Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3382 word(s) 3382 2021-07-22 07:54:29 |
2 format correct Meta information modification 3382 2021-08-03 11:24:33 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Varol, M. ROS in Cancer Progression. Encyclopedia. Available online: https://encyclopedia.pub/entry/12690 (accessed on 25 April 2024).
Varol M. ROS in Cancer Progression. Encyclopedia. Available at: https://encyclopedia.pub/entry/12690. Accessed April 25, 2024.
Varol, Mehmet. "ROS in Cancer Progression" Encyclopedia, https://encyclopedia.pub/entry/12690 (accessed April 25, 2024).
Varol, M. (2021, August 03). ROS in Cancer Progression. In Encyclopedia. https://encyclopedia.pub/entry/12690
Varol, Mehmet. "ROS in Cancer Progression." Encyclopedia. Web. 03 August, 2021.
ROS in Cancer Progression
Edit

Reactive oxygen species (ROS) play a pivotal role in biological processes and continuous ROS production in normal cells is controlled by the appropriate regulation between the silver lining of low and high ROS concentration mediated effects. Interestingly, ROS also dynamically influences the tumor microenvironment and is known to initiate cancer angiogenesis, metastasis, and survival at different concentrations. At moderate concentration, ROS activates the cancer cell survival signaling cascade involving mitogen-activated protein kinase/extracellular signal-regulated protein kinases 1/2 (MAPK/ERK1/2), p38, c-Jun N-terminal kinase (JNK), and phosphoinositide-3-kinase/ protein kinase B (PI3K/Akt), which in turn activate the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), matrix metalloproteinases (MMPs), and vascular endothelial growth factor (VEGF). At high concentrations, ROS can cause cancer cell apoptosis. Hence, it critically depends upon the ROS levels, to either augment tumorigenesis or lead to apoptosis.

reactive oxygen species (ROS) oxidative stress inflammation angiogenesis metastasis miRNA

1. Introduction

Despite continuous efforts in the development of novel treatment modalities, cancer still remains one of the most dreadful diseases in both developed and, developing countries; being an indomitable conundrum for scientists worldwide. Based on the recently published GLOBOCAN 2018 report, there was a prediction of 18.1 million new cancer cases and 9.6 million cases with cancer mortality in women and men. If the industrial development accompanied by environmental pollution and alterations in the human lifestyle, continue as fast as hitherto, predictions suggest by 2030, almost 13 million people will die from different cancers each year [1]. This would be a huge burden to the whole society and needs urgent intervention at different levels, including not only scientific contribution but also political statutes in implementation of diverse preventive measures.
The complexity of cancer is attributed to its multifaceted and multifactorial presentation. In the past few decades, the disruption of redox balance has been illustrated to be one of the most important reasons underlying cancer development, its progression, and metastasis in human cells [2]. This disproportionality in redox homeostasis is documented to be induced via increased free radicals, predominantly ROS. These highly active radicals originate from both intrinsic and extrinsic sources. Intrinsic sources of ROS mostly include mitochondria, inflammatory cells, and several enzymatic cellular complexes; extrinsic sources of ROS include pro-oxidant environmental toxins, radiation, [3] and diverse chemical compounds, including alcohol, tobacco smoking, and certain drugs [4]. Such free radicals can cause damage to various important biomolecules, including lipids, proteins, and nucleic acids, leading to oxidative stress and damaging different human tissues.
Elevated ROS levels, accompanied with down-regulation of cellular antioxidant enzyme systems, result in malignant transformation via different molecular targets, such as NF-κB and nuclear factor (erythroid-derived 2)-like-2 factor (Nrf2) [5][6]. Signaling cascades regulated by these key factors generate an inflammatory environment leading to the suppression of apoptotic cell death, tumor proliferation, angiogenesis, and metastasis; which cumulatively augment initiation, development, and progression of malignant neoplasms (Table 1). This review article summarizes the present up-to-date knowledge of the molecular mechanisms of ROS generation, its role in tumor growth and metastasis, along with highlighting the potential targets of the ROS cascade for potential therapeutic strategies.
Table 1. Role of ROS in cancer progression.
Effect Mechanism Cell Line References
Oxidative Stress Aquaporin AQP5-mediated H2O2 influx rate indicates the presence of a highly efficient peroxiporin activity and consequently activates signaling networks related to cell survival and cancer progression Pancreatic Carcinoma line-3 (BxPC3) [7]
  PCB118 promotes hepatocellular carcinoma cell (HCC) proliferation via Pyruvate kinase M2 (PKM2)-dependent up-regulation of glycolysis, which is mediated by Aryl hydrocarbon receptor/Nicotinamide adenine dinucleotide phosphate oxidase (AhR/NADPH oxidase)-induced ROS prouction SMMC-7721 [8]
  Enhanced ROS of exposed cells alters the mitochondrial metabolic activities in terms of increased mitochondrial mass and DNA content and initiates cancer progression through modifying cellar biomarkers MOE1A [9]
Inflammatory markers Serum ROS and damaged mtDNA may be markers of mitochondrial metabolism through oxygenation of the primary tumor and results in systemic inflammation and adverse outcomes of locally advanced rectal cancer (LARC) HCT-116, HT-29, and LoVo [10]
  Inflammation in the stroma induces TNF-α signaling and the NOX1/ROS signaling pathway is activated downstream with expression of TLR2 which is an important tumor-promoting mechanism stimulated by inflammation Mouse Model [11]
  Alkylating agents may evoke inflammatory responses that seem to contribute to malignant progression in specific breast cancer cells MDA-MB231, Hs578T, SKBR3 and MCF7 [12]
Metastasis ROS induce epithelial-mesenchymal transition (EMT), the glycolytic switch, and mitochondrial repression by activating the Distal-less homeobox-2 (Dlx-2)/Snail axis, thereby playing crucial roles in metastasis MCF-7 [13]
  Elevated mitochondrial ROS via fatty acid β-oxidation, activates the MAPK cascades, results in EMT process of ROS high tumor spheres (RH-TS) cells, and enhances metastasis 4 T1, SW480, HCT116 and HT29 [14]
  Loss of TMEM126A induces ROS production with mitochondrial dysfunction and subsequently metastasis by activating extracellular matrix (ECM) remodeling and EMT MDA-MB-231HM [15]
  PM2.5 exposure induces ROS, which activates loc146880 expression and promotes the malignant behavior A549 [16]
Angiogenesis ROS-ERK1/2-HIF-1α-VEGF-induces angiogenesis by increased level of RRM2 C33A and MCF-7 [17]
  High glucose increases angiogenesis and decreases apoptosis due to activation of the NF-κB pathway by increasing ROS MCF-7 [18]
  27-Hydroxycholesterol (27HC) enhanced the generation of ROS and activates the STAT-3/VEGF signaling in an ER independent manner which results in induced angiogenesis Breast Cancer Cells [19]

2. Role of ROS in Cancer Progression

2.1. ROS-Mediated Induction of Oxidative Stress

Oxygen is a multifaceted molecule, which is crucial for life sustainability but can be harmful when it is converted to ROS through oxidation-reduction reactions. The distinct aspect of ROS was first identified by Gerschmann in 1954 [20][21] and its free radical potential was described by Denham Harman in the free radical theory of aging in 1956 [21][22]. This initial work from the group; steadily triggered further investigations in the dimension of ROS to decipher their precise role in biological systems [21]. The discovery of the first described antioxidant enzyme, superoxide dismutase (SOD), by McCord [21][23] postulated the instrumental role of ROS through free radical generation. Following the initial studies on the substantial potential role of ROS, the focus shifted to delineate the beneficial and harmful effects of ROS in numerous pathological and physiological processes and their mechanism of action.
In a normal cell, ROS levels are balanced through numerous detoxification processes regulated through antioxidant enzymes. Hence, ROS homeostasis is well sustained, which contributes to the maintenance of redox balance in healthy cells. The complex I and III of the mitochondrial respiratory chain under high membrane potential, are contemplated to be the point of origin of ROS particularly, but numerous other resources may also play a pivotal role in elevated ROS generations, such as α-ketoglutarate dehydrogenase, monoamine oxidase, mitochondrial p66Shc, sirtuins, Nrf2, and forkhead box O3 (FOXO3) besides the underlying redox cycling reactions [24]. This elevated ROS production or defective defense mechanism may also lead to elevated oxidative stress levels leading to diverse pathological conditions. Examples of endogenous sources of ROS include mitochondrial oxidative phosphorylation, p450 metabolism, peroxisomes, and from the activation of inflammatory cells such as macrophages and neutrophils. It is thought that during the mitochondrial respiratory process, 1–2% of molecular oxygen is converted to ROS through one to three electron reductions and this leads to the formation of hydroxyl, hydrogen peroxide, superoxide, and peroxynitrite radicals [25].
Oxidative stress is one of the main leading causes of toxicity which is attributed to the interactions of ROS as well as reactive nitrogen species (RNS) with cellular macromolecules such as DNA, lipid, and proteins, which interfere with the signal transduction pathways such as protein kinases, phosphatases, and transduction mechanisms (Figure 1). The pathological consequences of oxidative stress are characterized by impaired glucose tolerance due to mitochondrial oxidative stress. This is attributed to pro-oxidants changing their thiol/disulphide redox state which leads to diabetes mellitus and cancer or through the augmented action of either NAD(P)H oxidase leading to inflammatory oxidative conditions which is associated with chronic inflammation and atherosclerosis or through the action of xanthine oxidase-induced ROS formation which has been associated with reperfusion injury and ischemia [21]. Further, the ageing process can be attributed to the detrimental magnitude of free radicals leading to DNA damage, lipid peroxidation, and protein oxidation [21][22].
Figure 1. Oxidative stress and production of reactive oxygen species. Intracellular ROS and environmental factors (exogenous ROS) initiates ROS production leading to oxidative stress which in turn leads to DNA/lipid/protein degradation resulting in apoptosis, autophagy, necrosis and production of pro-inflammatory cytokines.
The connotation of oxidative and nitrosative stress with chronic and acute disease presentation is based on the biomarkers validated for oxidative stress. In an excellent review by Dalle-Donne and coworkers [21][26], the group summarized the biomarkers of oxidative stress and their correlation with human disease presentation. Natural antioxidants can protect the cell via scavenging the ROS. They may be separated into three classes—endogenous antioxidants (bilirubin, catalase (CAT), ferritin, SOD, glutathione, coenzyme Q, l-carnitine, alpha lipoic acid, glutathione peroxidase (GPx), melatonin, metallothionein, thioredoxins, peroxiredoxins, and uric acid), natural antioxidants (ascorbic acid, polyphenol metabolites, β-carotene, vitamin E, and vitamin A), and synthetic antioxidants (Nrf2, tiron, pyruvate, selenium, and N-acetyl cysteine (NAC)). Protecting the organism against harmful oxidants is a complex interaction between these antioxidants. High intracellular ROS concentration is important for damage, but another important fact is the equilibrium between ROS and antioxidant systems. The ROS production/antioxidant defense system balance is required for homeostasis [25]. Under normal conditions antioxidants outbalance oxidants but under oxidative conditions pro-oxidants prevail antioxidants [27].

2.2. Inflammatory Markers and ROS

The intricate relationship between cancer and, prolonged inflammation has been thoroughly investigated since Virchow’s hypothesis [28]. In 1863, Rudolf Virchow propounded that the “lymphoreticular infiltrate” reflected the origin of cancer at the locations of chronic inflammation [29]. The obtained epidemiological and experimental data from the numerous studies supported Virchow’s hypothesis and revealed that inflammatory processes regulate the course of cancer based on the level of inflammation-related factors, inflammatory cytokines, and chemokines, in the tumor microenvironment, either by producing an antitumor response or by inducing cell transformation and malignancy [30][31][32]. One of the major regulatory components in the relationship between cancer and chronic inflammation is ROS that has an ability to affect the type, presence, and levels of inflammation-modulating factors such as activator protein 1 (AP-1), β-catenin/Wnt (wingless related integration site), HIF-1α (hypoxia-inducible factor-1 alpha), NF-κB, PPAR-γ (peroxisome proliferator-activated receptor gamma), p53, inflammatory cytokines, chemokines, and growth factors [27][33][34][35]. However, it should be noted that there is a complex crosstalk between chronic inflammation, ROS accumulation, and cancer progression (Figure 2). The inflammatory cells in the tumor microenvironment lead to a massive production of ROS by activating the oxidant-generating enzymes such as inducible nitric oxide synthase (iNOS), myeloperoxidase (MPO), NADPH oxidase, and xanthine oxidase (XO), and by up-regulating cyclooxygenase 2 (COX2) and lipoxygenase (LOX) to remove and disrupt the biological, chemical, and physical factors. The abundant accumulation of ROS also produces oxidative damage to the DNA, RNA, proteins, lipids, and mitochondria.
Figure 2. Schematic illustration of mechanism of action of reactive oxygen species (ROS) leading to inflammation. ADAM17 (ADAM metallopeptidase domain 17); ASC (Activating signal co-integrator 1); BMP4 (Bone morphogenetic protein 4); IKB-α (Inhibitor of nuclear factor kappa B kinase regulatory subunit alpha); IKK (Inhibitor of nuclear factor kappa-B kinase); IP3R (Inositol 1,4,5-trisphosphate receptor type 3); JNK (c-Jun N-terminal kinase); LPC (Lysophosphatidylcholine); LPS (Lipopolysaccharide); NF-кB (Nuclear factor kappa subunit B); NLRP3 (NLR family pyrin domain containing 3); NOX (NADPH oxidase); OxPL (Oxidized phospholipids); PAR (Par family cell polarity regulator); PAK (p21 (RAC1) activated kinase); SOD (Superoxide dismutase); TLR4 (Toll like receptor 4); TNF-α (Tumor necrosis factor alpha); TNFR (TNF receptor superfamily); TNFR1 (TNF receptor superfamily 1); TXNIP (Thioredoxin interacting protein); Ub (Ubiquitin).
This causes an increased mutation load, defects in signal-transduction, inactivation of apoptosis, and overpowered generation of additional ROS that activate the inflammation-modulating factors, inflammatory cytokines, and chemokines [34][35][36][37]. Apart from the induction of chronic inflammation by ROS-mediated NF-κB activation, the active NF-κB is considered to be a key component in the rise of therapy-resistant cancers toward fractional gamma-irradiation therapy and chemotherapeutic agents such as 5-fluorouracil, bortezomib, cisplatin, daunorubicin, doxorubicin, paclitaxel, vinblastine, vincristine, and tamoxifen, through the transcriptional up-regulation of Akt, Bcl-2 (B-cell lymphoma 2), Bcl-xL (B-cell lymphoma- extra-large), cyclin D1, COX-2, survivin, and XIAP (X-linked inhibitor of apoptosis) [38][39][40][41][42][43]. Targeting ROS consequently seems to be a very promising way to modulate cancer related chronic inflammation and the hallmarks for cancer development such as sustaining proliferative signaling, evading growth suppressors, activating invasion and metastasis, enabling replicative immortality, inducing angiogenesis, and resisting cell death [37][44].

2.3. Cancer Metastasis and ROS

Metastasis involves the spread of cancer cells from the primary tumor to the surrounding tissues and to distant organs, and is the primary cause of morbidity and mortality [45]. Studies reveal that tumor metastasis is not an autonomous program but a complex and multifaceted event, occurring due to the intrinsic mutational burden of cancerous cells and bidirectional interaction between nonmalignant and malignant cells [46]. It occurs due to the up-regulation of several transcriptional factors such as NF-ĸB, ETS-1 (ETS proto-oncogene 1, transcription factor), Twist, Snail, AP-1, and Zeb (zinc finger E-box binding homeobox); metalloproteases viz. MMP-9 and, MMP-2; and chemokines or cytokines like transforming growth factor beta (TGF-β) (Figure 3) [47]. ROS plays an important role in the migration and invasion of cancerous cells. ROS are mainly produced as byproducts during mitochondrial electron transport in aerobic respiration and have numerous deleterious effects [48]. Epithelial to mesenchymal transition (EMT) is the major cause of tumor metastasis, where epithelial cells lose their polarity, cell-cell adhesion, and gain mobility.
Figure 3. Reactive oxygen species and metastasis. High levels of reactive oxygen species leads to metastasis through the stimulation of phosphoinositide-3-kinase regulatory subunit/AKT serine/threonine kinases/mechanistic target of rapamycin kinase (PI3K/Akt/mTOR), and MAPK (Mitogen-activated protein kinases) signaling pathways which activates downstream SNAIL, MMP2 (metalloproteinase 2), and MMP9 (metalloproteinase 9) enzymes initiating epithelial-mesenchymal transition (EMT) leading to metastasis.
Several studies have proved ROS to be a major cause of EMT. TGF-β1 regulates uPA (Urokinase type Plasminogen Activator) and MMP-9 to facilitate cell migration and invasion through ROS-dependent mechanisms [49]. Another study revealed that ROS increases tumor migration by inducing hypoxia mediated MMPs and, cathepsin expression [50][51]. According to a study reported by Zhang, NADPH oxidase 4 (NOX4)-dependent ROS production is necessary for TGF β1-induced EMT in MDAMB-231C and MCG-10A cell lines [52]. P53 also plays a major role in cell migration using cytokines TGF β1. Pelicano et al. suggested that mitochondrial dysfunction can lead to increased ROS production, which further up-regulates C-X-C motif chemokine 14 (CXCL14) expression through the AP-1 signaling pathway and, enhances cell mobility by elevating cytosolic Ca2+ levels [53]. ROS activate Nrf2 that stimulates Klf9 (Krupple like factor 9), thus activating ERK1/2; and results in an increased ROS production in cancer cells. Thus, a premalignant growth can be suppressed by using topical antioxidants that target Klf9 [54][55].
Mitochondrial Ca2+ also plays an important role in cancer metastasis. Jin and his colleagues observed that MCUR1 (Mitochondrial Calcium Uniporter Regulator 1) is up-regulated in hepatocellular carcinoma (HCC) which promotes EMT by activating ROS/Notch1/Nrf2 pathways. Thus, MCUR1 can be a potential target for the treatment of HCC [56]. Aydin et al. analyzed that NOX2 generates ROS, which influence metastasis by down-modulating the function of natural killer (NK) cells, and its inhibition can restore the IFNγ (interferon gamma)-dependent NK cell-mediated clearance of myeloma cells [57]. Vimentin protein also plays a major role in cancer initiation and progression such as EMT, and metastasis. Oxidative stress caused by HIF-1 regulates vimentin gene transcription, which helps in the formation of invadopodia during cancer cell invasion and migration [58]. Inhibition of vimentin expression by RNAi can reduce cell metastasis and hence decrease tumor volume [59]. ROS also induces epigenetic changes in the promoter region of E-cadherin and various other tumor suppressor genes, hence leading to tumor progression and metastasis. It causes hyper-methylation of the promoter gene by increasing Snail expression. Snail induces DNA methylation with the help of histone deacetylase 1 (HDAC1) and DNA methyltransferase 1 (DNMT1) [60].

2.4. Angiogenesis and ROS

During the initial stages of tumorigenesis, new blood vessels are formed from the pre-existing vasculatures, a process known as angiogenesis, which supports tumor proliferation and survival [61][62][63][64]. ROS-dependent angiogenesis is initiated through cancer proliferation, which in turn increases the metabolic rate leading to the generation of high ROS levels [63][64]. These elevated ROS levels lead to oxidative stress in the tumor microenvironment, which initiates secretion of angiogenic modulators [65]. Both endogenous and exogenous ROS spearheads stimulation of growth factors, cytokines, and transcription factors like VEGF and HiF-1α, which promote tumor migration and proliferation through ROS-dependent cellular signaling [62][65][66]. The signaling cascade through ROS mediation has been documented to perpetuate VEGF secretion and activate the PI3K/Akt/mammalian target of the rapamycin (mTOR) pathway through hypoxia independent or dependent mechanisms (through stabilization of HIF-1α which increases production of VEGF) (Figure 4), [67]. Additionally, the Ras signaling pathway has also been reported to up-regulate VEGF secretion [68]. Recently, mutant p53 was also recognized to modulate the angiogenic response in tumor proliferation through the ROS-mediated activation of VEGF-A and HiF-1 in HCT116 human colon carcinoma cells [69]. The mechanism of ROS-mediated angiogenesis has been extensively studied to understand the signaling cascade modulating cancer progression. In a study carried out using MDA-MB-231 breast cancer cells, deferoxamine (DFO) induced HIF-1α through ERK1/2 phosphorylation which promoted tumor migration and metastasis [70].
Figure 4. Angiogenesis activation through reactive oxygen species (ROS) via hypoxia dependent and hypoxia independent pathways. The hypoxia dependent pathway increases vascular endothelial growth factor (VEGF) expression via the phosphoinositide-3-kinase regulatory subunit/AKT serine/threonine kinases/mechanistic target of rapamycin kinase (PI3K/Akt/mTOR), PTEN (phosphatase and tensin homolog), and MAPK (Mitogen-activated protein kinases) signaling cascades via HIF-1α (Hypoxia-inducible factor1-alpha) and p70S6K1 (ribosomal protein S6 kinase B1), which release various cytokines, growth factors, and up-regulation of MMPs (matrix metalloproteinases) leading to angiogenesis. The hypoxia independent pathway leads to angiogenesis through oxidative lipid ligands which activates NF-кB (Nuclear factor kappa subunit B) via Toll-like receptors (TLRs).
Han and colleagues demonstrated that the elevated levels of epidermal growth factor (EGF) triggered hydrogen peroxide production, which stimulated p70S6K1 via the PI3K/Akt signaling pathway, leading to the activation of downstream VEGF and HiF-1α [71]. On similar lines, Liu et al. reported that EGF lead to increased production of hydrogen peroxide in OVCAR-3 ovarian cancer cells, which activated the AKT/p70S6K1 pathway, thereby resulting in increased VEGF expression [72]. The group further documented that catalase overexpression and rapamycin inhibited angiogenesis. Hydrogen peroxide was also illustrated to inactivate phosphatase and tension homolog (PTEN) through the reversible oxidation of phosphatases in the cysteine thiol group and promote activation of the PI3K/Akt/mTORsignaling cascade and Ras [73].
In another study on ovarian cancer cells, Xia and colleagues documented that NOX4 knockdown lead to a reduction of VEGF and HIF-1α, which in turn regulated tumor angiogenesis [74][75]. A similar mechanism of action of ROS was illustrated through experiments in WM35 melanoma cells, where Akt induced the expression of NOX4 [75][76]. NADPH oxidase 2 (Nox2)-derived ROS was also reported to induce cancer progression and migration, modulated through the ERK/PI3K/AKT/Src (Proto-oncogene tyrosine -protein kinase)-dependent pathway leading to the activation of endothelial cells and induction of angiogenesis [77][78][79]. Nox1 (NADPH oxidase 1) was also reported to mediate the Ras-dependent up-regulation of VEGF expression and, angiogenesis via the Ras/ERK-dependent Sp1 phosphorylation and activation in CaCO-2 colon cancer cells [80][81]. In human umbilical vein endothelial cell (HUVECs), Angiopoietin -1 (Ang1) induced transient ROS through the activation of endothelial specific tyrosine kinase receptor, Tie-2, and p44/42, MAPK leading to vascular remodeling [82].
Moreover, copper was reported to increase the ROS-mediated expression of VEGF, HiF-1α, and G-protein estrogen receptor (GPER) in HepG2 hepatocellular carcinoma cells and SkBr3 breast cancer cells via activation of the EGFR/ERK/c-Fos pathway [83][84]. Similarly, the cadmium activated ERK/Akt pathway induced ROS expression and HiF-1; its downstream pro-angiogenic molecule in BEAS-2B bronchial epithelial cells [85]. In addition, several other extracellular remodeling proteins and transcription factors (p53, HiF-1α, VEGF, and MMPs) have been documented to be regulated by ROS [86][87][88][89][90][91]. Also, numerous studies in cancer cell lines (MCF-7, HepG2, H-1299, PC-3) showed that ROS acted through the PI3K/Akt signaling cascade, thereby enhancing HiF-1α expression and angiogenesis [92][93][94]. In view of literature supporting the angiogenic potential of ROS, therapeutic targets with antioxidant potential and targeting subsequent signaling cascades will be of clinical significance in the management and treatment of patients through down-regulation of neovascularization. Different strategies to restore redox imbalance is another avenue to improve outcome of diseases. As the interventional trials with small anti-oxidants have not been effective, further research to explore disease-specific ROS may be of clinical relevance for futuristic drug development.

References

  1. The Lancet. GLOBOCAN 2018: Counting the toll of cancer. Lancet 2018, 392.
  2. Saikolappan, S.; Kumar, B.; Shishodia, G.; Koul, S.; Koul, H.K. Reactive oxygen species and cancer: A complex interaction. Cancer Lett. 2019, 452, 132–143.
  3. Nourazarian, A.R.; Kangari, P.; Salmaninejad, A. Roles of oxidative stress in the development and progression of breast cancer. Asian Pac. J. Cancer Prev. 2014, 15, 4745–4751.
  4. Zhang, J.; Ahn, K.S.; Kim, C.; Shanmugam, M.K.; Siveen, K.S.; Arfuso, F.; Samym, R.P.; Deivasigamanim, A.; Lim, L.H.K.; Wang, L.; et al. Nimbolide-induced oxidative stress abrogates STAT3 signaling cascade and inhibits tumor growth in transgenic adenocarcinoma of mouse prostate model. Antioxid. Redox Signal. 2016, 24, 575–589.
  5. Morgan, M.J.; Liu, Z.-g. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115.
  6. Puar, Y.; Shanmugam, M.; Fan, L.; Arfuso, F.; Sethi, G.; Tergaonkar, V. Evidence for the involvement of the master transcription factor NF-κB in cancer initiation and progression. Biomedicines 2018, 6, 82.
  7. Rodrigues, C.; Pimpão, C.; Mósca, A.F.; Coxixo, A.S.; Lopes, D.; da Silva, I.V.; Pedersen, P.A.; Antunes, F.; Soveral, G. Human Aquaporin-5 Facilitates Hydrogen Peroxide Permeation Affecting Adaption to Oxidative Stress and Cancer Cell Migration. Cancers 2019, 11, 932.
  8. Liang, W.; Zhang, Y.; Song, L.; Li, Z. 2, 3′4, 4′, 5-Pentachlorobiphenyl induces hepatocellular carcinoma cell proliferation through pyruvate kinase M2-dependent glycolysis. Toxicol. Lett. 2019, 313, 108–119.
  9. Sarkar, R.; Kishida, S.; Kishida, M.; Nakamura, N.; Kibe, T.; Karmalar, D.; Chaudhuri, C.R.; Barui, A. Effect of cigarette smoke extract on mitochondrial heme-metabolism: An in vitro model of oral cancer progression. Toxicol. In Vitro 2019, 60, 336–346.
  10. Bousquet, P.A.; Meltzer, S.; Sønstevold, L.; Esbensen, Y.; Dueland, S.; Flatmark, K.; Sitter, B.; Bathen, T.F.; Seierstad, T.; Redalen, K.R.; et al. Markers of mitochondrial metabolism in tumor hypoxia, systemic inflammation, and adverse outcome of rectal cancer. Transl. Oncol. 2019, 12, 76–83.
  11. Echizen, K.; Oshima, H.; Nakayama, M.; Oshima, M. The inflammatory microenvironment that promotes gastrointestinal cancer development and invasion. Adv. Biol. Regul. 2018, 68, 39–45.
  12. Zanotto-Filho, A.; Rajamanickam, S.; Loranc, E.; Masamsetti, V.P.; Gorthi, A.; Romero, J.C.; Tonapi, S.; Gonçalves, R.M.; Reddick, R.L.; Benavides, R.; et al. Sorafenib improves alkylating therapy by blocking induced inflammation, invasion and angiogenesis in breast cancer cells. Cancer Lett. 2018, 425, 101–115.
  13. Lee, S.Y.; Ju, M.K.; Jeon, H.M.; Lee, Y.J.; Kim, C.H.; Park, H.G.; Han, S.I.; Kang, H.S. Reactive oxygen species induce epithelial--mesenchymal transition, glycolytic switch, and mitochondrial repression through the Dlx--2/Snail signaling pathways in MCF--7 cells. Mol. Med. Rep. 2019, 20, 2339–2346.
  14. Wang, C.; Shao, L.; Pan, C.; Ye, J.; Ding, Z.; Wu, J.; Du, Q.; Ren, Y.; Zhu, C. Elevated level of mitochondrial reactive oxygen species via fatty acid β-oxidation in cancer stem cells promotes cancer metastasis by inducing epithelial–mesenchymal transition. Stem Cell Res. Ther. 2019, 10.
  15. Sun, H.F.; Yang, X.L.; Zhao, Y.; Tian, Q.; Chen, M.T.; Zhao, Y.Y.; Jin, W. Loss of TMEM126A promotes extracellular matrix remodeling, epithelial-to-mesenchymal transition, and breast cancer metastasis by regulating mitochondrial retrograde signaling. Cancer Lett. 2019, 440, 189–201.
  16. Deng, X.; Feng, N.; Zheng, M.; Ye, X.; Lin, H.; Yu, X.; Gan, Z.; Fang, Z.; Zhang, H.; Gao, M.; et al. PM2.5 exposure-induced autophagy is mediated by lncRNA loc146880 which also promotes the migration and invasion of lung cancer cells. Biochimica Biophysica Acta Gen. Subj. 2017, 1861, 112–125.
  17. Wang, N.; Zhan, T.; Ke, T.; Huang, X.; Ke, D.; Wang, Q.; Li, H. Increased expression of RRM2 by human papillomavirus E7 oncoprotein promotes angiogenesis in cervical cancer. Br. J. Cancer 2014, 110, 1034–1044.
  18. Kansestani, A.N.; Mansouri, K.; Hemmati, S.; Zare, M.E.; Moatafaei, A. High Glucose-reduced Apoptosis in Human Breast Cancer Cells Is Mediated by Activation of NF-κB. Iran. J. Allergy Asthma Immunol. 2019, 18, 153–162.
  19. Zhu, D.; Shen, Z.; Liu, J.; Chen, J.; Liu, Y.; Hu, C.; Li, Z.; Li, Y. The ROS-mediated activation of STAT-3/VEGF signaling is involved in the 27-hydroxycholesterol-induced angiogenesis in human breast cancer cells. Toxicol. Lett. 2016, 264, 79–86.
  20. Gerschmann, R.; Gilbert, D.; Nye, S.W.; Dwyer, P.; Fenn, W.O. Oxygen poisoning and X-irradiation: A mechanism in common.1954. Nutrition 2001, 17, 162.
  21. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84.
  22. Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956, 11, 298–300.
  23. McCord, J.M.; Fridovich, I. Superoxide dismutase an enzymic function for erythrocuprein (hemocuprein). J. Biol. Chem. 1969, 244, 6049–6055.
  24. Lenaz, G. Mitochondria and reactive oxygen species. Which role in physiology and pathology? Adv. Exp. Med. Biol. 2012, 942, 93–136.
  25. Franco, R.; Schoneveld, O.; Georgakilas, A.G.; Panayiotidis, M.I. Oxidative stress, DNA methylation and carcinogenesis. Cancer Lett. 2008, 266, 6–11.
  26. Dalle-Donne, I.; Rossi, R.; Colombo, R.; Giustarini, D.; Milzani, A. Biomarkers of oxidative damage in human disease. Clin. Chem. 2006, 52, 601–623.
  27. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616.
  28. Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From inflammation to cancer. Ir. J. Med. Sci. 2017, 186, 57–62.
  29. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545.
  30. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899.
  31. Gonda, T.A.; Tu, S.; Wang, T.C. Chronic inflammation, the tumor microenvironment and carcinogenesis. Cell Cycle 2009, 8, 2005–2013.
  32. Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic inflammation and cytokines in the tumor microenvironment. J. Immunol. Res. 2014.
  33. Wu, Y.; Antony, S.; Meitzler, J.L.; Doroshow, J.H. Molecular mechanisms underlying chronic inflammation-associated cancers. Cancer Lett. 2014, 345, 164–173.
  34. Kashyap, D.; Tuli, H.S.; Sak, K.; Garg, V.K.; Goel, N.; Punia, S.; Chaudhary, A. Role of Reactive Oxygen Species in Cancer Progression. Curr. Pharmacol. Rep. 2019, 5, doi.
  35. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive oxygen species in metabolic and inflammatory signaling. Circ. Res. 2018, 122, 877–902.
  36. Azad, N.; Rojanasakul, Y.; Vallyathan, V. Inflammation and lung cancer: Roles of reactive oxygen/nitrogen species. J. Toxicol. Environ. Health B Crit. Rev. 2008, 11, 1–15.
  37. Liu, X.; Chen, Z. The pathophysiological role of mitochondrial oxidative stress in lung diseases. J. Transl. Med. 2017, 15.
  38. Li, F.; Sethi, G. Targeting transcription factor NF-κB to overcome chemoresistance and radioresistance in cancer therapy. Biochimica Biophysica Acta 2010, 1805, 167–180.
  39. Zheng, H.C. The molecular mechanisms of chemoresistance in cancers. Oncotarget 2017, 8, 59950–59964.
  40. Chua, A.W.L.; Hay, H.S.; Rajendran, P.; Shanmugam, M.K.; Li, F.; Bist, P.; Koay, E.S.; Lim, L.H.; Kumar, A.P.; Sethi, G. Butein downregulates chemokine receptor CXCR4 expression and function through suppression of NF-κB activation in breast and pancreatic tumor cells. Biochem. Pharmacol. 2010, 80, 1553–1562.
  41. Nair, A.S.; Shishodia, S.; Ahn, K.S.; Kunnumakkara, A.B.; Sethi, G.; Aggarwal, B.B. Deguelin, an Akt inhibitor, suppresses IκBα kinase activation leading to suppression of NF-κB-regulated gene expression, potentiation of apoptosis, and inhibition of cellular invasion. J. Immunol. 2006, 177, 5612–5622.
  42. Ahn, K.S.; Sethi, G.; Chaturvedi, M.M.; Aggarwal, B.B. Simvastatin, 3--hydroxy--3--methylglutaryl coenzyme A reductase inhibitor, suppresses osteoclastogenesis induced by receptor activator of nuclear factor--κB ligand through modulation of NF--κB pathway. Int. J. Cancer 2008, 123, 1733–1740.
  43. Manna, S.K.; Aggarwal, R.S.; Sethi, G.; Aggarwal, B.B.; Ramesh, G.T. Morin (3, 5, 7, 2′, 4′-pentahydroxyflavone) abolishes nuclear factor-κB activation induced by various carcinogens and inflammatory stimuli, leading to suppression of nuclear factor-κB–regulated gene expression and up-regulation of apoptosis. Clin. Cancer Res. 2007, 13, 2290–2297.
  44. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  45. Seyfried, T.N.; Huysentruyt, L.C. On the origin of cancer metastasis. Crit. Rev. Onco. 2013, 18, 43–73.
  46. Brooks, S.A.; Lomax-Browne, H.J.; Carter, T.M.; Kinch, C.E.; Hall, D.M. Molecular interactions in cancer cell metastasis. Acta Histochemica 2010, 112, 3–25.
  47. Guerrini, L.; Garcia-Rico, E.; Alvarez-Puebla, R. The Role of Nanoscience in Cancer Diagnosis. In Handbook of Nanomaterials for Cancer Theranostics; Conde, J., Ed.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 177–197.
  48. Chitty, J.L.; Filipe, E.C.; Lucas, M.C.; Herrmann, D.; Cox, T.R.; Timpson, P. Recent advances in understanding the complexities of metastasis. F1000Research 2018.
  49. Liao, Z.; Chua, D.; Tan, N.S. Reactive oxygen species: A volatile driver of field cancerization and metastasis. Mol. Cancer 2019, 18.
  50. Kamiya, T.; Goto, A.; Kurokawa, E.; Hara, H.; Adachi, T. Cross talk mechanism among EMT, ROS, and histone acetylation in phorbol ester-treated human breast cancer MCF-7 cells. Oxid. Med. Cell. Longev. 2016.
  51. Shin, D.H.; Dier, U.; Melendez, J.A.; Hempel, N. Regulation of MMP-1 expression in response to hypoxia is dependent on the intracellular redox status of metastatic bladder cancer cells. Biochim. Biophys. Acta 2015, 1852, 2593–2602.
  52. Zhang, B.; Liu, Z.; Hu, X. Inhibiting cancer metastasis via targeting NAPDH oxidase 4. Biochem. Pharmacol. 2013, 86, 253–266.
  53. Pelicano, H.; Lu, W.; Zhou, Y.; Zhang, W.; Chen, Z.; Hu, Y.; Huang, P. Mitochondrial dysfunction and reactive oxygen species imbalance promote breast cancer cell motility through a CXCL14-mediated mechanism. Cancer Res. 2009, 69, 2375–2383.
  54. Bagati, A.; Moparthy, S.; Fink, E.E.; Bianchi-Smiraglia, A.; Yun, D.H.; Kolesnikova, M.; Udartseva, O.O.; Wolff, D.W.; Roll, M.V.; Lipchick, B.C.; et al. KLF9-dependent ROS regulate melanoma progression in stage-specific manner. Oncogene 2019, 38, 3585–3597.
  55. Zucker, S.N.; Fink, E.E.; Bagati, A.; Mannava, S.; Bianchi-Smiraglia, A.; Bogner, P.N.; Wawrzyniak, J.A.; Foley, C.; Leonova, K.I.; Grimm, M.J.; et al. Nrf2 amplifies oxidative stress via induction of Klf9. Mol. Cell 2014, 53, 916–928.
  56. Jin, M.; Wang, J.; Ji, X.; Cao, H.; Zhu, J.; Chen, Y.; Yang, J.; Zhao, Z.; Ren, T.; Xing, J. MCUR1 facilitates epithelial-mesenchymal transition and metastasis via the mitochondrial calcium dependent ROS/Nrf2/Notch pathway in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2019, 38.
  57. Aydin, E.; Johansson, J.; Nazir, F.H.; Hellstrand, K.; Martner, A. Role of NOX2-derived reactive oxygen species in NK cell–mediated control of murine melanoma metastasis. Cancer Immunol. Res. 2017, 5, 804–811.
  58. Kidd, M.E.; Shumaker, D.K.; Ridge, K.M. The role of vimentin intermediate filaments in the progression of lung cancer. Am. J. Respir. Cell Mol. Biol. 2014, 50, 1–6.
  59. Paccione, R.J.; Miyazaki, H.; Patel, V.; Waseem, A.; Gutkind, J.S.; Zehner, Z.E.; Yeudall, W.A. Keratin down-regulation in vimentin-positive cancer cells is reversible by vimentin RNA interference, which inhibits growth and motility. Mol. Cancer Ther. 2008, 7, 2894–2903.
  60. Lim, S.O.; Gu, J.M.; Kim, M.S.; Kim, H.S.; Park, Y.N.; Park, C.K.; Cho, J.W.; Park, Y.M.; Jung, G. Epigenetic changes induced by reactive oxygen species in hepatocellular carcinoma: Methylation of the E-cadherin promoter. Gastroenterology 2008, 135, 2128–2140.
  61. Senger, D.R.; Brown, L.F.; Claffey, K.P.; Dvorak, H.F. Vascular permeability factor, tumor angiogenesis and stroma generation. Invasion Metastasis 1994, 14, 385–394.
  62. Ushio-Fukai, M.; Nakamura, Y. Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 2008, 266, 37–52.
  63. de Sa Junior, P.L.; Camara, D.A.D.; Porcacchia, A.S.; Fonseca, P.M.M.; Jorge, S.D.; Araldi, R.P.; Ferreira, A.K. The Roles of ROS in Cancer Heterogeneity and Therapy. Oxid. Med. Cell. Longev. 2017.
  64. Schito, L. Hypoxia-Dependent Angiogenesis and Lymphangiogenesis in Cancer. Adv. Exp. Med. Biol. 2019, 1136, 71–85.
  65. Dewhirst, M.W.; Cao, Y.; Moeller, B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat. Rev. Cancer 2008, 8, 425–437.
  66. Ushio-Fukai, M.; Alexander, R.W. Reactive oxygen species as mediators of angiogenesis signaling: Role of NAD(P)H oxidase. Mol. Cell. Biochem. 2004, 264, 85–97.
  67. Karar, J.; Maity, A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front. Mol. Neurosci. 2011, 4.
  68. Rezatabar, S.; Karimian, A.; Rameshknia, V.; Parsian, H.; Majidinia, M.; Kopi, T.A.; Bishayee, A.; Sadeghinia, A.; Yousefi, M.; Monirialamdari, M.; et al. RAS/MAPK signaling functions in oxidative stress, DNA damage response and cancer progression. J. Cell. Physiol. 2019.
  69. Khromova, N.V.; Kopnin, P.B.; Stepanova, E.V.; Agapova, L.S.; Kopnin, B.P. p53 hot-spot mutants increase tumor vascularization via ROS-mediated activation of the HIF1/VEGF-A pathway. Cancer Lett. 2009, 276, 143–151.
  70. Liu, Y.; Cui, Y.; Shi, M.; Zhang, Q.; Wang, Q.; Chen, X. Deferoxamine promotes MDA-MB-231 cell migration and invasion through increased ROS-dependent HIF-1alpha accumulation. Cell. Physiol. Biochem. 2014, 33, 1036–1046.
  71. Han, X.; Sun, S.; Zhao, M.; Cheng, X.; Chen, G.; Lin, S.; Guan, Y.; Yu, X. Celastrol stimulates hypoxia-inducible factor-1 activity in tumor cells by initiating the ROS/Akt/p70S6K signaling pathway and enhancing hypoxia-inducible factor-1alpha protein synthesis. PLoS One 2014, 9.
  72. Liu, L.Z.; Hu, X.W.; Xia, C.; He, J.; Zhou, Q.; Shi, X.; Fang, J.; Jiang, B.H. Reactive oxygen species regulate epidermal growth factor-induced vascular endothelial growth factor and hypoxia-inducible factor-1alpha expression through activation of AKT and P70S6K1 in human ovarian cancer cells. Free Radic. Biol. Med. 2006, 41, 1521–1533.
  73. Brewer, T.F.; Garcia, F.J.; Onak, C.S.; Carroll, K.S.; Chang, C.J. Chemical approaches to discovery and study of sources and targets of hydrogen peroxide redox signaling through NADPH oxidase proteins. Annu. Rev. Biochem. 2015, 84, 765–790.
  74. Xia, C.; Meng, Q.; Liu, L.Z.; Rojanasakul, Y.; Wang, X.R.; Jiang, B.H. Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor. Cancer Res. 2007, 67, 10823–10830.
  75. Jin, F.; Wu, Z.; Hu, X.; Zhang, J.; Gao, Z.; Han, X.; Qin, J.; Li, C.; Wang, Y. The PI3K/Akt/GSK-3beta/ROS/eIF2B pathway promotes breast cancer growth and metastasis via suppression of NK cell cytotoxicity and tumor cell susceptibility. Cancer Biol. Med. 2019, 16, 38–54.
  76. Govindarajan, B.; Sligh, J.E.; Vincent, B.J.; Li, M.; Canter, J.A.; Nickoloff, B.J.; Rodenburg, R.J.; Smeitink, J.A.; Oberley, L.; Zhang, Y.; et al. Overexpression of Akt converts radial growth melanoma to vertical growth melanoma. J. Clin. Invest. 2007, 117, 719–729.
  77. Giannoni, E.; Fiaschi, T.; Ramponi, G.; Chiarugi, P. Redox regulation of anoikis resistance of metastatic prostate cancer cells: Key role for Src and EGFR-mediated pro-survival signals. Oncogene 2009, 28, 2074–2086.
  78. Aydin, E.; Hallner, A.; Grauers Wiktorin, H.; Staffas, A.; Hellstrand, K.; Martner, A. NOX2 inhibition reduces oxidative stress and prolongs survival in murine KRAS-induced myeloproliferative disease. Oncogene 2019, 38, 1534–1543.
  79. Bino, L.; Vesela, I.; Papezikova, I.; Prochazkova, J.; Vasicek, O.; Stefkova, K.; Kucera, J.; Hanackova, M.; Kubala, L.; Pachernik, J. The depletion of p38alpha kinase upregulates NADPH oxidase 2/NOX2/gp91 expression and the production of superoxide in mouse embryonic stem cells. Arch. Biochem. Biophys. 2019, 671, 18–26.
  80. Komatsu, D.; Kato, M.; Nakayama, J.; Miyagawa, S.; Kamata, T. NADPH oxidase 1 plays a critical mediating role in oncogenic Ras-induced vascular endothelial growth factor expression. Oncogene 2008, 27, 4724–4732.
  81. De Bessa, T.C.; Pagano, A.; Moretti, A.I.S.; Oliveira, P.V.S.; Mendonca, S.A.; Kovacic, H.; Laurindo, F.R.M. Subverted regulation of Nox1 NADPH oxidase-dependent oxidant generation by protein disulfide isomerase A1 in colon carcinoma cells with overactivated KRas. Cell Death Dis. 2019, 10.
  82. Kim, Y.M.; Kim, K.E.; Koh, G.Y.; Ho, Y.S.; Lee, K.J. Hydrogen peroxide produced by angiopoietin-1 mediates angiogenesis. Cancer Res. 2006, 66, 6167–6174.
  83. Gupte, A.; Mumper, R.J. Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat. Rev. 2009, 35, 32–46.
  84. Rigiracciolo, D.C.; Scarpelli, A.; Lappano, R.; Pisano, A.; Santolla, M.F.; De Marco, P.; Cirillo, F.; Cappello, A.R.; Dolce, V.; Belfiore, A.; et al. Copper activates HIF-1alpha/GPER/VEGF signalling in cancer cells. Oncotarget 2015, 6, 34158–34177.
  85. Jing, Y.; Liu, L.Z.; Jiang, Y.; Zhu, Y.; Guo, N.L.; Barnett, J.; Rojanasakul, Y.; Agani, F.; Jiang, B.H. Cadmium increases HIF-1 and VEGF expression through ROS, ERK, and AKT signaling pathways and induces malignant transformation of human bronchial epithelial cells. Toxicol. Sci. 2012, 125, 10–19.
  86. Coso, S.; Harrison, I.; Harrison, C.B.; Vinh, A.; Sobey, C.G.; Drummond, G.R.; Williams, E.D.; Selemidis, S. NADPH oxidases as regulators of tumor angiogenesis: Current and emerging concepts. Antioxid. Redox Signal. 2012, 16, 1229–1247.
  87. Sauer, H.; Wartenberg, M. Reactive oxygen species as signaling molecules in cardiovascular differentiation of embryonic stem cells and tumor-induced angiogenesis. Antioxid. Redox Signal. 2005, 7, 1423–1434.
  88. Harrison, I.P.; Vinh, A.; Johnson, I.R.D.; Luong, R.; Drummond, G.R.; Sobey, C.G.; Tiganis, T.; Williams, E.D.; JJ, O.L.; Brooks, D.A.; et al. NOX2 oxidase expressed in endosomes promotes cell proliferation and prostate tumour development. Oncotarget 2018, 9, 35378–35393.
  89. Mori, K.; Uchida, T.; Yoshie, T.; Mizote, Y.; Ishikawa, F.; Katsuyama, M.; Shibanuma, M. A mitochondrial ROS pathway controls matrix metalloproteinase 9 levels and invasive properties in RAS-activated cancer cells. FEBS J. 2019, 286, 459–478.
  90. Guo, D.; Wang, Q.; Li, C.; Wang, Y.; Chen, X. VEGF stimulated the angiogenesis by promoting the mitochondrial functions. Oncotarget 2017, 8, 77020–77027.
  91. Matsuda, S.; Nakagawa, Y.; Kitagishi, Y.; Nakanishi, A.; Murai, T. Reactive Oxygen Species, Superoxide Dimutases, and PTEN-p53-AKT-MDM2 Signaling Loop Network in Mesenchymal Stem/Stromal Cells Regulation. Cells 2018, 7, 36.
  92. Ma, L.; Fu, Q.; Xu, B.; Zhou, H.; Gao, J.; Shao, X.; Xiong, J.; Gu, Q.; Wen, S.; Li, F.; et al. Breast cancer-associated mitochondrial DNA haplogroup promotes neoplastic growth via ROS-mediated AKT activation. Int. J. Cancer 2018, 142, 1786–1796.
  93. Monteiro, H.P.; Rodrigues, E.G.; Amorim Reis, A.K.C.; Longo, L.S., Jr.; Ogata, F.T.; Moretti, A.I.S.; da Costa, P.E.; Teodoro, A.C.S.; Toledo, M.S.; Stern, A. Nitric oxide and interactions with reactive oxygen species in the development of melanoma, breast, and colon cancer: A redox signaling perspective. Nitric Oxide 2019, 89, 1–13.
  94. Zhou, X.; Yue, G.G.; Chan, A.M.; Tsui, S.K.; Fung, K.P.; Sun, H.; Pu, J.; Lau, C.B. Eriocalyxin B, a novel autophagy inducer, exerts anti-tumor activity through the suppression of Akt/mTOR/p70S6K signaling pathway in breast cancer. Biochem. Pharmacol. 2017, 142, 58–70.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 515
Revisions: 2 times (View History)
Update Date: 03 Aug 2021
1000/1000