but whether and how future experiments can test this prediction rigorously are worth delving into.
The diverse trajectories seen in the EMT landscape can not only underlie the heterogeneity of hybrid E/M states but also explain the “exit” from hybrid E/M phenotypes stochastically toward a more epithelial or mesenchymal one. Upon experiencing homogeneous physical compression, the H1975 lung cancer cells, identified as a hybrid E/M phenotype,55 exhibited increased gene expression noise and consequent diversification into either an epithelial or a mesenchymal phenotype, as identified through single-cell sequencing and single-molecule fluorescent in situ hybridization (smFISH)56 of canonical EMT and MET markers. smFISH measurements of gene expression, together with lineage trees, have been instrumental in inferring reversible and stochastic dynamics of cell-state transition in multiple biological contexts.57,58 Similar approaches can facilitate measuring transition rates along different trajectories in the multidimensional EMP landscape.
Reversibility and Hysteretic Behavior of EMP Dynamics
EMT is a reversible process during many stages of embryonic development. Similarly, during metastasis, the reverse of EMT—MET—is considered to drive metastatic colonization.59 In vitro MET is often studied via treatment of epithelial cells with an EMT-inducing signal (such as TGFβ or inducible expression of EMT-inducing transcription factors such as SNAIL), followed by its withdrawal. For instance, inducible SNAIL expression in LNCAP prostate cancer cells drove transcriptomic changes concomitant with EMT. Also, while the EMT-induced cells exhibited reduced proliferation and increased invasion, the reverting cells reinitiated proliferation and formed multicellular spheroids, thus recapitulating traits often driving metastatic colonization. Interestingly, upon reversal, not all genes returned to their pretreatment expression levels,11 thus enabling a “transcriptional memory”. However, because this analysis was at a population level, it could not be distinguished as to whether this “memory” was restricted to a subpopulation of cells or more equitably distributed across cells. Other experimental analyses have shown that the extent of memory can depend on at least these factors—the heterogeneity in the degree of EMT induced in different cells in a population,60 the duration after which MET was assessed,61 and markers used for MET.
Single-cell investigations are well-positioned to address some of these questions. For instance, through TGFβ treatment and its subsequent withdrawal, eight distinct cell states were identified in lung cancer, using a mass cytometry-based time-course analysis with single-cell resolution in vitro.62 Using this high-dimensional temporal data, they developed an algorithm to project the malignant cells on a lung cancer reference map called PHENOtypic STAte MaP (PHENOSTAMP) (Figure 2). They observed that cells undergoing MET took a different trajectory in this map as compared to EMT trajectories, showcasing hysteresis—a hallmark feature of EMT predicted by many computational models.63,64 Such hysteretic response can enable a short-term stimulus to trigger cell-state transition in a subpopulation, as witnessed in flow cytometry-based single-cell analysis for TGFβ-driven EMT. Hysteresis is often seen in multistable systems; in the NmuMG mouse mammary epithelial cells undergoing TGFβ-driven EMT, for TGFβ concentrations between 10 and 25pM, flow cytometry and immunofluorescence experiments revealed two subpopulations: E-cadherin-low (mesenchymal) and E-cadherin-high (epithelial),65 thus explaining the underlying basis of hysteretic behavior. Intriguingly, upon CRISPR/Cas9-mediated disruption of the ZEB/miR-200 feedback loop that enabled such multistability, the bimodality of E-cadherin and hysteretic response was largely lost in vitro. Further, the mutant cells with the disrupted ZEB/miR-200 loop had dramatically reduced metastatic potential as compared to the wild-type (with an intact ZEB/miR-200 feedback loop) one. This decrease in metastasis in vivo was observed despite no significant reduction in migration on invasion in vitro, demonstrating the key importance of multistability and consequent cell plasticity in establishing metastasis in vivo. This analysis further endorses previous in vitro observations about the role of the miR-200/ZEB1 feedback loop in mediating the reversibility of EMT66,67 and explains how ZEB1-mediated epigenetic rewiring can control the rate of MET.61,68,69
Single-cell time-course observations have also helped map the extent of reversibility of EMT as a function of the duration of induction of EMT. For instance, MCF10A cells were cotransduced with two sensors—destabilized green fluorescent protein (GFP) that is regulated by EMT-transcription factor ZEB1 and red fluorescent protein (RFP) which is driven by the promoter of E-cadherin (CDH1). Counting the number of GFP+ and RFP+ cells when exposing cells to varying durations of TGF-β revealed that cells exposed for a shorter duration (3–6 days) reverted to being epithelial in a similar time frame. However, for cells exposed for a longer duration (12–15 days), they underwent a stronger degree of EMT measured by CDH1 and ZEB1 levels, and not all of them reverted to being epithelial even after 15 days post-TGF-β removal, suggesting that at least a subpopulation of cells exhibited an “effectively irreversible” EMT; i.e., they can maintain a mesenchymal state without exogenous TGF-β.70 Such “irreversibility” and consequent lack of plasticity displayed attenuated metastatic outgrowth, while coexpression of epithelial and mesenchymal programs enabled cell plasticity (i.e., more reversible) and consequent metastasis.60 Further longer-term wait time experiments later unraveled that cells post-EMT induction can revert back to being epithelial after an initial delay.61
Computational modeling simulations suggest nongenetic heterogeneity in terms of reversal time, a prediction that remains to be tested experimentally through single-cell dynamical investigations. Epigenetic reprogramming has been proposed to underlie such effectively irreversible EMT, and longer waiting periods may allow for sufficient cell divisions causing gradual loss of such memory (epigenetic reprogramming). Given that most of such analysis has been at a population level,61 future studies should track dynamic changes in the epigenetic status driven by key EMT/MET regulators at the single-cell level to establish causative connection(s) between the rate of EMT reversibility and chromatin reprogramming.
Another important aspect while investigating MET dynamics is to design experiments where MET is induced in a dose- and/or duration-dependent manner, similar to analysis of EMT in MCF10A cells.31 Given that EMT and MET may not follow the same trajectories and that many MET drivers such as GRHL2 can drive epigenetic remodeling,71 the trajectories of EMT reversal and of MET induction may not necessarily overlap, especially in a high-dimensional space.
Spatial Heterogeneity along the EMP Spectrum
Most in vitro investigations on EMP have considered only temporal dynamics, ignoring the spatial component. However, heterogeneity along the EMP status is observed spatially in tumors, with the mesenchymal cells preferentially located at the invasive edge while epithelial ones are present in the interior, thus indicating an “EMT gradient”.72,73 Computational models have revealed how diffusing molecules such as TGF-β, secreted mostly by stromal cells, coupled with cell–cell communication (for instance, through Notch-Jagged signaling), can explain the existence of such gradients. Importantly, Notch-Jagged signaling can stabilize cells in hybrid E/M phenotypes,74 thus possibly explaining their observed strong cell–cell communication traits as seen in scRNA-seq by quantifying the degree to which a given cell type can send and receive signals.32
Similar to pseudotime, pseudospatial trajectories have been generated for cells seeded within cloning rings in the center of a tissue culture dish; these rings were removed to allow for spontaneous EMT.75 These trajectories showed opposing gradients of CDH1 and VIM, reflecting a cell-state continuum. Recent advancements in in vivo intravital imaging have allowed a more comprehensive mapping of spatial heterogeneity in the EMP status. Using Fsp1 (fibroblast specific protein 1) as a marker to identify cells that have undergone EMT in vivo, cells exhibiting a more mesenchymal state were found to localize preferentially closer to blood vessels and also had longer membrane extensions oriented toward the vessels.76 How accurate and sensitive Fsp1 is as a marker of cells in hybrid epithelial/mesenchymal states is still under debate,77 given the multifaceted context-specific display of EMP.
Association of EMP with Patient Survival
Initial reports, positing EMT as a binary process, proposed EMT and its associated transcription factors to be associated with worse clinical outcomes.78,79 However, further pan-cancer analysis revealed that association of EMT with patient survival metrics was largely cancer subtype-specific.80,81 Particularly, coexpression of epithelial and mesenchymal signatures was found to be associated with the worst patient outcomes in luminal and basal breast cancer,20 thus highlighting the aggressive behavior of hybrid E/M cells. Consistently, a recent sequential immunohistochemistry analysis demonstrated that the presence of hybrid E/M cells was strongly associated with poor prognosis. This analysis suggested that the presence of a minimum percentage of tumor cells in the hybrid E/M state (E/M score <2%) was sufficient to confer poor overall and disease-free survival in patients.82 Such strong association of hybrid E/M cells with patient survival can be at least partly attributed to their salient features—enhanced stemness, drug-resistance, and immune-evasion.
Beyond these prognostic observations based on a few markers, computational methods to connect single-cell RNA-sequencing data to EMT have been developed, such as scPrognosis. It integrates EMT-associated pseudotime data and dynamic gene coexpression networks and suggests that most breast cancer signature genes so identified a peak during hybrid E/M state(s) acquisition.83 Similarly, another metric overlays single-cell RNA-sequencing data on targets of specific miRNAs to improve patient risk stratification in colorectal cancer.84 Future attempts to test the prognostic power of hybrid E/M signatures shall depend on identifying the tissue-specific signatures for EMT.
Conclusion
Single-cell investigations of EMP have begun to elucidate the complexity of phenotypic plasticity in the context of cancer metastasis. With the advent of single-cell technologies at transcriptomic, proteomic, live-cell imaging, and lineage-tracing levels, there has been a burst in employing them to improve our understanding of underlying dynamical patterns involved in cell decision-making. Although the emphasis to study EMP has been steadily growing in the past few years, EMP-based studies still form a minor portion of the total set of single-cell-based studies (Figure 3). We envision that further single-cell analysis of EMP will be instrumental in uncovering the mechanistic basis of the complex dynamics of EMP and its functional relevance in tumor progression and metastasis.
Figure 3.
Bar-plot depicting the increasing focus on EMP single-cell research documents in a decade. (A) Light blue bars represent the number of articles with major keywords as “single-cell”, and dark blue bars denote articles with keywords “EMT”, “EMP”, “Epithelial–Mesenchymal Transition”, or “Epithelial–Mesenchymal Plasticity” within the “single-cell” domain (right), zoomed-in view (left). (B) Light brown bars represent number of articles with major keywords “EMT”, “EMP”, “Epithelial–Mesenchymal Transition”, or “Epithelial–Mesenchymal Plasticity”; dark brown bars represent those with keyword “single-cell” within the EMT domain (right); zoomed-in view (left) (data taken Scopus on Nov. 8, 2022).
Acknowledgments
The authors would like to acknowledge the Ramanujan Fellowship (SB/S2/RJN-049/2018) awarded to M.K.J. by the Science and Engineering Research Board (SERB), Department of Science & Technology, Government of India. Additionally, we would like to acknowledge the Prime Ministers’ Research Fellowship, Government of India, received by S.S. and S.S.
The authors declare no competing financial interest.
References
- Xue B. K.; Leibler S. Benefits of Phenotypic Plasticity for Population Growth in Varying Environments. Proc. Natl. Acad. Sci. U. S. A. 2018, 115 (50), 12745–12750. 10.1073/pnas.1813447115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajagopal J.; Stanger B. Z. Plasticity in the Adult: How Should the Waddington Diagram Be Applied to Regenerating Tissues?. Dev Cell 2016, 36 (2), 133–137. 10.1016/j.devcel.2015.12.021. [DOI] [PubMed] [Google Scholar]
- Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov 2022, 12 (1), 31–46. 10.1158/2159-8290.CD-21-1059. [DOI] [PubMed] [Google Scholar]
- Tripathi S.; Levine H.; Jolly M. K. The Physics of Cellular Decision-Making during Epithelial-Mesenchymal Transition. Annu. Rev. Biophys 2020, 49, 1–18. 10.1146/annurev-biophys-121219-081557. [DOI] [PubMed] [Google Scholar]
- Han Y.; Villarreal-Ponce A.; Gutierrez G.; Nguyen Q.; Sun P.; Wu T.; Sui B.; Berx G.; Brabletz T.; Kessenbrock K.; Zeng Y. A.; Watanabe K.; Dai X. Coordinate Control of Basal Epithelial Cell Fate and Stem Cell Maintenance by Core EMT Transcription Factor Zeb1. Cell Rep 2022, 38 (2), 110240. 10.1016/j.celrep.2021.110240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nieto M. A.; Huang R. Y. Y. J.; Jackson R. A. A.; Thiery J. P. P. EMT: 2016. Cell 2016, 166 (1), 21–45. 10.1016/j.cell.2016.06.028. [DOI] [PubMed] [Google Scholar]
- Li Q.; Hutchins A. P.; Chen Y.; Li S.; Shan Y.; Liao B.; Zheng D.; Shi X.; Li Y.; Chan W. Y.; Pan G.; Wei S.; Shu X.; Pei D. A Sequential EMT-MET Mechanism Drives the Differentiation of Human Embryonic Stem Cells towards Hepatocytes. Nat. Commun. 2017, 8, 15166. 10.1038/ncomms15166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia D.; Li X.; Bocci F.; Tripathi S.; Deng Y.; Jolly M. K.; Onuchic J. N.; Levine H. Quantifying Cancer Epithelial-Mesenchymal Plasticity and Its Association with Stemness and Immune Response. J. Clin Med. 2019, 8 (5), 725. 10.3390/jcm8050725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pei D.; Shu X.; Gassama-Diagne A.; Thiery J. P. Mesenchymal-Epithelial Transition in Development and Reprogramming. Nature Cell Biology 2019 21:1 2019, 21 (1), 44–53. 10.1038/s41556-018-0195-z. [DOI] [PubMed] [Google Scholar]
- Brabletz S.; Schuhwerk H.; Brabletz T.; Stemmler M. P. Dynamic EMT: A Multi-tool for Tumor Progression. EMBO J. 2021, 40 (18), e108647 10.15252/embj.2021108647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stylianou N.; Lehman M. L.; Wang C.; Fard A. T.; Rockstroh A.; Fazli L.; Jovanovic L.; Ward M.; Sadowski M. C.; Kashyap A. S.; Buttyan R.; Gleave M. E.; Westbrook T. F.; Williams E. D.; Gunter J. H.; Nelson C. C.; Hollier B. G. A Molecular Portrait of Epithelial-Mesenchymal Plasticity in Prostate Cancer Associated with Clinical Outcome. Oncogene 2019, 38 (7), 913–934. 10.1038/s41388-018-0488-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taube J. H.; Herschkowitz J. I.; Komurov K.; Zhou A. Y.; Gupta S.; Yang J.; Hartwell K.; Onder T. T.; Gupta P. B.; Evans K. W.; Hollier B. G.; Ram P. T.; Lander E. S.; Rosen J. M.; Weinberg R. A.; Mani S. A. Core Epithelial-to-Mesenchymal Transition Interactome Gene-Expression Signature Is Associated with Claudin-Low and Metaplastic Breast Cancer Subtypes. Proc. Natl. Acad. Sci. U. S. A. 2010, 107 (35), 15449–15454. 10.1073/pnas.1004900107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jain A. P.; Sambath J.; Sathe G.; George I. A.; Pandey A.; Thompson E. W.; Kumar P. Pan-Cancer Quantitation of Epithelial-Mesenchymal Transition Dynamics Using Parallel Reaction Monitoring-Based Targeted Proteomics Approach. J. Transl Med. 2022, 20 (1), 84. 10.1186/s12967-021-03227-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson K. S.; Hussein S.; Chakraborty P.; Muruganantham A.; Mikhail S.; Gonzalez G.; Song S.; Jolly M. K.; Toneff M. J.; Benton M. L.; Lin Y. C.; Taube J. H. CTCF Expression and Dynamic Motif Accessibility Modulates Epithelial-Mesenchymal Gene Expression. Cancers (Basel) 2022, 14 (1), 209. 10.3390/cancers14010209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mandal M.; Ghosh B.; Rajput M.; Chatterjee J. Impact of intercellular connectivity on epithelial mesenchymal transition plasticity. BBA Mol. Cell Res. 2020, 1867, 118784. 10.1016/j.bbamcr.2020.118784. [DOI] [PubMed] [Google Scholar]
- Huang S.; Eichler G.; Bar-Yam Y.; Ingber D. E. Cell Fates as High-Dimensional Attractor States of a Complex Gene Regulatory Network. Phys. Rev. Lett. 2005, 94 (12), 128701. 10.1103/PhysRevLett.94.128701. [DOI] [PubMed] [Google Scholar]
- Hari K.; Sabuwala B.; Subramani B. V.; La Porta C. A. M.; Zapperi S.; Font-Clos F.; Jolly M. K. Identifying Inhibitors of Epithelial-Mesenchymal Plasticity Using a Network Topology Based Approach. NPJ. Syst. Biol. Appl. 2020, 6 (1), 15. 10.1038/s41540-020-0132-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J.; Tian X. J.; Zhang H.; Teng Y.; Li R.; Bai F.; Elankumaran S.; Xing J. TGF-β-Induced Epithelial-to-Mesenchymal Transition Proceeds through Stepwise Activation of Multiple Feedback Loops. Sci. Signal 2014, 7 (345), ra91. 10.1126/scisignal.2005304. [DOI] [PubMed] [Google Scholar]
- Brown M. S.; Abdollahi B.; Wilkins O. M.; Lu H.; Chakraborty P.; Ognjenovic N. B.; Muller K. E.; Jolly M. K.; Christensen B. C.; Hassanpour S.; Pattabiraman D. R. Phenotypic Heterogeneity Driven by Plasticity of the Intermediate EMT State Governs Disease Progression and Metastasis in Breast Cancer. Sci. Adv. 2022, 8 (31), eabj8002 10.1126/sciadv.abj8002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grosse-Wilde A.; Fouquier d’ Herouel A.; McIntosh E.; Ertaylan G.; Skupin A.; Kuestner R. E.; del Sol A.; Walters K.-A.; Huang S. Stemness of the Hybrid Epithelial/Mesenchymal State in Breast Cancer and Its Association with Poor Survival. PLoS One 2015, 10 (5), e0126522 10.1371/journal.pone.0126522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jolly M. K.; Somarelli J. A.; Sheth M.; Biddle A.; Tripathi S. C. C.; Armstrong A. J. J.; Hanash S. M. M.; Bapat S. A. A.; Rangarajan A.; Levine H. Hybrid Epithelial/Mesenchymal Phenotypes Promote Metastasis and Therapy Resistance across Carcinomas. Pharmacol Ther 2019, 194, 161–184. 10.1016/j.pharmthera.2018.09.007. [DOI] [PubMed] [Google Scholar]
- Steinbichler T. B.; Dudas J.; Ingruber J.; Glueckert R.; Sprung S.; Fleischer F.; Cidlinsky N.; Dejaco D.; Kofler B.; Giotakis A. I.; Skvortsova I. I.; Riechelmann H. Slug Is a Surrogate Marker of Epithelial to Mesenchymal Transition (EMT) in Head and Neck Cancer. J. Clin Med. 2020, 9 (7), 2061. 10.3390/jcm9072061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Subbalakshmi A. R.; Sahoo S.; Biswas K.; Jolly M. K. A Computational Systems Biology Approach Identifies SLUG as a Mediator of Partial Epithelial-Mesenchymal Transition (EMT). Cells Tissues Organs 2022, 211 (6), 689–702. 10.1159/000512520. [DOI] [PubMed] [Google Scholar]
- Aggarwal V.; Sahoo S.; Donnenberg V. S.; Chakraborty P.; Jolly M. K.; Sant S. P4HA2: A Link between Tumor-Intrinsic Hypoxia, Partial EMT and Collective Migration. Advances in Cancer Biology - Metastasis 2022, 5, 100057. 10.1016/j.adcanc.2022.100057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puram S. V.; Tirosh I.; Parikh A. S.; Patel A. P.; Yizhak K.; Gillespie S.; Rodman C.; Luo C. L.; Mroz E. A.; Emerick K. S.; Deschler D. G.; Varvares M. A.; Mylvaganam R.; Rozenblatt-Rosen O.; Rocco J. W.; Faquin W. C.; Lin D. T.; Regev A.; Bernstein B. E. Single-Cell Transcriptomic Analysis of Primary and Metastatic Tumor Ecosystems in Head and Neck Cancer. Cell 2017, 171 (7), 1611–1624. 10.1016/j.cell.2017.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pastushenko I.; Brisebarre A.; Sifrim A.; Fioramonti M.; Revenco T.; Boumahdi S.; Van Keymeulen A.; Brown D.; Moers V.; Lemaire S.; De Clercq S.; Minguijón E.; Balsat C.; Sokolow Y.; Dubois C.; De Cock F.; Scozzaro S.; Sopena F.; Lanas A.; D’Haene N.; Salmon I.; Marine J.-C.; Voet T.; Sotiropoulou P. A.; Blanpain C. Identification of the Tumour Transition States Occurring during EMT. Nature 2018, 556 (7702), 463–468. 10.1038/s41586-018-0040-3. [DOI] [PubMed] [Google Scholar]
- Pastushenko I.; Mauri F.; Song Y.; de Cock F.; Meeusen B.; Swedlund B.; Impens F.; Van Haver D.; Opitz M.; Thery M.; Bareche Y.; Lapouge G.; Vermeersch M.; Van Eycke Y. R.; Balsat C.; Decaestecker C.; Sokolow Y.; Hassid S.; Perez-Bustillo A.; Agreda-Moreno B.; Rios-Buceta L.; Jaen P.; Redondo P.; Sieira-Gil R.; Millan-Cayetano J. F.; Sanmatrtin O.; D’Haene N.; Moers V.; Rozzi M.; Blondeau J.; Lemaire S.; Scozzaro S.; Janssens V.; De Troya M.; Dubois C.; Pérez-Morga D.; Salmon I.; Sotiriou C.; Helmbacher F.; Blanpain C. Fat1 Deletion Promotes Hybrid EMT State, Tumour Stemness and Metastasis. Nature 2021, 589 (7842), 448–455. 10.1038/s41586-020-03046-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hari K.; Ullanat V.; Balasubramanian A.; Gopalan A.; Jolly M. K. Landscape of Epithelial Mesenchymal Plasticity as an Emergent Property of Coordinated Teams in Regulatory Networks. Elife 2022, 11, e76535 10.7554/eLife.76535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lüönd F.; Sugiyama N.; Bill R.; Bornes L.; Hager C.; Tang F.; Santacroce N.; Beisel C.; Ivanek R.; Bürglin T.; Tiede S.; van Rheenen J.; Christofori G. Distinct Contributions of Partial and Full EMT to Breast Cancer Malignancy. Dev Cell 2021, 56 (23), 3203–3221. 10.1016/j.devcel.2021.11.006. [DOI] [PubMed] [Google Scholar]
- Deshmukh A. P.; Vasaikar S. V.; Tomczak K.; Tripathi S.; Den Hollander P.; Arslan E.; Chakraborty P.; Soundararajan R.; Jolly M. K.; Rai K.; Levine H.; Mani S. A. Identification of EMT Signaling Cross-Talk and Gene Regulatory Networks by Single-Cell RNA Sequencing. Proc. Natl. Acad. Sci. U. S. A. 2021, 118 (19), e2102050118 10.1073/pnas.2102050118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Panchy N.; Watanabe K.; Takahashi M.; Willems A.; Hong T. Comparative Single-Cell Transcriptomes of Dose and Time Dependent Epithelial-Mesenchymal Spectrums. NAR Genom Bioinform 2022, 4 (3), 36159174 10.1093/nargab/lqac072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bocci F.; Zhou P.; Nie Q. Single-Cell RNA-Seq Analysis Reveals the Acquisition of Cancer Stem Cell Traits and Increase of Cell-Cell Signaling during EMT Progression. Cancers (Basel) 2021, 13 (22), 5726. 10.3390/cancers13225726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahoo S.; Duddu A. S.; Biddle A.; Jolly M. K.; Ashraf B. Interconnected High-Dimensional Landscapes of Epithelial-Mesenchymal Plasticity and Stemness. Clin Exp Metastasis 2022, 39 (2), 279–290. 10.1007/s10585-021-10139-2. [DOI] [PubMed] [Google Scholar]
- Zou H.; Yang Z.; Chan Y. S.; Yeung S.-k. A.; Alam M. K.; Si T.; Xu T.; Yang M. Single Cell Analysis of Mechanical Properties and EMT-Related Gene Expression Profiles in Cancer Fingers. iScience 2022, 25 (3), 103917. 10.1016/j.isci.2022.103917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basu A.; Paul M. K.; Alioscha-Perez M.; Grosberg A.; Sahli H.; Dubinett S. M.; Weiss S. Statistical Parametrization of Cell Cytoskeleton Reveals Lung Cancer Cytoskeletal Phenotype with Partial EMT Signature. Communications Biology 2022 5:1 2022, 5 (1), 1–11. 10.1038/s42003-022-03358-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leggett S. E.; Sim J. Y.; Rubins J. E.; Neronha Z. J.; Williams E. K.; Wong I. Y. Morphological Single Cell Profiling of the Epithelial-Mesenchymal Transition. Integr. Biol. 2016, 8 (11), 1133–1144. 10.1039/C6IB00139D. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mandal M.; Ghosh B.; Anura A.; Mitra P.; Pathak T.; Chatterjee J. Modeling Continuum of Epithelial Mesenchymal Transition Plasticity. Integr. Biol. 2016, 8 (2), 167–176. 10.1039/C5IB00219B. [DOI] [PubMed] [Google Scholar]
- Devaraj V.; Bose B. Morphological State Transition Dynamics in EGF-Induced Epithelial to Mesenchymal Transition. J. Clin Med. 2019, 8 (7), 911. 10.3390/jcm8070911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- You L.; Su P.-R.; Betjes M.; Rad R. G.; Beerens C.; van Oosten E.; Leufkens F.; Gasecka P.; Muraro M.; van Tol R.; Chou T.-C.; van Steenderen D.; Farooq S.; Hardillo J. A. U.; de Jong R. B.; Brinks D.; Chien M.-P. Functional Single Cell Selection and Annotated Profiling of Dynamically Changing Cancer Cells. bioRxiv 2021, 464052. 10.1101/2021.10.12.464054. [DOI] [Google Scholar]
- Carstens J. L.; Yang S.; Correa de Sampaio P.; Zheng X.; Barua S.; McAndrews K. M.; Rao A.; Burks J. K.; Rhim A. D.; Kalluri R. Stabilized Epithelial Phenotype of Cancer Cells in Primary Tumors Leads to Increased Colonization of Liver Metastasis in Pancreatic Cancer. Cell Rep 2021, 35 (2), 108990. 10.1016/j.celrep.2021.108990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steinway S. N.; Zañudo J. G. T.; Michel P. J.; Feith D. J.; Loughran T. P.; Albert R. Combinatorial Interventions Inhibit TGFβ-Driven Epithelial-to-Mesenchymal Transition and Support Hybrid Cellular Phenotypes. NPJ. Syst. Biol. Appl. 2015, 1 (1), 15014. 10.1038/npjsba.2015.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Font-Clos F.; Zapperi S.; La Porta C. A. M. Topography of Epithelial-Mesenchymal Plasticity. Proc. Natl. Acad. Sci. U. S. A. 2018, 115 (23), 5902–5907. 10.1073/pnas.1722609115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goetz H.; Melendez-Alvarez J. R.; Chen L.; Tian X. J. A Plausible Accelerating Function of Intermediate States in Cancer Metastasis. PLoS Comput. Biol. 2020, 16 (3), e1007682 10.1371/journal.pcbi.1007682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pillai M.; Jolly M. K. Systems-Level Network Modeling Deciphers the Master Regulators of Phenotypic Plasticity and Heterogeneity in Melanoma. iScience 2021, 24 (10), 103111. 10.1016/j.isci.2021.103111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nordick B.; Yu P. Y.; Liao G.; Hong T. Nonmodular Oscillator and Switch Based on RNA Decay Drive Regeneration of Multimodal Gene Expression. Nucleic Acids Res. 2022, 50 (7), 3693–3708. 10.1093/nar/gkac217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burkhardt D. B.; San Juan B. P.; Lock J. G.; Krishnaswamy S.; Chaffer C. L. Mapping Phenotypic Plasticity upon the Cancer Cell State Landscape Using Manifold Learning. Cancer Discov 2022, 12 (8), 1847–1859. 10.1158/2159-8290.CD-21-0282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cook D. P.; Vanderhyden B. C. Context Specificity of the EMT Transcriptional Response. Nat. Commun. 2020, 11, 2142. 10.1038/s41467-020-16066-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cook D. P.; Vanderhyden B. C. Transcriptional Census of Epithelial-Mesenchymal Plasticity in Cancer. Sci. Adv. 2022, 8 (1), eabi7640 10.1126/sciadv.abi7640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyler M.; Tirosh I. Decoupling Epithelial-Mesenchymal Transitions from Stromal Profiles by Integrative Expression Analysis. Nat. Commun. 2021, 12 (1), 2592. 10.1038/s41467-021-22800-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mandal S.; Tejaswi T.; Janivara R.; Srikrishnan S.; Thakur P.; Sahoo S.; Chakraborty P.; Sohal S. S.; Levine H.; George J. T.; Jolly M. K. Transcriptomic-Based Quantification of the Epithelial-Hybrid-Mesenchymal Spectrum across Biological Contexts. Biomolecules 2022, 12 (1), 29. 10.3390/biom12010029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bronsert P.; Enderle-Ammour K.; Bader M.; Timme S.; Kuehs M.; Csanadi A.; Kayser G.; Kohler I.; Bausch D.; Hoeppner J.; Hopt U. T.; Keck T.; Stickeler E.; Passlick B.; Schilling O.; Reiss C. P.; Vashist Y.; Brabletz T.; Berger J.; Lotz J.; Olesch J.; Werner M.; Wellner U. F. Cancer Cell Invasion and EMT Marker Expression: A Three-Dimensional Study of the Human Cancer-Host Interface. J. Pathol 2014, 234 (3), 410–422. 10.1002/path.4416. [DOI] [PubMed] [Google Scholar]
- Wang W.; Poe D.; Yang Y.; Hyatt T.; Xing J. Epithelial-to-Mesenchymal Transition Proceeds through Directional Destabilization of Multidimensional Attractor. Elife 2022, 11, e74866 10.7554/eLife.74866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su Y.; Ko M. E.; Cheng H.; Zhu R.; Xue M.; Wang J.; Lee J. W.; Frankiw L.; Xu A.; Wong S.; Robert L.; Takata K.; Yuan D.; Lu Y.; Huang S.; Ribas A.; Levine R.; Nolan G. P.; Wei W.; Plevritis S. K.; Li G.; Baltimore D.; Heath J. R. Multi-Omic Single-Cell Snapshots Reveal Multiple Independent Trajectories to Drug Tolerance in a Melanoma Cell Line. Nat. Commun. 2020, 11 (1), 2345. 10.1038/s41467-020-15956-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pillai M.; Chen Z.; Jolly M. K.; Li C. Quantitative Landscapes Reveal Trajectories of Cell-State Transitions Associated with Drug Resistance in Melanoma. iScience 2022, 25 (12), 105499. 10.1016/j.isci.2022.105499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jolly M. K.; Tripathi S. C.; Jia D.; Mooney S. M.; Celiktas M.; Hanash S. M.; Mani S. A.; Pienta K. J.; Ben-Jacob E.; Levine H. Stability of the Hybrid Epithelial/Mesenchymal Phenotype. Oncotarget 2016, 7 (19), 27067–27084. 10.18632/oncotarget.8166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao X.; Hu J.; Li Y.; Guo M. Volumetric Compression Develops Noise-Driven Single-Cell Heterogeneity. Proc. Natl. Acad. Sci. U. S. A. 2021, 118 (51), e2110550118 10.1073/pnas.2110550118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hormoz S.; Singer Z. S.; Linton J. M.; Antebi Y. E.; Shraiman B. I.; Elowitz M. B. Inferring Cell-State Transition Dynamics from Lineage Trees and Endpoint Single-Cell Measurements. Cell Syst 2016, 3 (5), 419–433. 10.1016/j.cels.2016.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wheat J. C.; Sella Y.; Willcockson M.; Skoultchi A. I.; Bergman A.; Singer R. H.; Steidl U. Single-Molecule Imaging of Transcription Dynamics in Somatic Stem Cells. Nature 2020, 583 (7816), 431–436. 10.1038/s41586-020-2432-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gunasinghe N. P. A. D.; Wells A.; Thompson E. W.; Hugo H. J. Mesenchymal-Epithelial Transition (MET) as a Mechanism for Metastatic Colonisation in Breast Cancer. Cancer and Metastasis Reviews 2012, 31 (3–4), 469–478. 10.1007/s10555-012-9377-5. [DOI] [PubMed] [Google Scholar]
- Eichelberger L.; Saini M.; Moreno H. D.; Klein C.; Bartsch J. M.; Falcone M.; Reitberger M.; Espinet E.; Vogel V.; Graf E.; Schwarzmayr T.; Strom T.-M.; Lehmann M.; Königshoff M.; Pfarr N.; Würth R.; Donato E.; Haas S.; Spaich S.; Sütterlin M.; Schneeweiss A.; Weichert W.; Schotta G.; Trumpp A.; Sprick M. R.; Scheel C. H. Maintenance of Epithelial Traits and Resistance to Mesenchymal Reprogramming Promote Proliferation in Metastatic Breast Cancer. bioRxiv 2020, 998823. 10.1101/2020.03.19.998823. [DOI] [Google Scholar]
- Jain P.; Corbo S.; Mohammad K.; Sahoo S.; Ranganathan S.; George J. T.; Levine H.; Taube J.; Toneff M.; Jolly M. K. Epigenetic Memory Acquired during Long-Term EMT Induction Governs the Recovery to the Epithelial State. J. R Soc. Interface 2023, 20 (198), 20220627 10.1098/rsif.2022.0627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karacosta L. G.; Anchang B.; Ignatiadis N.; Kimmey S. C.; Benson J. A.; Shrager J. B.; Tibshirani R.; Bendall S. C.; Plevritis S. K. Mapping Lung Cancer Epithelial-Mesenchymal Transition States and Trajectories with Single-Cell Resolution. Nat. Commun. 2019, 10, 5587. 10.1038/s41467-019-13441-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu M.; Jolly M. K.; Levine H.; Onuchic J. N.; Ben-Jacob E. MicroRNA-Based Regulation of Epithelial-Hybrid-Mesenchymal Fate Determination. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (45), 18144–18149. 10.1073/pnas.1318192110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong T.; Watanabe K.; Ta C. H.; Villarreal-Ponce A.; Nie Q.; Dai X. An Ovol2-Zeb1Mutual Inhibitory Circuit Governs Bidirectional and Multi-Step Transition between Epithelial and Mesenchymal States. PLoS Comput. Biol. 2015, 11 (11), e1004569 10.1371/journal.pcbi.1004569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Celià-Terrassa T.; Bastian C.; Liu D. D.; Ell B.; Aiello N. M.; Wei Y.; Zamalloa J.; Blanco A. M.; Hang X.; Kunisky D.; Li W.; Williams E. D.; Rabitz H.; Kang Y. Hysteresis Control of Epithelial-Mesenchymal Transition Dynamics Conveys a Distinct Program with Enhanced Metastatic Ability. Nat. Commun. 2018, 9, 5005. 10.1038/s41467-018-07538-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe K.; Panchy N.; Noguchi S.; Suzuki H.; Hong T. Combinatorial Perturbation Analysis Reveals Divergent Regulations of Mesenchymal Genes during Epithelial-to-Mesenchymal Transition. NPJ. Syst. Biol. Appl. 2019, 5, 21. 10.1038/s41540-019-0097-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gregory P. A.; Bracken C. P.; Smith E.; Bert A. G.; Wright J. a.; Roslan S.; Morris M.; Wyatt L.; Farshid G.; Lim Y.-Y.; Lindeman G. J.; Shannon M. F.; Drew P. a.; Khew-Goodall Y.; Goodall G. J. An Autocrine TGF-Beta/ZEB/MiR-200 Signaling Network Regulates Establishment and Maintenance of Epithelial-Mesenchymal Transition. Mol. Biol. Cell 2011, 22 (10), 1686–1698. 10.1091/mbc.e11-02-0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Somarelli J. A.; Shetler S.; Jolly M. K.; Wang X.; Bartholf Dewitt S.; Hish A. J.; Gilja S.; Eward W. C.; Ware K. E.; Levine H.; Armstrong A. J.; Garcia-Blanco M. A. Mesenchymal-Epithelial Transition in Sarcomas Is Controlled by the Combinatorial Expression of MicroRNA 200s and GRHL2. Mol. Cell. Biol. 2016, 36 (19), 2503–2513. 10.1128/MCB.00373-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dumont N.; Wilson M. B.; Crawford Y. G.; Reynolds P. A.; Sigaroudinia M.; Tlsty T. D. Sustained Induction of Epithelial to Mesenchymal Transition Activates DNA Methylation of Genes Silenced in Basal-like Breast Cancers. Proc. Natl. Acad. Sci. U. S. A. 2008, 105 (39), 14867–14872. 10.1073/pnas.0807146105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia W.; Deshmukh A.; Mani S. A.; Jolly M. K.; Levine H. A Possible Role for Epigenetic Feedback Regulation in the Dynamics of the Epithelial-Mesenchymal Transition (EMT). Phys. Biol. 2019, 16 (6), 066004. 10.1088/1478-3975/ab34df. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung V. Y.; Tan T. Z.; Ye J.; Huang R.-L.; Lai H.-C.; Kappei D.; Wollmann H.; Guccione E.; Huang R. Y.-J. The Role of GRHL2 and Epigenetic Remodeling in Epithelial-Mesenchymal Plasticity in Ovarian Cancer Cells. Commun. Biol. 2019, 2, 272. 10.1038/s42003-019-0506-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brabletz T.; Jung A.; Reu S.; Porzner M.; Hlubek F.; Kunz-Schughart L. A.; Knuechel R.; Kirchner T. Variable β-Catenin Expression in Colorectal Cancers Indicates Tumor Progression Driven by the Tumor Environment. Proc. Natl. Acad. Sci. U. S. A. 2001, 98 (18), 10356–10361. 10.1073/pnas.171610498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu S.; Cong Y.; Wang D.; Sun Y.; Deng L.; Liu Y.; Martin-Trevino R.; Shang L.; McDermott S. P.; Landis M. D.; Hong S.; Adams A.; D’Angelo R.; Ginestier C.; Charafe-Jauffret E.; Clouthier S. G.; Birnbaum D.; Wong S. T.; Zhan M.; Chang J. C.; Wicha M. S. Breast Cancer Stem Cells Transition between Epithelial and Mesenchymal States Reflective of Their Normal Counterparts. Stem Cell Reports 2014, 2 (1), 78–91. 10.1016/j.stemcr.2013.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boareto M.; Jolly M. K.; Goldman A.; Pietilä M.; Mani S. A.; Sengupta S.; Ben-Jacob E.; Levine H.; Onuchic J. N. Notch-Jagged Signalling Can Give Rise to Clusters of Cells Exhibiting a Hybrid Epithelial/Mesenchymal Phenotype. J. R Soc. Interface 2016, 13 (118), 20151106. 10.1098/rsif.2015.1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McFaline-Figueroa J. L.; Hill A. J.; Qiu X.; Jackson D.; Shendure J.; Trapnell C. A Pooled Single-Cell Genetic Screen Identifies Regulatory Checkpoints in the Continuum of the Epithelial-to-Mesenchymal Transition. Nat. Genet. 2019, 51 (9), 1389–1398. 10.1038/s41588-019-0489-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Z.; Zhu X.; Cui K.; Mancuso J.; Federley R.; Fischer K.; Teng G.; Mittal V.; Gao D.; Zhao H.; Wong S. T. C. In Vivo Visualization and Characterization of Epithelial-Mesenchymal Transition in Breast Tumors. Cancer Res. 2016, 76 (8), 2094–2104. 10.1158/0008-5472.CAN-15-2662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bornes L.; van Scheppingen R. H.; Beerling E.; Schelfhorst T.; Ellenbroek S. I. J.; Seinstra D.; van Rheenen J. Fsp1-Mediated Lineage Tracing Fails to Detect the Majority of Disseminating Cells Undergoing EMT. Cell Rep 2019, 29 (9), 2565–2569. 10.1016/j.celrep.2019.10.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang J.; Mani S. A.; Donaher J. L.; Ramaswamy S.; Itzykson R. A.; Come C.; Savagner P.; Gitelman I.; Richardson A.; Weinberg R. A. Twist, a Master Regulator of Morphogenesis, Plays an Essential Role in Tumor Metastasis. Cell 2004, 117 (7), 927–939. 10.1016/j.cell.2004.06.006. [DOI] [PubMed] [Google Scholar]
- Mani S. A.; Yang J.; Brooks M.; Schwaninger G.; Zhou A.; Miura N.; Kutok J. L.; Hartwell K.; Richardson A. L.; Weinberg R. A. Mesenchyme Forkhead 1 (FOXC2) Plays a Key Role in Metastasis and Is Associated with Aggressive Basal-like Breast Cancers. Proc. Natl. Acad. Sci. U. S. A. 2007, 104 (24), 10069–10074. 10.1073/pnas.0703900104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan T. Z.; Miow Q. H.; Miki Y.; Noda T.; Mori S.; Huang R. Y.-J.; Thiery J. P. Epithelial-Mesenchymal Transition Spectrum Quantification and Its Efficacy in Deciphering Survival and Drug Responses of Cancer Patients. EMBO Mol. Med. 2014, 6 (10), 1279–1293. 10.15252/emmm.201404208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- George J. T.; Jolly M. K.; Xu S.; Somarelli J. A.; Levine H. Survival Outcomes in Cancer Patients Predicted by a Partial EMT Gene Expression Scoring Metric. Cancer Res. 2017, 77 (22), 6415–6428. 10.1158/0008-5472.CAN-16-3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Godin L.; Balsat C.; van Eycke Y.; Allard J.; Royer C.; Remmelink M.; Pastushenko I.; D'Haene N.; Blanpain C.; Salmon I.; Rorive S.; Decaestecker C. A Novel Approach for Quantifying Cancer Cells Showing Hybrid Epithelial/Mesenchymal States in Large Series of Tissue Samples : Towards a New Prognostic Marker. Cancers (Basel) 2020, 12, 906. 10.3390/cancers12040906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X.; Liu L.; Goodall G. J.; Schreiber A.; Xu T.; Li J.; Le T. D. A Novel Single-Cell Based Method for Breast Cancer Prognosis. PLoS Comput. Biol. 2020, 16 (8), e1008133 10.1371/journal.pcbi.1008133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willems A.; Panchy N.; Hong T. Using Single-Cell RNA Sequencing and MicroRNA Targeting Data to Improve Colorectal Cancer Survival Prediction. Cells 2023, 12 (2), 228. 10.3390/cells12020228. [DOI] [PMC free article] [PubMed] [Google Scholar]


