Skip to main content

Cancer stem cell fate determination: mito-nuclear communication

Abstract

Cancer stem cells (CSCs) are considered to be responsible for tumor recurrence and metastasis. Therefore, clarification of the mechanisms involved in CSC stemness maintenance and cell fate determination would provide a new strategy for cancer therapy. Unregulated cellular energetics has been accepted as one of the hallmarks of cancer cells, but recent studies have revealed that mitochondrial metabolism can also actively determine CSC fate by affecting nuclear stemness gene expression. Herein, from the perspective of mito-nuclear communication, we review recent progress on the influence of mitochondria on CSC potential from four aspects: metabolism, dynamics, mitochondrial homeostasis, and reactive oxygen species (ROS).

Video Abstract

Introduction

Cancer stem cells (CSCs) are a subpopulation of cancer cells with the potential for self-renewal and multidifferentiation and thus drive carcinogenesis, chemoresistance, recurrence and metastasis [1,2,3,4,5,6]. CSCs are also known as cancer stem cell-like cells [7], tumorigenic cells [8], tumor stem-like cells (TSCs) [9], and cancer- or tumor-initiating cells (CICs or TICs) [10, 11]. In 1997, CSCs were successfully isolated from the blood of leukemia patients for the first time [12]. Subsequently, CSCs were further identified in solid tumors, such as colon cancer [13], breast cancer [14], skin squamous cell cancer [15] and glioblastoma (GBM) [16]. However, CSCs are as heterogeneous as cancer cells, include metastatic cancer stem cell (MeCSC) or chemoresistant cancer stem cell (CRCSC) subsets, and have a quiescent or proliferative status and epithelial or mesenchymal status [17]. In response to various stimuli, the subsets or status of CSCs can be altered accordingly; therefore, CSCs are also plastic [18]. Moreover, upon attack by chemo/radiotherapy [19,20,21,22], hypoxia [23] and detachment [24], cancer cells can acquire stemness potential. Thus, plasticity is an important feature of CSCs and the key point to understanding CSC stemness maintenance and fate determination.

Mitochondrial energy metabolism is essential to CSCs in various intracellular activities, especially for nuclear stemness gene expression. Under different functional statuses or environmental conditions, plastic CSCs might adopt different metabolic patterns accordingly. Thus, the acquisition of CSC potential is accompanied by a reprogramming of cellular metabolism [25, 26]. Furthermore, cellular metabolism has been reported to determine CSC fate by epigenetically modifying nuclear stemness genes via metabolites. Therefore, mitochondrial energy metabolism is not merely a phenotype of CSCs but also a determinant of CSC fate. In this review, we focus on the regulation of CSC plasticity from the view of mitochondrial signals, that is, the regulation of mitochondrial metabolism, dynamics, mitochondrial homeostasis, and reactive oxygen species (ROS) on CSC potentials and the involvement of mito-nuclear communication.

Cancer stem cell plasticity and fate determination

The CSC theory holds that cancer cells are heterogeneous, and rare CSCs are the major driver of tumor initiation, metastasis and therapeutic resistance and thus the target for eradicating tumors [27,28,29,30]. More recently, the plastic CSC model has become widely accepted, and plasticity has become the main challenge of CSC targeted therapy [31]. Effective therapy must focus on the key regulatory factors that both maintain and induce the stemness of CSCs.

Cancer stem cell plasticity

Cell plasticity is defined as the ability of cells to quickly adapt to the changing microenvironment by dynamically switching between different cellular statuses or phenotypes in a reversible manner, which hijacks the program of dedifferentiation or transdifferentiation in cells [32,33,34]. The plasticity of CSCs, however, is exhibited as the dynamic and reversible transitions between quiescent and proliferative CSCs, epithelial and mesenchymal CSCs, CSCs and non-CSCs, or the evolution from primary CSCs to MeCSCs or CRCSCs, which respond to the adverse tumor microenvironment (Fig. 1). Higher CSC plasticity is likely to facilitate tumor progression and is associated with poor patient clinical outcomes.

Fig. 1
figure 1

Phenotypic plasticity of cancer stem cells. The plasticity of CSCs is exhibited as the dynamic and reversible transitions between quiescent and proliferative CSCs, epithelial and mesenchymal CSCs and CSCs and non-CSCs (A) or the evolution from primary CSCs to MeCSCs or CRCSCs (B). CSCs Cancer stem cells, MeCSCs Metastatic cancer stem cells, CRCSCs Chemoresistant cancer stem cells

Due to changing microenvironment signals, CSCs are not always in a quiescent status but can be activated and enter a proliferative status and then initiate tumor growth and recurrence [35]. In bladder cancer, for example, combined treatment with gemcitabine and cisplatin (GC) induces the production of a high level of prostaglandin E2 (PGE2), which further forces CSCs to exit from a quiescent status and enter a proliferative status and then causes tumor progression [36]. In addition, Prx1+ mesenchymal progenitor-derived CXCL12 maintains leukemic stem cells (LSCs) in a quiescent and tyrosine kinase inhibitor (TKI)-resistant status, whereas CXCL12 deletion forces LSCs to enter a proliferative (cycling) status and makes them subsequently sensitive to TKI [37] (Fig. 1A).

Another important manifestation of CSC plasticity is the transition between epithelial-like phenotypes and mesenchymal-like phenotypes, that is, epithelial-to-mesenchymal transition (EMT). In squamous cell carcinoma, two different CSC subsets are found: CD44highESAhigh CSCs (non-EMT subset), which have an epithelial phenotype and proliferate rapidly; and CD44highESAlow CSCs (EMT subset), which have a mesenchymal phenotype and migrate extensively [38]. When migrating to a secondary site, EMT CSCs recover into a proliferative mesenchymal-to-epithelial transition (MET) phenotype and then form metastatic tumors. Similarly, CD24-CD44+ breast cancer stem cells (BCSCs) located at the tumor invasive frontier display a quiescent mesenchymal phenotype, while aldehyde dehydrogenase-positive (ALDH+) BCSCs distributed more centrally show a proliferative epithelial phenotype. Moreover, BCSCs switch between a mesenchymal phenotype and an epithelial phenotype [39] (Fig. 1A).

Moreover, CSCs and non-CSCs can interconvert and achieve a dynamic balance [40, 41]. It has been reported that bulk breast cancer cells can convert into BCSCs under certain conditions, such as chemo/radiotherapy [42], detachment [24], and IL-6 induction [43]. During this process, breast cancer cells are switched between non-CSCs and CSCs by selective splicing of CD44 into CD44v or CD44s, and cancer cells with high levels of CD44v lose CSC stemness, while those with high levels of CD44s gain CSC stemness [44] (Fig. 1A).

In addition, CSCs exhibit phenotypic evolution during the process of tumor progression. During the early stage of tumorigenesis, primary CSCs are present in tumor cells with gene mutations. As a tumor develops into an advanced stage, MeCSCs with the potential to disseminate from the primary site, survive in the circulation, and seed and expand in the new microenvironment evolve (Fig. 1B). These MeCSCs can be organ specific [45]. When patients receive targeted therapy and/or chemotherapy, primary CSCs survive long-term administration of drugs; that is, CRCSCs or drug-resistant CSCs are developed [17] (Fig. 1B). It has been reported that MeCSCs share similar stemness potentials and epigenetic regulation mechanisms with primary CSCs. The first MeCSCs were identified as CD133+CXCR4+ subpopulations in CD133+ pancreatic CSCs with the potential to metastasize to the liver [46]. MeCSC subpopulations such as CD44v6 CSCs or CD26+ CSCs in colorectal cancer [47, 48], CD44+ CSCs in breast cancer [49], and CXCR4+ CSCs in prostate cancer [50] have also been found. Regarding CRCSCs, the CD44+CD133+ subpopulation with the potential for drug resistance was identified in CSC-like SKOV3 ovarian tumorspheres treated with cisplatin and/or paclitaxel [51]. In addition, IGF1R+ chemoresistant TSCs are found in lymphoma hepatocellular carcinoma (HCC) or Lewis lung cancer cells treated with doxorubicin, 5-fluorouracil or cisplatin [9] (Fig. 1B).

The plasticity of phenotype and status indicates that the developmental fate of cells can be changed; for example, it can be shifted toward a more undifferentiated status via dedifferentiation or to other lineages via transdifferentiation. Plasticity enables CSCs to better survive in adverse environments by easily switching their status and quickly altering phenotypes in response to various internal or external signals [52].

Cancer stem cell fate determination

Although the alteration of plastic cellular phenotype or status is triggered by environmental cues, the decision of CSC fate is actually determined by the expression of nuclear stemness genes, which are regulated by specific transcription factors (TFs) [53]. In glioblastoma stem cells (GSCs), for example, the expression levels of key stemness-related TFs (POU3F2, SOX2, SALL2, and OLIG2) are significantly higher than those in more differentiated tumor cells. These four TFs can dedifferentiate and reprogram GBM cells into GSCs in in vitro cultured cells and in vivo animal models [54]. In contrast, dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) promotes the differentiation of GSCs and inhibits the acquisition of stemness potential by decreasing the expression of SOX2 [55].

With the gain or loss of stemness potential, the expression of stemness-related TFs is increased or decreased and is regulated at the posttranscriptional or translational level via epigenetic modifications. Growing evidence reveals that nonmutational and reversible epigenetic events, such as histone and chromatin modifications or DNA methylation, significantly contribute to CSC plasticity and carcinogenesis. For instance, SIRT1-mediated deacetylation of β-catenin maintains its stability, and the resulting nuclear accumulation of β-catenin increases the transcriptional level of NANOG and promotes the stemness potential of liver CSCs [56]. In another study, the zinc finger and homeobox 2 (ZHX2) protein was reported to eliminate liver CSC features by transcriptionally repressing KDM2A and inhibiting KDM2A-mediated demethylation of histone H3 lysine 36 in the promoter regions of stemness-related TFs (NANOG, SOX4, OCT4) [57].

Many factors inside and outside the cells, such as growth factors, inflammatory mediators, intracellular pH, mitochondrial metabolites and ROS, can influence the activity of stemness-related TFs at the level of epigenetic modification and then alter cell fates [58, 59]. Recently, an increasing number of studies have suggested that mitochondria play important roles not only in maintaining CSC stemness but also in determining CSC fate [60,61,62,63,64,65,66]. Therefore, we summarize CSC fate determination from a new view of mito-nuclear communication.

Mito-nuclear communication and CSC fate determination

Mitochondria have their own genetic material (mtDNA) and corresponding gene transcription and protein translation systems. However, the majority (>99%) of proteins in mitochondria are not encoded by the mitochondrial genome but by the nuclear genome [67]. To harmonize nuclear-encoded protein synthesis with appropriate mitochondrial biogenesis or energy metabolism, the crosstalk between mitochondria and nucleus, in other words, mito-nuclear communication, has evolved. Proper communication between mitochondria and the nucleus allows mutual benefits and ensures the overall fitness of cells.

Ordinarily, mitochondria are under tight control by the nucleus through anterograde regulation signaling (from nucleus to mitochondria) according to cellular energy needs [68]. However, more recent studies have demonstrated that mitochondria can also generate retrograde signals to the nucleus via mito-to-nuclear communication mediated by small molecules, metabolites, peptides, mtDNA and ions through physical contact or signal transmission [69,70,71]. This concept expands the previous understanding that mitochondria are not merely semiautonomous organelles. Moreover, mitochondria can actively influence the expression of nuclear stemness genes, reprogram cell metabolism and phenotype, and thus determine CSC fate [68, 72, 73].

First, mitochondrial energy metabolism-derived metabolites or ROS are reported to play important roles in CSC fate determination through metabolic and epigenetic modification of stemness genes. In addition, mitochondrial dynamics or the balance between biogenesis and mitophagy could exert an influence on CSCs. In the following section, we discuss the influences of mitochondrial retrograde signaling on CSC fate determination in four aspects: energy metabolism, dynamics, mitochondrial homeostasis, and ROS.

Mitochondrial energy metabolism

Mitochondrial energy metabolism has profound impacts on the fate of CSCs [74,75,76] (Fig. 2). Recently, mitochondrial metabolites, which are generally involved in energy support, were reported to act as signaling molecules and to play critical roles in controlling gene expression [77]. In detail, mitochondrial metabolites, such as acetyl-coenzyme A (acetyl-CoA), β-hydroxybutyric acid (βHB), S-adenosylmethionine (SAM), NAD+, succinate, α-ketoglutarate (α-KG), ATP, and FAD, act as cofactors of epigenetic modifying enzymes and drive the acquisition of stemness potential by affecting stemness gene expression [72, 78]. For example, by enhancing the acetylation levels of histone H4K8ac, H4K12ac, and H4K16ac, acetyl-CoA activates the protein expression of stemness-related TFs (c-MYC, OCT4, KLF4, SOX2), which further increases the tumorsphere formation of CSCs in triple-negative breast cancer [79] (Fig. 2A). Furthermore, in hepatocellular carcinoma, accumulated βHB upregulates the expression levels of CD44, CD133, SOX9 and EpCAM by increasing the β-hydroxybutyrylation level of histone H3K9bhb and then significantly improves the proportions of CSCs for in vivo tumor formation and increases the abilities of colony formation and tumorsphere formation [80] (Fig. 2A). However, in acute myeloid leukemia (AML), low SAM levels decrease global DNA methylation, causing increased expression of differentiated myeloid genes (CD11b, CD14) but decreased expression of stemness genes, thereby enhancing differentiation but inhibiting self-renewal in LSCs [81].

Fig. 2
figure 2

Mitochondrial energy metabolism and cancer stem cell stemness. A Acetyl-CoA promotes the expression of stemness-related transcription factors by enhancing the acetylation levels of histone H4K8ac, H4K12ac, and H4K16ac. βHB upregulates the expression levels of CSC marker genes by increasing the β-hydroxybutyrylation level of histone H3K9bhb. B Metformin suppresses mitochondrial complex I of oxidative phosphorylation and downregulates the expression of CSC-related genes. Inhibitors of the key methionine cycle enzymes MAT2A, cycloleucine and FIDAS-5 reduce CSC stemness by decreasing SAM levels and the expression of the methylation marks H3K4me3 alone or H3K9me3, H3K27me3, H3K36me2, H3K36me3 and H3K79me3. acetyl-CoA acetyl-coenzyme A, βHB β-hydroxybutyric acid, CSC Cancer stem cell, TFs Transcription factors, MAT2A Methionine adenosyltransferase 2A, SAM S-adenosylmethionine

Mitochondrial dynamics

Mitochondrial dynamics, which involve vigorous changes in mitochondrial morphology between fission and fusion, are essential events that maintain the distribution, function and vitality of mitochondria [82,83,84,85]. Mitochondrial fission is the process by which a single mitochondrion divides into two short, round, balloon-shaped or fragmented daughter mitochondria and is mainly regulated by dynamin-related protein 1 (DRP1), DRP1 receptor mitochondrial fission factor (MFF), and fission factor 1 (FIS1) [86, 87]. Mitochondrial fusion is a phenomenon in which two closely contacted mitochondria are coordinately fused in the outer and inner membranes, forming an elongated, large, and interconnected mitochondrial network. The process of mitochondrial fusion is mainly regulated by the outer membrane fusion proteins mitofusins 1 and 2 (MFN1, MFN2) and the inner membrane fusion protein optic atrophy 1 (OPA1) [88]. To meet the cellular energy requirements in response to environmental changes, mitochondria dynamically switch between tubular and fragmented forms by balancing the process of fission and fusion.

Both fusion and fission can enhance CSC stemness and maintain self-renewal, depending on the tissue type of the tumor. Bonnay et al found that mitochondrial fusion helps to sustain CSC fates or phenotypes in neuroblastoma [89]. In detail, mitochondrial fusion induced by brat knockdown increased oxidative phosphorylation (OXPHOS) and NAD+ levels and then drove the immortalization and tumorigenicity of TICs [89] (Fig. 3A). However, some other studies have indicated that mitochondrial fission promotes the potential and self-renewal of CSCs [90]. In liver cancer, the complex formed by T-box transcription factor 19 (TBX19) and PRMT1 induces MFF expression by increasing histone H4R3me2a/H3K9ac levels; then, MFF promotes mitochondrial fission, increases the expression level of OCT4, enhances the formation of tumorspheres and enriches the side populations (SPs) [91] (Fig. 3B). In glioblastomas, cycle-dependent kinase 5 (CDK5) phosphorylates DRP1 at Ser616 and increases mitochondrial cleavage, which then induces the expression of stemness genes (OLIG2, OCT4, NANOG, NESTIN, POU3F2, CD133, SSEA1) [92] (Fig. 3B). Contrary to popular views, recent studies have shown evidence that mitochondrial fission inhibits the stemness of CSCs. For example, the activation of mitochondrial fission by overexpressing MFF impairs BCSC propagation through methods such as reducing the capacity for tumorsphere formation and the enzyme activity of the stem cell marker ALDH [93].

Fig. 3
figure 3

Mitochondrial dynamics and cancer stem cell stemness. A Mitochondrial fusion induced by brat knockdown increased OXPHOS and NAD+ levels and then drove the immortalization and tumorigenicity of tumor-initiating cells (TICs). B CDK5 phosphorylates DRP1 and increases mitochondrial cleavage, which then induces the expression of stemness genes. Similarly, MFF promotes mitochondrial fission and then increases OCT4 expression levels. Mdivi-1 reduces the expression of stemness genes by inhibiting DRP1; selective COX-2 inhibitors resveratrol, celecoxib, and etodolac reduce the expression level of stemness genes by inhibiting DRP1 indirectly. OXPHOS Oxidative phosphorylation, TICs Tumor-initiating cells, CDK5 Cycle-dependent kinase 5, MFF Mitochondrial fission factor

Mitochondrial homeostasis

Mitochondrial biogenesis and mitophagy are two balanced processes in cells that control the quantity and quality of mitochondria [94]. Mitochondrial biogenesis is the process of replenishing new healthy mitochondria, while mitophagy is the activity by which cells spontaneously phagocytose or selectively degrade dysfunctional or redundant aging mitochondria under conditions of stress such as hypoxia or nutritional deficiency [95,96,97,98]. Mitochondrial biogenesis and mitophagy are both key events in regulating the stemness of CSCs.

Promoting mitochondrial biogenesis by PGC-1α [99] or its cofactor estrogen-related receptor α (ERRα) [100] increases tumorsphere formation in pancreatic cancer and breast cancer, respectively (Fig. 4A). In contrast, PGC-1α knockdown in glioblastoma cells leads to the attenuation of the neoplastic phenotype and loss of stem-like features, which was reflected by reduced expression of the stemness gene SOX2 but prolonged survival of nude mice [101].

Fig. 4
figure 4

Mitochondrial homeostasis and cancer stem cell stemness. A PGC-1α or its cofactor ERRα increases mitochondrial biogenesis, which further promotes the maintenance of the CSC phenotype. Azithromycin, doxycycline, and tigecycline block mitochondrial biogenesis by targeting mitochondrial ribosomes 39S and 28S and then inhibit CSC self-renewal capability. In addition, the selective PGC-1α inhibitor SR-18292 or ERRα inverse agonist XCT790 downregulates the expression of stemness genes and reduces the ratio of CSCs. B AMPK-FIS1 signaling promotes mitophagy, thereby enhancing CSC self-renewal by inhibiting ROS production. CCCP increases CSC ratios by recruiting PINK1 and enhancing mitophagy-mediated removal of phosphorylated p53 and then increases NANOG expression. In contrast, the combination of melatonin and verteporfin reduces CSC stemness by inhibiting the expression of PINK1/Parkin, while mefloquine hydrochloride reduces the ratio of colon CSCs by inhibiting mitophagy and lysosomal activity. In addition, 188Re-liposomes reduced the protein levels of mitophagy markers, which further decreased the function of CSCs. CSC Cancer stem cell, AMPK Adenosine 5'-monophosphate-activated protein, FIS1 Fission factor 1, CCCP Carbonyl cyanide chlorophenylhydrazone, PINK1 PTEN-induced kinase 1

Regarding mitophagy, an increase in mitophagy by the depression of oxidative stress or an attenuation of mitophagy by parkin interference can lead to the generation or loss of CD44highCD24-/low esophageal CSCs [102]. For example, adenosine 5'-monophosphate-activated protein (AMPK)-FIS1 signaling-mediated mitophagy leads to the elimination of damaged mitochondria, thereby inhibiting intracellular ROS production and promoting the self-renewal and survival of LSCs [103] (Fig. 4B). At the molecular level, mitophagy promotes the formation of CSCs by altering the subcellular location of phosphorylated p53 between mitochondria and the nucleus. In detail, when mitophagy is enhanced by carbonyl cyanide chlorophenylhydrazone (CCCP), PTEN-induced kinase 1 (PINK1) recruits and phosphorylates p53 at serine-392 and entraps p53 on mitochondria; subsequently, p53 is removed by mitophagy, and thus, the expression of NANOG and the ratio of the CD133+ liver CSC population are increased (Fig. 4B). Conversely, when mitophagy is inhibited by a mitochondrial fission inhibitor (Mdivi-1), PINK1-phosphorylated p53 is rapidly translocated to the nucleus, which results in the suppressed expression of NANOG and a reduction in the CD133+ CSC ratio [104].

Mitochondrial ROS

ROS, the main byproducts of oxidative metabolism, include superoxide radical anions, hydroxyl radicals, hydrogen peroxide, and lipid hydrogen peroxide [105, 106]. The roles of ROS in regulating CSC self-renewal and survival occur in a context- and tissue-dependent manner. Generally, ROS are reported to maintain CSC properties and to induce CSC proliferation and tumorigenicity. In colorectal cancer (CRC), RAC1 activation upon Apc loss triggers the production of high levels of ROS in the intestines of vil-Cre-ERT2 Apcfl/fl Rac1fl/fl mice; then, ROS further increase the expression of stemness genes (LGR5, OLFM4, RGMB), confer LGR5 CSC phenotypes, and thus initiate CRC [107]. Conversely, as ROS levels decrease following a sublethal dose of H2O2, ESA+CD44+CD24- BCSCs lose the ability to form tumorspheres and colonies [108]. However, many studies have found that increasing intracellular ROS levels can induce CSC death and that a low level of ROS is required for the maintenance of cancer stemness [109]. In pancreatic cancer, lncRNA SLC7A11-AS1 promoted stemness potential by scavenging ROS, which functioned by interacting with β-TRCP1 and then blocking nuclear factor erythroid-2-related factor 2 (NRF2) degradation [110] (Fig. 5A). In GSCs, high levels of prohibitin promote GSC self-renewal by stabilizing PRDX3 and maintaining low levels of mitochondrial ROS [111] (Fig. 5A).

Fig. 5
figure 5

Mitochondrial ROS signaling and cancer stem cell stemness. A SLC7A11-AS1/β-TRCP1 or prohibitin promote CSC stemness potential by blocking NRF2 degradation and thus scavenging ROS or by interacting with PRDX3 and maintaining low mitochondrial ROS levels. B By blocking the role of iron in electron transport, deferiprone induces mitochondrial ROS and disrupts CSC stemness. KS10076, 13 R, 20-diHDHA, and PAA induce the production of ROS, degrade STAT3 or decrease the expression levels of CSC self-renewal genes, while TiOxNPs reduce CSC marker expression levels by inducing ROS levels and inactivating AKT signaling. CSC Cancer stem cell, 13 R 20-diHDHA, 13R 20-dihydroxydocosahexaenoic acid, PAA Phenylacetaldehyde, TiOxNPs Titanium peroxide nanoparticles

Blocking mito-nuclear communication as an emerging strategy for anti-CSC therapy

Despite significant improvements in anticancer drug development, CSC-derived chemoresistance and recurrence are still major challenges for cancer treatments. As CSCs and non-CSCs have different metabolic characteristics and mitochondrial metabolism governs CSC fates, blocking mito-nuclear communication would be an effective and innovative strategy. In the following section, we summarize the recent progress in gene interventions and therapeutic agents targeting mito-nuclear communication for CSC eradication (Tables 1 and 2).

Table 1 Genetic inhibition for blocking mito-nuclear communication in CSCs
Table 2 Therapeutic agents for blocking mito-nuclear communication in CSCs

Targeting mitochondrial energy metabolism

The well-known means of disrupting the maintenance of CSC features by interfering with mitochondrial energy metabolism is the inhibition of mitochondrial complex I of oxidative phosphorylation. One such example is the effective drug metformin, which downregulates the expression of CSC-related genes, decreases the ratios of CD44highALDHhigh cells as well as the sizes and numbers of tumorspheres, and thus reduces the volume of tumors in cholangiocarcinoma and head and neck squamous cell cancer (HNSCC) [116, 117] (Fig. 2B).

In addition, mitochondrial-derived metabolites are popular choices for targeting CSCs by interfering with mitochondrial energy metabolism. For example, SAM is the universal donor for DNA and histone methylation and has been linked to CSC self-renewal; therefore, decreasing SAM levels by blocking the methionine cycle key enzyme adenosyltransferase 2A (MAT2A) is considered an ideal strategy for eradicating CSCs. In lung cancer, it was reported that the MAT2A inhibitor FIDAS-5 strongly decreases the intracellular level of SAM and significantly inhibits the expression of methylation marks such as H3K4me3, H3K9me3, H3K27me3, H3K36me2, H3K36me3 and H3K79me3 in CSCs, thereby greatly lowering the tumorigenic potential of CSCs, which includes reducing the mass and volume of xenograft tumors and diminishing the size of NOD. Cg-Prkdcscid Il2rgtm1Wjl/SzJ mouse lung lesions and decreased the ratio of CD166+ CSCs [118] (Fig. 2B). Another MAT2A inhibitor, cycloleucine, has been found to enhance the suppressive effect of methionine depletion on BCSCs by inhibiting the protein expression of the stemness transcription factor SOX9 by suppressing the demethylation of H3K4me3 (Fig. 2B). In addition, the combination of MAT2A inhibition and methionine depletion could more effectively reduce mammospheres in vitro and the burden of primary and lung metastases in vivo [119].

Targeting mitochondrial dynamics

Some studies have found that inhibiting mitochondrial fission-related proteins may be another potential therapeutic strategy for targeting the stemness potential of CSCs. For example, knockdown of mitochondrial fission-related genes, such as DRP1, MFF, BRD4 or FIS1, reduces the expression levels of stemness genes and the capability of forming tumorspheres and tumors, especially the self-renewal of CSCs in brain, prostate and lung cancers [92, 112, 113] (Table 1).

For pharmacological intervention, Mdivi-1 is the most common and generally recognized DRP1-selective inhibitor that has been reported to reduce the percentage of SP+ or CD44+ CSC subpopulations, the expression of stemness genes, and the formation capacity of tumorspheres in vitro and in tumors in vivo in the context of nasopharyngeal carcinoma (NPC), liver cancer, ovarian cancer, etc. [86, 91, 120, 121] (Fig. 3B). Moreover, Mdivi-1 directly suppresses the function of CSCs that are isolated by stem cell markers. In detail, Mdivi-1 inhibits the self-renewal and tumor initiation capacities of CD133+CD15+ brain tumor-initiating cells (BTICs) [92] and decreases the levels of stemness genes in EpCAM+CD133+ liver cancer stem cells (LCSCs) [91]. As COX-2 maintains the CSC phenotype by activating DRP1, selective COX-2 inhibitors, such as resveratrol, celecoxib, and etodolac, are reported to reduce the expression levels of stemness genes, the ratio of SP+ subpopulations, and the tumorsphere formation capacity in NPC, bladder cancer, and glioblastoma [120, 122, 123] (Fig. 3B). In addition, BRD4 or PRMT1 can act as a transcription regulator or an epigenetic activator for MFF, respectively; therefore, targeting BRD4 or PRMT1 can be an attractive therapeutic option for eradicating CSCs by blocking mitochondrial fission. Therefore, the BRD4 inhibitor OTX015 suppresses tumorigenicity and self-renewal ability in prostate CSCs, and the PRMT1 inhibitor furamidine blocks TBX19-induced mitochondrial fission and decreases the capacity for tumorsphere formation and tumorigenesis in LCSCs [91].

Targeting mitophagy or mitochondrial biosynthesis.

Because mitophagy actively promotes the production of CSCs, targeting mitophagy-related genes could counteract CSCs. In AML, oral squamous cell cancer, and liver cancer, the knockdown of mitophagy-related regulator genes, such as PINK1, TBC1D15, ATG14, and BNIP3, decreases the levels of self-renewal and stemness, the number of tumorspheres, and the percentage of SP+ subpopulations [103, 114, 115] (Table 1).

Unfortunately, there are few reports on targeting CSCs by mitophagy-specific inhibitors but indirect mitophagy inhibition. For example, the combination of the circadian rhythm-regulating molecule melatonin and the YAP/TAZ inhibitor verteporfin was shown to decrease the expression of PINK1/parkin and then to reduce the capacity for tumorsphere formation and the numbers of CD44+CD24- and CD133+ CSCs in HNSCC [124] (Fig. 4B). However, some reports have revealed that targeting mitophagy-associated lysosomes could interfere with CSC function. As RAB5/7 (the regulators of early and late lysosome biogenesis) and LAMP1/2 (lysosomal/late endosomal marker and lysosomal receptor, respectively) were able to facilitate PINK1/parkin-dependent mitophagy, targeting RAB5/7 or LAMP1/2 could be a potential strategy for eradicating CSCs (Fig. 4B). Mefloquine hydrochloride, a novel RAB5/7 inhibitor, was reported to disrupt CD44v9+CD133+ colon CSCs by inhibiting lysosomal activity and mitophagy and thus could be a promising colorectal CSC-targeting drug [125] (Fig. 4B). In addition, the nanomedicine 188Re-liposome was shown to effectively suppress the expression of stemness markers and reduce the protein levels of lysosome (Lamp-1 and cathepsin-B) and autophagy/mitophagy (LC3B, Atg16L and Beclin-1) markers, which then resulted in decreased tumor growth in xenograft mouse models, lowered CA-125 levels and prolonged ovarian cancer patient survival in a clinical phase I trial [126, 127] (Fig. 4B).

In contrast to inhibitors of mitophagy, many antibiotics that inhibit mitochondrial biogenesis have become a prospective means of depleting CSCs. Azithromycin, doxycycline, and tigecycline, which target the 39S and 28S mitochondrial ribosomes, were shown to inhibit tumorsphere formation, tumorigenicity and self-renewal ability in breast cancer, AML, etc. [128, 129] (Fig. 4A). More specifically, drugs targeting PGC-1α and related genes are another attractive therapeutic choice for CSC elimination. SR-18292, a selective PGC-1α inhibitor, reduces tumorsphere formation, shrinks tumor size, and downregulates the expression of genes involved in stemness maintenance and self-renewal in cholangiocarcinoma [116] (Fig. 4A). XCT790, an ERRα inverse agonist, inhibits the formation of mammospheres in a concentration-dependent manner and decreases the percentage, survival and propagation of CD44+CD24- BCSCs [100] (Fig. 4A).

Induction of mitochondrial ROS production

CSCs maintain mitochondrial ROS at a low level; thus, inducing mitochondrial ROS is considered a novel option for anti-CSC-based therapy. Due to the critical roles of iron in electron transport and ROS generation, an iron chelator is considered a candidate for eradicating CSCs. FDA-approved deferiprone is such an example that dose-dependently decreases the tumorsphere numbers of CSCs and ALDH+ BCSCs by inducing mitochondrial ROS and is now being recommended for phase II clinical trials [130] (Fig. 5B). By inducing ROS-mediated STAT3 degradation or downregulation, the metal chelator KS10076, a novel dihydroxy-DHA derivative 13R, 20-dihydroxydocosahexaenoic acid (13 R, 20-diHDHA), and flower flavor phenylacetaldehyde (PAA) were reported to be potential agents for targeting CSCs by reducing the size of the tumorsphere and tumor formation, decreasing the expression of CSC self-renewal genes, and decreasing the ratios of subpopulations of ALDH+ or CD44highCD24low (CD44+CD24−) CSCs in colon and breast cancers [131,132,133] (Fig. 5B). In addition, by producing intolerable levels of ROS and inactivating AKT signaling, titanium peroxide nanoparticles (TiOxNPs) sensitize radioresistant CSCs to ionizing radiation through the decline of tumorsphere number and CSC marker expression and the reduction of pancreatic CSC self-renewal ability, which then leads to decreases in tumor growth rate and necrosis area while improving mouse survival rate [134] (Fig. 5B).

Conclusions and perspectives

Mitochondria have emerged as a regulatory hub of energy and signaling that can alter the fate of CSCs through the mito-nuclear communication process described above. Therefore, targeting mito-nuclear communication would eradicate CSCs and provide therapeutic benefits for cancer patients. To date, although a few studies that focus on blocking mito-nuclear communication have made great breakthroughs in inhibiting CSC potential, there are some challenges that still need to be overcome in the future.

First, the functional activities of the mitochondria are essential to both normal cells and CSCs. Thus, normal cells may be impaired by therapeutic agents that target mitochondria, which results in some side effects. However, the sensitivity of normal cells and CSCs to drugs may be different. More importantly, real-time monitoring of drug concentrations and metabolite levels would help to improve the specificity of targeting CSC agents and to reduce their toxicity to normal cells.

Second, during the process of tumorigenesis, the metabolic requirements of cancer cells change continuously. As these changes are caused by the input and changes in microenvironment signals, we need to target microenvironment signaling and CSCs together. Normal cells and CSCs may have different responses to changes in microenvironment signaling; thus, it is possible to eradicate CSCs while avoiding damaging normal cells.

Availability of data and materials

Not applicable.

Abbreviations

CSCs:

Cancer stem cells

ROS:

Reactive oxygen species

TSCs:

Tumor stem-like cells

CICs:

Cancer initiating cells

TICs:

Tumor-initiating cells

MeCSC:

Metastatic cancer stem cell

CRCSC:

Chemo-resistant cancer stem cell

GBM:

Glioblastoma

GC:

Gemcitabine and cisplatin

PGE2:

Prostaglandin E2

BCSCs:

Breast cancer stem cells

EMT:

Epithelial-to-mesenchymal transition

MET:

Mesenchymal-to-epithelial transition

ALDH+ :

Aldehyde dehydrogenase positive

HCC:

Hepatocellular carcinoma

TFs:

Transcription factors

GSCs:

Glioblasoma stem cells

ZHX2:

Zinc-finger and homeoboxe 2

mtDNA:

Mitochondrial DNA

acetyl-CoA:

Acetyl-coenzyme A

βHB:

β-Hydroxybutyric acid

SAM:

S-adenosylmethionine

α-KG:

α-Ketoglutarate

AML:

Acute myeloid leukemia

LSC:

Leukemia stem cell

TKI:

Tyrosine kinase inhibitor

DRP1:

Dynamin-related protein 1

MFF:

Mitochondrial fission factor

FIS1:

Fission factor 1

MFN1:

Mitofusin 1

MFN2:

Mitofusin 2

OPA1:

Optic atrophy 1

OXPHOS:

Oxidative phosphorylation

TBX19:

T-box transcription factor 19

SP:

Side population

CDK5:

Cyclin-dependent kinase 5

ERRα:

Estrogen-Related Receptor α

CCCP:

Carbonyl cyanide m-chlorophenylhydrazone

PINK1:

PTEN-induced kinase 1

Mdivi-1:

Mitochondrial fission inhibitor

AMPK:

Adenosine 5'-monophos-phate-activated protein kinase

CRC:

Colorectal cancer

NRF2:

Nuclear factor erythroid-2-related factor 2

NPC:

Nasopharyngeal carcinoma

BTICs:

Brain tumor-initiating cells

LCSCs:

Liver cancer stem cells

13 R:

20-DiHDHA: 13R: 20-dihydroxydocosahexaenoic acid

MAT2A:

Methionine adenosyltransferase 2A

STAT3:

Signal transducer and activator of transcription 3

HNSCC:

Head and neck squamous cell carcinoma

References

  1. Wang SY, Hu QC, Wu T, Xia J, Tao XA, Cheng B. Abnormal lipid synthesis as a therapeutic target for cancer stem cells. World j stem cells. 2022;14(2):146–62.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Swati K, Agrawal K, Raj S, Kumar R, Prakash A, Kumar D. Molecular mechanism(s) of regulations of cancer stem cell in brain cancer propagation. Med Res Rev. 2023;43(3):441–63.

    Article  CAS  PubMed  Google Scholar 

  3. Zhou HM, Zhang JG, Zhang X, Li Q. Targeting cancer stem cells for reversing therapy resistance: mechanism, signaling, and prospective agents. Signal Transduct Target Ther. 2021;6(1):62.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Paul S, Sinha S, Kundu CN. Targeting cancer stem cells in the tumor microenvironment: An emerging role of PARP inhibitors. Pharmacol Res. 2022;184: 106425.

    Article  CAS  PubMed  Google Scholar 

  5. Shrestha S, Banstola A, Jeong JH, Seo JH, Yook S. Targeting Cancer Stem Cells: Therapeutic and di-agnostic strategies by the virtue of nanoparticles. J control release. 2022;348:518–36.

    Article  CAS  PubMed  Google Scholar 

  6. Sher G, Masoodi T, Patil K, et al. Dysregulated FOXM1 signaling in the regulation of cancer stem cells. Semin Cancer Biol. 2022;86:107–21.

    Article  CAS  PubMed  Google Scholar 

  7. Yang F, Cui P, Lu Y, Zhang X. Requirement of the transcription factor YB-1 for maintaining the stemness of cancer stem cells and reverting differentiated cancer cells into cancer stem cells. Stem Cell Res Ther. 2019;10(1):233.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Luo M, Wicha MS. Targeting Cancer Stem Cell Redox Metabolism to Enhance Therapy Responses. Semin Radiat Oncol. 2019;29(1):42–54.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Cao Z, Scandura JM, Inghirami GG, Shido K, Ding BS, Rafii S. Molecular Checkpoint Decisions Made by Subverted Vascular Niche Transform Indolent Tumor Cells into Chemoresistant Cancer Stem Cells. Cancer Cell. 2017;31(1):110–26.

    Article  CAS  PubMed  Google Scholar 

  10. Qian Y, Zhang L, Cai M, et al. The prostate cancer risk variant rs55958994 regulates multiple gene ex-pression through extreme long-range chromatin interaction to control tumor progression. Sci Adv. 2019;5(7):eaaw6710.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang C, Shao L, Pan C, et al. Elevated level of mitochondrial reactive oxygen species via fatty acid β-oxidation in cancer stem cells promotes cancer metastasis by inducing epithelial-mesenchymal transi-tion. Stem Cell Res Ther. 2019;10(1):175.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3(7):730–7.

    Article  CAS  PubMed  Google Scholar 

  13. Vaiopoulos AG, Kostakis ID, Koutsilieris M, Papavassiliou AG. Colorectal cancer stem cells. Stem cells. 2012;30(3):363–71.

    Article  CAS  PubMed  Google Scholar 

  14. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100(7):3983–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Han J, Fujisawa T, Husain SR, Puri RK. Identification and characterization of cancer stem cells in human head and neck squamous cell carcinoma. BMC Cancer. 2014;14:173.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Singh SK, Clarke ID, Terasaki M, et al. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63(18):5821–8.

    CAS  PubMed  Google Scholar 

  17. Baccelli I, Trumpp A. The evolving concept of cancer and metastasis stem cells. J Cell Biol. 2012;198(3):281–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Thankamony AP, Saxena K, Murali R, Jolly MK, Nair R. Cancer Stem Cell Plasticity - A Deadly Deal. Front Mol Biosci. 2020;7:79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yang Q, Zhao S, Shi Z, et al. Chemotherapy-elicited exosomal miR-378a-3p and miR-378d promote breast cancer stemness and chemoresistance via the activation of EZH2/STAT3 signaling. J Exp Clin Cancer Res. 2021;40(1):120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shen M, Dong C, Ruan X, et al. Chemotherapy-Induced Extracellular Vesicle miRNAs Promote Breast Cancer Stemness by Targeting ONECUT2. Cancer Res. 2019;79(14):3608–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liu L, Yang L, Yan W, et al. Chemotherapy Induces Breast Cancer Stemness in Association with Dysregulated Monocytosis. Clin cancer res. 2018;24(10):2370–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Arnold KM, Opdenaker LM, Flynn NJ, Appeah DK, Sims-Mourtada J. Radiation induces an inflam-matory response that results in STAT3-dependent changes in cellular plasticity and radioresistance of breast cancer stem-like cells. Int J Radiat Biol. 2020;96(4):434–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen S, Zhang J, Chen J, et al. RER1 enhances carcinogenesis and stemness of pancreatic cancer under hypoxic environment. J Exp Clin Cancer Res. 2019;38(1):15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Meng Y, Fan XY, Yang LJ, et al. Detachment Activated CyPA/CD147 Induces Cancer Stem Cell Po-tential in Non-stem Breast Cancer Cells. Front Cell Dev Biol. 2020;8: 543856.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mello T, Simeone I, Galli A. Mito-Nuclear Communication in Hepatocellular Carcinoma Metabolic Rewiring. Cells. 2019;8(5):417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li H, Feng Z, He ML. Lipid metabolism alteration contributes to and maintains the properties of cancer stem cells. Theranostics. 2020;10(16):7053–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Qi W, Liang W, Jiang H, Miuyee WM. The function of miRNA in hepatic cancer stem cell. Biomed Res Int. 2013;2013: 358902.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Naz F, Shi M, Sajid S, Yang Z, Yu C. Cancer stem cells: a major culprit of intra-tumor heterogeneity. Am J Cancer Res. 2021;11(12):5782–811.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Kapoor-Narula U, Lenka N. Cancer stem cells and tumor heterogeneity: Deciphering the role in tumor progression and metastasis. Cytokine. 2022;157: 155968.

    Article  CAS  PubMed  Google Scholar 

  30. Zeng X, Liu C, Yao J, et al. Breast cancer stem cells, heterogeneity, targeting therapies and therapeutic implications. Pharmacol Res. 2021;163: 105320.

    Article  CAS  PubMed  Google Scholar 

  31. Olivares-Urbano MA, Griñán-Lisón C, Marchal JA, Núñez MI. CSC Radioresistance: A Therapeutic Challenge to Improve Radiotherapy Effectiveness in Cancer. Cells. 2020;9(7):1651.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sarnella A, D’Avino G, Hill BS, et al. A Novel Inhibitor of Carbonic Anhydrases Prevents Hypox-ia-Induced TNBC Cell Plasticity. Int J Mol Sci. 2020;21(21):8405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ma SKY, Chan ASF, Rubab A, Chan WCW, Chan D. Extracellular Matrix and Cellular Plasticity in Musculoskeletal Development. Front Cell Dev Biol. 2020;8:781.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Paul R, Dorsey JF, Fan Y. Cell plasticity, senescence, and quiescence in cancer stem cells: Biological and therapeutic implications. Pharmacol Ther. 2021;231: 107985.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017;23(10):1124–34.

    Article  CAS  PubMed  Google Scholar 

  36. Kurtova AV, Xiao J, Mo Q, et al. Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance. Nature. 2015;517(7533):209–13.

    Article  CAS  PubMed  Google Scholar 

  37. Agarwal P, Isringhausen S, Li H, et al. Mesenchymal Niche-Specific Expression of Cxcl12 Controls Quiescence of Treatment-Resistant Leukemia Stem Cells. Cell Stem Cell. 2019;24(5):769-84.e766.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Biddle A, Liang X, Gammon L, et al. Cancer stem cells in squamous cell carcinoma switch between two distinct phenotypes that are preferentially migratory or proliferative. Cancer Res. 2011;71(15):5317–26.

    Article  CAS  PubMed  Google Scholar 

  39. Liu S, Cong Y, Wang D, et al. Breast cancer stem cells transition between epithelial and mesenchymal states reflective of their normal counterparts. Stem cell reports. 2014;2(1):78–91.

    Article  CAS  PubMed  Google Scholar 

  40. Ibragimova M, Tsyganov M, Litviakov N. Tumour stem cells in breast cancer. Int J Mol Sci. 2022;23(9):5058.

  41. Das PK, Pillai S, Rakib MA, et al. Plasticity of Cancer Stem Cell: Origin and Role in Disease Progression and Therapy Resistance. Stem cell rev rep. 2020;16(2):397–412.

    Article  PubMed  Google Scholar 

  42. Chen X, Liao R, Li D, Sun J. Induced cancer stem cells generated by radiochemotherapy and their therapeutic implications. Oncotarget. 2017;8(10):17301–12.

    Article  PubMed  Google Scholar 

  43. Iliopoulos D, Hirsch HA, Wang G, Struhl K. Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion. Proc Natl Acad Sci USA. 2011;108(4):1397–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang H, Brown RL, Wei Y, et al. CD44 splice isoform switching determines breast cancer stem cell state. Genes Dev. 2019;33(3–4):166–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ganesh K, Massagué J. Targeting metastatic cancer. Nat Med. 2021;27(1):34–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hermann PC, Huber SL, Herrler T, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1(3):313–23.

    Article  CAS  PubMed  Google Scholar 

  47. Todaro M, Gaggianesi M, Catalano V, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell. 2014;14(3):342–56.

    Article  CAS  PubMed  Google Scholar 

  48. Pang R, Law WL, Chu AC, et al. A subpopulation of CD26+ cancer stem cells with metastatic capacity in human colorectal cancer. Cell Stem Cell. 2010;6(6):603–15.

    Article  CAS  PubMed  Google Scholar 

  49. Liu H, Patel MR, Prescher JA, et al. Cancer stem cells from human breast tumors are involved in spon-taneous metastases in orthotopic mouse models. Proc Natl Acad Sci USA. 2010;107(42):18115–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Dubrovska A, Elliott J, Salamone RJ, et al. CXCR4 expression in prostate cancer progenitor cells. PLoS ONE. 2012;7(2): e31226.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Liu CL, Chen YJ, Fan MH, Liao YJ, Mao TL. Characteristics of CD133-Sustained Chemoresistant Cancer Stem-Like Cells in Human Ovarian Carcinoma. Int J Mol Sci. 2020;21(18):6467.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zheng X, Yu C, Xu M. Linking Tumor Microenvironment to Plasticity of Cancer Stem Cells: Mecha-nisms and Application in Cancer Therapy. Front Oncol. 2021;11: 678333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wainwright EN, Scaffidi P. Epigenetics and Cancer Stem Cells: Unleashing, Hijacking, and Restricting Cellular Plasticity. Trends in cancer. 2017;3(5):372–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Suvà ML, Rheinbay E, Gillespie SM, et al. Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells. Cell. 2014;157(3):580–94.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Chen B, McCuaig-Walton D, Tan S, et al. DYRK1A Negatively Regulates CDK5-SOX2 Pathway and Self-Renewal of Glioblastoma Stem Cells. Int J Mol Sci. 2021;22(8):4011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chen X, Huan H, Liu C, et al. Deacetylation of β-catenin by SIRT1 regulates self-renewal and onco-genesis of liver cancer stem cells. Cancer Lett. 2019;463:1–10.

    Article  CAS  PubMed  Google Scholar 

  57. Lin Q, Wu Z, Yue X, et al. ZHX2 restricts hepatocellular carcinoma by suppressing stem cell-like traits through KDM2A-mediated H3K36 demethylation. EBioMedicine. 2020;53: 102676.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Tatapudy S, Aloisio F, Barber D, Nystul T. Cell fate decisions: emerging roles for metabolic signals and cell morphology. EMBO Rep. 2017;18(12):2105–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Stadhouders R, Filion GJ, Graf T. Transcription factors and 3D genome conformation in cell-fate deci-sions. Nature. 2019;569(7756):345–54.

    Article  CAS  PubMed  Google Scholar 

  60. Genovese I, Vezzani B, Danese A, et al. Mitochondria as the decision makers for cancer cell fate: from signaling pathways to therapeutic strategies. Cell Calcium. 2020;92: 102308.

    Article  CAS  PubMed  Google Scholar 

  61. Filippi MD, Ghaffari S. Mitochondria in the maintenance of hematopoietic stem cells: new perspectives and opportunities. Blood. 2019;133(18):1943–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Chiu HY, Tay EXY, Ong DST, Taneja R. Mitochondrial Dysfunction at the Center of Cancer Therapy. Antioxid Redox Signal. 2020;32(5):309–30.

    Article  CAS  PubMed  Google Scholar 

  63. Bahat A, Gross A. Mitochondrial plasticity in cell fate regulation. J Biol Chem. 2019;294(38):13852–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Mohareer K, Medikonda J, Vadankula GR, Banerjee S. Mycobacterial Control of Host Mitochondria: Bioenergetic and Metabolic Changes Shaping Cell Fate and Infection Outcome. Front Cell Infect Microbiol. 2020;10:457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Skoda J, Borankova K, Jansson PJ, Huang ML, Veselska R, Richardson DR. Pharmacological targeting of mitochondria in cancer stem cells: An ancient organelle at the crossroad of novel anti-cancer therapies. Pharmacol Res. 2019;139:298–313.

    Article  CAS  PubMed  Google Scholar 

  66. Vasconcelos ESJ, Simão D, Terrasso AP, et al. Unveiling dynamic metabolic signatures in human in-duced pluripotent and neural stem cells. PLoS Comput Biol. 2020;16(4): e1007780.

    Article  Google Scholar 

  67. Walker BR, Moraes CT. Nuclear-mitochondrial interactions. Biomolecules. 2022;12(3):427.

  68. Ludikhuize MC, Meerlo M, Gallego MP, et al. Mitochondria Define Intestinal Stem Cell Differentiation Downstream of a FOXO/Notch Axis. Cell Metab. 2020;32(5):889-900.e887.

    Article  CAS  PubMed  Google Scholar 

  69. English J, Son JM, Cardamone MD, Lee C, Perissi V. Decoding the rosetta stone of mitonuclear com-munication. Pharmacol Res. 2020;161: 105161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Mottis A, Herzig S, Auwerx J. Mitocellular communication: Shaping health and disease. Science. 2019;366(6467):827–32.

    Article  CAS  PubMed  Google Scholar 

  71. Quiros PM, Mottis A, Auwerx J. Mitonuclear communication in homeostasis and stress. Nat Rev Mol Cell Biol. 2016;17(4):213–26.

    Article  CAS  PubMed  Google Scholar 

  72. Chakrabarty RP, Chandel NS. Mitochondria as Signaling Organelles Control Mammalian Stem Cell Fate. Cell Stem Cell. 2021;28(3):394–408.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Egan G, Khan DH, Lee JB, Mirali S, Zhang L, Schimmer AD. Mitochondrial and Metabolic Pathways Regulate Nuclear Gene Expression to Control Differentiation, Stem Cell Function, and Immune Re-sponse in Leukemia. Cancer Discov. 2021;11(5):1052–66.

    Article  CAS  PubMed  Google Scholar 

  74. Zhang Q, Li W. Correlation between amino acid metabolism and self-renewal of cancer stem cells: Perspectives in cancer therapy. World j stem cells. 2022;14(4):267–86.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Menendez JA. Metabolic control of cancer cell stemness: Lessons from iPS cells. Cell cycle. 2015;14(24):3801–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Chen F. Linking metabolism to epigenetics in stem cells and cancer stem cells. Semin Cancer Biol. 2019;57:iii–v.

    Article  CAS  PubMed  Google Scholar 

  77. Martínez-Reyes I, Chandel NS. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 2020;11(1):102.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Sun L, Zhang H, Gao P. Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell. 2022;13(12):877–919.

    Article  CAS  PubMed  Google Scholar 

  79. Yang D, Peng M, Hou Y, et al. Oxidized ATM promotes breast cancer stem cell enrichment through energy metabolism reprogram-mediated acetyl-CoA accumulation. Cell Death Dis. 2020;11(7):508.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zhang H, Chang Z, Qin LN, et al. MTA2 triggered R-loop trans-regulates BDH1-mediated β-hydroxybutyrylation and potentiates propagation of hepatocellular carcinoma stem cells. Signal Transduct Target Ther. 2021;6(1):135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Singh RP, Jeyaraju DV, Voisin V, et al. Disrupting Mitochondrial Copper Distribution Inhibits Leukemic Stem Cell Self-Renewal. Cell Stem Cell. 2020;26(6):926-37.e910.

    Article  CAS  PubMed  Google Scholar 

  82. Miwa S, Kashyap S, Chini E, von Zglinicki T. Mitochondrial dysfunction in cell senescence and aging. J Clin Investig. 2022;132(13): e158447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Du F, Yu Q, Yan S, et al. PINK1 signalling rescues amyloid pathology and mitochondrial dysfunction in Alzheimer’s disease. Brain. 2017;140(12):3233–51.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Sessions DT, Kashatus DF. Mitochondrial dynamics in cancer stem cells. Cell mol life sci. 2021;78(8):3803–16.

    Article  CAS  PubMed  Google Scholar 

  85. Lee JH, Mourkioti F. Fission impossible: Mitochondrial dynamics direct muscle stem cell fates. Cell Stem Cell. 2022;29(9):1287–9.

    Article  CAS  PubMed  Google Scholar 

  86. Courtois S, de Luxán-Delgado B, Penin-Peyta L, et al. Inhibition of Mitochondrial Dynamics Preferen-tially Targets Pancreatic Cancer Cells with Enhanced Tumorigenic and Invasive Potential. Cancers. 2021;13(4):698.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. van der Bliek AM, Shen Q, Kawajiri S. Mechanisms of mitochondrial fission and fusion. Cold Spring Harb Perspect Biol. 2013;5(6):a011072.

  88. Wang S, Tan J, Miao Y, Zhang Q. Mitochondrial Dynamics, Mitophagy, and Mitochondria-Endoplasmic Reticulum Contact Sites Crosstalk Under Hypoxia. Front Cell Dev Biol. 2022;10:848214.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Bonnay F, Veloso A, Steinmann V, et al. Oxidative Metabolism Drives Immortalization of Neural Stem Cells during Tumorigenesis. Cell. 2020;182(6):1490-1507 e1419.

    Article  CAS  PubMed  Google Scholar 

  90. Chen H, Chan DC. Mitochondrial Dynamics in Regulating the Unique Phenotypes of Cancer and Stem Cells. Cell Metab. 2017;26(1):39–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Tang M, Yang M, Wu G, et al. Epigenetic Induction of Mitochondrial Fission Is Required for Mainte-nance of Liver Cancer-Initiating Cells. Cancer Res. 2021;81(14):3835–48.

    Article  CAS  PubMed  Google Scholar 

  92. Xie Q, Wu Q, Horbinski CM, et al. Mitochondrial control by DRP1 in brain tumor initiating cells. Nat Neurosci. 2015;18(4):501–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Sánchez-Alvarez R, De Francesco EM, Fiorillo M, Sotgia F, Lisanti MP. Mitochondrial Fission Factor (MFF) Inhibits Mitochondrial Metabolism and Reduces Breast Cancer Stem Cell (CSC) Activity. Front oncol. 2020;10:1776.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Liu L, Li Y, Wang J, et al. Mitophagy receptor FUNDC1 is regulated by PGC-1α/NRF1 to fine tune mitochondrial homeostasis. EMBO Rep. 2021;22(3): e50629.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Roberts FL, Markby GR. New Insights into Molecular Mechanisms Mediating Adaptation to Exercise; A Review Focusing on Mitochondrial Biogenesis, Mitochondrial Function, Mitophagy and Autophagy. Cells. 2021;10(10):2639.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Praharaj PP, Panigrahi DP, Bhol CS, et al. Mitochondrial rewiring through mitophagy and mitochondrial biogenesis in cancer stem cells: A potential target for anti-CSC cancer therapy. Cancer Lett. 2021;498:217–28.

    Article  CAS  PubMed  Google Scholar 

  97. Poole LP, Macleod KF. Mitophagy in tumorigenesis and metastasis. Cell mol life sci. 2021;78(8):3817–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ma K, Chen G, Li W, Kepp O, Zhu Y, Chen Q. Mitophagy, Mitochondrial Homeostasis, and Cell Fate. Front Cell Dev Biol. 2020;8:467.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Sancho P, Burgos-Ramos E, Tavera A, et al. MYC/PGC-1α Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells. Cell Metab. 2015;22(4):590–605.

    Article  CAS  PubMed  Google Scholar 

  100. De Luca A, Fiorillo M, Peiris-Pagès M, et al. Mitochondrial biogenesis is required for the anchor-age-independent survival and propagation of stem-like cancer cells. Oncotarget. 2015;6(17):14777–95.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Bruns I, Sauer B, Burger MC, et al. Disruption of peroxisome proliferator-activated receptor γ coactivator (PGC)-1α reverts key features of the neoplastic phenotype of glioma cells. J Biol Chem. 2019;294(9):3037–50.

    Article  CAS  PubMed  Google Scholar 

  102. Whelan KA, Chandramouleeswaran PM, Tanaka K, et al. Autophagy supports generation of cells with high CD44 expression via modulation of oxidative stress and Parkin-mediated mitochondrial clearance. Oncogene. 2017;36(34):4843–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Pei S, Minhajuddin M, Adane B, et al. AMPK/FIS1-Mediated Mitophagy Is Required for Self-Renewal of Human AML Stem Cells. Cell Stem Cell. 2018;23(1):86-100.e106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Liu K, Lee J, Kim JY, et al. Mitophagy Controls the Activities of Tumor Suppressor p53 to Regulate Hepatic Cancer Stem Cells. Mol Cell. 2017;68(2):281-92.e285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Jovanović M, Podolski-Renić A, Krasavin M, Pešić M. The Role of the Thioredoxin Detoxification System in Cancer Progression and Resistance. Front Mol Biosci. 2022;9: 883297.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Glasauer A, Chandel NSROS. Curr Biol. 2013;23(3):R100–2.

    Article  CAS  PubMed  Google Scholar 

  107. Myant KB, Cammareri P, McGhee EJ, et al. ROS production and NF-κB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell. 2013;12(6):761–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhong G, Qin S, Townsend D, Schulte BA, Tew KD, Wang GY. Oxidative stress induces senescence in breast cancer stem cells. Biochem Biophys Res Commun. 2019;514(4):1204–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Culp-Hill R, D’Alessandro A, Pietras EM. Extinguishing the Embers: Targeting AML Metabolism. Trends Mol Med. 2021;27(4):332–44.

    Article  CAS  PubMed  Google Scholar 

  110. Yang Q, Li K, Huang X, et al. lncRNA SLC7A11-AS1 Promotes Chemoresistance by Blocking SCF(β-TRCP)-Mediated Degradation of NRF2 in Pancreatic Cancer. Mol Ther Nucleic Acids. 2020;19:974–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Huang H, Zhang S, Li Y, et al. Suppression of mitochondrial ROS by prohibitin drives glioblastoma progression and therapeutic resistance. Nat Commun. 2021;12(1):3720.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Civenni G, Bosotti R, Timpanaro A, et al. Epigenetic Control of Mitochondrial Fission Enables Self-Renewal of Stem-like Tumor Cells in Human Prostate Cancer. Cell Metab. 2019;30(2):303-18.e306.

    Article  CAS  PubMed  Google Scholar 

  113. Liu D, Sun Z, Ye T, et al. The mitochondrial fission factor FIS1 promotes stemness of human lung cancer stem cells via mitophagy. FEBS Open Bio. 2021;11(7):1997–2007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Naik PP, Mukhopadhyay S, Panda PK, et al. Autophagy regulates cisplatin-induced stemness and chemoresistance via the upregulation of CD44, ABCB1 and ADAM17 in oral squamous cell carcinoma. Cell Prolif. 2018;51(1): e12411.

    Article  PubMed  Google Scholar 

  115. Chen YY, Wang WH, Che L, et al. BNIP3L-Dependent Mitophagy Promotes HBx-Induced Cancer Stemness of Hepatocellular Carcinoma Cells via Glycolysis Metabolism Reprogramming. Cancers. 2020;12(3):655.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Raggi C, Taddei ML, Sacco E, et al. Mitochondrial oxidative metabolism contributes to a cancer stem cell phenotype in cholangiocarcinoma. J Hepatol. 2021;74(6):1373–85.

    Article  CAS  PubMed  Google Scholar 

  117. Wu X, Yeerna H, Goto Y, et al. Metformin Inhibits Progression of Head and Neck Squamous Cell Car-cinoma by Acting Directly on Carcinoma-Initiating Cells. Cancer Res. 2019;79(17):4360–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Wang Z, Yip LY, Lee JHJ, et al. Methionine is a metabolic dependency of tumor-initiating cells. Nat Med. 2019;25(5):825–37.

    Article  CAS  PubMed  Google Scholar 

  119. Strekalova E, Malin D, Weisenhorn EMM, et al. S-adenosylmethionine biosynthesis is a targetable metabolic vulnerability of cancer stem cells. Breast Cancer Res Treat. 2019;175(1):39–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Zhou TJ, Zhang SL, He CY, et al. Downregulation of mitochondrial cyclooxygenase-2 inhibits the stemness of nasopharyngeal carcinoma by decreasing the activity of dynamin-related protein 1. Theranostics. 2017;7(5):1389–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Prasad P, Ghosh S, Roy SS. Glutamine deficiency promotes stemness and chemoresistance in tumor cells through DRP1-induced mitochondrial fragmentation. Cell mol life sci. 2021;78(10):4821–45.

    Article  CAS  PubMed  Google Scholar 

  122. Ooki A, Del Carmen Rodriguez Pena M, Marchionni L, et al. YAP1 and COX2 Coordinately Regulate Urothelial Cancer Stem-like Cells. Cancer Res. 2018;78(1):168–81.

    Article  CAS  PubMed  Google Scholar 

  123. Wu M, Guan J, Li C, et al. Aberrantly activated Cox-2 and Wnt signaling interact to maintain cancer stem cells in glioblastoma. Oncotarget. 2017;8(47):82217–30.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Shin YY, Seo Y, Oh SJ, et al. Melatonin and verteporfin synergistically suppress the growth and stemness of head and neck squamous cell carcinoma through the regulation of mitochondrial dynamics. J Pineal Res. 2022;72(1): e12779.

    Article  CAS  PubMed  Google Scholar 

  125. Takeda M, Koseki J, Takahashi H, et al. Disruption of Endolysosomal RAB5/7 Efficiently Eliminates Colorectal Cancer Stem Cells. Cancer Res. 2019;79(7):1426–37.

    Article  CAS  Google Scholar 

  126. Chang CM, Lan KL, Huang WS, et al. (188)Re-Liposome Can Induce Mitochondrial Autophagy and Reverse Drug Resistance for Ovarian Cancer: From Bench Evidence to Preliminary Clinical Proof-of-Concept. Int J Mol Sci. 2017;18(5):903.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Shen YA, Lan KL, Chang CH, et al. Intraperitoneal (188)Re-Liposome delivery switches ovarian cancer metabolism from glycolysis to oxidative phosphorylation and effectively controls ovarian tumour growth in mice. Radiother oncol. 2016;119(2):282–90.

    Article  CAS  PubMed  Google Scholar 

  128. Lamb R, Ozsvari B, Lisanti CL, et al. Antibiotics that target mitochondria effectively eradicate cancer stem cells, across multiple tumor types: treating cancer like an infectious disease. Oncotarget. 2015;6(7):4569–84.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Skrtić M, Sriskanthadevan S, Jhas B, et al. Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia. Cancer Cell. 2011;20(5):674–88.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Fiorillo M, Tóth F, Brindisi M, Sotgia F, Lisanti MP. Deferiprone (DFP) Targets Cancer Stem Cell (CSC) Propagation by Inhibiting Mitochondrial Metabolism and Inducing ROS Production. Cells. 2020;9(6):1529.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Kim J, Park A, Hwang J, et al. KS10076, a chelator for redox-active metal ions, induces ROS-mediated STAT3 degradation in autophagic cell death and eliminates ALDH1(+) stem cells. Cell Rep. 2022;40(3): 111077.

    Article  CAS  PubMed  Google Scholar 

  132. Wang L, Choi HS, Lee B, Choi JH, Jang YS, Seo JW. 13R,20-Dihydroxydocosahexaenoic Acid, a Novel Dihydroxy- DHA Derivative, Inhibits Breast Cancer Stemness through Regulation of the Stat3/IL-6 Signaling Pathway by Inducing ROS Production. Antioxidants. 2021;10(3):457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Choi HS, Kim SL, Kim JH, Ko YC, Lee DS. Plant Volatile, Phenylacetaldehyde Targets Breast Cancer Stem Cell by Induction of ROS and Regulation of Stat3 Signal. Antioxidants. 2020;9(11):1119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Salah M, Akasaka H, Shimizu Y, et al. Reactive oxygen species-inducing titanium peroxide nanoparticles as promising radiosensitizers for eliminating pancreatic cancer stem cells. J Exp Clin Cancer Res. 2022;41(1):146.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank American Journal Experts for their professional editing and proofreading (W8ZY69GC).

Funding

This work was supported by the National Natural Science Foundation of China (NSFC) [grant No. 31571469 & 81872349]; Key Project Program of the State Key Laboratory of Cancer Biology, Fourth Military Medical University [grant No. CBSKL2022ZZ09]; General Project of Natural Science Basic Research Program of Shaanxi [grant No. 2022JQ-874]; Science and Technology Innovation Team of Tumor Development and Antitumor Drug Development of Yan'an, and the Special Support Plan 'Regional Development Talents' Project of Shaanxi.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, L.L. and Y.S.; writing-original draft preparation, M.F.; writing-review and editing, J.Z. and L.L.; supervision, L.L.; funding acquisition, L.L. and Y.S. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jumei Zhao or Ling Li.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors have given their consent for publication.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fan, M., Shi, Y., Zhao, J. et al. Cancer stem cell fate determination: mito-nuclear communication. Cell Commun Signal 21, 159 (2023). https://doi.org/10.1186/s12964-023-01160-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-023-01160-x

Keywords