Next Article in Journal
Exclusion Zone Phenomena in Water—A Critical Review of Experimental Findings and Theories
Next Article in Special Issue
MicroRNAs and Apoptosis in Colorectal Cancer
Previous Article in Journal
Combinatory Treatment with miR-7-5p and Drug-Loaded Cubosomes Effectively Impairs Cancer Cells
Previous Article in Special Issue
Mechanisms of Apoptosis Resistance to NK Cell-Mediated Cytotoxicity in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cellular Mechanisms of Circulating Tumor Cells During Breast Cancer Metastasis

1
Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, P.O. Box 870311, Tuscaloosa, AL 35487, USA
2
Department of Chemical and Biological Engineering, College of Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(14), 5040; https://doi.org/10.3390/ijms21145040
Submission received: 30 June 2020 / Revised: 13 July 2020 / Accepted: 14 July 2020 / Published: 17 July 2020
(This article belongs to the Special Issue Survival Pathways Involved in Resistance to Apoptosis in Cancer)

Abstract

:
Circulating tumor cells (CTCs) are cancer cells that detach from the primary site and travel in the blood stream. A higher number of CTCs increases the risk of breast cancer metastasis, and it is inversely associated with the survival rates of patients with breast cancer. Although the numbers of CTCs are generally low and the majority of CTCs die in circulation, the survival of a few CTCs can seed the development of a tumor at a secondary location. An increasing number of studies demonstrate that CTCs undergo modification in response to the dynamic biophysical environment in the blood due in part to fluid shear stress. Fluid shear stress generates reactive oxygen species (ROS), triggers redox-sensitive cell signaling, and alters the function of intracellular organelles. In particular, the mitochondrion is an important target organelle in determining the metastatic phenotype of CTCs. In healthy cells, mitochondria produce adenosine triphosphate (ATP) via oxidative phosphorylation in the electron transport chain, and during oxidative phosphorylation, they produce physiological levels of ROS. Mitochondria also govern death mechanisms such as apoptosis and mitochondrial permeability transition pore opening to, in order eliminate unwanted or damaged cells. However, in cancer cells, mitochondria are dysregulated, causing aberrant energy metabolism, redox homeostasis, and cell death pathways that may favor cancer invasiveness. In this review, we discuss the influence of fluid shear stress on CTCs with an emphasis on breast cancer pathology, then discuss alterations of cellular mechanisms that may increase the metastatic potentials of CTCs.

1. Breast Cancer and Circulating Tumor Cells

Breast cancer is the most common cancer in women. According to Cancer Facts & Figures 2020, published by the American Cancer Society, an estimated 279,100 new cases of invasive breast cancer and 42,690 deaths will occur in 2020 [1]. Both genetic and lifestyle factors contribute to the development of breast cancer. Mutations of tumor suppressor genes such as BRCA1 and BRCA2 are strongly associated with hereditary breast cancer, and abnormalities of other genes such as CHECK2, ATM, PALB2, PTEN, and PT53 also increase risk [2,3]. Lifestyle factors such as obesity, hormone treatment, and a high-fat diet are positively correlated with breast cancer risk, whereas physical activity and a diet rich in vitamins, minerals, and phytochemicals may reduce the risk of breast cancer [4,5]. The breast cancer mortality rate was 33.2 per 100,000 in 1989, but this has declined to 19.8 since 2017 due in part to increased screening and advancements in diagnostic and therapeutic technologies [1]. Currently, the 5-year survival rate for those with non-metastatic breast cancer is 99%, whereas this declines steeply for metastatic breast cancer to just 27% [1]. Therefore, localized breast cancer is considered more manageable, and strategies to prevent metastasis are vital to reducing breast cancer mortality.
Metastatic progression is a primary cause of breast cancer-associated death [6,7]. Breast cancer cells may spread to the bone, lung, liver, and brain. However, metastatic patterns are not uniform and can vary by type of breast cancer. Especially, the distributions of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) influence the metastatic potential of breast cancer. Therefore, elucidating receptor-mediated signaling and differential cellular outcomes is crucial to understanding the molecular mechanisms of breast cancer cell metastatic behavior. To progress to clinically detectable metastasis, cancer cells must undergo a metastasis cascade, as follows: primary tumor formation, local invasion, intravasation into blood or lymph, survival during circulation, implantation at a distant organ site, initial survival in a foreign microenvironment, and finally metastatic colonization [8,9]. Each step of the metastasis cascade acts as a biological barrier; thus, the majority of cells die before progressing to metastasis. In particular, when cancer cells detach from the primary site and enter the bloodstream as circulating tumor cells (CTCs), they are challenged with anoikis, a type of apoptosis caused by loss of attachment to the extracellular matrix. However, a few CTCs survive this challenge and, when coupled with a favorable microenvironment, develop into metastasis [10,11]. Although cutoffs can vary by type of tumor, five or more CTCs in a 7.5 mL blood sample is considered CTC positive in breast cancer [12,13]. An increasing number of studies have emphasized the significance of CTCs in mediating breast cancer metastasis. The presence of CTCs increases the risk of metastasis, and higher numbers of CTCs are inversely associated with progression-free survival and overall survival in patients with breast cancer [12,14,15]. CTCs have been suggested as a prognostic tool for monitoring metastasis or the efficacy of chemotherapy [16,17,18,19]. Studies have shown that potential diagnostic biomarkers representing stemness [20], immunogenic CTC [18], and signaling molecules that promote breast cancer metastasis [19] are found in CTCs. The mutation and expression levels of breast cancer-associated genes such as BRCA 1/2 are also detectable by liquid biopsy [15,21].

2. In Vitro Models of Circulating Tumor Cells for Studying Metastasis

Due to our current inability to observe and study metastasizing cells in vivo, it is necessary to engineer and model the cells’ dynamic environment in vitro. These models allow for the examination and analysis of the mechanobiology of the cells as they experience physiologically relevant stresses and the consequences thereof. The most significant aspect of this dynamic environment is the fluid shear stress (FSS) that CTCs experience in the blood stream. Here, cells encounter a moving, heterogeneous environment that is unfriendly to most cells, resulting in either their destruction or dormancy [22,23]. Conversely, CTCs that survive such stress become especially endowed with high metastatic potential. Physiological fluid flow is typically identified as blood, lymphoid, and interstitial flow [24], with blood and interstitial flow affecting CTCs the most. Physiological FSS ranges from 1 to 30 dyn/cm2, depending on the location (capillaries, veins, or arteries) [25]. As such, models for applying FSS need to achieve these levels of shear stress, while also considering the diameter of the simulated vessels. Moreover, the type of FSS induced is also relevant to the study target, as laminar shear stress and oscillatory shear stress do not produce the same effects [24,26].
Currently, there are several strategies for applying FSS to cancer cells, several of which are summarized in Table 1. Each of these models has been utilized to gain insight into various aspects of the mechanobiology of the CTCs, including their stemness [27], the generation of reactive oxygen species [28], drug resistance [29,30], promotion of the epithelial-mesenchymal transition (EMT) [31], fluctuation in hormone receptor expression [32], and facilitation of apoptosis [33], to name a few.
The syringe pump model is the simplest yet most robust CTC model. This device allows for the application of shear rates ranging from low (1 dyn/cm2) to high (>60 dyn/cm2), as per physiological standards [27,30]. The syringe pump is simple to use and stands on its own as a model for CTCs, but can also be incorporated into other models, such as perfusion and microfluidic devices for inducing flow. A primary objection to using the syringe pump exclusively is that it is a single-pass system that does not maintain a high residence time. For observations of the consequences of prolonged exposure to FSS, a peristaltic pump system would be more appropriate. This would allow a circulatory system for cells to experience shear stress over a wider range of exposure time [28,29,34,35]. Furthermore, the pulsatile flow resulting from the peristaltic pumps more closely mimics the blood flow generated by the cardiovascular system, so its physiological relevance is increased as compared to the previously discussed apparatus. Cone and plate viscometers calculate FSS in relation to torque and angular viscosity, as opposed to flow rate, as syringes and peristaltic pumps do. These constructs permit a larger number of cells to be exposed to FSS in a single experiment as the entire culture plate can be utilized, similarly to the rotational shakers [31,32,33]. These devices introduce a uniform application of FSS and therefore provide for more whole-population analysis, since all the cells undergo the same, proportional shear experience. Perfusion and microfluidic devices are smaller-scale FSS applications, but they allow for the introduction of greater complexity into the exposure to stress [36,37,38]. Microfluidic devices in particular can be designed with more complex tortuous flow paths for cells. The significance of this is reflected in some studies of complex extracellular matrices, as the layout of the cells’ path dictates their progression through the course [39]. Microfluidic devices have also been further developed as means of detecting CTC in patient blood samples, which has to the potential to carry clinical significance [40]. Finally, computational modeling allows the investigator to build the desired biological environment and introduce known variables and responses, in order to observe what is not actually visible on the clinical stage. Notably, the designs of the perfusion and microfluidic devices as well as the computational simulations create the opportunity for single-cell analysis, granting deeper insight into cell behaviors and the cues to which it responds. With these advances, it is conceivable to identify what makes CTCs capable of surviving the heterogeneous environment and progress through the metastatic cascade, rather than entering dormancy or being destroyed.

3. Oxidative Stress and Mitochondrial Dysfunction

In recent decades, increasing numbers of studies have suggested an association between of FSS-induced oxidative stress with CTC pathology. Loss of extracellular matrix in non-tumorigenic human mammary epithelial MCF10A cells is shown to increase ROS production [42]. Similarly, breast cancer cells treated with FSS increased the 2′, 7′-dichlorodihydrofluorescein (DCF) positive signal indicating accumulation of hydrogen peroxide [28,29,31,43]. FSS-induced ROS accumulation contributes to increased invasiveness of breast cancer cells by promoting migration and EMT, a process by which epithelial cells lose polarity but gain the motile and invasive characteristics of mesenchymal cells [31]. Oxidative stress enhances the transition to the HER2 negative phenotype in CTCs [44]. In addition, other non-cancer cells and extracellular vesicles found in the bloodstream also incorporate with CTC and enhance metastatic potentials. Sprouse et al. showed that the formation of clusters of non-tumor, myeloid-derived suppressor cells (MDSCs) and peripheral mononuclear cells (PMNs) enhances ROS production in CTCs and promotes ROS-mediated mitogenic signaling in breast cancer [45]. Fu et al. demonstrated a positive correlation between ROS and SMAD3, essential during cell adhesion via the mediation of transforming growth factor beta (TGF-β) signaling [46]. The authors also showed that primary tumor-derived exosomes (PTDEs) increase ROS production, and PTDEs directly communicate with CTCs, enhancing their survival and adhesion [46]. FSS-treated breast cancer cells isolated from stage III breast cancer patients showed an upregulation of antioxidant enzyme genes such as superoxide dismutase, catalase, and glutathione peroxidase [31], which potentially support cancer cell survival. Circulating tumor cells isolated from patients with leukemia showed a high expression of nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that binds to genes containing antioxidant response element (ARE) upon oxidative stress, is associated with drug resistance [47]. FSS-induced ROS may signal a change the intracellular antioxidant profiles of CTCs.
The mitochondrion is the central organelle that consumes oxygen to produce adenosine triphosphate (ATP) via oxidative phosphorylation. Electrons produced during metabolic processes such as the TCA cycle and fatty acid oxidation are processed by complex I, complex II, complex III, and complex IV in the electron transport chain. The electron transport creates an electrochemical gradient that forces ATP production via the F1Fo ATP synthase. Although the majority of oxygen is reduced to water by complex IV, 1–3% of the oxygen leaked from complex I and III is responsible for producing ROS such as superoxide [48,49]. Inefficient operation of the electron transport chain in the mitochondria increases ROS production, contributing to pathological processes. Accumulation of ROS also damages mitochondria, altering energy metabolism, mitochondrial DNA expression, and mitochondria-dependent death mechanisms. Tumorigenic breast cancer cells showed significantly high baseline ROS levels, associated with mitochondrial fragmentation and mitochondrial membrane potential loss, compared to the non-tumorigenic control [50]. Mitochondrial dysfunctions, including the fragmentation of mitochondrial DNA and the alteration of mitochondrial apoptosis, are reported for CTCs of various cancers [51,52]. Mitochondrially produced free radicals such as the superoxide anion are also greater in FSS-induced breast cancer cells [29]. Cancer cells exhibit altered metabolic characteristics with decreased reliance on mitochondrial energy metabolism, referred to as the Warburg effect (decreased ATP production via oxidative phosphorylation, but increased ATP production via glycolysis [53]). Glycolysis is an alternative source of ATP when mitochondrial function is devoted to other cellular functions such as the biosynthesis of macromolecules for proliferation; thus, high glycolytic activity supports rapid cancer cell proliferation.

4. Cell Signaling Pathways in Circulating Tumor Cells

ROS play dual roles regulating both the death and survival of cancer cells [54,55]. A surge of ROS potentiates breast cancer cell death during treatment with chemotherapeutics such as cisplatin and doxorubicin [56,57]. FSS can enhance the effects of ROS-generating drugs promoting the apoptosis of breast cancer cells [58], and the application of mitochondrial antioxidants reverses the pro-death effect of chemotherapy drugs [29]. However, moderate levels of ROS also contribute to cancer cell growth and survival by manipulating cell signaling pathways.
The extracellular signaling-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway regulates the cell cycle and proliferation of cancer cells. ERK/MAPK is one of the most studied pathways in FSS-associated cancer pathology [28,31], and it is a highly activated pathway in triple negative breast cancer [59,60]. ERK is activated by ErbB family receptors including the epidermal growth factor receptor (EGFR/HER1) and other HERs. HER2, a key receptor responsible for over 20% of breast cancer [61], undergoes heterodimerization, and amplifies ERK signaling [62]. EGFR and HER2 are types of receptor tyrosine kinases, and phosphorylation of tyrosine residues recruits growth factor receptor-bound protein 2 (GRB2) and Son of Sevenless (SOS). SOS is a guanine nucleotide exchange factor that activates Ras, a small GTPase found in the membrane. GTP-bound activated Ras initiates the activation of Raf, a serine/threonine-specific protein kinase, and its downstream kinase cascades including mitogen-activated protein kinase kinase (MEK/MAPKK) followed by ERK. ERK phosphorylates cytoplasmic targets including ribosomal protein S6 kinase (RSK) and Bcl-2 family proteins such as Bcl-2, Bcl-xL and Mcl-1 [63]: RSK converts non-transformed epithelial cells to motile, mesenchymal carcinoma cells [64], and the application of an RSK inhibitor or siRNA blocks breast cancer cell initiation and proliferation [65,66]. Bcl-2, Bcl-xL, and Mcl-1 are pro-survival proteins that inhibit the formation of pro-death oligomers on the mitochondrial membrane. In addition to regulating cytoplasmic proteins, phosphorylated ERK translocated into the nucleus and activates transcription factors such as Elk-1, c-Jun, c-Fos, c-Myc, Mnk, and cAMP response element binding protein (CREB) which control metabolism, cell cycle, proliferation, and aggressiveness [67,68].
Ma et al. showed that MDA-MB-231 cells exposed to 5, 15, and 30 dyn/cm2 FSS increased the production of hydrogen peroxide and the migration of cancer cells [28]. Higher FSS increased levels of ERK1/2 phosphorylation [28]. The authors further demonstrated that the application of antioxidants such as N-acetyl-cysteine and propyl gallate, or MEK inhibitor U0126 reversed FSS-mediated breast cancer cell migration [28]. Ejaeidi et al. showed that CTCs derived from breast cancer patients increased protein levels of ERK, Akt, and survivin [69]. Choi et al. showed that MDA-MB-231 cells exposed to 5 dyn/cm2 bi-directional, oscillating shear stress increased stemness factors (e.g., Nanog, Oct4B, and Sox2) and EMT markers (e.g., cadherin, Twist, and Snail1) [31]. Breast cancer cells grown on an orbital shaker at 60 rpm, equivalent to 4.5 dyn/cm2, increased the production of ROS and reactive nitrogen species [31] upregulating ROS-responsive genes such as Sod1, Cat, Nox1, and Gpx1 and NO-responsive genes such as Nos1 and Nos2 [31]. Co-treatment with FSS and a ROS scavenger, N-acetylcysteine, inhibited ROS-induced transcriptional change indicating an association between cancer cell redox status and FSS-mediated EMT [31]. Notably, Choi et al. showed the inhibitory effects of shear stress on ERK pathways, including decreased ERK phosphorylation [31]. c-Jun NH2-terminal kinase (JNK) is a downstream kinase activated by the ERK pathway. Although the ERK pathway did not lead to the activation of JNK in this study [31], Takabe et al. showed that FSS activates JNK, promoting mitochondrial ROS production [70]. The ERK pathway is an important player during FSS-induced alteration of survival, EMT, and stemness in breast cancer cells. However, ERK activation or inhibition may vary by cell type, duration, or intensity of FSS.
Phosphoinositide 3-kinase (PI3K) is a family of p85/p110 heterodimeric lipid kinases that regulates cancer cell proliferation, growth, motility, and survival via the activation of mammalian target of rapamycin (mTOR) [71]. Class IA PI3Ks contain regulatory (p85) and catalytic subunits (p110), continuing the signaling transduction activated by cell membrane receptors, including receptor tyrosine kinases and G-protein coupled receptors [72,73,74,75]. PI3K phosphorylates phosphatidylinositol 4,5 bisphosphate (PIP2) at the 3 position producing phosphatidylinositol-3,4,5-triphosphate (PIP3) in the membrane, whereas phosphatase and tension homolog (PTEN) dephosphorylates PIP3 inhibiting the PI3K/Akt pathway. PIP3 recruits phosphoinositide-dependent kinase 1 (PDK1), and PDK1 activates Akt, also called protein kinase B (PKB), via phosphorylation at the Thr 308 residue [76,77]. Akt is also phosphorylated by rapamycin complex 2 (mTORC2) at Ser 473 [78]. Activated Akt phosphorylates multiple downstream targets including tuberous sclerosis protein 2 (TSC2). TSC2 forms a heterodimer with TSC1 to act as a tumor suppressor, and TSC2 activates Rheb, a GTP-binding protein. However, Akt-mediated phosphorylation inhibits the formation of the TSC1-TSC2 complex causing the accumulation of GTP-bound Rheb. The Rheb-GTP activates mTORC1, which inhibits 4E-BP1 and activates S6K1/2, regulators of translation [77]. The targets of Akt also include glycogen synthase kinase 3β (GSK3β), NF-κB, and Bad, regulating cancer cell growth, death, and energy metabolism [79,80].
Activation of the PI3K/Akt pathways occurs in FSS-exposed cancer cells [38,81,82]. ROS generated during FSS [28,29,31,43] may signal activation of the PI3K/Akt pathway in breast cancer [83,84,85]. Application of a low level of FSS (1.8 dyn/cm2) to MDA-MB-231 breast cancer cells increased phosphorylation of the regulatory subunit of PI3K, p85, followed by phosphorylation of the Ser 473 site of Akt [38]. Activation of PI3K/Akt increased the protein levels of membrane type 1-matrix metalloproteinase (MT1-MMP) [38], a key player during extracellular matrix degradation [86], allowing cancer cell migration. Treatment with LY2294002, a PI3K inhibitor, MK-2206, an Akt inhibitor, or rapamycin, an mTOR inhibitor, reversed FSS-induced MT1-MMP upregulation [38]. FSS exposed colorectal cancer cells showed Akt-mediated upregulation of atonal bHLH transcription factor 8 (ATOH8) and yes-associated protein 1 (YAP1), transcription factors that enhance cancer cell survival, growth, metastasis, and metabolic remodeling [81,82]. In addition to FSS-induced ROS, changes in the microenvironment also contribute to ROS generation. Cancer cells are hypoxic; thus, oxygenation occurring during their entrance into the bloodstream increases the production of ROS. MCF7 breast cancer cells subjected to hypoxia (1% oxygen) followed by reoxygenation (10% oxygen) increased phosphorylation of Akt and Erk1/2 [87]. ROS regulates the PI3K/Akt pathways, but PI3K/Akt also regulates redox status in breast cancer cells [88]. Akt enhances glutathione biosynthesis via Nrf2-mediated upregulation of glutathione synthase and glutathione reductase [88].

5. Alteration of Mitochondrial Death Mechanism

Alteration of apoptotic death including impaired pro-apoptotic or excess anti-apoptotic mechanisms is commonly found in cancer cells. Under normal physiological conditions, unwanted or damaged cells undergo apoptotic death. Binding of a ligand to a death receptor such as the Fas receptor and tumor necrosis factor-α (TNF-α) receptor initiates extrinsic apoptosis activating an initiator caspase followed by activator caspases, or integrates with mitochondria-mediated intrinsic apoptosis. Signaling proteins activated by an extrinsic pathway such as Bid, or accumulation of intracellular stimuli such as ROS and calcium activate the pro-apoptotic Bcl-2 proteins, Bax and Bak. These pro-death proteins are oligomerized on the mitochondrial membrane, causing the mitochondrial membrane to become permeable. Cytochrome c is released from mitochondria upon loss of mitochondrial membrane integrity, and forms the apoptosome, a protein complex with an apoptotic protease activating factor 1 (Apaf-1), activating caspase 9 and downstream activator caspases including caspase 3 [89,90].
The application of gene set enrichment analysis showed that CTC derived from breast cancer patients decreased apoptosis [91,92], but increased the pathways involved in metastasis, cholesterol biosynthesis, and angiogenesis [91]. Human breast cancer cells MDA-MB-231, MCF7, and ZR75-1 cultured in suspension, mimicking the bloodstream environment, lowered protein levels of Fas, TNF-α receptor, and death receptor 5 (DR5), death receptors that initiate extrinsic apoptosis [93], and this impaired the activation of caspase 3 compared to the adherent group [93]. Consistently, the application of low shear stress (2 dyn/cm2) to MDA-MB-231 cells blocks Fas-induced extrinsic apoptosis [52]. FSS exposure impaired the conversion of pro- to the active form of caspase-8 preventing truncation of Bid. This impairment led to the failure of downstream executor caspase activation [52]. However, the inhibitory effects of FSS on apoptosis may vary by differential receptor profiles the breast cancer cells. Fu et al. showed that triple negative MDA-MB-231 breast cancer cells were more resistant against FSS-mediated oxidative stress and caspase 3/7 activation than hormonal receptor-expressing MCF7 cells [29].
Previous clinical research using CTCs isolated from patients with metastatic breast cancer suggested B-cell lymphoma 2 (Bcl-2) as a potential tool for assessing therapeutic efficacy [94,95]. Bcl-2 is an anti-apoptotic member of the Bcl-2 family of proteins along with Bcl-xL and Mcl-1. These anti-apoptotic Bcl-2 proteins are expressed on the mitochondrial membrane and bind directly to pro-death Bax and Bak, thus preventing apoptotic cell death. Thangavel et al. showed upregulation of the Bcl-2 protein in CTC collected from mice transplanted with triple negative human breast cancer [91]. ROS upregulate the mitochondrial anti-apoptotic proteins including Bcl-xL and Bcl-2 via activation of the transcription factors NF-κB, Nrf-2, and HIF-1α [96,97,98]. ROS also manipulate expression of the pro-apoptotic proteins, including Bad and Bim, via the ERK/MAPK and PI3K/Akt pathways [99,100,101]. Thus, FSS-induced ROS may change the proportions of pro- and anti-apoptotic gene expression, altering the apoptotic pathway. Although it is still unclear whether high or low levels of anti-apoptotic Bcl-2 proteins in CTCs is correlated with breast cancer metastasis in humans [94,95], mice intravenously injected with MDA-MB-435 breast cancer cells overexpressing Bcl-xL increased formation of secondary tumors [102]. CTCs expressing Bcl-2 were resistant to anoikis [103]. This study also showed that higher Bcl-2 expression on CTCs is correlated with higher levels of adhesion molecules including E-selectin, ICAM-1, and VCAM-1 [103]. Strategies enhancing apoptosis such as caspase 3 activation and Bcl-xL depletion are correlated with a decreased number of CTCs and metastatic colonization [104].
Previous clinical research using CTCs isolated from patients with metastatic breast cancer suggested B-cell lymphoma 2 (Bcl-2) as a potential tool for assessing therapeutic efficacy [94,95]. Bcl-2 is an anti-apoptotic member of the Bcl-2 family of proteins along with Bcl-xL and Mcl-1. These anti-apoptotic Bcl-2 proteins are expressed on the mitochondrial membrane and bind directly to pro-death Bax and Bak, thus preventing apoptotic cell death. Thangavel et al. showed upregulation of the Bcl-2 protein in CTC collected from mice transplanted with triple negative human breast cancer [91]. ROS upregulate the mitochondrial anti-apoptotic proteins including Bcl-xL and Bcl-2 via activation of the transcription factors NF-κB, Nrf-2, and HIF-1α [96,97,98]. ROS also manipulate expression of the pro-apoptotic proteins, including Bad and Bim, via the ERK/MAPK and PI3K/Akt pathways [99,100,101]. Thus, FSS-induced ROS may change the proportions of pro- and anti-apoptotic gene expression, altering the apoptotic pathway. Although it is still unclear whether high or low levels of anti-apoptotic Bcl-2 proteins in CTCs is correlated with breast cancer metastasis in humans [94,95], mice intravenously injected with MDA-MB-435 breast cancer cells overexpressing Bcl-xL increased formation of secondary tumors [102]. CTCs expressing Bcl-2 were resistant to anoikis [103]. This study also showed that higher Bcl-2 expression on CTCs is correlated with higher levels of adhesion molecules including E-selectin, ICAM-1, and VCAM-1 [103]. Strategies enhancing apoptosis such as caspase 3 activation and Bcl-xL depletion are correlated with a decreased number of CTCs and metastatic colonization [104].

6. Alteration of Mitochondrial Energy Metabolism

Metabolic remodeling such as decreased oxidative phosphorylation in mitochondria and increased glycolysis in the cytoplasm (Warburg effect), is a hallmark of cancer [53,105,106,107]. Breast cancer cells can compensate energy deficits during impaired oxidative phosphorylation by upregulating genes involved in glycolysis [108]. Glycolysis provides ATP more rapidly than oxidative phosphorylation, and high glycolytic activity supports rapid cancer cell proliferation. Indeed, invasive cells show higher glycolysis than less invasive cancer cells [109,110]. While the Warburg effect is an important metabolic phenotype in cancer, cancer cells are also known to have high metabolic flexibility in the presence of shear stress. FSS can shift energy metabolism to glycolysis via the upregulation of hexokinase in colorectal cancer [82]. In addition to glucose metabolism, metastatic breast cancer cells show a higher dependency on fatty acid oxidation [111]. FSS has been previously reported to alter mitochondrial bioenergetic profiles in vascular endothelium [112]. Recently, Huang et al. demonstrated that the application of a physiological range of FSS (5–20 dyn/cm2) is capable of shifting cancer cell energy metabolism to glycolysis in colorectal cancer models. This study suggested that FSS-induced Akt signaling increases the expression of ATOH8, and the upregulation of ATOH8 ultimately promotes glycolysis by activating hexokinase transcription [82]. Similarly, Chen et al. showed that advanced tumor states and EMT phenotypes are associated with increased expression of phosphoglycerate kinase 1 and glucose-6-phosphate dehydrogenase in CTCs, enzymes that conduct cytoplasmic glycolysis and the pentose phosphate pathway, respectively [113].
F1Fo ATP synthase, an enzyme complex located in the mitochondrial inner membrane, plays a central role in cellular energy metabolism. F1Fo ATP synthase has multiple subunits, including the F1 complex with a, b, c, α, β, γ, δ and ε subunits and the oligomycin sensitivity-conferring protein (OSCP). The Fo complex has a, b, and c subunits. Studies have shown an association between breast cancer and mutations on ATP6 and ATP8 genes that encode the a-subunit of the F1Fo ATP synthase [114,115,116]. The a-subunit is critical in the rotation of F1Fo ATP synthase, a necessary process during ATP production; therefore, ATP6 and ATP8 mutations negatively influence mitochondrial energy metabolism [117]. In addition to mutation, breast cancer pathology alters the abundance of other subunits of the F1Fo ATP synthase [118]. Tissue biopsy samples obtained from patients with breast, gastric, and esophageal cancer showed depletion of the β-subunit of the F1Fo ATP synthase [118]. The β-subunit interacts with ADP and releases ATP, and it also binds to various proteins that regulate the efficiency of mitochondrial energy metabolism [119,120]. The F1Fo ATP synthase inhibitor (IF1) is a protein that targets the α and β subunits of F1Fo ATP synthase, blocking ATP production [121]. The upregulation of IF1 in human breast cancer cells is associated with impaired mitochondrial energy metabolism, enhanced glycolysis, lactate production, and fatty acid oxidation [122]. Analysis with CTCs isolated from breast cancer patients showed downregulation of acetyl-CoA carboxylase, a key enzyme during fatty acid synthesis [91] suggesting the alteration of fat metabolism also occurs in CTCs. The c-subunit ring of F1Fo ATP synthase exhibits a large non-selective mitochondrial channel activity hindering mitochondrial energy metabolism [120,123,124,125], and inhibition or mutation of the c-subunit prevents cell death associated with oxidative stress [123]. Interestingly, F1Fo ATP synthase is also found on the surface (ecto) of human breast cancer cells, MDA-MB-231 [126]. The ecto F1Fo ATP synthase acts as an ApoA1 receptor and regulates cellular uptake of lipoproteins and angiogenesis [127]. The application of angiostatin, an inhibitor of cell migration and the proliferation, binds to the α-subunit of the ecto F1Fo ATP synthase and inhibits angiogenesis [128]. The expression of the β-subunit of the ecto F1Fo ATP synthase is also higher in breast cancer cells than normal cells [129], and application of an aptamer or an antibody targeting the β-subunit induced cytotoxicity in various epithelial cells including breast cancer cells [129,130]. Inhibition of the β-subunit increased apoptosis and blocked phosphorylation of ERK and Akt, thus the ecto β-subunit may regulate survival genes that are under the control of the ERK and Akt pathways [130].

7. Antioxidant and Breast Cancer Metastasis

Due to the roles of ROS in manipulating the signaling pathways, energy metabolism and death mechanisms of breast cancer cells, treatments with antioxidants potentially exhibit therapeutic effects against CTC-associated metastasis [131]. Tam et al. injected luciferase labeled-human breast cancer cells, MDA-MB-231, into the mammary fat pads of immunodeficient mice to develop primary tumors. The animals were then fed with a normal vs. an antioxidant-rich diet, including vitamin E (99 IU/kg) and pterostilbene (40 μg/kg) [131]. The mice in the experimental group demonstrated a significantly reduced number of CTCs, and these dietary antioxidants also reduced the size of the primary tumor [131]. Vitamin Es including both tocopherols and tocotrienols are well-studied antioxidants. Vitamin E scavenges singlet oxygen [132] and prevents mitochondrial dysfunction associated with the accumulation of superoxide [133,134,135]. Therefore, vitamin E may be beneficial in preventing the mitochondrial remodeling that occurs during an FSS-induced ROS challenge. Treatment with vitamin E attenuated estrogen-induced nitrosative and oxidative stress in both in vivo and in vitro breast cancer models, while it enhanced expression of antioxidant enzymes such as superoxide dismutase, glutathione peroxidase, and catalase via upregulation of Nrf2 [136,137]. Vitamin E manipulates redox-sensitive cell signaling pathways: Vitamin E decreases phosphorylation of ERK and Akt in breast cancer cells [131,138,139] changing the activities of downstream targets that regulate glycolysis, apoptosis, and cell cycles [131,138,140,141]. In addition to its traditional antioxidant functions, vitamin E also regulates mitochondrial genes and exhibits anticancer effects. The application of mitochondrially targeted vitamin E lowered mitochondrial DNA transcription in mouse model of HER2high breast cancer [142]. Vitamin E may bind directly to Bcl-2 family proteins regulating their activities or mitochondrial permeability transition [133,143]. Mitochondrial vitamin E increases the loss of mitochondrial membrane potential and the production of ROS potentiating breast cancer cell death [142]. Vitamin E prevents the production of inflammatory cytokines that are causative for breast cancer development [144].
Vitamin C donates an electron, to impart antioxidant properties and the ability to neutralize free radicals. Low levels of plasma vitamin C and high levels of lipid peroxidation are associated with metastatic breast cancer [145], while increased dietary vitamin C is associated with a decreased risk of breast cancer related mortality [146]. In addition to its well-studied antioxidant function, vitamin C can also act as a pro-oxidant and cause cytotoxicity in cancer cells [147]. Supratherapeutic concentrations of vitamin C decrease the viability of both non-metastatic and metastatic breast cancer cells and support chemotherapy treatment [148,149,150]. Loss of extracellular matrix adhesion in primary cancer cells leads to intravasation, releasing CTCs. Studies suggested potential roles of vitamin C in preventing breast cancer metastasis via the regulation of the extracellular matrix [151,152]. An electron from vitamin C can be used to reduce ferric iron to ferrous iron, and this supports the action of lysyl hydroxylase and prolyl hydroxylase, iron binding enzymes required during collagen synthesis. Transgenic mice lacking gulonolactone oxidase, a key enzyme for vitamin C synthesis, showed a poorly defined collagenous barrier, whereas vitamin C supplemented mice showed optimal extracellular matrix formation [151,153]. Treatment with a high (millimolar) concentration of vitamin C increased protein levels of E-cadherin, a key protein for cell-extracellular matrix adhesion in the BCap-37 human breast cancer cell [152]. Micromolar levels of vitamin C impaired the assembly of the actin filament via downregulation of YAP1, and this decreased motility of MDA-MB-231 cells [154]. YAP1 is a transcriptional co-activator that regulates angiogenesis, apoptosis, DNA repair, and energy metabolism [155]. FSS is shown to increase the nuclear translocation of YAP1 in epithelial cells [156].
Carotenoids, pigments found in fruits, vegetables, and algae, play a role in redox homeostasis. Examples include β-carotene, lycopene, lutein, astaxanthin, and fucoxanthin. Some carotenoids such as β-carotene are converted into vitamin A. Vitamin A regulates genes that control proliferation and differentiation by interacting with nuclear receptors such as the retinoic acid receptor (RAR) and retinoid X receptor (RXR). Clinical studies with breast cancer patients showed an association between stage III metastatic cancer, metabolic alteration, and decreased serum concentrations of β-carotene and vitamin A [157,158]. β-carotene regulates the expression of oxidative stress-sensitive genes via Nrf2 [159]. β-carotene also enhances the apoptosis of breast cancer cells by enhancing the activation of caspase 3 [159]. A combination of vitamin A with a chemotherapy drug or pro-apoptotic molecules potentiates breast cancer cell death by decreasing pro-survival Bcl-2, increasing pro-death Bax, and activating caspases [160,161,162]. CTCs isolated from breast cancer patients demonstrate an altered apoptotic pathway [91,92], and FSS exposed breast cancer cells decrease the abundance of pro-apoptotic proteins such as caspase 3 [29,52]. Therefore, treatment with β-carotene or vitamin A may prevent FSS-mediated resistance against apoptotic death. Treatment with fucoxanthin showed concentration-dependent suppression of PI3K/Akt phosphorylation and NF-κB protein levels in human breast cancer cells [163]. The activation of PI3K/Akt contributes to metastatic phenotypes of breast cancer. In particular, FSS increases the phosphorylation of PI3K and Akt, increasing the motility of breast cancer [38]. NF-κB is a proinflammatory transcription factor that promotes the development of invasive breast cancer. NF-κB has previously been shown to bind to the promoter region of Bcl-xL and Bcl-2, resulting in a pro-survival response [164,165]. Therefore, treatment with fucoxanthin may help to decrease the malignant phenotypes of CTCs and prevent the metastasis of breast cancer. Despite the anti-metastatic properties of antioxidants, opposing effects of antioxidants have also been documented. Zhen et al. show that treatment with N-acetyl cysteine protects CTCs from oxidative stress-associated damage and promotes the survival of breast cancer cells [166]. Studies also suggest that supplementation with antioxidants including vitamin E, C and carotenoids may interfere with the potency of chemotherapy drugs and enhance the recurrence of breast cancer [167,168,169]. Due to these mixed results, further investigation into specific conditions like concentration and duration of antioxidant treatment, severity of cancer, and cancer cell characteristics may be useful to determine the exact therapeutic role of antioxidants in CTC-mediated metastasis.

8. Conclusions

Breast cancer cells traveling in the bloodstream undergo various modifications. FSS occurs in a dynamic circulation environment that can challenge cancer cells; however, it can also activate survival mechanisms that potentially increase the risk of metastasis. In this review, cellular mechanisms that may explain CTC-induced metastasis in breast cancer were discussed, including of redox homeostasis, cell signaling pathways, energy metabolism, and death mechanisms (Table 2). The effect of mitochondria on metastasis via their role in generating ROS, performing oxidative phosphorylation, and governing death mechanisms like apoptosis and mitochondrial permeability transition pore opening was highlighted. Mitochondrial dysfunction that occurs in cancer cells during circulation may influence ROS-sensitive cell signaling, energy remodeling, growth, and survival of these cancer cells. Therefore, further investigations that elucidate the cellular and molecular mechanisms by which CTCs gain the metastatic phenotype and new strategies that inhibit CTC modification will be important in preventing breast cancer metastasis.

Funding

We gratefully acknowledge funding for this research from The University of Alabama’s Alabama Life Research Institute grant number ALRI-14565 (Y.K., and H.-A.P.) and from The Bridge to the Doctorate: Alabama LSAMP Program (S.R.B.) funded by the National Science Foundation grant number HRD-1702406.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. National Cancer Institute. Cancer facts and figures. In American Cancer Society; National Cancer Institute: Bethesda, MD, USA, 2020. [Google Scholar]
  2. Ripperger, T.; Gadzicki, D.; Meindl, A.; Schlegelberger, B. Breast cancer susceptibility: Current knowledge and implications for genetic counselling. Eur. J. Hum. Genet. 2009, 17, 722–731. [Google Scholar] [CrossRef]
  3. Slavin, T.P.; Maxwell, K.N.; Lilyquist, J.; Vijai, J.; Neuhausen, S.L.; Hart, S.N.; Ravichandran, V.; Thomas, T.; Maria, A.; Villano, D.; et al. The contribution of pathogenic variants in breast cancer susceptibility genes to familial breast cancer risk. NPJ Breast Cancer 2017, 3, 22. [Google Scholar] [CrossRef] [PubMed]
  4. Niehoff, N.M.; Nichols, H.B.; Zhao, S.; White, A.J.; Sandler, D.P. Adult physical activity and breast cancer risk in women with a family history of breast cancer. Cancer Epidemiol. Biomark. Prev. 2019, 28, 51–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Seiler, A.; Chen, M.A.; Brown, R.L.; Fagundes, C.P. Obesity, dietary factors, nutrition, and breast cancer risk. Curr. Breast Cancer Rep. 2018, 10, 14–27. [Google Scholar] [CrossRef]
  6. Chia, S.K.; Speers, C.H.; D’Yachkova, Y.; Kang, A.; Malfair-Taylor, S.; Barnett, J.; Coldman, A.; Gelmon, K.A.; O’Reilly S, E.; Olivotto, I.A. The impact of new chemotherapeutic and hormone agents on survival in a population-based cohort of women with metastatic breast cancer. Cancer 2007, 110, 973–979. [Google Scholar] [CrossRef]
  7. Gennari, A.; Conte, P.; Rosso, R.; Orlandini, C.; Bruzzi, P. Survival of metastatic breast carcinoma patients over a 20-year period: A retrospective analysis based on individual patient data from six consecutive studies. Cancer 2005, 104, 1742–1750. [Google Scholar] [CrossRef]
  8. Jin, X.; Mu, P. Targeting breast cancer metastasis. Breast Cancer (Auckl) 2015, 9, 23–34. [Google Scholar] [CrossRef] [Green Version]
  9. Valastyan, S.; Weinberg, R.A. Tumor metastasis: Molecular insights and evolving paradigms. Cell 2011, 147, 275–292. [Google Scholar] [CrossRef] [Green Version]
  10. Liu, Q.; Zhang, H.; Jiang, X.; Qian, C.; Liu, Z.; Luo, D. Factors involved in cancer metastasis: A better understanding to ”seed and soil” hypothesis. Mol. Cancer 2017, 16, 176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Paget, S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev. 1989, 8, 98–101. [Google Scholar] [CrossRef] [Green Version]
  12. Cristofanilli, M.; Pierga, J.Y.; Reuben, J.; Rademaker, A.; Davis, A.A.; Peeters, D.J.; Fehm, T.; Nole, F.; Gisbert-Criado, R.; Mavroudis, D.; et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): International expert consensus paper. Crit. Rev. Oncol. Hematol. 2019, 134, 39–45. [Google Scholar] [CrossRef] [PubMed]
  13. Deutsch, T.M.; Stefanovic, S.; Feisst, M.; Fischer, C.; Riedel, F.; Fremd, C.; Domschke, C.; Pantel, K.; Hartkopf, A.D.; Sutterlin, M.; et al. Cut-off analysis of CTC change under systemic therapy for defining early therapy response in metastatic breast cancer. Cancers 2020, 12, 1055. [Google Scholar] [CrossRef] [PubMed]
  14. Bidard, F.C.; Peeters, D.J.; Fehm, T.; Nole, F.; Gisbert-Criado, R.; Mavroudis, D.; Grisanti, S.; Generali, D.; Garcia-Saenz, J.A.; Stebbing, J.; et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: A pooled analysis of individual patient data. Lancet Oncol. 2014, 15, 406–414. [Google Scholar] [CrossRef]
  15. Riebensahm, C.; Joosse, S.A.; Mohme, M.; Hanssen, A.; Matschke, J.; Goy, Y.; Witzel, I.; Lamszus, K.; Kropidlowski, J.; Petersen, C.; et al. Clonality of circulating tumor cells in breast cancer brain metastasis patients. Breast Cancer Res. 2019, 21, 101. [Google Scholar] [CrossRef] [Green Version]
  16. Smerage, J.B.; Barlow, W.E.; Hortobagyi, G.N.; Winer, E.P.; Leyland-Jones, B.; Srkalovic, G.; Tejwani, S.; Schott, A.F.; O’Rourke, M.A.; Lew, D.L.; et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J. Clin. Oncol. 2014, 32, 3483–3489. [Google Scholar] [CrossRef]
  17. Gennari, A.; Foca, F.; Zamarchi, R.; Rocca, A.; Amadori, D.; De Censi, A.; Bologna, A.; Cavanna, L.; Gianni, L.; Scaltriti, L.; et al. Insulin-like growth factor-1 receptor (IGF-1R) expression on circulating tumor cells (CTCs) and metastatic breast cancer outcome: Results from the TransMYME trial. Breast Cancer Res. Treat. 2020, 181, 61–68. [Google Scholar] [CrossRef]
  18. Papadaki, M.A.; Koutsopoulos, A.V.; Tsoulfas, P.G.; Lagoudaki, E.; Aggouraki, D.; Monastirioti, A.; Koutoulaki, C.; Apostolopoulou, C.A.; Merodoulaki, A.C.; Papadaki, C.; et al. Clinical relevance of immune checkpoints on circulating tumor cells in breast cancer. Cancers 2020, 12, 376. [Google Scholar] [CrossRef] [Green Version]
  19. Pantano, F.; Rossi, E.; Iuliani, M.; Facchinetti, A.; Simonetti, S.; Ribelli, G.; Zoccoli, A.; Vincenzi, B.; Tonini, G.; Zamarchi, R.; et al. Dynamic changes of Receptor activator of nuclear factor-kappaB expression in Circulating Tumor Cells during Denosumab predict treatment effectiveness in Metastatic Breast Cancer. Sci. Rep. 2020, 10, 1288. [Google Scholar] [CrossRef]
  20. Savelieva, O.E.; Tashireva, L.A.; Kaigorodova, E.V.; Buzenkova, A.V.; Mukhamedzhanov, R.K.; Grigoryeva, E.S.; Zavyalova, M.V.; Tarabanovskaya, N.A.; Cherdyntseva, N.V.; Perelmuter, V.M. Heterogeneity of stemlike circulating tumor cells in invasive breast cancer. Int. J. Mol. Sci. 2020, 21, 2780. [Google Scholar] [CrossRef]
  21. Mego, M.; Karaba, M.; Sedlackova, T.; Benca, J.; Repiska, G.; Krasnicanova, L.; Macuch, J.; Sieberova, G.; Jurisova, S.; Pindak, D.; et al. Circulating tumor cells and breast cancer-specific mutations in primary breast cancer. Mol. Clin. Oncol. 2020, 12, 565–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Fidler, I.J.; Yano, S.; Zhang, R.D.; Fujimaki, T.; Bucana, C.D. The seed and soil hypothesis: Vascularisation and brain metastases. Lancet Oncol. 2002, 3, 53–57. [Google Scholar] [CrossRef]
  23. Wirtz, D.; Konstantopoulos, K.; Searson, P.C. The physics of cancer: The role of physical interactions and mechanical forces in metastasis. Nat. Rev. Cancer 2011, 11, 512–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Huang, Q.; Hu, X.; He, W.; Zhao, Y.; Hao, S.; Wu, Q.; Li, S.; Zhang, S.; Shi, M. Fluid shear stress and tumor metastasis. Am. J. Cancer Res. 2018, 8, 763–777. [Google Scholar] [PubMed]
  25. Papaioannou, T.G.; Stefanadis, C. Vascular wall shear stress: Basic principles and methods. Hell. J. Cardiol. 2005, 46, 9–15. [Google Scholar]
  26. Lien, S.C.; Chang, S.F.; Lee, P.L.; Wei, S.Y.; Chang, M.D.; Chang, J.Y.; Chiu, J.J. Mechanical regulation of cancer cell apoptosis and autophagy: Roles of bone morphogenetic protein receptor, Smad1/5, and p38 MAPK. Biochim. Biophys. Acta 2013, 1833, 3124–3133. [Google Scholar] [CrossRef] [Green Version]
  27. Triantafillu, U.L.; Park, S.; Klaassen, N.L.; Raddatz, A.D.; Kim, Y. Fluid shear stress induces cancer stem cell-like phenotype in MCF7 breast cancer cell line without inducing epithelial to mesenchymal transition. Int. J. Oncol. 2017, 50, 993–1001. [Google Scholar] [CrossRef] [Green Version]
  28. Ma, S.; Fu, A.; Chiew, G.G.; Luo, K.Q. Hemodynamic shear stress stimulates migration and extravasation of tumor cells by elevating cellular oxidative level. Cancer Lett. 2017, 388, 239–248. [Google Scholar] [CrossRef]
  29. Fu, A.; Ma, S.; Wei, N.; Tan, B.X.; Tan, E.Y.; Luo, K.Q. High expression of MnSOD promotes survival of circulating breast cancer cells and increases their resistance to doxorubicin. Oncotarget 2016, 7, 50239–50257. [Google Scholar] [CrossRef]
  30. Triantafillu, U.L.; Park, S.; Kim, Y. Fluid shear stress induces drug resistance to doxorubicin and paclitaxel in the breast cancer cell line MCF7. Adv. Ther. 2019, 2, 1800112. [Google Scholar] [CrossRef]
  31. Choi, H.Y.; Yang, G.M.; Dayem, A.A.; Saha, S.K.; Kim, K.; Yoo, Y.; Hong, K.; Kim, J.H.; Yee, C.; Lee, K.M.; et al. Hydrodynamic shear stress promotes epithelial-mesenchymal transition by downregulating ERK and GSK3beta activities. Breast Cancer Res. 2019, 21, 6. [Google Scholar] [CrossRef]
  32. Akutagawa, T.; Aoki, S.; Yamamoto-Rikitake, M.; Iwakiri, R.; Fujimoto, K.; Toda, S. Cancer-adipose tissue interaction and fluid flow synergistically modulate cell kinetics, HER2 expression, and trastuzumab efficacy in gastric cancer. Gastric Cancer 2018, 21, 946–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Mitchell, M.J.; King, M.R. Fluid shear stress sensitizes cancer cells to receptor-mediated apoptosis via trimeric death receptors. New J. Phys. 2013, 15, 015008. [Google Scholar] [CrossRef] [Green Version]
  34. Ma, S.; Fu, A.; Lim, S.; Chiew, G.G.Y.; Luo, K.Q. MnSOD mediates shear stress-promoted tumor cell migration and adhesion. Free Radic. Biol. Med. 2018, 129, 46–58. [Google Scholar] [CrossRef] [PubMed]
  35. Regmi, S.; Fu, A.; Luo, K.Q. High shear stresses under exercise condition destroy circulating tumor cells in a microfluidic system. Sci. Rep. 2017, 7, 39975. [Google Scholar] [CrossRef] [Green Version]
  36. Fan, R.; Emery, T.; Zhang, Y.; Xia, Y.; Sun, J.; Wan, J. Circulatory shear flow alters the viability and proliferation of circulating colon cancer cells. Sci. Rep. 2016, 6, 27073. [Google Scholar] [CrossRef]
  37. Jabbar, A.A.; Kazarian, T.; Hakobyan, N.; Valentino, L.A. Gangliosides promote platelet adhesion and facilitate neuroblastoma cell adhesion under dynamic conditions simulating blood flow. Pediatr. Blood Cancer 2006, 46, 292–299. [Google Scholar] [CrossRef] [PubMed]
  38. Yang, H.; Guan, L.; Li, S.; Jiang, Y.; Xiong, N.; Li, L.; Wu, C.; Zeng, H.; Liu, Y. Mechanosensitive caveolin-1 activation-induced PI3K/Akt/mTOR signaling pathway promotes breast cancer motility, invadopodia formation and metastasis in vivo. Oncotarget 2016, 7, 16227–16247. [Google Scholar] [CrossRef] [Green Version]
  39. Mosier, J.A.; Rahman-Zaman, A.; Zanotelli, M.R.; VanderBurgh, J.A.; Bordeleau, F.; Hoffman, B.D.; Reinhart-King, C.A. Extent of cell confinement in microtracks affects speed and results in differential matrix strains. Biophys. J. 2019, 117, 1692–1701. [Google Scholar] [CrossRef]
  40. Burinaru, T.A.; Avram, M.; Avram, A.; Marculescu, C.; Tincu, B.; Tucureanu, V.; Matei, A.; Militaru, M. Detection of circulating tumor cells using microfluidics. ACS Comb. Sci. 2018, 20, 107–126. [Google Scholar] [CrossRef]
  41. Mitchell, M.J.; King, M.R. Computational and experimental models of cancer cell response to fluid shear stress. Front. Oncol. 2013, 3, 44. [Google Scholar] [CrossRef] [Green Version]
  42. Schafer, Z.T.; Grassian, A.R.; Song, L.; Jiang, Z.; Gerhart-Hines, Z.; Irie, H.Y.; Gao, S.; Puigserver, P.; Brugge, J.S. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature 2009, 461, 109–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gong, C.; Liu, B.; Yao, Y.; Qu, S.; Luo, W.; Tan, W.; Liu, Q.; Yao, H.; Zou, L.; Su, F.; et al. Potentiated DNA damage response in circulating breast tumor cells confers resistance to chemotherapy. J. Biol. Chem. 2015, 290, 14811–14825. [Google Scholar] [CrossRef] [Green Version]
  44. Jordan, N.V.; Bardia, A.; Wittner, B.S.; Benes, C.; Ligorio, M.; Zheng, Y.; Yu, M.; Sundaresan, T.K.; Licausi, J.A.; Desai, R.; et al. HER2 expression identifies dynamic functional states within circulating breast cancer cells. Nature 2016, 537, 102–106. [Google Scholar] [CrossRef] [PubMed]
  45. Sprouse, M.L.; Welte, T.; Boral, D.; Liu, H.N.; Yin, W.; Vishnoi, M.; Goswami-Sewell, D.; Li, L.; Pei, G.; Jia, P.; et al. PMN-MDSCs enhance CTC metastatic properties through reciprocal interactions via ROS/Notch/Nodal signaling. Int. J. Mol. Sci. 2019, 20, 1916. [Google Scholar] [CrossRef] [Green Version]
  46. Fu, Q.; Zhang, Q.; Lou, Y.; Yang, J.; Nie, G.; Chen, Q.; Chen, Y.; Zhang, J.; Wang, J.; Wei, T.; et al. Primary tumor-derived exosomes facilitate metastasis by regulating adhesion of circulating tumor cells via SMAD3 in liver cancer. Oncogene 2018, 37, 6105–6118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Weniger, M.A.; Rizzatti, E.G.; Perez-Galan, P.; Liu, D.; Wang, Q.; Munson, P.J.; Raghavachari, N.; White, T.; Tweito, M.M.; Dunleavy, K.; et al. Treatment-induced oxidative stress and cellular antioxidant capacity determine response to bortezomib in mantle cell lymphoma. Clin. Cancer Res. 2011, 17, 5101–5112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Chance, B.; Sies, H.; Boveris, A. Hydroperoxide metabolism in mammalian organs. Physiol. Rev. 1979, 59, 527–605. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, Q.; Vazquez, E.J.; Moghaddas, S.; Hoppel, C.L.; Lesnefsky, E.J. Production of reactive oxygen species by mitochondria: Central role of complex III. J. Biol. Chem. 2003, 278, 36027–36031. [Google Scholar] [CrossRef] [Green Version]
  50. Sarmiento-Salinas, F.L.; Delgado-Magallon, A.; Montes-Alvarado, J.B.; Ramirez-Ramirez, D.; Flores-Alonso, J.C.; Cortes-Hernandez, P.; Reyes-Leyva, J.; Herrera-Camacho, I.; Anaya-Ruiz, M.; Pelayo, R.; et al. Breast cancer subtypes present a differential production of reactive oxygen species (ROS) and susceptibility to antioxidant treatment. Front. Oncol. 2019, 9, 480. [Google Scholar] [CrossRef] [Green Version]
  51. An, Q.; Hu, Y.; Li, Q.; Chen, X.; Huang, J.; Pellegrini, M.; Zhou, X.J.; Rettig, M.; Fan, G. The size of cell-free mitochondrial DNA in blood is inversely correlated with tumor burden in cancer patients. Precis. Clin. Med. 2019, 2, 131–139. [Google Scholar] [CrossRef] [Green Version]
  52. Li, S.; Chen, Y.; Zhang, Y.; Jiang, X.; Jiang, Y.; Qin, X.; Yang, H.; Wu, C.; Liu, Y. Shear stress promotes anoikis resistance of cancer cells via caveolin-1-dependent extrinsic and intrinsic apoptotic pathways. J. Cell. Physiol. 2019, 234, 3730–3743. [Google Scholar] [CrossRef] [PubMed]
  53. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
  54. Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef] [PubMed]
  55. Yang, H.; Villani, R.M.; Wang, H.; Simpson, M.J.; Roberts, M.S.; Tang, M.; Liang, X. The role of cellular reactive oxygen species in cancer chemotherapy. J. Exp. Clin. Cancer Res. 2018, 37, 266. [Google Scholar] [CrossRef]
  56. Marullo, R.; Werner, E.; Degtyareva, N.; Moore, B.; Altavilla, G.; Ramalingam, S.S.; Doetsch, P.W. Cisplatin induces a mitochondrial-ROS response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS ONE 2013, 8, e81162. [Google Scholar] [CrossRef] [PubMed]
  57. Pilco-Ferreto, N.; Calaf, G.M. Influence of doxorubicin on apoptosis and oxidative stress in breast cancer cell lines. Int. J. Oncol. 2016, 49, 753–762. [Google Scholar] [CrossRef] [Green Version]
  58. Regmi, S.; Fung, T.S.; Lim, S.; Luo, K.Q. Fluidic shear stress increases the anti-cancer effects of ROS-generating drugs in circulating tumor cells. Breast Cancer Res. Treat. 2018, 172, 297–312. [Google Scholar] [CrossRef]
  59. Cancer Genome Atlas, N. Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [Google Scholar] [CrossRef] [Green Version]
  60. Craig, D.W.; O’Shaughnessy, J.A.; Kiefer, J.A.; Aldrich, J.; Sinari, S.; Moses, T.M.; Wong, S.; Dinh, J.; Christoforides, A.; Blum, J.L.; et al. Genome and transcriptome sequencing in prospective metastatic triple-negative breast cancer uncovers therapeutic vulnerabilities. Mol. Cancer Ther. 2013, 12, 104–116. [Google Scholar] [CrossRef] [Green Version]
  61. Ross, J.S.; Slodkowska, E.A.; Symmans, W.F.; Pusztai, L.; Ravdin, P.M.; Hortobagyi, G.N. The HER-2 receptor and breast cancer: Ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist 2009, 14, 320–368. [Google Scholar] [CrossRef] [Green Version]
  62. Hendriks, B.S.; Orr, G.; Wells, A.; Wiley, H.S.; Lauffenburger, D.A. Parsing ERK activation reveals quantitatively equivalent contributions from epidermal growth factor receptor and HER2 in human mammary epithelial cells. J. Biol. Chem. 2005, 280, 6157–6169. [Google Scholar] [CrossRef] [Green Version]
  63. Boucher, M.J.; Morisset, J.; Vachon, P.H.; Reed, J.C.; Laine, J.; Rivard, N. MEK/ERK signaling pathway regulates the expression of Bcl-2, Bcl-X(L), and Mcl-1 and promotes survival of human pancreatic cancer cells. J. Cell. BioChem. 2000, 79, 355–369. [Google Scholar] [CrossRef]
  64. Doehn, U.; Hauge, C.; Frank, S.R.; Jensen, C.J.; Duda, K.; Nielsen, J.V.; Cohen, M.S.; Johansen, J.V.; Winther, B.R.; Lund, L.R.; et al. RSK is a principal effector of the RAS-ERK pathway for eliciting a coordinate promotile/invasive gene program and phenotype in epithelial cells. Mol. Cell 2009, 35, 511–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ludwik, K.A.; Campbell, J.P.; Li, M.; Li, Y.; Sandusky, Z.M.; Pasic, L.; Sowder, M.E.; Brenin, D.R.; Pietenpol, J.A.; O’Doherty, G.A.; et al. Development of a RSK inhibitor as a novel therapy for triple-negative breast cancer. Mol. Cancer Ther. 2016, 15, 2598–2608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Stratford, A.L.; Reipas, K.; Hu, K.; Fotovati, A.; Brough, R.; Frankum, J.; Takhar, M.; Watson, P.; Ashworth, A.; Lord, C.J.; et al. Targeting p90 ribosomal S6 kinase eliminates tumor-initiating cells by inactivating Y-box binding protein-1 in triple-negative breast cancers. STEM CELLS 2012, 30, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  67. Liu, F.; Yang, X.; Geng, M.; Huang, M. Targeting ERK, an Achilles’ Heel of the MAPK pathway, in cancer therapy. Acta Pharm. Sin. B 2018, 8, 552–562. [Google Scholar] [CrossRef]
  68. Smalley, I.; Smalley, K.S.M. ERK inhibition: A new front in the war against MAPK pathway-driven cancers? Cancer Discov. 2018, 8, 140–142. [Google Scholar] [CrossRef] [Green Version]
  69. Ejaeidi, A.A.; Craft, B.S.; Puneky, L.V.; Lewis, R.E.; Cruse, J.M. Hormone receptor-independent CXCL10 production is associated with the regulation of cellular factors linked to breast cancer progression and metastasis. Exp. Mol. Pathol. 2015, 99, 163–172. [Google Scholar] [CrossRef]
  70. Takabe, W.; Jen, N.; Ai, L.; Hamilton, R.; Wang, S.; Holmes, K.; Dharbandi, F.; Khalsa, B.; Bressler, S.; Barr, M.L.; et al. Oscillatory shear stress induces mitochondrial superoxide production: Implication of NADPH oxidase and c-Jun NH2-terminal kinase signaling. Antioxid. Redox Signal. 2011, 15, 1379–1388. [Google Scholar] [CrossRef]
  71. Verret, B.; Cortes, J.; Bachelot, T.; Andre, F.; Arnedos, M. Efficacy of PI3K inhibitors in advanced breast cancer. Ann. Oncol. 2019, 30 (Suppl. 10), x12–x20. [Google Scholar] [CrossRef]
  72. Hennessy, B.T.; Smith, D.L.; Ram, P.T.; Lu, Y.; Mills, G.B. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug Discov. 2005, 4, 988–1004. [Google Scholar] [CrossRef] [PubMed]
  73. Law, N.C.; White, M.F.; Hunzicker-Dunn, M.E. G protein-coupled receptors (GPCRs) That Signal via Protein Kinase A (PKA) Cross-talk at Insulin Receptor Substrate 1 (IRS1) to Activate the phosphatidylinositol 3-kinase (PI3K)/AKT Pathway. J. Biol. Chem. 2016, 291, 27160–27169. [Google Scholar] [CrossRef] [Green Version]
  74. Vadas, O.; Dbouk, H.A.; Shymanets, A.; Perisic, O.; Burke, J.E.; Abi Saab, W.F.; Khalil, B.D.; Harteneck, C.; Bresnick, A.R.; Nurnberg, B.; et al. Molecular determinants of PI3Kgamma-mediated activation downstream of G-protein-coupled receptors (GPCRs). Proc. Natl. Acad. Sci. USA 2013, 110, 18862–18867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Butti, R.; Das, S.; Gunasekaran, V.P.; Yadav, A.S.; Kumar, D.; Kundu, G.C. Receptor tyrosine kinases (RTKs) in breast cancer: Signaling, therapeutic implications and challenges. Mol. Cancer 2018, 17, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Stokoe, D.; Stephens, L.R.; Copeland, T.; Gaffney, P.R.; Reese, C.B.; Painter, G.F.; Holmes, A.B.; McCormick, F.; Hawkins, P.T. Dual role of phosphatidylinositol-3,4,5-trisphosphate in the activation of protein kinase B. Science 1997, 277, 567–570. [Google Scholar] [CrossRef] [Green Version]
  77. Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef] [Green Version]
  78. Sarbassov, D.D.; Guertin, D.A.; Ali, S.M.; Sabatini, D.M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005, 307, 1098–1101. [Google Scholar] [CrossRef] [Green Version]
  79. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [Green Version]
  80. Koundouros, N.; Poulogiannis, G. Phosphoinositide 3-Kinase/Akt signaling and redox metabolism in cancer. Front. Oncol. 2018, 8, 160. [Google Scholar] [CrossRef]
  81. Hagihara, T.; Kondo, J.; Endo, H.; Ohue, M.; Sakai, Y.; Inoue, M. Hydrodynamic stress stimulates growth of cell clusters via the ANXA1/PI3K/AKT axis in colorectal cancer. Sci. Rep. 2019, 9, 20027. [Google Scholar] [CrossRef]
  82. Huang, Q.; Li, S.; Hu, X.; Sun, M.; Wu, Q.; Dai, H.; Tan, Y.; Sun, F.; Wang, C.; Rong, X.; et al. Shear stress activates ATOH8 via autocrine VEGF promoting glycolysis dependent-survival of colorectal cancer cells in the circulation. J. Exp. Clin. Cancer Res. 2020, 39, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zibara, K.; Zeidan, A.; Bjeije, H.; Kassem, N.; Badran, B.; El-Zein, N. ROS mediates interferon gamma induced phosphorylation of Src, through the Raf/ERK pathway, in MCF-7 human breast cancer cell line. J. Cell Commun. Signal. 2017, 11, 57–67. [Google Scholar] [CrossRef] [Green Version]
  84. Deng, W.; Wang, Y.; Zhao, S.; Zhang, Y.; Chen, Y.; Zhao, X.; Liu, L.; Sun, S.; Zhang, L.; Ye, B.; et al. MICAL1 facilitates breast cancer cell proliferation via ROS-sensitive ERK/cyclin D pathway. J. Cell Mol. Med. 2018, 22, 3108–3118. [Google Scholar] [CrossRef] [PubMed]
  85. Ma, Z.; Liu, X.; Zhang, Q.; Yu, Z.; Gao, D. Carvedilol suppresses malignant proliferation of mammary epithelial cells through inhibition of the ROSmediated PI3K/AKT signaling pathway. Oncol. Rep. 2019, 41, 811–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Poincloux, R.; Lizarraga, F.; Chavrier, P. Matrix invasion by tumour cells: A focus on MT1-MMP trafficking to invadopodia. J. Cell Sci. 2009, 122, 3015–3024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Bartkowiak, K.; Koch, C.; Gartner, S.; Andreas, A.; Gorges, T.M.; Pantel, K. In vitro modeling of reoxygenation effects on mRNA and protein levels in hypoxic tumor cells upon entry into the bloodstream. Cells 2020, 9, 1316. [Google Scholar] [CrossRef] [PubMed]
  88. Lien, E.C.; Lyssiotis, C.A.; Juvekar, A.; Hu, H.; Asara, J.M.; Cantley, L.C.; Toker, A. Glutathione biosynthesis is a metabolic vulnerability in PI(3)K/Akt-driven breast cancer. Nat. Cell Biol. 2016, 18, 572–578. [Google Scholar] [CrossRef] [Green Version]
  89. Tait, S.W.; Green, D.R. Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat. Rev. Mol. Cell Biol. 2010, 11, 621–632. [Google Scholar] [CrossRef] [PubMed]
  90. Suhaili, S.H.; Karimian, H.; Stellato, M.; Lee, T.H.; Aguilar, M.I. Mitochondrial outer membrane permeabilization: A focus on the role of mitochondrial membrane structural organization. Biophys. Rev. 2017, 9, 443–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Thangavel, H.; De Angelis, C.; Vasaikar, S.; Bhat, R.; Jolly, M.K.; Nagi, C.; Creighton, C.J.; Chen, F.; Dobrolecki, L.E.; George, J.T.; et al. A CTC-cluster-specific signature derived from OMICS analysis of patient-derived xenograft tumors predicts outcomes in basal-like breast cancer. J. Clin. Med. 2019, 8, 1772. [Google Scholar] [CrossRef] [Green Version]
  92. Lang, J.E.; Scott, J.H.; Wolf, D.M.; Novak, P.; Punj, V.; Magbanua, M.J.; Zhu, W.; Mineyev, N.; Haqq, C.M.; Crothers, J.R.; et al. Expression profiling of circulating tumor cells in metastatic breast cancer. Breast Cancer Res. Treat. 2015, 149, 121–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Twomey, J.D.; Zhang, B. Circulating tumor cells develop resistance to TRAIL-induced apoptosis through autophagic removal of death receptor 5: Evidence from an in vitro model. Cancers 2019, 11, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Smerage, J.B.; Budd, G.T.; Doyle, G.V.; Brown, M.; Paoletti, C.; Muniz, M.; Miller, M.C.; Repollet, M.I.; Chianese, D.A.; Connelly, M.C.; et al. Monitoring apoptosis and Bcl-2 on circulating tumor cells in patients with metastatic breast cancer. Mol. Oncol. 2013, 7, 680–692. [Google Scholar] [CrossRef] [PubMed]
  95. Smerage, J.B.; Doyle, G.V.; Budd, G.T.; Repollet, M.I.; Miller, M.C.; Terstappen, L.W.; Hayes, D.F. Detection of Bcl-2 and apoptosis in circulating tumor cells during treatment of metastatic breast cancer. J. Clin. Oncol. 2008, 26, 11016. [Google Scholar] [CrossRef]
  96. Chen, N.; Chen, X.; Huang, R.; Zeng, H.; Gong, J.; Meng, W.; Lu, Y.; Zhao, F.; Wang, L.; Zhou, Q. BCL-xL is a target gene regulated by hypoxia-inducible factor-1{alpha}. J. Biol. Chem. 2009, 284, 10004–10012. [Google Scholar] [CrossRef] [Green Version]
  97. Chen, C.; Edelstein, L.C.; Gelinas, C. The Rel/NF-kappaB family directly activates expression of the apoptosis inhibitor Bcl-x(L). Mol. Cell Biol. 2000, 20, 2687–2695. [Google Scholar] [CrossRef]
  98. Niture, S.K.; Jaiswal, A.K. Nrf2-induced antiapoptotic Bcl-xL protein enhances cell survival and drug resistance. Free Radic. Biol. Med 2013, 57, 119–131. [Google Scholar] [CrossRef] [Green Version]
  99. Eisenmann, K.M.; VanBrocklin, M.W.; Staffend, N.A.; Kitchen, S.M.; Koo, H.M. Mitogen-activated protein kinase pathway-dependent tumor-specific survival signaling in melanoma cells through inactivation of the proapoptotic protein bad. Cancer Res. 2003, 63, 8330–8337. [Google Scholar]
  100. Goldstein, N.B.; Johannes, W.U.; Gadeliya, A.V.; Green, M.R.; Fujita, M.; Norris, D.A.; Shellman, Y.G. Active N-Ras and B-Raf inhibit anoikis by downregulating Bim expression in melanocytic cells. J. Investig. Dermatol. 2009, 129, 432–437. [Google Scholar] [CrossRef] [Green Version]
  101. Son, Y.; Cheong, Y.K.; Kim, N.H.; Chung, H.T.; Kang, D.G.; Pae, H.O. Mitogen-activated protein kinases and reactive oxygen species: How can ROS activate MAPK pathways? J. Signal Transduct. 2011, 2011, 792639. [Google Scholar] [CrossRef]
  102. Fernandez, Y.; Espana, L.; Manas, S.; Fabra, A.; Sierra, A. Bcl-xL promotes metastasis of breast cancer cells by induction of cytokines resistance. Cell Death Differ. 2000, 7, 350–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yadav, A.; Kumar, B.; Yu, J.G.; Old, M.; Teknos, T.N.; Kumar, P. Tumor-associated endothelial cells promote tumor metastasis by chaperoning circulating tumor cells and protecting them from anoikis. PLoS ONE 2015, 10, e0141602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Na, T.Y.; Schecterson, L.; Mendonsa, A.M.; Gumbiner, B.M. The functional activity of E-cadherin controls tumor cell metastasis at multiple steps. Proc. Natl. Acad. Sci. USA 2020, 117, 5931–5937. [Google Scholar] [CrossRef]
  105. Zhang, Y.; Yang, J.M. Altered energy metabolism in cancer: A unique opportunity for therapeutic intervention. Cancer Biol. Ther. 2013, 14, 81–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lu, J. The Warburg metabolism fuels tumor metastasis. Cancer Metastasis Rev. 2019, 38, 157–164. [Google Scholar] [CrossRef] [PubMed]
  107. Spurlock, B.; Gupta, P.; Basu, M.K.; Mukherjee, A.; Hjelmeland, A.B.; Darley-Usmar, V.; Parker, D.; Foxall, M.E.; Mitra, K. New quantitative approach reveals heterogeneity in mitochondrial structure-function relations in tumor-initiating cells. J. Cell Sci. 2019, 132. [Google Scholar] [CrossRef] [Green Version]
  108. Raninga, P.V.; Lee, A.; Sinha, D.; Dong, L.F.; Datta, K.K.; Lu, X.; Kalita-de Croft, P.; Dutt, M.; Hill, M.; Pouliot, N.; et al. Marizomib suppresses triple-negative breast cancer via proteasome and oxidative phosphorylation inhibition. Theranostics 2020, 10, 5259–5275. [Google Scholar] [CrossRef]
  109. Zancan, P.; Sola-Penna, M.; Furtado, C.M.; Da Silva, D. Differential expression of phosphofructokinase-1 isoforms correlates with the glycolytic efficiency of breast cancer cells. Mol. Genet. Metab. 2010, 100, 372–378. [Google Scholar] [CrossRef]
  110. Daurio, N.A.; Tuttle, S.W.; Worth, A.J.; Song, E.Y.; Davis, J.M.; Snyder, N.W.; Blair, I.A.; Koumenis, C. AMPK activation and metabolic reprogramming by tamoxifen through estrogen receptor-independent mechanisms suggests new uses for this therapeutic modality in cancer treatment. Cancer Res. 2016, 76, 3295–3306. [Google Scholar] [CrossRef] [Green Version]
  111. Van Weverwijk, A.; Koundouros, N.; Iravani, M.; Ashenden, M.; Gao, Q.; Poulogiannis, G.; Jungwirth, U.; Isacke, C.M. Metabolic adaptability in metastatic breast cancer by AKR1B10-dependent balancing of glycolysis and fatty acid oxidation. Nat. Commun. 2019, 10, 2698. [Google Scholar] [CrossRef] [Green Version]
  112. Breton-Romero, R.; Acin-Perez, R.; Rodriguez-Pascual, F.; Martinez-Molledo, M.; Brandes, R.P.; Rial, E.; Enriquez, J.A.; Lamas, S. Laminar shear stress regulates mitochondrial dynamics, bioenergetics responses and PRX3 activation in endothelial cells. Biochim. Biophys. Acta 2014, 1843, 2403–2413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Chen, J.; Cao, S.; Situ, B.; Zhong, J.; Hu, Y.; Li, S.; Huang, J.; Xu, J.; Wu, S.; Lin, J.; et al. Metabolic reprogramming-based characterization of circulating tumor cells in prostate cancer. J. Exp. Clin. Cancer Res. 2018, 37, 127. [Google Scholar] [CrossRef] [PubMed]
  114. Grzybowska-Szatkowska, L.; Slaska, B.; Rzymowska, J.; Brzozowska, A.; Florianczyk, B. Novel mitochondrial mutations in the ATP6 and ATP8 genes in patients with breast cancer. Mol. Med. Rep. 2014, 10, 1772–1778. [Google Scholar] [CrossRef] [Green Version]
  115. Niedzwiecka, K.; Kabala, A.M.; Lasserre, J.P.; Tribouillard-Tanvier, D.; Golik, P.; Dautant, A.; di Rago, J.P.; Kucharczyk, R. Yeast models of mutations in the mitochondrial ATP6 gene found in human cancer cells. Mitochondrion 2016, 29, 7–17. [Google Scholar] [CrossRef]
  116. Maximo, V.; Soares, P.; Lima, J.; Cameselle-Teijeiro, J.; Sobrinho-Simoes, M. Mitochondrial DNA somatic mutations (point mutations and large deletions) and mitochondrial DNA variants in human thyroid pathology: A study with emphasis on Hurthle cell tumors. Am. J. Pathol. 2002, 160, 1857–1865. [Google Scholar] [CrossRef]
  117. Boominathan, A.; Vanhoozer, S.; Basisty, N.; Powers, K.; Crampton, A.L.; Wang, X.; Friedricks, N.; Schilling, B.; Brand, M.D.; O’Connor, M.S. Stable nuclear expression of ATP8 and ATP6 genes rescues a mtDNA Complex V null mutant. Nucleic Acids Res. 2016, 44, 9342–9357. [Google Scholar] [CrossRef] [Green Version]
  118. Isidoro, A.; Martinez, M.; Fernandez, P.L.; Ortega, A.D.; Santamaria, G.; Chamorro, M.; Reed, J.C.; Cuezva, J.M. Alteration of the bioenergetic phenotype of mitochondria is a hallmark of breast, gastric, lung and oesophageal cancer. BioChem. J. 2004, 378, 17–20. [Google Scholar] [CrossRef] [Green Version]
  119. Chen, R.; Park, H.A.; Mnatsakanyan, N.; Niu, Y.; Licznerski, P.; Wu, J.; Miranda, P.; Graham, M.; Tang, J.; Boon, A.J.W.; et al. Parkinson’s disease protein DJ-1 regulates ATP synthase protein components to increase neuronal process outgrowth. Cell Death Dis. 2019, 10, 469. [Google Scholar] [CrossRef] [Green Version]
  120. Alavian, K.N.; Li, H.; Collis, L.; Bonanni, L.; Zeng, L.; Sacchetti, S.; Lazrove, E.; Nabili, P.; Flaherty, B.; Graham, M.; et al. Bcl-xL regulates metabolic efficiency of neurons through interaction with the mitochondrial F1FO ATP synthase. Nat. Cell Biol. 2011, 13, 1224–1233. [Google Scholar] [CrossRef]
  121. Cabezon, E.; Montgomery, M.G.; Leslie, A.G.; Walker, J.E. The structure of bovine F1-ATPase in complex with its regulatory protein IF1. Nat. Struct. Biol. 2003, 10, 744–750. [Google Scholar] [CrossRef]
  122. Wang, Y.Y.; Attane, C.; Milhas, D.; Dirat, B.; Dauvillier, S.; Guerard, A.; Gilhodes, J.; Lazar, I.; Alet, N.; Laurent, V.; et al. Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight 2017, 2, e87489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Alavian, K.N.; Beutner, G.; Lazrove, E.; Sacchetti, S.; Park, H.A.; Licznerski, P.; Li, H.; Nabili, P.; Hockensmith, K.; Graham, M.; et al. An uncoupling channel within the c-subunit ring of the F1FO ATP synthase is the mitochondrial permeability transition pore. Proc. Natl. Acad. Sci. USA 2014, 111, 10580–10585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Mnatsakanyan, N.; Llaguno, M.C.; Yang, Y.; Yan, Y.; Weber, J.; Sigworth, F.J.; Jonas, E.A. A mitochondrial megachannel resides in monomeric F1FO ATP synthase. Nat. Commun. 2019, 10, 5823. [Google Scholar] [CrossRef]
  125. Neginskaya, M.A.; Solesio, M.E.; Berezhnaya, E.V.; Amodeo, G.F.; Mnatsakanyan, N.; Jonas, E.A.; Pavlov, E.V. ATP synthase C-subunit-deficient mitochondria have a small cyclosporine A-sensitive channel, but lack the permeability transition pore. Cell Rep. 2019, 26, 11–17.e12. [Google Scholar] [CrossRef] [Green Version]
  126. Zhang, X.; Gao, F.; Yu, L.L.; Peng, Y.; Liu, H.H.; Liu, J.Y.; Yin, M.; Ni, J. Dual functions of a monoclonal antibody against cell surface F1F0 ATP synthase on both HUVEC and tumor cells. Acta Pharmacol. Sin. 2008, 29, 942–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Gonzalez-Pecchi, V.; Valdes, S.; Pons, V.; Honorato, P.; Martinez, L.O.; Lamperti, L.; Aguayo, C.; Radojkovic, C. Apolipoprotein A-I enhances proliferation of human endothelial progenitor cells and promotes angiogenesis through the cell surface ATP synthase. Microvasc. Res. 2015, 98, 9–15. [Google Scholar] [CrossRef]
  128. Moser, T.L.; Stack, M.S.; Asplin, I.; Enghild, J.J.; Hojrup, P.; Everitt, L.; Hubchak, S.; Schnaper, H.W.; Pizzo, S.V. Angiostatin binds ATP synthase on the surface of human endothelial cells. Proc. Natl. Acad. Sci. USA 1999, 96, 2811–2816. [Google Scholar] [CrossRef] [Green Version]
  129. Speransky, S.; Serafini, P.; Caroli, J.; Bicciato, S.; Lippman, M.E.; Bishopric, N.H. A novel RNA aptamer identifies plasma membrane ATP synthase beta subunit as an early marker and therapeutic target in aggressive cancer. Breast Cancer Res. Treat. 2019, 176, 271–289. [Google Scholar] [CrossRef] [Green Version]
  130. Wang, W.J.; Ma, Z.; Liu, Y.W.; He, Y.Q.; Wang, Y.Z.; Yang, C.X.; Du, Y.; Zhou, M.Q.; Gao, F. A monoclonal antibody (Mc178-Ab) targeted to the ecto-ATP synthase beta-subunit-induced cell apoptosis via a mechanism involving the MAPKase and Akt pathways. Clin. Exp. Med. 2012, 12, 3–12. [Google Scholar] [CrossRef]
  131. Tam, K.W.; Ho, C.T.; Tu, S.H.; Lee, W.J.; Huang, C.S.; Chen, C.S.; Wu, C.H.; Lee, C.H.; Ho, Y.S. Alpha-tocopherol succinate enhances pterostilbene anti-tumor activity in human breast cancer cells in vivo and in vitro. Oncotarget 2018, 9, 4593–4606. [Google Scholar] [CrossRef] [Green Version]
  132. Fukuzawa, K.; Matsuura, K.; Tokumura, A.; Suzuki, A.; Terao, J. Kinetics and dynamics of singlet oxygen scavenging by alpha-tocopherol in phospholipid model membranes. Free Radic. Biol. Med. 1997, 22, 923–930. [Google Scholar] [CrossRef]
  133. Park, H.A.; Mnatsakanyan, N.; Broman, K.; Davis, A.U.; May, J.; Licznerski, P.; Crowe-White, K.M.; Lackey, K.H.; Jonas, E.A. Alpha-tocotrienol prevents oxidative stress-mediated post-translational cleavage of Bcl-xL in primary hippocampal neurons. Int. J. Mol. Sci. 2019, 21, 220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Cuddihy, S.L.; Ali, S.S.; Musiek, E.S.; Lucero, J.; Kopp, S.J.; Morrow, J.D.; Dugan, L.L. Prolonged alpha-tocopherol deficiency decreases oxidative stress and unmasks alpha-tocopherol-dependent regulation of mitochondrial function in the brain. J. Biol. Chem. 2008, 283, 6915–6924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Chow, C.K. Vitamin E regulation of mitochondrial superoxide generation. Neurosignals 2001, 10, 112–124. [Google Scholar] [CrossRef]
  136. Das Gupta, S.; So, J.Y.; Wall, B.; Wahler, J.; Smolarek, A.K.; Sae-Tan, S.; Soewono, K.Y.; Yu, H.; Lee, M.J.; Thomas, P.E.; et al. Tocopherols inhibit oxidative and nitrosative stress in estrogen-induced early mammary hyperplasia in ACI rats. Mol. Carcinog. 2015, 54, 916–925. [Google Scholar] [CrossRef] [Green Version]
  137. Bak, M.J.; Das Gupta, S.; Wahler, J.; Lee, H.J.; Li, X.; Lee, M.J.; Yang, C.S.; Suh, N. Inhibitory effects of gamma- and delta-tocopherols on estrogen-stimulated breast cancer in vitro and in vivo. Cancer Rev. Res. 2017, 10, 188–197. [Google Scholar] [CrossRef] [Green Version]
  138. Gu, W.; Prasadam, I.; Yu, M.; Zhang, F.; Ling, P.; Xiao, Y.; Yu, C. Gamma tocotrienol targets tyrosine phosphatase SHP2 in mammospheres resulting in cell death through RAS/ERK pathway. BMC Cancer 2015, 15, 609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Yang, P.; Zhao, J.; Hou, L.; Yang, L.; Wu, K.; Zhang, L. Vitamin E succinate induces apoptosis via the PI3K/AKT signaling pathways in EC109 esophageal cancer cells. Mol. Med. Rep. 2016, 14, 1531–1537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Tiwari, R.V.; Parajuli, P.; Sylvester, P.W. Synergistic anticancer effects of combined gamma-tocotrienol and oridonin treatment is associated with the induction of autophagy. Mol. Cell. BioChem. 2015, 408, 123–137. [Google Scholar] [CrossRef] [PubMed]
  141. Dronamraju, V.; Ibrahim, B.A.; Briski, K.P.; Sylvester, P.W. Gamma-tocotrienol suppression of the warburg effect is mediated by AMPK activation in human breast cancer cells. Nutr. Cancer 2019, 71, 1214–1228. [Google Scholar] [CrossRef] [PubMed]
  142. Truksa, J.; Dong, L.F.; Rohlena, J.; Stursa, J.; Vondrusova, M.; Goodwin, J.; Nguyen, M.; Kluckova, K.; Rychtarcikova, Z.; Lettlova, S.; et al. Mitochondrially targeted vitamin E succinate modulates expression of mitochondrial DNA transcripts and mitochondrial biogenesis. Antioxid. Redox Signal. 2015, 22, 883–900. [Google Scholar] [CrossRef]
  143. Park, H.A.; Jonas, E.A. DeltaN-Bcl-xL, a therapeutic target for neuroprotection. Neural Regen. Res. 2017, 12, 1791–1794. [Google Scholar] [CrossRef] [PubMed]
  144. Larouche, D.; Hanna, M.; Chang, S.L.; Jacob, S.; Tetu, B.; Diorio, C. Evaluation of antioxidant intakes in relation to inflammatory markers expression within the normal breast tissue of breast cancer patients. Integr. Cancer Ther. 2017, 16, 485–495. [Google Scholar] [CrossRef]
  145. Khanzode, S.S.; Muddeshwar, M.G.; Khanzode, S.D.; Dakhale, G.N. Antioxidant enzymes and lipid peroxidation in different stages of breast cancer. Free Radic. Res. 2004, 38, 81–85. [Google Scholar] [CrossRef] [PubMed]
  146. Harris, H.R.; Orsini, N.; Wolk, A. Vitamin C and survival among women with breast cancer: A meta-analysis. Eur. J. Cancer 2014, 50, 1223–1231. [Google Scholar] [CrossRef] [PubMed]
  147. Pawlowska, E.; Szczepanska, J.; Blasiak, J. Pro- and antioxidant effects of vitamin c in cancer in correspondence to its dietary and pharmacological concentrations. Oxid. Med. Cell. Longev. 2019, 2019, 7286737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Raymond, Y.C.; Glenda, C.S.; Meng, L.K. Effects of high doses of vitamin c on cancer patients in singapore: Nine cases. Integr. Cancer Ther. 2016, 15, 197–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Vollbracht, C.; Schneider, B.; Leendert, V.; Weiss, G.; Auerbach, L.; Beuth, J. Intravenous vitamin C administration improves quality of life in breast cancer patients during chemo-/radiotherapy and aftercare: Results of a retrospective, multicentre, epidemiological cohort study in Germany. In Vivo 2011, 25, 983–990. [Google Scholar]
  150. Lee, S.J.; Jeong, J.H.; Lee, I.H.; Lee, J.; Jung, J.H.; Park, H.Y.; Lee, D.H.; Chae, Y.S. Effect of high-dose vitamin c combined with anti-cancer treatment on breast cancer cells. Anticancer Res. 2019, 39, 751–758. [Google Scholar] [CrossRef]
  151. Cha, J.; Roomi, M.W.; Kalinovsky, T.; Niedzwiecki, A.; Rath, M. Lipoprotein(a) and vitamin C impair development of breast cancer tumors in Lp(a)+; Gulo−/− mice. Int. J. Oncol. 2016, 49, 895–902. [Google Scholar] [CrossRef] [Green Version]
  152. Zeng, L.H.; Wang, Q.M.; Feng, L.Y.; Ke, Y.D.; Xu, Q.Z.; Wei, A.Y.; Zhang, C.; Ying, R.B. High-dose vitamin C suppresses the invasion and metastasis of breast cancer cells via inhibiting epithelial-mesenchymal transition. Onco Targets Ther. 2019, 12, 7405–7413. [Google Scholar] [CrossRef] [Green Version]
  153. Cha, J.; Roomi, M.W.; Ivanov, V.; Kalinovsky, T.; Niedzwiecki, A.; Rath, M. Ascorbate supplementation inhibits growth and metastasis of B16FO melanoma and 4T1 breast cancer cells in vitamin C-deficient mice. Int. J. Oncol. 2013, 42, 55–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Gan, L.; Camarena, V.; Mustafi, S.; Wang, G. Vitamin C inhibits triple-negative breast cancer metastasis by affecting the expression of YAP1 and synaptopodin 2. Nutrients 2019, 11, 2997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Wang, Y.; Xu, X.; Maglic, D.; Dill, M.T.; Mojumdar, K.; Ng, P.K.; Jeong, K.J.; Tsang, Y.H.; Moreno, D.; Bhavana, V.H.; et al. Comprehensive molecular characterization of the hippo signaling pathway in cancer. Cell Rep. 2018, 25, 1304–1317.e1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Nakajima, H.; Yamamoto, K.; Agarwala, S.; Terai, K.; Fukui, H.; Fukuhara, S.; Ando, K.; Miyazaki, T.; Yokota, Y.; Schmelzer, E.; et al. Flow-dependent endothelial YAP regulation contributes to vessel maintenance. Dev. Cell 2017, 40, 523–536.e526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Rosa, C.; Franca, C.; Lanes Vieira, S.; Carvalho, A.; Penna, A.; Nogueira, C.; Lessa, S.; Ramalho, A. Reduction of Serum Concentrations and Synergy between Retinol, beta-Carotene, and Zinc According to Cancer Staging and Different Treatment Modalities Prior to Radiation Therapy in Women with Breast Cancer. Nutrients 2019, 11, 2953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Wu, J.; Yang, R.; Zhang, L.; Li, Y.; Liu, B.; Kang, H.; Fan, Z.; Tian, Y.; Liu, S.; Li, T. Metabolomics research on potential role for 9-cis-retinoic acid in breast cancer progression. Cancer Sci. 2018, 109, 2315–2326. [Google Scholar] [CrossRef] [Green Version]
  159. Sowmya Shree, G.; Yogendra Prasad, K.; Arpitha, H.S.; Deepika, U.R.; Nawneet Kumar, K.; Mondal, P.; Ganesan, P. Beta-carotene at physiologically attainable concentration induces apoptosis and down-regulates cell survival and antioxidant markers in human breast cancer (MCF-7) cells. Mol. Cell. BioChem. 2017, 436, 1–12. [Google Scholar] [CrossRef]
  160. Lin, G.; Zhu, S.; Wu, Y.; Song, C.; Wang, W.; Zhang, Y.; Chen, Y.L.; He, Z. Omega-3 free fatty acids and all-trans retinoic acid synergistically induce growth inhibition of three subtypes of breast cancer cell lines. Sci. Rep. 2017, 7, 2929. [Google Scholar] [CrossRef] [Green Version]
  161. Sabzichi, M.; Mohammadian, J.; Ghorbani, M.; Saghaei, S.; Chavoshi, H.; Ramezani, F.; Hamishehkar, H. Fabrication of all-trans-retinoic acid-loaded biocompatible precirol: A strategy for escaping dose-dependent side effects of doxorubicin. Colloids Surf. B Biointerfaces 2017, 159, 620–628. [Google Scholar] [CrossRef]
  162. Reinhardt, A.; Liu, H.; Ma, Y.; Zhou, Y.; Zang, C.; Habbel, J.P.; Possinger, K.; Eucker, J. Tumor cell-selective synergism of TRAIL- and ATRA-induced cytotoxicity in breast cancer cells. Anticancer Res. 2018, 38, 2669–2682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Wang, J.; Ma, Y.; Yang, J.; Jin, L.; Gao, Z.; Xue, L.; Hou, L.; Sui, L.; Liu, J.; Zou, X. Fucoxanthin inhibits tumour-related lymphangiogenesis and growth of breast cancer. J. Cell. Mol. Med. 2019, 23, 2219–2229. [Google Scholar] [CrossRef] [PubMed]
  164. Khoshnan, A.; Tindell, C.; Laux, I.; Bae, D.; Bennett, B.; Nel, A.E. The NF-kappa B cascade is important in Bcl-xL expression and for the anti-apoptotic effects of the CD28 receptor in primary human CD4+ lymphocytes. J. Immunol. 2000, 165, 1743–1754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Catz, S.D.; Johnson, J.L. Transcriptional regulation of bcl-2 by nuclear factor kappa B and its significance in prostate cancer. Oncogene 2001, 20, 7342–7351. [Google Scholar] [CrossRef] [Green Version]
  166. Zheng, Y.; Miyamoto, D.T.; Wittner, B.S.; Sullivan, J.P.; Aceto, N.; Jordan, N.V.; Yu, M.; Karabacak, N.M.; Comaills, V.; Morris, R.; et al. Expression of beta-globin by cancer cells promotes cell survival during blood-borne dissemination. Nat. Commun. 2017, 8, 14344. [Google Scholar] [CrossRef] [PubMed]
  167. Peralta, E.A.; Viegas, M.L.; Louis, S.; Engle, D.L.; Dunnington, G.L. Effect of vitamin E on tamoxifen-treated breast cancer cells. Surgery 2006, 140, 607–614, discussion 614–605. [Google Scholar] [CrossRef]
  168. Peralta, E.A.; Brewer, A.T.; Louis, S.; Dunnington, G.L. Vitamin E increases biomarkers of estrogen stimulation when taken with tamoxifen. J. Surg. Res. 2009, 153, 143–147. [Google Scholar] [CrossRef]
  169. Ambrosone, C.B.; Zirpoli, G.R.; Hutson, A.D.; McCann, W.E.; McCann, S.E.; Barlow, W.E.; Kelly, K.M.; Cannioto, R.; Sucheston-Campbell, L.E.; Hershman, D.L.; et al. Dietary supplement use during chemotherapy and survival outcomes of patients with breast cancer enrolled in a cooperative group clinical trial (SWOG S0221). J. Clin. Oncol. 2020, 38, 804–814. [Google Scholar] [CrossRef]
  170. LeBleu, V.S.; O’Connell, J.T.; Gonzalez Herrera, K.N.; Wikman, H.; Pantel, K.; Haigis, M.C.; de Carvalho, F.M.; Damascena, A.; Domingos Chinen, L.T.; Rocha, R.M.; et al. PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat. Cell Biol. 2014, 16, 992–1003, 1001–1015. [Google Scholar] [CrossRef] [Green Version]
  171. Cognart, H.A.; Viovy, J.L.; Villard, C. Fluid shear stress coupled with narrow constrictions induce cell type-dependent morphological and molecular changes in SK-BR-3 and MDA-MB-231 cells. Sci. Rep. 2020, 10, 6386. [Google Scholar] [CrossRef] [Green Version]
  172. Landwehr, G.M.; Kristof, A.J.; Rahman, S.M.; Pettigrew, J.H.; Coates, R.; Balhoff, J.B.; Triantafillu, U.L.; Kim, Y.; Melvin, A.T. Biophysical analysis of fluid shear stress induced cellular deformation in a microfluidic device. Biomicrofluidics 2018, 12, 054109. [Google Scholar] [CrossRef] [PubMed]
Table 1. Methods for applying fluid shear stress to cancer cells.
Table 1. Methods for applying fluid shear stress to cancer cells.
Flow ApparatusApplicationReferences
Syringe pumpSingle-pass expulsion of cells from syringe through attached tubing into collection tube[27,30]
Peristaltic pumpCirculatory system that permits multiple passes of cells through a closed loop, permitting the application of wall shear stress and laminar shear stress[28,29,34,35]
Cone and plate viscometerStationary plate positioned beneath a rotating cone in a circulating water bath, permitting a uniform shear rate applied to the cell suspension[31,33]
Orbital/rotary shakerCells in culture containers placed on rotating shakers at a programmed speed (rpm), permitting continuous exposure to fluid shear stress[31,32]
Microfluidic devicesPolymeric devices with inlet and outlet ports that permit the flow of cells through designed channels, ranging in complexity, permitting the observation of cellular behavior[36]
Parallel plate perfusion deviceStationary device with a polymeric distributor, a silicon gasket and a glass coverslip; the distributor contains the inlet and outlet ports, as well as the vacuum slot[37,38]
Computational modeling systemSimulation of metastasizing cells in a 3D environment[41]
Table 2. Current research reporting cellular the mechanisms of the FSS-associated alteration of breast cancer cells applying in vitro and in vivo models.
Table 2. Current research reporting cellular the mechanisms of the FSS-associated alteration of breast cancer cells applying in vitro and in vivo models.
MechanismsReferences
Oxidative stress and antioxidant[28,29,31,43,50,131]
Cell Signaling
MAPK/ERK [28,31,131]
PI3K/Akt [38,86,131]
JNK[70]
RANK[19]
Apoptotic Pathway[29,52,91,93,94,95,102,103,104]
Energy Metabolism[170]
EMT[20,21,27,31,104,170,171]
Stemness[31]
Morphology[171,172]

Share and Cite

MDPI and ACS Style

Park, H.-A.; Brown, S.R.; Kim, Y. Cellular Mechanisms of Circulating Tumor Cells During Breast Cancer Metastasis. Int. J. Mol. Sci. 2020, 21, 5040. https://doi.org/10.3390/ijms21145040

AMA Style

Park H-A, Brown SR, Kim Y. Cellular Mechanisms of Circulating Tumor Cells During Breast Cancer Metastasis. International Journal of Molecular Sciences. 2020; 21(14):5040. https://doi.org/10.3390/ijms21145040

Chicago/Turabian Style

Park, Han-A, Spenser R. Brown, and Yonghyun Kim. 2020. "Cellular Mechanisms of Circulating Tumor Cells During Breast Cancer Metastasis" International Journal of Molecular Sciences 21, no. 14: 5040. https://doi.org/10.3390/ijms21145040

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

Article Metrics

Back to TopTop