Review

Cell division symmetry control and cancer stem cells

  • Received: 15 February 2020 Accepted: 26 April 2020 Published: 06 May 2020
  • Stem cells including cancer stem cells (CSC) divide symmetrically or asymmetrically. Usually symmetric cell division makes two daughter cells of the same fate, either as stem cells or more differentiated progenies; while asymmetric cell division (ACD) produces daughter cells of different fates. In this review, we first provide an overview of ACD, and then discuss more molecular details of ACD using the well-characterized Drosophila neuroblast system as an example. Aiming to explore the connections between cell heterogeneity in cancers and the critical need of ACD for self-renewal and generating cell diversity, we then examine how cell division symmetry control impacts common features associated with CSCs, including niche competition, cancer dormancy, drug resistance, epithelial-mesenchymal transition (EMT) and its reverse process mesenchymal-epithelial transition (MET), and cancer stem cell plasticity. As CSC may underlie resistance to therapy and cancer metastasis, understanding how cell division mode is selected and executed in these cells will provide possible strategies to target CSC.

    Citation: Sreemita Majumdar, Song-Tao Liu. Cell division symmetry control and cancer stem cells[J]. AIMS Molecular Science, 2020, 7(2): 82-101. doi: 10.3934/molsci.2020006

    Related Papers:

    [1] Laura Serna . Crosstalk among hormones and signaling networks during stomatal development in Arabidopsis hypocotyls. AIMS Molecular Science, 2016, 3(4): 550-559. doi: 10.3934/molsci.2016.4.550
    [2] Kishore Kumar Meenakshi Sundaram, Giridharan Bupesh, Konda Mani Saravanan . Instrumentals behind embryo and cancer: a platform for prospective future in cancer research. AIMS Molecular Science, 2022, 9(1): 25-45. doi: 10.3934/molsci.2022002
    [3] Jin-Yih Low, Helen D. Nicholson . Investigating the role of caveolin-2 in prostate cancer cell line. AIMS Molecular Science, 2017, 4(1): 82-88. doi: 10.3934/molsci.2017.1.82
    [4] Michela Illiano, Luigi Sapio, Ilaria Caiafa, Emilio Chiosi, Annamaria Spina, Silvio Naviglio . Forskolin sensitizes pancreatic cancer cells to gemcitabine via Stat3 and Erk1/2 inhibition. AIMS Molecular Science, 2017, 4(2): 224-240. doi: 10.3934/molsci.2017.2.224
    [5] Margarethe Draga, Martin Scaal, Felicitas Pröls . Signaling filopodia in avian embryogenesis: formation and function. AIMS Molecular Science, 2016, 3(4): 683-691. doi: 10.3934/molsci.2016.4.683
    [6] Karen E. Livermore, Jennifer Munkley, David J. Elliott . Androgen receptor and prostate cancer. AIMS Molecular Science, 2016, 3(2): 280-299. doi: 10.3934/molsci.2016.2.280
    [7] Andrew V. Nguyen, Yuan-Yuan Wu, Elaine Y. Lin . The regulatory function of sphingosine-1-phosphate signaling axis on regulatory T cells in colorectal cancer. AIMS Molecular Science, 2015, 1(1): 34-47. doi: 10.3934/molsci.2015.1.34
    [8] Kannan Mayuri, Sundaram Vickram, Thirunavukarasou Anand, Konda Mani Saravanan . MicroRNA-mediated regulation of BCL-2 in breast cancer. AIMS Molecular Science, 2025, 12(1): 32-48. doi: 10.3934/molsci.2025003
    [9] Chunzheng Li, Chenyu Wei, Gongke Zhao, Xianguang Yang . Cancer cells remodeling and quality control are inextricably linked to autophagy. AIMS Molecular Science, 2023, 10(2): 109-126. doi: 10.3934/molsci.2023009
    [10] Jin-Yih Low, Helen D. Nicholson . The up-stream regulation of polymerase-1 and transcript release factor(PTRF/Cavin-1) in prostate cancer: an epigenetic analysis. AIMS Molecular Science, 2016, 3(3): 466-478. doi: 10.3934/molsci.2016.3.466
  • Stem cells including cancer stem cells (CSC) divide symmetrically or asymmetrically. Usually symmetric cell division makes two daughter cells of the same fate, either as stem cells or more differentiated progenies; while asymmetric cell division (ACD) produces daughter cells of different fates. In this review, we first provide an overview of ACD, and then discuss more molecular details of ACD using the well-characterized Drosophila neuroblast system as an example. Aiming to explore the connections between cell heterogeneity in cancers and the critical need of ACD for self-renewal and generating cell diversity, we then examine how cell division symmetry control impacts common features associated with CSCs, including niche competition, cancer dormancy, drug resistance, epithelial-mesenchymal transition (EMT) and its reverse process mesenchymal-epithelial transition (MET), and cancer stem cell plasticity. As CSC may underlie resistance to therapy and cancer metastasis, understanding how cell division mode is selected and executed in these cells will provide possible strategies to target CSC.


    Stem cells are capable of long-term self-renewal while also producing differentiated progeny. The mode of cell division plays a critical role in the activities of stem cells [1]. One distinguishing hallmark of stem cells is to undergo asymmetric cell division (ACD), during which stem cells give rise to daughter cells of different fates, proliferative potential, size, or other characteristics. ACD of stem cells increases the diversity of cell types during development. However, stem cells can also engage in symmetric cell division (SCD) to expand the pool of either stem cells or more differentiated progenies (Figure 1A). In the past decade, molecular understandings about stem cells have evolved significantly, and the concept of “stem cell plasticity” has been developed. In the updated view, stemness is appreciated not to be a fixed privilege of certain cells but can be gained and lost depending on signaling from the microenvironment and intrinsic lineage history [2][5].

    The original theory of cancer stem cells (CSC) suggests that there is a hierarchy in cancer cells, in which CSC lies at the top level [6]. A small number of CSCs could reconstitute a tumor in animal models because CSCs retain the capability of self-renewal and differentiation [6],[7] (Figure 1B). The CSC model, which emphasizes epigenetic changes, is not mutually exclusive from the classical “clonal evolution” model which underscores genetic mutations during cancer development [8]. Together they provide good explanation of genetic, epigenetic, and functional heterogeneity in cancer tissues [9],[10]. Concurring with a better understanding of normal stem cells in recent years, evidence has been accumulated to suggest CSC plasticity [5],[11],[12]. At different stages of cancer development or under different therapeutic treatments, presumed CSC and more differentiated progenies can be inter-convertible depending on the overall signal input from the microenvironment. In the extreme scenario, a continuum of states from stem to differentiated cells exists, with each state more transitory or conditional, increasing the adaptability for cancer cells and the difficulty for cancer treatment [9],[13][16] (Figure 1C).

    Figure 1.  Diagrams on cell division symmetry and cancer stem cells (CSC).

    Even in light of CSC plasticity, ACD is still an efficient mechanism to simultaneously preserve self and produce a daughter at a different state. ACD was therefore traditionally construed to be incompatible with rapid cell proliferation—a hallmark of cancer tissues. Rapid proliferation is usually thought to be carried out through SCD. However, as cell heterogeneity and cell proliferation are both required for cancer development, ACD and SCD must co-exist for cancer cell survival under stress conditions such as during therapeutic treatment and metastasis. Better elucidation of how cell division symmetry is controlled is therefore critical for designing more efficient cancer treatment strategies.

    In this review, we first discuss general features of ACD and then provide more molecular details about the ACD in the well-characterized Drosophila neuroblast system. Although mutations in many ACD regulators induced tumor-like growth in the fly, more complicated relationships exist between major ACD regulators and cancer development in vertebrates. Motivated by the critical need of ACD in self-renewal and generating diversity, we then focus on discussing how cell division symmetry control can impact common features associated with CSCs, including niche competition, cancer dormancy, drug resistance, epithelial-mesenchymal transition (EMT) and its reverse process mesenchymal-epithelial transition (MET), and cancer stem cell plasticity. We conclude the review with a brief summary and some ideas for future studies.

    The canonical ACD of stem cells indicates that only one of the two daughter cells maintains the stemness while the other becomes more differentiated [17],[18]. Fate differentiation can be achieved “extrinsically” or “intrinsically”. The two daughter cells can appear identical initially after birth, but are placed at different distances from the “niche”. The cell-cell junctions with and spatially restricted signals from the niche help maintain the proximal daughter cell staying in the undifferentiated state (Figure 2A, “Extrinsic asymmetry”). The good examples for this mode are male and female germline stem cells in Drosophila [5],[19]. Alternatively, intracellular fate determinants are asymmetrically distributed in a dividing stem cell and the two daughter cells inherit different fate determinants that affect the direction of their development (Figure 2B, “Intrinsic asymmetry”). The “cues” for polarizing fate determinants intrinsically can be the polarity of neighboring cells (e.g. sensory organ precursor cells in Drosophila), basement membrane (e.g. basal cells in skin epidermis), or the sperm entry site in the case of one-cell embryo of C. elegans, although the nature or even presence of the “cues” might sometimes be hard to track, and the asymmetry seems built in the lineage history of stem cells [18][21]. Spindle orientation in the dividing stem cell is critical for both “extrinsically” and “intrinsically” controlled ACD. Misplacement of the spindle relative to the niche or the fate determinant polarity leads to an increase of the stem cell population [22],[23] (Figure 2C&D). In summary, proper ACD requires niche-stem cell interaction or intrinsic polarity establishment, mitotic spindle alignment with the polarity cues and subsequent daughters gaining different fates.

    Figure 2.  Extrinsic and intrinsic mechanisms of asymmetric cell division (ACD).

    There have been excellent reviews on the mechanisms of ACD in detail [19],[24][26]. Major known regulators of ACD are summarized in Supplemental Table 1. In Drosophila a neuroblast (i.e. neural stem cell) divides asymmetrically to form a neuroblast and a ganglion mother cell (GMC) which divides further to give neurons or glia (Figure 3). We will use this well-characterized model to illustrate the general principles and introduce several specific proteins involved in ACD.

    The polarity establishment in this system involves signaling between neuroblasts and the neuroectoderm from which neuroblasts delaminate. The Par3 (Bazooka)/Par6/aPKC protein kinase complex is localized at the apical cortex of the dividing neuroblast, with fate determinants such as Numb, Prospero (Pros), Staufen, and Brain tumor (Brat), and adaptor proteins such as Miranda accumulate near the basal membrane (Figure 3). Drosophila Numb is the first recognized cell fate determinant that partitions differentially between two daughter cells to drive their distinct developmental identities [27]. Numb is an endocytosis adapter protein that inhibits Notch signaling pathway and promotes differentiation [28]. The basal surface localization of Numb depends on Aurora A kinase [29],[30].

    In Drosophila Aurora A also phosphorylates Par6, which activates aPKC and recruits Par3 to form the Par3/Par6/aPKC complex. The Par3/Par6/aPKC complex, when enriched at the apical cortex through interaction with membrane bound CDC42, works with another apical cortex-localized complex, the Gαi/Partner of Inscuteable (Pins)/Mud complex, to align spindle with the apical-basal axis. Inscuteable bridges the two complexes through direct binding with both Pins and Par3 [26],[31] (Figure 3). The interactions between these proteins could be more complicated and dynamic as demonstrated in recent results [32]. Nonetheless, cell cortex localized Mud recruits dynein to capture and move astral microtubules so as to orient and pull the mitotic spindle (Supplemental Table 1; Figure 3). Kinesin Khc73 also contributes to spindle positioning through interaction with Dlg protein, which is recruited also by Pins [33]. The mitotic spindle is symmetric in metaphase but in anaphase, the apical half spindle becomes more extended with longer astral microtubules. This places the cleavage furrow closer to the basal cortex, so a larger neuroblast and a smaller GMC are produced. The outcome of the cell division is to distribute differentiation-promoting fate determinants asymmetrically into the daughter destined to become GMC [25]. A spindle independent but myosin based membrane contraction mechanism also plays a role in the neuroblast ACD [34].

    Figure 3.  Asymmetric division of Drosophila neuroblasts.

    Gateff first showed that 12 recessive-lethal larval mutants of Drosophila exhibited tumor-like growth in neuroblasts and other tissues [35]. The malignant cells were undifferentiated and invasive, causing lethal growth when transplanted to wild type hosts. Caussinus and Gozalez directly tested the contribution of ACD regulators to cancer development in Drosophila using similar tissue transplantation techniques [36]. They found that within 2 weeks larval brain tissue transplants carrying neuroblasts with mutations in Pins, Miranda, Numb, and Pros grew to over 100 times their initial size and invaded neighboring tissues. It should be noted that although pins mutant neuroblasts exhibited symmetric division in the fly tumors, cells with mutations in Miranda, Numb, and Pros still maintained certain features of ACD, despite uncontrolled proliferation [36]. Tumor-promoting activities were also shown in Drosophila neuroblasts after mutating other ACD regulators including Brat and Aurora A [37],[38]. These results supported tumor-suppressing roles of ACD regulators and fate determinants and indicated disruption of ACD regulators in fly stem cells lead to tumorigenesis.

    Most ACD regulators and fate determinants are conserved through evolution (Supplemental Table 1). However, the seemingly straightforward relationship between mutations in ACD regulators and tumorigenesis in Drosophila could not be simply applied to vertebrate systems [39]. The simplistic view that human cancers arise from the loss of ACD in mutated adult stem cells turned out not true. Nevertheless, there were many studies on whether and how ACD polarizing factors, spindle alignment regulators, and cell fate determinants contribute to tumorigenesis, metastasis, or drug resistance. We discuss a few examples to illustrate the complicated relationship between alterations in ACD regulators and cancer development in vertebrates.

    Par3 expression is frequently lost in human breast cancers and squamous cell carcinoma [40],[41]. Mammary glands in Par3 depleted mice expanded progenitor population that expresses both keratin 8 and keratin 14, the markers for luminal and basal epithelial cells [42], indicating a possible SCD-based increase of bipotent precursors [24]. The depletion of Par3 from primary mammary epithelial cells (MECs) in mice of certain oncogenic backgrounds also led to invasive or metastatic breast cancers [40]. Although the above results supported a role of Par3 loss in breast cancers, the Par3/Par6/aPKC complex was overexpressed in other cancers [30],[43]. Therefore, ACD regulators including the Par3/Par6/aPKC complex could have tissue-specific effects in vertebrates.

    As mentioned above, Aurora A mutation promotes tumor growth in Drosophila neuroblasts [37]. However, in mouse embryonic stem cells, Aurora A loss negatively impacts self-renewal and triggers differentiation [44]. In addition, Aurora A (encoded by STK11 gene) overexpression is well documented in human cancers [45]. Aurora A has been further indicated as a positive regulator of CSC and EMT in glioma, breast cancer, and colorectal cancer cells [46]. The role of Aurora A in vertebrate cancers was often ascribed to its well-characterized role in centrosome maturation [47]. Aurora A overexpression leads to centrosome amplification and genomic instability [47]. As a multi-functional protein, the respective weight of Aurora A in ACD regulation or other biological processes could be varied in different species.

    The cell cortex localized Gα-LGN-NUMA complex in vertebrates is equivalent to the Gαi-Pins-Mud complex in the Drosophila neuroblasts, and plays a conserved role in positioning spindles [48]. The dynamics of spindle orientation determine symmetric or asymmetric division in many organisms across a range of cell types. Defective expression of the Gα-LGN-NUMA complex or spindle orientation, in general, has also been correlated to tumorigenesis in mammals, but whether the defects have causative roles still needs further assessment [39].

    The fate-determining proteins/RNAs usually promote cell differentiation, therefore molecules such as Numb, Prospero, Brat and Staufen are segregated to the basal GMC after ACD of Drosophila neuroblasts (Figure 3). As mentioned above, brain tissues containing mutations in fate-determining genes develop tumor-like growth in Drosophila [36]. Similarly, the homologs of these fate determinants have been reported as potential tumor suppressors in human cancers [43],[49]. Numb is a well-characterized tumor suppressor in mammals [49]. Inactivation of Numb and its close homolog Numb-L in the mouse dorsal forebrain resulted in neural progenitor hyper-proliferation [50]. Downregulation of Numb is seen in breast cancer, salivary gland carcinoma, non-small-cell lung carcinoma, and medulloblastoma [51]. In addition to its role antagonizing Notch, recent work showed that in mouse mammary stem cells, Numb controls asymmetric division by positively modulating p53 activity [52]. Inactive Numb leads to inactive p53 and symmetric stem cell division, causing EMT, hyperplasiaand tumorigeneses in mouse mammary epithelium [52].

    The development of malignant cancers, despite its nature as a caricature of normal tissue development, is a multi-step process that entails cell diversification and cell proliferation. As discussed above, the roles of many ACD regulators in cancer development, including the Par3/Par6/aPKC complex and the Gα-LGN-NUMA complex, may be context-dependent. However, the intrinsic connections of ACD with CSC, and CSC with self-renewal and cancer cell heterogeneity, seem too important to be overlooked in our endeavors to understand cancer progression and design novel cancer therapeutics. Therefore we will attempt to examine what is known about the cell division control mechanisms in CSC, and explore how ACD/SCD could impact different features commonly associated with CSC.

    Characterizing the properties associated with CSCs was intricately linked with the assays developed to identify and isolate CSCs. Despite the long-known genetic and epigenetic heterogeneity in cancers and similarity of cancer cells to adult stem cells [6], CSCs were first experimentally identified only in the 1990s in leukemia through limiting dilutions and engraftment of cancer cells with stem-like surface markers to immune-compromised mice [53],[54]. CSCs from glioma, breast cancers, and other solid tumors were subsequently identified [7],[55],[56]. Tumor reconstitution using a small number of CSCs in animal models has now become a gold-standard functional assay. In vitro tumorsphere formation was a surrogate assay to examine CSC, its self-renewal and differentiation [57],[58]. Knowledge of adult stem cells and progenitor cells catalyzed the adoption of cell surface markers to characterize CSCs, for example, CD44+CD24lowLin for breast CSC [55], and CD34+CD38 for leukemia stem cells [54]. An analogy to usual quiescence of normal stem cells led to several label-retention techniques, especially the use of an irreversible fluorescent lipid dye PKH26, to identify CSC. Reduced proliferation resulted that CSC retains the dye while it's more proliferative progenies go through several rounds of mitosis and dilute the dye [59],[60]. The tendency of quiescence may also render CSC more resistant to conventional radiotherapy or chemotherapy which primarily targets DNA replication and mitosis in dividing cells [6],[61]. When CSC does divide, ACD is expected, so the daughter cells exhibit size differences or differential inheritance of fate determinants such as Numb [60],[62]. CSCs may also exhibit metabolic rewiring compared to normal cells and the bulk of cancer cells, such as higher expression of ALDH1 and drug transporters [63],[64]. In addition, EMT is usually associated with stem cell-like states [65]. Table 1 summarizes these CSC features. It should be noted that not all features can be observed in all CSCs, even CSCs from the same tissue origin.

    Table 1.  Features of Cancer Stem Cells.
    Assays of cancer stem cells Underlying CSC property
    Asymmetric cell division Self-renewal and differentiation
    Cancer reconstitution in mice Self-renewal and differentiation
    Tumorsphere formation in vitro Self-renewal and differentiation
    Label retention (e.g. nucleotide analog or lipid dye PKH26) Cell quiescence
    Radiotherapy/Drug resistance Cell quiescence
    EMT/MET Stem cell plasticity
    Cell surface markers Self-renewal and differentiation; Genetic and epigenetic features; Rarity?
    Metabolic markers (e.g. ALDH1, drug transporters, etc) Self-renewal and differentiation; Genetic and epigenetic features; Rarity?

     | Show Table
    DownLoad: CSV

    ACD can occur through extrinsic or intrinsic mechanisms (Figure 2). Isolated human CSC from various cancers, when cultured in vitro, often exhibited asymmetric distribution of fate determinants such as Numb or microRNA miR-34a by immunofluorescence [60],[66],[67]. In contrast, the bulk of cancer cells primarily divided symmetrically. As stem cell niche was not easily identified in these experiments, the CSCs seemed to retain the capability to carry out ACD based on intrinsic asymmetry. The details about such ACD remain to be characterized. However, these results further confirmed that human cancers were not simply caused by the amplification of erratic adult stem cells through SCD. There was a suggestion that ACD is linked with CSC in early stage and well differentiated colon cancers, but ACD suppression is associated with late stage, highly proliferating cancers [62]. The caveat of such an explanation is that markers to indicate asymmetry in early stage cancers could have got lost in late stage cancers. Besides, it is hard to imagine ACD does not happen in late stage cancers which usually exhibit even higher cell heterogeneity.

    The gold-standard functional assay of CSC is still serial transplantation in immune compromised mice [10],[68]. Presumable CSC should not only reconstitute cancers by supporting cell proliferation and generation of different lineages but also show self-renewal. Self-renewal is a distinctive feature of stem cells as compared to progenitor cells, and killing cells that sustain long-term self-renewal should be the ultimate goal of any cancer therapy. In tumor reconstitution experiments, ACD (or self-renewal and cell diversification) is usually postulated based on fluorescence activated cell sorting. For example, purified CD34+CD38 leukemia stem cells regenerated cells with the same markers as well as distinct subpopulations carrying CD34+CD38+ markers [54].

    It has been commonly assumed that CSCs are rare in cancer tissues. The idea might be rooted in results from studying normal hematopoietic stem cells [69], and was consistent with earlier nude mice engraftment experiments to identify CSCs [7],[53][56]. However, using modified xenograft protocols on more immunocompromised mouse models, ∼27% of single cell transplants of unsorted patient melanoma cells successfully formed tumors [70]. The improvement of xenograft protocols and mouse models aside, this result exemplified the variation of CSC frequency in different cancers. Recent results indicated that even normal stem cells are not that rare in solid tissues, especially when stem cell plasticity is taken into consideration [71].

    In vitro tumorsphere formation tests the capability of presumable CSC to grow into a sphere in low-attachment growth conditions [57],[58]. The serial passage of tumorspheres also confirmed the self-renewal capability of CSC. Recent improvement significantly reduced the problem caused by cell agammaegation [72]. As studying ACD in vivo is still technically challenging, tumorspheres are a more accessible choice to assess ACD of CSCs in vitro in the presence of cell-cell interactions.

    In extrinsically controlled ACD of any stem cell, usually the neighboring cells, extracellular matrix, and spatially restricted signaling molecules form the “niche” to maintain the stemness of at least one daughter cell (Figure 2). Tumor microenvironment sometimes was suggested to provide “niches” for CSCs, and some factors such as inflammatory cytokines or hypoxic conditions are indeed inductive for CSC survival and proliferation [15],[73]. However, the general description of the tumor microenvironment seems to lack polarity cues that guide ACD of CSCs. There could be several possible solutions to this paradox. First, some remaining tissue structure or resident cell types in the tumor microenvironment could provide polarized cues for ACD. In recent years, the sinusoidal and arteriolar endothelia were found to serve as the niches for hematopoietic stem cells in bone marrow [74],[75]. Endothelial cells in cancers could have similar roles. Second, “niche competition” could apply if the niche is limited and the interactions between CSC and the “niche” are not stably maintained. Only the progeny landing near the niche becomes CSC and CSC can be pushed out by more differentiated cells and loses its stemness. The “niche competition” or neutral drift concept has been described for normal stem cell homeostasis in tissues such as intestine crypt stem cells [76],[77]. Third, it is also possible that CSCs could regenerate their own niche by producing diverse lineage of cells that support stemness. Progenies of hematopoietic stem cells are known to provide feedback and regulate the population of stem cells [2]. Although in a solid tumor the origins of different cell lineages are not always clear, glioblastoma CSC could generate tumor endothelium [78]. As mentioned above, the endothelial cells, in turn, may function as a “niche” to guide CSC cell division. The CSC pool could also be composed of a group of interdependent cells at related but distinguishable states, for example, the epithelial-like and the mesenchymal-like states of breast cancer CSCs [15]. At least some of the polarity cues resulted in intrinsic asymmetry that can be observed in the ACD of CSCs in vitro [60],[66],[67].

    As described in Figure 1, CSC does not always undergo ACD. Tominaga et al showed that stimulation with a cytokine, semaphorin, activates monooxygenase MICAL3, a cytoplasmic signal transducer, through the neuropilin receptor that is specifically expressed on the breast CSC plasma membrane. The activation of MICAL3 induces symmetric division of breast CSCs [79].

    Adult stem cells are usually thought to stay in quiescence for long-term survival and their activation is only triggered when tissue homeostasis and repair is required. If CSC behaves similarly, quiescent CSCs would be intrinsically resistant to common cancer drugs or radiation therapy, as these treatments usually only target actively dividing cells [61]. As mentioned above, the property of quiescence has been used to isolate CSCs after ACD: daughter cells retaining labeled nucleotides or fluorescent lipid markers were regarded as quiescent and hence enriched CSCs [60],[80][82]. Some reports have provided experimental support that cancer dormancy might be explained by the label-retaining cancer cell population [61],[83]. In addition, Dey-Guha et al found that in long-established human breast and colon cancer cell lines there existed a small fraction of G0-like cells marked by AKTlo Roslo Hes1hi [84] [ROS: reactive oxygen species]. These cells arose from ACD but did not express widely used CSC surface markers (e.g., CD44high/CD24low for breast CSC). Inhibition of AKT increased the occurrence of this fraction which are resistant to drug treatment. Whether these cells show other features of CSC is unknown, but they may be a result of Notch/Numb imbalance as seen in many other cancers [85]. Cancer dormancy also reminds of immunological memory. Interestingly, T memory stem cells have recently been characterized [86]. Reactivation of T memory cells in face of secondary pathogens challenge is accompanied by ACD to retain memory cells and produce effector cells [87]. Cancer relapse from dormancy is likely also accompanied by ACD of CSC.

    In 2008, Mani et al first showed EMT is associated with stem cell-like states [65]. This stimulated further studies on the relationship between EMT and CSC. The current view holds that EMT is not exclusively associated with stemness [15]. Instead, transient activation of the EMT program and an E/M hybrid state seems critical for acquiring CSC states [88][90]. Various states of cancer cells exist spanning the epithelial-mesenchymal spectrum, and fully differentiated states, whether epithelial or mesenchymal, are endowed with reduced tumorigenicity [9].

    EMT and its reverse process MET are crucial events for cancer metastasis. Together with additional intermediate states revealed recently, they represent the dynamic plasticity of CSC [11],[91] (Figure 1C). Interestingly, during wound healing and tissue repair, adult stem cells exhibited “lineage infidelity” and backup stem cells, progenitor cells, or even more differentiated progenies could be mobilized and reverted to “stemness” [2]. Considered as “a wound never healed”, cancers could hijack the transient plasticity mechanism to sustain its malignancy [2]. Conceptually ACD has to be involved in producing the diverse “states” along the epithelial-mesenchymal spectrum, but it remains unclear whether any ACD regulator [section 2.2] plays a role in promoting EMT or MET.

    The concept of CSC provides a useful framework to explain the functional heterogeneity in cancer cells, especially those with apparently homogenous genetic background [10],[92]. ACD is an efficient route to generate heterogeneity while preserving CSC self-renewal. It was thus exciting when ACD disruption in Drosophila neuroblasts was found to result in malignant tumor-like growth. However, the relationship between evolutionarily conserved ACD regulators and general cancer occurrences seems more complicated in vertebrates. At present we still lack a good assessment of the role ACD plays in CSC activities during different stages of human cancer development.

    The recent appreciation of stem cell plasticity poses more challenges and more opportunities to study how cell division symmetry control (ACD/SCD) affects CSC subpopulations and functions. On the one hand, the moving target nature of CSC seems exacerbated with the revelation of multiple “transient” states [10],[68]. On the other hand, more markers available that distinguish different “states” (such as epithelial and mesenchymal markers) can be used to monitor how cell division symmetry control mechanisms respond to ever-changing external signaling to sculpt the cancer cell populations during tumorigenesis, metastasis and drug responses [12],[93].

    Technical innovations will continue to drive ACD research especially in the context of CSC. ACD has been traditionally evaluated based on asymmetric distribution of one or a few intracellular or cell surface markers (such as Numb or CD44/CD24) by microscopy or FACS. In some cases, the size differences of two daughter cells are observed as in Drosophila neuroblasts (Figure 3). Single cell genome-seq and RNA-seq or multiplex RT-PCR analysis, various 3D or organoid cultures, advanced lineage tracking, and long time live cell imaging, will provide deep insights into the fates and determinants of symmetric and asymmetric cell divisions of CSC in vitro and in vivo. Mapping genetic and epigenetic landscapes of both daughter cells after ACD will generate unprecedented insights into both ACD and CSC [92],[94].

    Emphasizing epigenetic and functional heterogeneity in response to signals from the tumor microenvironment, the CSC theory has clinical significance. It provides a useful working model to explain drug resistance, cancer relapse, and metastasis. Even in light of recent development in CSC plasticity, the general importance of cell division symmetry control in CSC is still valid: ACD favors balanced self-renewal and differentiation, and SCD amplifies one or more types of daughters. Future efforts are needed to understand how the choice of SCD and ACD is selected under specific circumstances.


    Abbreviation ACD: asymmetric cell division; CSC: cancer stem cells; EMT: epithelial-mesenchymal transition; GMC: Ganglion mother cell; MET: mesenchymal-epithelial transition; SCD: symmetric cell division;
    Acknowledgments



    The work in the Liu lab is funded by National Institutes of Health (R15CA238894) and UToledo Biomedical Research Innovation Award.

    Conflict of interest



    The authors declare there is no conflict of interest.

    [1] Morrison SJ, Kimble J (2006) Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 441: 1068-1074. doi: 10.1038/nature04956
    [2] Ge Y, Fuchs E (2018) Stretching the limits: from homeostasis to stem cell plasticity in wound healing and cancer. Nat Rev Genet 19: 311-325. doi: 10.1038/nrg.2018.9
    [3] Yousefi M, Li L, Lengner CJ (2017) Hierarchy and Plasticity in the Intestinal Stem Cell Compartment. Trends Cell Biol 27: 753-764. doi: 10.1016/j.tcb.2017.06.006
    [4] Tetteh PW, Farin HF, Clevers H (2015) Plasticity within stem cell hierarchies in mammalian epithelia. Trends Cell Biol 25: 100-108. doi: 10.1016/j.tcb.2014.09.003
    [5] Losick VP, Morris LX, Fox DT, et al. (2011) Drosophila stem cell niches: a decade of discovery suggests a unified view of stem cell regulation. Dev Cell 21: 159-171. doi: 10.1016/j.devcel.2011.06.018
    [6] Reya T, Morrison SJ, Clarke MF, et al. (2001) Stem cells, cancer, and cancer stem cells. Nature 414: 105-111. doi: 10.1038/35102167
    [7] Bajaj J, Diaz E, Reya T (2020) Stem cells in cancer initiation and progression. J Cell Biol 219: e201911053. doi: 10.1083/jcb.201911053
    [8] Greaves M, Maley CC (2012) Clonal evolution in cancer. Nature 481: 306-313. doi: 10.1038/nature10762
    [9] Brooks MD, Burness ML, Wicha MS (2015) Therapeutic Implications of Cellular Heterogeneity and Plasticity in Breast Cancer. Cell Stem Cell 17: 260-271. doi: 10.1016/j.stem.2015.08.014
    [10] Kreso A, Dick JE (2014) Evolution of the cancer stem cell model. Cell Stem Cell 14: 275-291. doi: 10.1016/j.stem.2014.02.006
    [11] Ye X, Weinberg RA (2015) Epithelial-Mesenchymal Plasticity: A Central Regulator of Cancer Progression. Trends Cell Biol 25: 675-686. doi: 10.1016/j.tcb.2015.07.012
    [12] Najafi M, Mortezaee K, Ahadi R (2019) Cancer stem cell (a)symmetry & plasticity: Tumorigenesis and therapy relevance. Life Sci 231: 116520. doi: 10.1016/j.lfs.2019.05.076
    [13] Lee G, Hall RR, Ahmed AU (2016) Cancer Stem Cells: Cellular Plasticity, Niche, and its Clinical Relevance. J Stem Cell Res Ther 6: 363.
    [14] Skrypek N, Goossens S, De Smedt E, et al. (2017) Epithelial-to-Mesenchymal Transition: Epigenetic Reprogramming Driving Cellular Plasticity. Trends Genet 33: 943-959. doi: 10.1016/j.tig.2017.08.004
    [15] Hung KF, Yang T, Kao SY (2019) Cancer stem cell theory: Are we moving past the mist? J Chin Med Assoc 82: 814-818. doi: 10.1097/JCMA.0000000000000186
    [16] Batlle E, Clevers H (2017) Cancer stem cells revisited. Nat Med 23: 1124-1134. doi: 10.1038/nm.4409
    [17] Horvitz HR, Herskowitz I (1992) Mechanisms of asymmetric cell division: two Bs or not two Bs, that is the question. Cell 68: 237-255. doi: 10.1016/0092-8674(92)90468-R
    [18] Knoblich JA (2001) Asymmetric cell division during animal development. Nat Rev Mol Cell Biol 2: 11-20. doi: 10.1038/35048085
    [19] Venkei ZG, Yamashita YM (2018) Emerging mechanisms of asymmetric stem cell division. J Cell Biol 217: 3785-3795. doi: 10.1083/jcb.201807037
    [20] Neumuller RA, Knoblich JA (2009) Dividing cellular asymmetry: asymmetric cell division and its implications for stem cells and cancer. Genes Dev 23: 2675-2699. doi: 10.1101/gad.1850809
    [21] Lechler T, Fuchs E (2005) Asymmetric cell divisions promote stratification and differentiation of mammalian skin. Nature 437: 275-280. doi: 10.1038/nature03922
    [22] Cabernard C, Doe CQ (2009) Apical/Basal Spindle Orientation Is Required for Neuroblast Homeostasis and Neuronal Differentiation in Drosophila. Developmental Cell 17: 134-141. doi: 10.1016/j.devcel.2009.06.009
    [23] Yamashita YM, Jones DL, Fuller MT (2003) Orientation of Asymmetric Stem Cell Division by the APC Tumor Suppressor and Centrosome. Science 301: 1547. doi: 10.1126/science.1087795
    [24] Santoro A, Vlachou T, Carminati M, et al. (2016) Molecular mechanisms of asymmetric divisions in mammary stem cells. EMBO Rep 17: 1700-1720. doi: 10.15252/embr.201643021
    [25] Knoblich JA (2008) Mechanisms of asymmetric stem cell division. Cell 132: 583-597. doi: 10.1016/j.cell.2008.02.007
    [26] Morin X, Bellaiche Y (2011) Mitotic spindle orientation in asymmetric and symmetric cell divisions during animal development. Dev Cell 21: 102-119. doi: 10.1016/j.devcel.2011.06.012
    [27] Rhyu MS, Jan LY, Jan YN (1994) Asymmetric distribution of numb protein during division of the sensory organ precursor cell confers distinct fates to daughter cells. Cell 76: 477-491. doi: 10.1016/0092-8674(94)90112-0
    [28] Guo M, Jan LY, Jan YN (1996) Control of daughter cell fates during asymmetric division: interaction of Numb and Notch. Neuron 17: 27-41. doi: 10.1016/S0896-6273(00)80278-0
    [29] Wirtz-Peitz F, Nishimura T, Knoblich JA (2008) Linking cell cycle to asymmetric division: Aurora-A phosphorylates the Par complex to regulate Numb localization. Cell 135: 161-173. doi: 10.1016/j.cell.2008.07.049
    [30] Campanale JP, Sun TY, Montell DJ (2017) Development and dynamics of cell polarity at a glance. J Cell Sci 130: 1201-1207. doi: 10.1242/jcs.188599
    [31] Doe CQ, Bowerman B (2001) Asymmetric cell division: fly neuroblast meets worm zygote. Curr Opin Cell Biol 13: 68-75. doi: 10.1016/S0955-0674(00)00176-9
    [32] Loyer N, Januschke J (2020) Where does asymmetry come from? Illustrating principles of polarity and asymmetry establishment in Drosophila neuroblasts. Curr Opin Cell Biol 62: 70-77. doi: 10.1016/j.ceb.2019.07.018
    [33] Johnston CA, Hirono K, Prehoda KE, et al. (2009) Identification of an Aurora-A/PinsLINKER/Dlg spindle orientation pathway using induced cell polarity in S2 cells. Cell 138: 1150-1163. doi: 10.1016/j.cell.2009.07.041
    [34] Cabernard C, Prehoda KE, Doe CQ (2010) A spindle-independent cleavage furrow positioning pathway. Nature 467: 91-94. doi: 10.1038/nature09334
    [35] Gateff E (1978) Malignant neoplasms of genetic origin in Drosophila melanogaster. Science 200: 1448-1459. doi: 10.1126/science.96525
    [36] Caussinus E, Gonzalez C (2005) Induction of tumor growth by altered stem-cell asymmetric division in Drosophila melanogaster. Nat Genet 37: 1125-1129. doi: 10.1038/ng1632
    [37] Wang H, Somers GW, Bashirullah A, et al. (2006) Aurora-A acts as a tumor suppressor and regulates self-renewal of Drosophila neuroblasts. Genes Dev 20: 3453-3463. doi: 10.1101/gad.1487506
    [38] Betschinger J, Mechtler K, Knoblich JA (2006) Asymmetric segregation of the tumor suppressor brat regulates self-renewal in Drosophila neural stem cells. Cell 124: 1241-1253. doi: 10.1016/j.cell.2006.01.038
    [39] Seldin L, Macara I (2017) Epithelial spindle orientation diversities and uncertainties: recent developments and lingering questions. F1000Res 6: 984. doi: 10.12688/f1000research.11370.1
    [40] McCaffrey LM, Montalbano J, Mihai C, et al. (2012) Loss of the Par3 polarity protein promotes breast tumorigenesis and metastasis. Cancer Cell 22: 601-614. doi: 10.1016/j.ccr.2012.10.003
    [41] Zen K, Yasui K, Gen Y, et al. (2009) Defective expression of polarity protein PAR-3 gene (PARD3) in esophageal squamous cell carcinoma. Oncogene 28: 2910-2918. doi: 10.1038/onc.2009.148
    [42] McCaffrey LM, Macara IG (2009) The Par3/aPKC interaction is essential for end bud remodeling and progenitor differentiation during mammary gland morphogenesis. Genes Dev 23: 1450-1460. doi: 10.1101/gad.1795909
    [43] Huang L, Muthuswamy SK (2010) Polarity protein alterations in carcinoma: a focus on emerging roles for polarity regulators. Curr Opin Genet Dev 20: 41-50. doi: 10.1016/j.gde.2009.12.001
    [44] Lee DF, Su J, Ang YS, et al. (2012) Regulation of embryonic and induced pluripotency by aurora kinase-p53 signaling. Cell Stem Cell 11: 179-194. doi: 10.1016/j.stem.2012.05.020
    [45] Zhou H, Kuang J, Zhong L, et al. (1998) Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation. Nat Genet 20: 189-193. doi: 10.1038/2496
    [46] Willems E, Dedobbeleer M, Digregorio M, et al. (2018) The functional diversity of Aurora kinases: a comprehensive review. Cell Div 13: 7. doi: 10.1186/s13008-018-0040-6
    [47] Sasai K, Parant JM, Brandt ME, et al. (2008) Targeted disruption of Aurora A causes abnormal mitotic spindle assembly, chromosome misalignment and embryonic lethality. Oncogene 27: 4122-4127. doi: 10.1038/onc.2008.47
    [48] di Pietro F, Echard A, Morin X (2016) Regulation of mitotic spindle orientation: an integrated view. EMBO Rep 17: 1106-1130. doi: 10.15252/embr.201642292
    [49] Mukherjee S, Kong J, Brat DJ (2015) Cancer stem cell division: when the rules of asymmetry are broken. Stem Cells Dev 24: 405-416. doi: 10.1089/scd.2014.0442
    [50] Li HS, Wang D, Shen Q, et al. (2003) Inactivation of Numb and Numblike in embryonic dorsal forebrain impairs neurogenesis and disrupts cortical morphogenesis. Neuron 40: 1105-1118. doi: 10.1016/S0896-6273(03)00755-4
    [51] Garcia-Heredia JM, Verdugo Sivianes EM, Lucena-Cacace A, et al. (2016) Numb-like (NumbL) downregulation increases tumorigenicity, cancer stem cell-like properties and resistance to chemotherapy. Oncotarget 7: 63611-63628. doi: 10.18632/oncotarget.11553
    [52] Tosoni D, Zecchini S, Coazzoli M, et al. (2015) The Numb/p53 circuitry couples replicative self-renewal and tumor suppression in mammary epithelial cells. J Cell Biol 211: 845-862. doi: 10.1083/jcb.201505037
    [53] Lapidot T, Sirard C, Vormoor J, et al. (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367: 645-648. doi: 10.1038/367645a0
    [54] Bonnet D, Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 3: 730-737. doi: 10.1038/nm0797-730
    [55] Al-Hajj M, Wicha MS, Benito-Hernandez A, et al. (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 100: 3983-3988. doi: 10.1073/pnas.0530291100
    [56] Singh SK, Clarke ID, Terasaki M, et al. (2003) Identification of a cancer stem cell in human brain tumors. Cancer Res 63: 5821-5828.
    [57] Dontu G, Abdallah WM, Foley JM, et al. (2003) In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev 17: 1253-1270. doi: 10.1101/gad.1061803
    [58] Reynolds BA, Weiss S (1996) Clonal and population analyses demonstrate that an EGF-responsive mammalian embryonic CNS precursor is a stem cell. Dev Biol 175: 1-13. doi: 10.1006/dbio.1996.0090
    [59] Lanzkron SM, Collector MI, Sharkis SJ (1999) Hematopoietic stem cell tracking in vivo: a comparison of short-term and long-term repopulating cells. Blood 93: 1916-1921. doi: 10.1182/blood.V93.6.1916.406k15_1916_1921
    [60] Pece S, Tosoni D, Confalonieri S, et al. (2010) Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell 140: 62-73. doi: 10.1016/j.cell.2009.12.007
    [61] Moore N, Lyle S (2011) Quiescent, slow-cycling stem cell populations in cancer: a review of the evidence and discussion of significance. J Oncol Article ID 396076.
    [62] Bu P, Chen KY, Lipkin SM, et al. (2013) Asymmetric division: a marker for cancer stem cells in early stage tumors? Oncotarget 4: 950-951. doi: 10.18632/oncotarget.1029
    [63] Ginestier C, Hur MH, Charafe-Jauffret E, et al. (2007) ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1: 555-567. doi: 10.1016/j.stem.2007.08.014
    [64] Greve B, Kelsch R, Spaniol K, et al. (2012) Flow cytometry in cancer stem cell analysis and separation. Cytometry A 81: 284-293. doi: 10.1002/cyto.a.22022
    [65] Mani SA, Guo W, Liao MJ, et al. (2008) The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133: 704-715. doi: 10.1016/j.cell.2008.03.027
    [66] Bu P, Chen KY, Chen JH, et al. (2013) A microRNA miR-34a-regulated bimodal switch targets Notch in colon cancer stem cells. Cell Stem Cell 12: 602-615. doi: 10.1016/j.stem.2013.03.002
    [67] Chen G, Kong J, Tucker-Burden C, et al. (2014) Human Brat ortholog TRIM3 is a tumor suppressor that regulates asymmetric cell division in glioblastoma. Cancer Res 74: 4536-4548. doi: 10.1158/0008-5472.CAN-13-3703
    [68] Clarke MF, Dick JE, Dirks PB, et al. (2006) Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res 66: 9339-9344. doi: 10.1158/0008-5472.CAN-06-3126
    [69] Till JE, Mc CE (1961) A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res 14: 213-222. doi: 10.2307/3570892
    [70] Quintana E, Shackleton M, Sabel MS, et al. (2008) Efficient tumour formation by single human melanoma cells. Nature 456: 593-598. doi: 10.1038/nature07567
    [71] Post Y, Clevers H (2019) Defining Adult Stem Cell Function at Its Simplest: The Ability to Replace Lost Cells through Mitosis. Cell Stem Cell 25: 174-183. doi: 10.1016/j.stem.2019.07.002
    [72] Bailey PC, Lee RM, Vitolo MI, et al. (2018) Single-Cell Tracking of Breast Cancer Cells Enables Prediction of Sphere Formation from Early Cell Divisions. iScience 8: 29-39. doi: 10.1016/j.isci.2018.08.015
    [73] Korkaya H, Liu S, Wicha MS (2011) Breast cancer stem cells, cytokine networks, and the tumor microenvironment. J Clin Invest 121: 3804-3809. doi: 10.1172/JCI57099
    [74] Kiel MJ, Yilmaz OH, Iwashita T, et al. (2005) SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121: 1109-1121. doi: 10.1016/j.cell.2005.05.026
    [75] Kunisaki Y, Bruns I, Scheiermann C, et al. (2013) Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502: 637-643. doi: 10.1038/nature12612
    [76] Snippert HJ, van der Flier LG, Sato T, et al. (2010) Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143: 134-144. doi: 10.1016/j.cell.2010.09.016
    [77] Lopez-Garcia C, Klein AM, Simons BD, et al. (2010) Intestinal stem cell replacement follows a pattern of neutral drift. Science 330: 822-825. doi: 10.1126/science.1196236
    [78] Wang R, Chadalavada K, Wilshire J, et al. (2010) Glioblastoma stem-like cells give rise to tumour endothelium. Nature 468: 829-833. doi: 10.1038/nature09624
    [79] Tominaga K, Minato H, Murayama T, et al. (2019) Semaphorin signaling via MICAL3 induces symmetric cell division to expand breast cancer stem-like cells. Proc Natl Acad Sci USA 116: 625-630. doi: 10.1073/pnas.1806851116
    [80] Huang S, Law P, Francis K, et al. (1999) Symmetry of initial cell divisions among primitive hematopoietic progenitors is independent of ontogenic age and regulatory molecules. Blood 94: 2595-2604. doi: 10.1182/blood.V94.8.2595.420k37_2595_2604
    [81] Braun KM, Watt FM (2004) Epidermal label-retaining cells: background and recent applications. J Investig Dermatol Symp Proc 9: 196-201. doi: 10.1111/j.1087-0024.2004.09313.x
    [82] Bickenbach JR, Mackenzie IC (1984) Identification and localization of label-retaining cells in hamster epithelia. J Invest Dermatol 82: 618-622. doi: 10.1111/1523-1747.ep12261460
    [83] Kusumbe AP, Bapat SA (2009) Cancer stem cells and aneuploid populations within developing tumors are the major determinants of tumor dormancy. Cancer Res 69: 9245-9253. doi: 10.1158/0008-5472.CAN-09-2802
    [84] Dey-Guha I, Wolfer A, Yeh AC, et al. (2011) Asymmetric cancer cell division regulated by AKT. Proc Natl Acad Sci USA 108: 12845-12850. doi: 10.1073/pnas.1109632108
    [85] Bajaj J, Zimdahl B, Reya T (2015) Fearful symmetry: subversion of asymmetric division in cancer development and progression. Cancer Res 75: 792-797. doi: 10.1158/0008-5472.CAN-14-2750
    [86] Gattinoni L, Speiser DE, Lichterfeld M, et al. (2017) T memory stem cells in health and disease. Nature Med 23: 18-27. doi: 10.1038/nm.4241
    [87] Ciocca ML, Barnett BE, Burkhardt JK, et al. (2012) Cutting edge: Asymmetric memory T cell division in response to rechallenge. J Immunol 188: 4145-4148. doi: 10.4049/jimmunol.1200176
    [88] Schmidt JM, Panzilius E, Bartsch HS, et al. (2015) Stem-cell-like properties and epithelial plasticity arise as stable traits after transient Twist1 activation. Cell Rep 10: 131-139. doi: 10.1016/j.celrep.2014.12.032
    [89] Kroger C, Afeyan A, Mraz J, et al. (2019) Acquisition of a hybrid E/M state is essential for tumorigenicity of basal breast cancer cells. Proc Natl Acad Sci USA 116: 7353-7362. doi: 10.1073/pnas.1812876116
    [90] Ye X, Tam WL, Shibue T, et al. (2015) Distinct EMT programs control normal mammary stem cells and tumour-initiating cells. Nature 525: 256-260. doi: 10.1038/nature14897
    [91] Dongre A, Weinberg RA (2019) New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 20: 69-84. doi: 10.1038/s41580-018-0080-4
    [92] Kreso A, O'Brien CA, van Galen P, et al. (2013) Variable clonal repopulation dynamics influence chemotherapy response in colorectal cancer. Science 339: 543-548. doi: 10.1126/science.1227670
    [93] Zhu Y, Luo M, Brooks M, et al. (2014) Biological and clinical significance of cancer stem cell plasticity. Clin Transl Med 3: 32.
    [94] Jasnos L, Aksoy FB, Hersi HM, et al. (2013) Identifying division symmetry of mouse embryonic stem cells: negative impact of DNA methyltransferases on symmetric self-renewal. Stem Cell Rep 1: 360-369. doi: 10.1016/j.stemcr.2013.08.005
  • molsci-07-02-006-s001.pdf
  • This article has been cited by:

    1. Amaris Guevara-Garcia, Matis Soleilhac, Nicolas Minc, Delphine Delacour, Regulation and functions of cell division in the intestinal tissue, 2023, 10849521, 10.1016/j.semcdb.2023.01.004
    2. Koangyong Sung, Kenji Hosoya, Yusuke Murase, Tatsuya Deguchi, Sangho Kim, Takafumi Sunaga, Masahiro Okumura, Visualizing the cancer stem‐like properties of canine tumour cells with low proteasome activity, 2022, 20, 1476-5810, 324, 10.1111/vco.12779
    3. Mina Lagzian, S. Ehsan Razavi, M. Goharimanesh, Investigation on tumor cells growth by Taguchi method, 2022, 77, 17468094, 103734, 10.1016/j.bspc.2022.103734
    4. Seung-Hee Seo, Shamrat Kumar Paul, Mita Shikder, Mushira Khanam, Popy Ghosh, Tasnin Al Hasib, Kazi Ahsan Ahmed, Suranjana Sikdar, Md Jamal Uddin, Youngjoo Kwon, An Insight into Pathophysiological Features and Therapeutic Advances on Ependymoma, 2021, 13, 2072-6694, 3221, 10.3390/cancers13133221
    5. Shaan N. Chhabra, Brian W. Booth, Asymmetric cell division of mammary stem cells, 2021, 16, 1747-1028, 10.1186/s13008-021-00073-w
    6. Burak Cem SONER, Eda AÇIKGÖZ, Gülperi ÖKTEM, Çağ ÇAL, Effect of Zoledronic Acid Treatment on Cell Adhesion Molecules in Prostate Cancer Stem Cells, 2022, 1300-414X, 10.32708/uutfd.1137962
    7. Hye Yeon Choi, Jaekwon Seok, Geun-Ho Kang, Kyung Min Lim, Ssang-Goo Cho, The role of NUMB/NUMB isoforms in cancer stem cells, 2021, 54, 1976-670X, 335, 10.5483/BMBRep.2021.54.7.048
    8. Diletta Rosati, Antonio Giordano, Single-cell RNA sequencing and bioinformatics as tools to decipher cancer heterogenicity and mechanisms of drug resistance, 2022, 195, 00062952, 114811, 10.1016/j.bcp.2021.114811
    9. Zizhu Li, Ying Yi Zhang, Haomiao Zhang, Jiaxuan Yang, Yongze Chen, Hezhe Lu, Asymmetric Cell Division and Tumor Heterogeneity, 2022, 10, 2296-634X, 10.3389/fcell.2022.938685
    10. Samuel Kogan, The Mesenchymal-Epithelial Transition and Metaplasia, 2022, 16, 1911-8902, 4, 10.33137/juls.v16i1.38941
    11. Hector Mayani, Antonieta Chávez-González, Karla Vázquez-Santillan, Jorge Contreras, Monica L Guzman, Cancer Stem Cells: Biology and Therapeutic Implications, 2022, 53, 01884409, 770, 10.1016/j.arcmed.2022.11.012
    12. Ana Salas, Silvia Beltrán-Flores, Carmen Évora, Ricardo Reyes, Francisco Montes de Oca, Araceli Delgado, Teresa A. Almeida, Stem Cell Growth and Differentiation in Organ Culture: New Insights for Uterine Fibroid Treatment, 2022, 10, 2227-9059, 1542, 10.3390/biomedicines10071542
    13. Elena L. Paley, 2023, 9780443191220, 267, 10.1016/B978-0-443-19122-0.00023-6
    14. Rosalia Moreddu, Nanotechnology and Cancer Bioelectricity: Bridging the Gap Between Biology and Translational Medicine, 2024, 11, 2198-3844, 10.1002/advs.202304110
    15. Anca-Narcisa Neagu, Danielle Whitham, Pathea Bruno, Aneeta Arshad, Logan Seymour, Hailey Morrissiey, Angiolina I. Hukovic, Costel C. Darie, Onco-Breastomics: An Eco-Evo-Devo Holistic Approach, 2024, 25, 1422-0067, 1628, 10.3390/ijms25031628
    16. Viviana Rubino, Michelle Hüppi, Sabine Höpner, Luigi Tortola, Noah Schnüriger, Hugo Legenne, Lea Taylor, Svenja Voggensperger, Irene Keller, Remy Bruggman, Marie-Noëlle Kronig, Ulrike Bacher, Manfred Kopf, Adrian F. Ochsenbein, Carsten Riether, IL-21/IL-21R signaling renders acute myeloid leukemia stem cells more susceptible to cytarabine treatment and CAR T cell therapy, 2024, 26663791, 101826, 10.1016/j.xcrm.2024.101826
    17. Nick A. Kuburich, Petra den Hollander, Maria Castaneda, Mika Pietilä, Ximing Tang, Harsh Batra, Francisco Martínez-Peña, Tanvi H. Visal, Tieling Zhou, Breanna R. Demestichas, Ritesh V. Dontula, Jojo Y. Liu, Joanna Joyce Maddela, Reethi S. Padmanabhan, Lan Thi Hanh Phi, Matthew J. Rosolen, Thiru Sabapathy, Dhiraj Kumar, Filippo G. Giancotti, Luke L. Lairson, Maria Gabriela Raso, Rama Soundararajan, Sendurai A. Mani, Stabilizing vimentin phosphorylation inhibits stem-like cell properties and metastasis of hybrid epithelial/mesenchymal carcinomas, 2023, 42, 22111247, 113470, 10.1016/j.celrep.2023.113470
    18. Jan Jakub Lica, Joanna Jakóbkiewicz-Banecka, Andrzej Hellmann, In Vitro models of leukemia development: the role of very small leukemic stem-like cells in the cellular transformation cascade, 2025, 12, 2296-634X, 10.3389/fcell.2024.1463807
    19. Sakshi Kevat, Archie Mistry, Naman Oza, Mohit Majmudar, Netra Patel, Rushabh Shah, A. V. Ramachandran, Ritu Chauhan, Shafiul Haque, Nidarshana Chaturvedi Parashar, Hardeep Singh Tuli, Gaurav Parashar, Cancer Stem Cell Regulation as a Target of Therapeutic Intervention: Insights into Breast, Cervical and Lung Cancer, 2025, 1559-0283, 10.1007/s12013-025-01666-w
    20. Georgios Stamatakos, Eleni Kolokotroni, Foteini Panagiotidou, Stamatia Tsampa, Christos Kyroudis, Simon Spohn, Anca-Ligia Grosu, Dimos Baltas, Constantinos Zamboglou, Ilias Sachpazidis, In silico oncology: a mechanistic multiscale model of clinical prostate cancer response to external radiation therapy as the core of a digital (virtual) twin. Sensitivity analysis and a clinical adaptation approach, 2025, 16, 1664-042X, 10.3389/fphys.2025.1434739
    21. Jie Huang, Shufeng Luo, Juan Shen, Maya Lee, Rachel Chen, Shenglin Ma, Lun-Quan Sun, Jian Jian Li, Cellular polarity pilots breast cancer progression and immunosuppression, 2025, 0950-9232, 10.1038/s41388-025-03324-0
    22. Zhenzhen Chen, Huanle Qi, Yapeng Xue, Yaqi Zhang, Zhuo Zhang, Shun Xu, Shixin Liao, Xiaoyu Zeng, Jiayi Wu, Xinrui Lv, Qiankun He, Pingping Zhu, Characteristic, Regulation and Targeting Strategies of Cancer Stem Cells and Their Niche in Digestive System Tumors, 2025, 4, 2769-6448, 10.1002/mog2.70015
    23. Shengze Li, Xinran Dong, Kexing Gao, Yiyang Wang, Min Li, Huayun Deng, Shuangyu Ma, Yaping Lv, Wei Jin, Quanfu Li, Yuming Wang, Xiaodong Liao, Kangjing Bian, Aiwu Zhou, Chengping Hu, Lei Huang, PP2A Promotes the Symmetric Division of MUC1‐Dominant Cancer Stem‐Like Cells in Small Cell Lung Cancer, 2025, 2198-3844, 10.1002/advs.202503545
    24. Ruotian Du, Yizhuo Wu, Min Zou, Meiying Luo, Yan Li, Yingfeng Shao, Jing Ji, Fan Song, Long Li, Intracellular pressure: Regulation, characterization and implication, 2025, 68, 1674-7348, 10.1007/s11433-025-2640-x
  • Reader Comments
  • © 2020 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(9169) PDF downloads(624) Cited by(24)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog