Cancer is a complex disorder extremely dependent on its microenvironment and highly regulated by multiple intracellular and extracellular stimuli. Studies show that reactive oxygen and nitrogen species (RONS) play key roles in cancer initiation and progression. Accumulation of RONS caused by imbalance between RONS generation and activity of antioxidant system (AOS) has been observed in many cancer types. This leads to alterations in gene expression levels, signal transduction pathways, and protein quality control machinery, that is, processes that regulate cancer cell proliferation, migration, invasion, and apoptosis. This review focuses on the latest advancements evidencing that RONS-induced modifications of key redox-sensitive residues in regulatory proteins, that is, cysteine oxidation/S-sulfenylation/S-glutathionylation/S-nitrosylation and tyrosine nitration, represent important molecular mechanisms underlying carcinogenesis. The oxidative/nitrosative modifications cause alterations in activities of intracellular effectors of MAPK- and PI3K/Akt-mediated signaling pathways, transcription factors (Nrf2, AP-1, NFκB, STAT3, and p53), components of ubiquitin/proteasomal and autophagy/lysosomal protein degradation systems, molecular chaperones, and cytoskeletal proteins. Redox-sensitive proteins, RONS-generating enzymes, and AOS components can serve as targets for relevant anticancer drugs. Chemotherapeutic agents exert their action via RONS generation and induction of cancer cell apoptosis, while drug resistance associates with RONS-induced cancer cell survival; this is exploited in selective anticancer therapy strategies. Cancer Res; 78(21); 6040–7. ©2018 AACR.

Cancer cells grow in low oxygen concentration environment denoted as hypoxia and adapt their metabolism to meet elevated requirements in energy and nutrients for proliferation and survival. Metabolic reprogramming is a hallmark of cancer cell phenotype that is characterized by increased anaerobic glycolysis, deficiency in oxidative phosphorylation and ATP generation, and the overall mitochondrial dysfunction (1).

In response to hypoxia, an elevated production of reactive oxygen and nitrogen species (RONS) has been observed in various cancer cells (2). Mitochondria and Nox family NADPH oxidases are major internal sources of primary endogenous reactive oxygen species (ROS), superoxide anion radical (O2). The latter spontaneously or enzymatically, by superoxide dismutases (SOD), gives rise to hydrogen peroxide (H2O2) that, in turn, in the presence of Fe2+ produces hydroxyl radical (HO•; refs. 3, 4). Nitric oxide synthases (NOS) use l-arginine to produce primary RNS type, nitric oxide radical, •NO, that interacts with O2 yielding another RNS, peroxynitrite (ONOO; ref. 5).

High reactivity of RONS and toxicity of their elevated concentrations for cells dictate the necessity of scavenging mechanisms. Under physiologic conditions, cellular redox homeostasis is maintained by balance between RONS generation and activity of endogenous antioxidant system (AOS; ref. 6). Both enzymatic and nonenzymatic AOS components terminate RONS propagation to result in low intracellular RONS concentrations that cause reversible oxidative/nitrosative modifications of redox-sensitive residues in regulatory proteins. This underlies redox switches in activities of intracellular effectors of cell signaling pathways and transcription factors. This phenomenon denoted as ROS/RNS signaling plays a key role in regulation of cell proliferation, differentiation, migration, and survival (7).

Oppositely, disbalance between RONS generation and AOS activity causes RONS accumulation and oxidative/nitrosative stress, which contribute to pathogenesis of various diseases including cancer (8). Studies evidence that oxidative/nitrosative stress causes alterations in signal transduction pathways, activity of transcription factors, ubiquitin/proteasomal and lysosomal/autophagy-mediated protein degradation, and cytoskeletal organization to contribute to disturbances in cellular homeostasis and higher level of proteotoxic stress in cancer cells as compared with normal cells (9). This review focuses on recent advancements in studying RONS-induced protein modifications as mechanisms underlying the abovementioned alterations to cause cancer initiation and progression.

Oxidative/nitrosative stress may both cause and modify tumor growth through multiple cellular and molecular events such as DNA damage and genome instability, shaping tumor microenvironment, and alteration in cell signaling, which transforms normal cells into malignant and neoplastic cells. Various human cancer types including ovarian, prostate, hepatic, bladder, breast, gastric, oral, and pharyngeal cancers can produce in vitro much greater amount of RONS and protein oxidation adducts and have decreased AOS activity as compared with nontransformed cells (10–13). Indeed, in HT29 colon carcinoma cell line, c-Src tyrosine kinase induces ROS generation by Nox1 through Rac-dependent mechanism (14). Hepatocellular carcinoma (HCC) cells overexpressed Nox1, DUOX1, and DUOX2, while normal cells failed to express all the three NADPH oxidases (15). Both DUOX1 and DOUX2 were highly expressed in MHCC-97H and MHCC-97L, but not in Bel-7402 cells. High Nox1/2/5 levels in patients with HCC correlated with expression of genes associated with cell survival and metastasis and poor prognosis (16).

Growth factors and cytokines can stimulate RONS production (Fig. 1). Indeed, TGFβ1 has been shown to activate Rac1 and stimulates ROS generation by NADPH oxidase accompanied by NFκB and MMP-2 activation, IL6 release, and enhanced SW1990 invasiveness (17). In addition, EGF stimulates NADPH oxidase and heme oxygenase-1 to activate NFκB via c-Src and PI3K/PKB/Akt-mediated signaling and to enhance HT-29 colon cancer cells' proliferation (18). Prolonged •NO production by iNOS promoted angiogenesis and oral squamous cell carcinoma progression (19). In addition, •NO generation by tumor-associated macrophages prevented tumor cells apoptosis and caused chemoresistance to cisplatin (20). However, in breast, colorectal, epidermoid, head, and neck tumors decrease in tetrahydrobiopterin level causes NOS uncoupling and production of O2 and NOO¬ instead of NO• to promote tumorigenesis (21). Reconstruction of coupled NOS activity shifted downstream signaling toward increased cGMP-dependent PKG activation, reduced β-catenin expression, and decreased NFκB activity.

Figure 1.

RONS-regulated cell signaling pathways and transcription factors involved in cancer initiation and progression. Reversible oxidative/nitrosative modifications of redox-sensitive residues including Cys oxidation/S-sulfenylation/S-glutathionylation/S-nitrosylation and Tyr oxidation/nitration in enzymes, intracellular effectors of signal transduction pathways, and transcription factors enable redox switches in their activities. This provides regulation of cancer cell proliferation, differentiation, migration and apoptosis, and cytoskeleton reorganization.

Figure 1.

RONS-regulated cell signaling pathways and transcription factors involved in cancer initiation and progression. Reversible oxidative/nitrosative modifications of redox-sensitive residues including Cys oxidation/S-sulfenylation/S-glutathionylation/S-nitrosylation and Tyr oxidation/nitration in enzymes, intracellular effectors of signal transduction pathways, and transcription factors enable redox switches in their activities. This provides regulation of cancer cell proliferation, differentiation, migration and apoptosis, and cytoskeleton reorganization.

Close modal

However, in some solid and hematologic malignancies, oxidative stress can induce apoptosis, and this may be exploited in selective anticancer therapy strategies (22). Ionizing radiation and chemotherapeutic agents work through either direct or indirect RONS generation followed by accumulation of oxidized proteins, DNA damage, and cell-cycle arrest. Indeed, tamoxifen, paclitaxel, and As2O3 cytotoxicity associates with O2, H2O2, and NO• accumulation, while cancer cell chemoresistance to these agents is proportional to activity of antioxidant genes (23, 24). Furthermore, docosahexaenoic acid (DHA) sensitizes As2O3-resistant HL-60 cells to ROS-inducing anticancer agents and enhances apoptosis through increased proapoptotic Bax protein expression, caspase-3 activation, and mitochondrial inner membrane disruption (25). DHA-induced cytotoxicity was prevented by intracellular GSH and glutathione peroxidase (GPx) elevation, which enhances cancer cell motility and metastatic capacity (26). Thus, both RONS-generating enzymes and AOS components can be targets for anticancer therapy.

RONS-MAPK signaling

Studies show that in cancer tissues, RONS activates all three members of MAPK family stress-responsible protein kinases including ERK1/2, JNK, and p38 (Fig. 1). Stimulation of RONS-MAPK signaling is involved in proliferation, migration, and invasion of human breast, liver, prostate, lung, skin, and pancreatic cancer cells. Indeed, dose-dependent O2 and H2O2-induced migration of MDA-MB-231 breast cancer cells via activation of lysophosphatidic acid–stimulated PI3K/PAK1/ERK signaling has been reported (27). ROS caused ERK1/2 and p38 activation, and IL1α-enhanced IL8 secretion in human pancreatic cancer cells (28), while HGF-induced H2O2 generation and JNK phosphorylation were observed in HCC cells (29).

In malignant lung epithelial cells, stimulation of caspase-9 and p38/MAPK associates with increased •NO and antiapoptotic Bcl-2 levels, suggesting that Bcl-2 S-nitrosylation is critical for tumorigenesis (30). Furthermore, TPA-dependent ROS generation induces PKCα activation, sustained ERK1/2 phosphorylation, and integrin-α5, -α6, -β1 expression followed by HepG2 cell migration (31). This involves FAK, Src, and paxillin phosphorylation to enhance tumor progression.

In addition, chemotherapeutic agents trigger cancer cell apoptosis and autophagy through dose-dependent stimulation of ROS-MAPK signaling. Indeed, ROS-ERK1/2 signaling was involved in HCT116 human colon cancer cell autophagy caused by short-term treatment with MS-275, histone deacetylase inhibitor (32). Long-term treatment with MS-275 led to ROS-p38/MAPK-mediated autophagy-to-apoptosis switch. Besides, salinomycin induces autophagy-to-apoptosis switch in chemoresistant PC-3 prostate cancer cells via both ROS-ERK/p38 and ROS-PI3K/Akt/mTOR signaling (33). Furthermore, AICAR-stimulated ROS-mediated AMPK, energy sensor, activation induces DU-145 prostate cancer cell apoptosis through JNK phosphorylation, caspase-3 activation, and mTOR inhibition (34).

Mechanisms underlying RONS-induced MAPK activation involve oxidative modification and inhibition of various PTPs that dephosphorylate MAPKs. Specific oxidation of redox-sensitive Cys215 in PTPB1 followed by its inhibition was observed in HepG2 and A341 cancer cells (35). At 0.1 mmol/L H2O2, Cys215 was oxidized both reversibly to sulfenic acid, and irreversibly to sulfinic/sulfonic acids, while at 10 mmol/L H2O2 only irreversible Cys215 oxidation occurs (Table 1). In addition, H2O2-induced PTEN catalytic domain oxidation/inactivation causes both ERK1/2 and Akt phosphorylation/activation to enhance DU145 cell migration and invasion (36).

Table 1.

Types of protein oxidative/nitrosative modifications responsible for redox switches in protein activities and relevant to cancer growth

RONS typesType of a protein modificationModified residuesModified proteinCellular responseReferences
H2O2 (dose-dependent) Oxidation/S-sulfenylation/sulfinylation/sulfonylation Cys215 PTPB1 phosphatase ROS-MAPK signaling 35 
O2- and H2O2 Oxidation/S-sulfenylation Cys124 Akt2 Cell migration, G1–S cell-cycle transition 40 
H2O2 Oxidation/S-sulfenylation, Cys71-Cys124 disulfide bonding Cys124 PTEN Stimulation of ROS-PI3K/Akt signaling and cell proliferation 36, 42 
•NO Oxidation/S-nitrosylation Cys118 Wild-type Ras Activation of PI3K/PKB/Akt pathway 43 
H2O2 Oxidation/S-glutathionylation Cys179 IKKβ Prevention of TNFα-induced NFκB activation 46 
•NO Oxidation/nitration Tyr181 IκBα IKK-independent activation of NFκB 47 
•NO Oxidation/S-nitrosylation Cys254 c-FLIP IKK-independent activation of NFκB 50 
•NO Oxidation/nitration Tyr1007/Tyr1008 JAK2 Progression of JAK/STAT/Akt signaling 51 
•NO, ONOO Oxidation/nitration Tyr327 Wild-type p53 p53 DNA-binding and nuclear accumulation 54 
H2O2 and HOCl Oxidation/Cys226-Cys613 disulfide bond formation Cys151, Cys226, Cys273, Cys288, Cys613 Keap1 Prevention of Nrf2 UPS-mediated degradation 57 
H2O2 Oxidation/S-sulfenylation Cys293 MKP-3 MKP-3 UPS-mediated degradation and stimulation of ROS-ERK1/2 signaling 62 
•NO Oxidation/S-nitrosylation Cys128, Cys226 Bcl-2 Prevention of Bcl-2 UPS-mediated degradation and Bcl-2 stabilization 30, 66 
H2O2, O2 Oxidation/S-sulfenylation Cys128, Cys226 Bcl-2 Bcl-2 UPS-mediated degradation 66 
H2O2 Oxidation/S-glutathionylation Cys442 HSP60 ERK1/2 phosphorylation and activation 69 
H2O2 Oxidation/S-glutathionylation Cys18 Protein disulfide isomerase ERK1/2 phosphorylation and activation 69 
H2O2 Oxidation/S-glutathionylation Cys374, Met44, Met47 Actin and vimentin Cytoskeleton remodeling 75, 76 
RONS typesType of a protein modificationModified residuesModified proteinCellular responseReferences
H2O2 (dose-dependent) Oxidation/S-sulfenylation/sulfinylation/sulfonylation Cys215 PTPB1 phosphatase ROS-MAPK signaling 35 
O2- and H2O2 Oxidation/S-sulfenylation Cys124 Akt2 Cell migration, G1–S cell-cycle transition 40 
H2O2 Oxidation/S-sulfenylation, Cys71-Cys124 disulfide bonding Cys124 PTEN Stimulation of ROS-PI3K/Akt signaling and cell proliferation 36, 42 
•NO Oxidation/S-nitrosylation Cys118 Wild-type Ras Activation of PI3K/PKB/Akt pathway 43 
H2O2 Oxidation/S-glutathionylation Cys179 IKKβ Prevention of TNFα-induced NFκB activation 46 
•NO Oxidation/nitration Tyr181 IκBα IKK-independent activation of NFκB 47 
•NO Oxidation/S-nitrosylation Cys254 c-FLIP IKK-independent activation of NFκB 50 
•NO Oxidation/nitration Tyr1007/Tyr1008 JAK2 Progression of JAK/STAT/Akt signaling 51 
•NO, ONOO Oxidation/nitration Tyr327 Wild-type p53 p53 DNA-binding and nuclear accumulation 54 
H2O2 and HOCl Oxidation/Cys226-Cys613 disulfide bond formation Cys151, Cys226, Cys273, Cys288, Cys613 Keap1 Prevention of Nrf2 UPS-mediated degradation 57 
H2O2 Oxidation/S-sulfenylation Cys293 MKP-3 MKP-3 UPS-mediated degradation and stimulation of ROS-ERK1/2 signaling 62 
•NO Oxidation/S-nitrosylation Cys128, Cys226 Bcl-2 Prevention of Bcl-2 UPS-mediated degradation and Bcl-2 stabilization 30, 66 
H2O2, O2 Oxidation/S-sulfenylation Cys128, Cys226 Bcl-2 Bcl-2 UPS-mediated degradation 66 
H2O2 Oxidation/S-glutathionylation Cys442 HSP60 ERK1/2 phosphorylation and activation 69 
H2O2 Oxidation/S-glutathionylation Cys18 Protein disulfide isomerase ERK1/2 phosphorylation and activation 69 
H2O2 Oxidation/S-glutathionylation Cys374, Met44, Met47 Actin and vimentin Cytoskeleton remodeling 75, 76 

Direct MAPK oxidation also contributes to cancer cell proliferative phenotype. Indeed, selective H2O2-induced oxidation/S-sulfenylation of ERK1/2, JNK, and p38/MAPK increases their binding to cognate upstream MAPKKs/MEKs and sustained LP07 murine tumor cell proliferation (37). Thus, in addition to intricate ways of MAPK spatiotemporal localization and phosphorylation dynamics, endogenous reversible oxidation/S-sulfenylation of redox-sensitive Cys in MAPKs and PTPs serve as fine-tuning mechanisms underlying regulation of MAPK-mediated signaling.

RONS-PI3K/Akt signaling

ROS-induced PI3K/Akt signaling plays a key role in acquisition of malignant phenotype by normal cells and survival of cancer cells through Akt activation or its negative regulator, PTEN, inactivation (Fig. 1). ROS trigger PI3K activation and PKB/Akt-mediated upregulation of cell-cycle genes (cdc2, PRC1, and PCNA) contributing to E2- and 4-OH-E2–induced malignant transformation of MCF-10A human mammary epithelial cells (38). In addition, exogenous LPA-induced ERK and Akt phosphorylation/stimulation, and NFκB activation promote SKOV3 ovarian cancer cell proliferation through O2 and H2O2 production (39). Mechanism of LPA-mediated cancer cell signaling involves Akt2 Cys124 oxidation/S-sulfenylation followed by loss of its kinase activity, G1–S cell-cycle transition and stimulated cell migration (40).

Indirect stimulation of ROS-Akt signaling occurs through oxidative modification/inactivation of tumor suppressors, PTEN, and CDC25A phosphatases. In E2-dependent MCF-7 cells, this causes NRF-1 activation, p27 inhibition, and stimulated cell proliferation (41). PTEN transient inactivation results from H2O2-induced Cys124 oxidation/S-sulfenylation and Cys71-Cys124 disulfide bonding (42). Oxidative regulation of PI3K/Akt signaling involves GSH, peroxiredoxins (Prx), and glutaredoxins (Grx) that control ROS and oxidized protein levels.

Stimulation of PI3K/Akt signaling and cancer cell proliferation can be achieved through eNOS and •NO-induced Cys118 oxidation/S-nitrosylation in wild-type Ras small GTPases (43). It is known that mutations in ras genes lead to permanently activated Ras proteins, and mutated human H-Ras, K-Ras, and N-Ras remain in GTP-bound oncogenic state. Induction of ROS production by Nox1 in K-Ras–dependent manner is considered as a key step in cellular malignant transformation and tumorigenesis (44).

Activation of transcription factors

Coordinated ROS-mediated activation of NFκB, p53, AP-1, HIF-1α, and STAT3 transcription factors is involved in expression of many genes controlling cancer cell metabolism and survival (Fig. 1). Indeed, EGF- and TNFα-induced ROS generation and MAPK-mediated signaling cause coordinated activation of NFκB and AP-1 during skin carcinogenesis (45). ROS increase DNA binding capacity of both NFκB and AP-1 and enhance proliferation of pancreatic cancer cells through IL8 secretion and ERK1/2 and p38 MAPK signaling (28).

NFκB is involved in cell response to various stress stimuli including oxidative stress, heavy metals, proinflammatory cytokines, and infectious agents to promote prosurvival gene expression. Canonical pathway of NFκB activation is triggered by phosphorylation of IκB, NFκB inhibitor, with IκB kinases (IKK) resulted in IκB release and UPS-mediated degradation. IKK-dependent mechanism can underlie association between inflammation and cancer growth because H2O2-induced IKKβ Cys179 oxidation/S-glutathionylation reversed by Grx1 prevents TNFα-enhanced NFκB activation (46).

IKK-independent NFκB activation involves •NO-induced IκBα Tyr181 nitration (47) and Tyr42 phosphorylation through H2O2-induced c-Src oxidation during hypoxia/reoxygenation followed by IκBα dissociation from its complex with NFκB (48). During hypoxia, mitochondrial ROS promote HIF-1α expression and HepG2 hepatoma, SH-SY5Y neuroblastoma, and DLD-1 colon carcinoma cell survival via c-Src stimulation and NFκB activation (49). In addition, NFκB can be activated by •NO-induced Cys254 S-nitrosylation in caspase-like domain of c-FLIP, an inhibitor of TNFα, FasL, and TRAIL-induced apoptosis (50).

NFκB can act cooperatively with STAT3 that is phosphorylated/activated by JAK tyrosine kinases. RONS altered phosphorylation of JAK2 through growth hormone–associated •NO-induced Tyr1007/Tyr1008 nitration and JAK/STAT/Akt signaling (51). ROS generation by NADPH oxidase activates JAK2/STAT3 pathway and IL6 synthesis to induce autophagy of starved or rapamycin-treated HeLa cells (52). In addition, antineoplastic agent, BITC, can inhibit pancreatic carcinogenesis via apoptosis induction and proliferation/migration inhibition through ROS-induced downregulation of c-Myc and STAT3/phosphorylated STAT3 (53).

Another transcription factor, p53 tumor suppressor, triggers cell-cycle arrest and apoptosis, but can be also involved in maintaining cellular redox homeostasis via promoting expression of genes encoding MnSOD and GPx1 to prevent DNA damage, genomic instability, and accumulation of oxidized proteins. Elevated RONS inhibit wild-type p53 through Cys oxidation and Tyr nitration by ONOO associated with creating tumor inflammatory microenvironment as observed in human malignant glioblastoma (54).

RONS-induced alterations in activities of another master regulator of antioxidant cell response, Nrf2, and its negative regulator, Keap1, also have been observed. Under nonstressed conditions, Nrf2 binds to Keap1 to undergo degradation through UPS-mediated pathway. Under oxidative stress conditions, Nrf2 releases from complex with Keap1, to translocate into the nucleus, where it heterodimerizes with small Maf protein and binds to antioxidant responsive element (ARE) of antioxidant genes (55). Mutations in KEAP1 and NRF2 genes disrupt Keap1–Nrf2 complex followed by persistent Nrf2 activation to create tumor cell prosurvival environment and anticancer drug resistance.

Keap1 inhibition due to oxidation of redox-sensitive Cys residues causes conformational changes in Nrf2-Keap1-Cul3 E3 ligase complex to prevent Nrf2 UPS-mediated degradation. Large Cys amount in Keap1 provides its role as a sensor to various electrophiles/oxidants named Nrf2 inducers (56). Keap1 exposure to RONS causes Cys226 oxidation and Cys226-Cys613 disulfide bonding (Table 1), while Cys151 oxidation triggers intermolecular disulfide bonding (57). Conserved Cys residues in Nrf2 itself are also critical to electrophile sensing to control Nrf2 binding to target ARE, UPS-mediated degradation, and coactivator recruitment (58).

Oxidative stress–mediated Nrf2 expression is implicated in ERα-positive breast cancer cell survival and tamoxifen resistance (24). However, RONS-induced controlled pharmacologic Nrf2 upregulation can inhibit cancer cell proliferation and migration. Indeed, bardoxolone-methyl causes mitochondrial ROS-induced Nrf2 activation to downregulate glycolysis and oxidative phosphorylation, to decrease intracellular GSH level and inhibit MCF-7 cell migration (59). In addition, resveratrol, a natural phytoestrogen, significantly upregulates Nrf2 causing SOD3, NQO1, and OGG1 antioxidant gene expression, and inhibited E2-induced breast cancer cell migration and apoptosis induction (60). Different cell response to pharmacologic Nrf2 activation may be explained by interplay with other transcription factors and diverse mechanisms underlying regulation of Keap1-Nrf2 signaling in target gene- and inducer-dependent manner.

UPS- and lysosomal/autophagy-mediated protein degradation

ROS-induced carcinogenesis may be caused by UPS-mediated degradation of regulators of ROS-generating enzymes and intracellular effectors of signaling pathways. For example, EGF-mediated NoxO1 phosphorylation triggers formation of enzymatically active complex with NoxA1 and ROS production. In human colon cancer cells, NoxO1 and Grb2 interaction causes Cbl E3 ligase recruitment, leading to NoxO1 UPS-mediated degradation and decrease in Nox1 activity (61). Intracellular H2O2 accumulation led to UPS-mediated degradation of MKP-3, negative regulator of ERK1/2, contributing to ovarian cancer cell survival and chemoresistance (62). Furthermore, glutamate-induced oxidative stress and PKC-δ activation, aberrant ERK1/2 stimulation, increased tumorigenicity, and chemoresistance involve UPS-mediated MKP-1 degradation (63). Thus, targeting key oncoproteins to induce their RONS-mediated UPS-dependent degradation can serve as a basis for anticancer therapy.

Variety of tissue-specific proteasomes are targets for inhibitors used in anticancer therapy, although proteasome subtypes have different susceptibility to different inhibitors. Indeed, a proteasome inhibitor, b-AP15, causes distinct cellular response as compared with clinically used bortezomib and leads to oxidative stress- and endoplasmic reticulum stress–induced tumor cell apoptosis through JNK/AP-1–mediated signaling (64). Increased cancer cell resistance to existing proteasome inhibitors requires novel proteomic and drug discovery approaches.

Approximately 10%–20% of damaged proteins undergo degradation through lysosomal/autophagy pathway that plays a dual role in cancer cell death and survival being implicated in crosstalk between diverse cellular pathways including oncogene Ras-induced carcinogenesis, mitochondrial metabolism, redox status, and energy production (65). Generally, at initial cancer stages, autophagy inhibition is observed, while at advanced stages, autophagy is upregulated to provide cell survival and proliferation under oxygen/nutrient starved conditions. •NO negatively regulates autophagy initiation through Bcl-2 oxidation/S-nitrosylation to prevent UPS-mediated degradation and to inhibit apoptosis of malignant lung epithelial cells (66). However, Bcl-2 exposure to O2 caused its downregulation and UPS-mediated degradation, suggesting a proapoptotic role of this ROS type (Fig. 1).

Mitochondrial ROS production depends on lysosomal ferritin degradation followed by redox-active iron release and cancer cell death in response to anticancer agent, artesunate (67). In addition, zinc protoporphyrin exhibits anticancer effects via induction of β-catenin lysosomal degradation followed by inhibition of Wnt/β-catenin signaling (68). Crosstalk between ROS generation and autophagy is an emerging challenge in targeting for anticancer chemotherapy in preclinical and clinical studies.

Dysfunction of molecular chaperone machinery

Additional mechanism underlying ROS-induced carcinogenesis is disruption of molecular chaperone machinery, which leads to impairment in cellular protein quality control. Indeed, HGF-induced ROS generation in HepG2 cells causes decrease in free thiol group amount in HSP60 and protein disulfide isomerase (PDI) resulted from HSP60 Cys442 and PDI Cys18 S-glutathionylation followed by ERK1/2 phosphorylation/activation and HCC cell migration (69). In addition, HSP70 is expressed at very low concentrations in mitochondria of normal cells, being abundant in mitochondria of tumor cells to play key roles in stress-inducible pathways. HSP70 inhibition causes multifaceted mitochondrial dysfunction, impaired mitochondrial integrity, loss of mitochondrial membrane potential, reduced oxygen consumption, oxidative phosphorylation, and ATP production (70).

Mitochondria of tumor cells substantially differ from mitochondria of nontransformed cells in metabolism, energy and ROS production, and membrane permeability. HSP90, a key player of protein quality control machinery, compartmentalizes in mitochondria to provide mitochondrial reprogramming and apoptosis suppression. This is achieved via inhibition of AMPK, preservation of cytoskeletal dynamics, and release of FAK from its complex with FIP200, autophagy initiator, to promote cancer cell adhesion and metastasis (71).

Because induction of HSP synthesis protects cancer cells from oxidative stress, effective HSP inhibitor–based anticancer therapy is of great concern. For example, ascorbate/menadione ROS-generating system used to induce cancer cell death exerts its action through complex mechanisms including glycolysis inhibition, dysregulation of calcium homeostasis, and impairment of HSP chaperoning functions (72). Additional strategy is an oxidative stress–induced HSP cleavage to cause degradation of their client proteins and cancer cell death (73).

Cytoskeletal disruption

Under physiologic conditions, cytoskeleton dynamics provides multiple cellular functions including migration, adhesion, and endocytosis. However, oxidative stress causes disruption in cytoskeleton assembly and dynamics, which facilitates cancer cell motility and aggressive behavior. RONS-induced cytoskeleton protein modifications change microtubule stability in a wide range of cancers. For example, treatment of pancreatic cancer cells with high H2O2 concentrations caused rapid loss of intracellular actin microfilaments and microtubules accompanied by increased NFκB activity and morphologic changes (74).

Cytoskeletal proteins, actin and vimentin, contain Cys and Met residues susceptible to oxidation. Cys oxidation/S-glutathionylation and both Cys and Met carbonylation (Table 1) in actin monomers control cytoskeleton dynamics (75). H2O2 significantly reduces cell adhesion on fibronectin, laminin, and collagen, and decreases FAK and paxillin and α5β1, αvβ3, β1 integrin levels (76). These changes associate with increased NFκB activity, and actin and vimentin microfilament reorganization.

Another mechanism underlying regulation of cytoskeleton organization may involve proteolysis and protein fragmentation. Indeed, RhoA, a member of Ras-related small GTPases, which regulates actin microfilament assembly and cytoskeleton dynamics, undergoes proteolysis/cleavage in response to oxidative stress (77). Oxidative stress–induced cytoskeletal disruption correlates with poor prognosis and chemoresistance, especially, in patients with solid and hematologic cancers.

Understanding cancer complexity requires elucidating molecular mechanisms underlying alterations in cellular functioning during carcinogenesis. An association of RONS generation with human cancer initiation and progression occurs via several mechanisms including oxidative/nitrosative modifications of key redox-sensitive residues in enzymes, intracellular effectors of signal transduction pathways, transcription factors, molecular chaperones, cytoskeleton, and proteasomal/lysosomal proteins. However, data on RONS-induced protein modifications during cancer growth are not complete; more investigations are required in this field. Novel redox proteomic approaches are being developed for high-throughput screening of redox-sensitive proteins to benefit discovery of novel molecular targets for anticancer therapy.

No potential conflicts of interest were disclosed.

Dedicated to the memory of Prof. Yuri S. Tatarinov (September 02, 1928–January 27, 2012) and Prof. Garry I. Abelev (January 10, 1928–December 23, 2013), who discovered the first cancer biomarker, α-fetoprotein (AFP), recognized as a golden standard in the field of molecular biomarkers used in cancer diagnosis and prognosis. We celebrate the 90th anniversary of the birth of both professors in 2018. This work was supported by I.M. Sechenov First Moscow State Medical University Strategic Development Program under the Russian Academic Excellence 5-100 Project.

1.
DeBerardinis
RJ
,
Chandel
NS
. 
Fundamentals of cancer metabolism
.
Sci Adv
2016
;
2
:
e1600200
.
2.
Smith
KA
,
Waypa
GB
,
Schumacker
PT
. 
Redox signaling during hypoxia in mammalian cells
.
Redox Biol
2017
;
13
:
228
34
.
3.
Jiang
F
,
Zhang
Y
,
Dusting
GJ
. 
NADPH oxidase-mediated redox signaling: roles in cellular stress response, stress tolerance, and tissue repair
.
Pharmacol Rev
2011
;
63
:
218
42
.
4.
Brand
MD
. 
Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling
.
Free Radic Biol Med
2016
;
100
:
14
31
.
5.
Förstermann
U
,
Sessa
WC
. 
Nitric oxide synthases: regulation and function
.
Eur Heart J
2012
;
33
:
829
37
.
6.
He
L
,
He
T
,
Farrar
S
,
Ji
L
,
Liu
T
,
Ma
X
. 
Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species
.
Cell Physiol Biochem
2017
;
44
:
532
53
.
7.
Moldogazieva
NT
,
Mokhosoev
IM
,
Feldman
NB
,
Lutsenko
SV
. 
ROS and RNS signalling: adaptive redox switches through oxidative/nitrosative protein modifications
.
Free Radic Res
2018
;
52
:
507
43
.
8.
Niki
E
. 
Oxidative stress and antioxidants: distress or eustress?
Arch Biochem Biophys
2016
;
595
:
19
24
.
9.
Klaunig
JE
,
Wang
Z
,
Pu
X
,
Zhou
S
. 
Oxidative stress and oxidative damage in chemical carcinogenesis
.
Toxicol Appl Pharmacol
2011
;
254
:
86
99
.
10.
Huang
T
,
Zhou
F
,
Wang-Johanning
F
,
Nan
K
,
Wei
Y
. 
Depression accelerates the development of gastric cancer through reactive oxygen species-activated ABL1
.
Oncol Rep
2016
;
36
:
2435
43
.
11.
Fu
N
,
Yao
H
,
Nan
Y
,
Qiao
L
. 
Role of oxidative stress in hepatitis C virus induced hepatocellular carcinoma
.
Curr Cancer Drug Targets
2017
;
17
:
498
504
.
12.
Mahalingaiah
PK
,
Singh
KP
. 
Chronic oxidative stress increases growth and tumorigenic potential of MCF-7 breast cancer cells
.
PLoS One
2014
;
9
:
e87371
.
13.
Kumar
B
,
Koul
S
,
Khandrika
L
,
Meacham
RB
,
Koul
HK
. 
Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype
.
Cancer Res
2008
;
68
:
1777
85
.
14.
Gianni
D
,
Bohl
B
,
Courtneidge
SA
,
Bokoch
GM
. 
The involvement of the tyrosine kinase c-Src in the regulation of reactive oxygen species generation mediated by NADPH oxidase-1
.
Mol Biol Cell
2008
;
19
:
2984
94
.
15.
Lu
CL
,
Qui
JL
,
Huang
PZ
,
Zou
RH
,
Hong
J
,
Li
BK
, et al
NADPH oxidase DUOX1 and DUOX2 but not NOX4 are independent predictors in hepatocellular carcinoma after hepatectomy
.
Tumour Biol
2011
;
32
:
1173
82
.
16.
Eun
HS
,
Cho
SY
,
Joo
JS
,
Kang
SH
,
Moon
HS
,
Lee
ES
, et al
Gene expression of NOX family members and their clinical significance in hepatocellular carcinoma
.
Sci Rep
2017
;
7
:
11060
.
17.
Binker
MG
,
Binker-Cosen
AA
,
Gaisano
HY
,
de Cosen
RH
,
Cosen-Binker
LI
. 
TGF- β1 increases invasiveness of SW1990 cells through Rac1/ROS/NF-κB/IL-6/MMP-2
.
Biochem Biophys Res Commun
2011
;
405
:
140
5
.
18.
Lien
GS
,
Wu
MS
,
Bien
MY
,
Chen
CH
,
Lin
CH
,
Chen
BC
. 
Epidermal growth factor stimulates nuclear factor-κB activation and heme oxygenase-1 expression via c-Src NADPH oxidase, PI3K, and Akt in human colon cancer cells
.
PLoS One
2014
;
9
:
e104891
.
19.
Varghese
SS
,
Sunil
PM
,
Madhavan
RN
. 
Expression of inducible nitric oxide synthase (iNOS) in oral precancer and oral squamous cell carcinoma: an immunohistochemical study
.
Cancer Biomark
2011
;
8
:
155
60
.
20.
Perrotta
C
,
Cervia
D
,
Di Renzo
I
,
Moscheni
C
,
Bassi
MT
,
Campana
L
, et al
Nitric oxide generated by tumor-associated macrophages is responsible for cancer resistance to cisplatin and correlated with syntaxin 4 and acid sphingomyelinase inhibition
.
Front Immunol
2018
;
9
:
1186
.
21.
Rabender
CS
,
Alam
A
,
Sundaresan
G
,
Cardnell
RJ
,
Yakovlev
VA
,
Mukhopadhyay
ND
, et al
The role of nitric oxide synthase uncoupling in tumor progression
.
Mol Cancer Res
2015
;
13
:
1034
43
.
22.
Liu
J
,
Wang
Z
. 
Increased oxidative stress as a selective anticancer therapy
.
Oxid Med Cell Longev
2015
;
2015
:
294303
.
23.
Ramanathan
B
,
Jan
KY
,
Chen
CH
,
Hour
TC
,
Yu
HJ
,
Pu
YS
. 
Resistance to paclitaxel is proportional to cellular total antioxidant capacity
.
Cancer Res
2005
;
65
:
8455
60
.
24.
Bekele
RT
,
Venkatraman
G
,
Liu
RZ
,
Tang
X
,
Mi
S
,
Benesch
MG
, et al
Oxidative stress contributes to the tamoxifen-induced killing of breast cancer cells: implications for tamoxifen therapy and resistance
.
Sci Rep
2016
;
6
:
21164
.
25.
Sturlan
S
,
Baumgartner
M
,
Roth
E
,
Bachleitner-Hofmann
T
. 
Docosahexaenoic acid enhances arsenic trioxide-mediated apoptosis in arsenic trioxide-resistant HL-60 cells
.
Blood
2003
;
101
:
4990
7
.
26.
Ding
WQ
,
Lind
SE
. 
Phospholipid hydroperoxide glutathione peroxidase plays a role in protecting cancer cells from docosahexaenoic acid-induced cytotoxicity
.
Mol Cancer Ther
2007
;
6
:
1467
74
.
27.
Du
J
,
Sun
C
,
Hu
Z
,
Yang
Y
,
Zhu
Y
,
Zheng
D
, et al
Lysophosphatidic acid induces MDA-MB-231 breast cancer cells migration through activation of PI3K/PAK1/ERK signaling
.
PLoS One
2010
;
5
:
e15940
.
28.
Sawai
H
,
Funahashi
H
,
Okada
Y
,
Matsuo
Y
,
Sakamoto
M
,
Yamamoto
M
, et al
Interleukin-1 alpha enhances IL-8 secretion through p38 mitogen-activated protein kinase and reactive oxygen species signaling in human pancreatic cancer cells
.
Med Sci Monit
2005
;
11
:
BR343
50
.
29.
Wu
JR
,
Hu
CT
,
You
RI
,
Pan
SM
,
Cheng
CC
,
Lee
MC
, et al
Hydrogen peroxide inducible clone-5 mediates reactive oxygen species signaling for hepatocellular carcinoma progression
.
Oncotarget
2015
;
6
:
32526
44
.
30.
Wright
C
,
Iver
AK
,
Kulkarni
Y
,
Azad
N
. 
S-nitrosylation of Bcl-2 negatively affects autophagy in lung epithelial cells
.
J Cell Biochem
2016
;
117
:
521
32
.
31.
Hu
CT
,
Wu
JR
,
Cheng
CC
,
Wang
S
,
Wang
HT
,
Lee
MC
, et al
Reactive oxygen species-mediated PKC and integrin signaling promotes tumor progression of human hepatoma HepG2
.
Clin Exp Metastasis
2011
;
28
:
851
63
.
32.
Zhan
Y
,
Gong
K
,
Chen
C
,
Wang
H
,
Li
W
. 
P38 MAP kinase functions as a switch in MS-275-induced reactive oxygen species-dependent autophagy and apoptosis in human colon cancer cells
.
Free Radic Biol Med
2012
;
53
:
532
43
.
33.
Kim
KY
,
Park
KI
,
Kim
SH
,
Yu
SN
,
Park
SG
,
Kim
YW
, et al
Inhibition of autophagy promotes salinomycin-induced apoptosis via reactive oxygen species-mediated PI3K/AKT/mTOR and ERK/p38 MAPK-dependent signaling in human prostate cancer cells
.
Int J Mol Sci
2017
;
18
:
1088
.
34.
Sauer
H
,
Engel
S
,
Milosevic
N
,
Sharifpanah
F
,
Wartenberg
M
. 
Activation of AMP-kinase by AICAR induces apoptosis of DU-145 prostate cancer cells through generation of reactive oxygen species and activation of c-Jun N-terminal kinase
.
Int J Oncol
2012
;
40
:
501
8
.
35.
Lou
YW
,
Chen
YY
,
Hsu
SF
,
Chen
RK
,
Lee
CL
,
Khoo
KH
, et al
Redox regulation of the protein phosphatase PTP1B in cancer cells
.
FEBS J
2008
;
275
:
69
88
.
36.
Chetram
MA
,
Don-Salu-Hewage
AS
,
Hinton
CV
. 
ROS enhances CXCR4-mediated functions through inactivation of PTEN in prostate cancer cells
.
Biochem Biophys Res Commun
2011
;
410
:
195
200
.
37.
Galli
S
,
Antico Arciuch
VG
,
Poderoso
C
,
Converso
DP
,
Zhou
Q
,
Bal de Kier Joffé
E
, et al
Tumor cell phenotype is sustained by selective MAPK oxidation in mitochondria
.
PLoS One
2008
;
3
:
e2379
.
38.
Okoh
VO
,
Felty
Q
,
Parkash
J
,
Poppiti
R
,
Roy
D
. 
Reactive oxygen species via redox signaling to PI3K/AKT pathway contribute to the malignant growth of 4-hydroxyestradiol-transformed mammary epithelial cells
.
PLoS One
2013
;
8
:
e54206
.
39.
Saunders
JA
,
Rogers
LC
,
Klomsiri
C
,
Poole
LB
,
Daniel
LW
. 
Reactive oxygen species mediate lysophosphatidic acid induced signaling in ovarian cancer cells
.
Free Radic Biol Med
2010
;
49
:
2058
67
.
40.
Klomsiri
C
,
Rogers
LC
,
Soito
L
,
McCauley
AK
,
King
SB
,
Nelson
KJ
, et al
Endosomal H2O2 production leads to localized cysteine sulfenic acid formation on proteins during lysophosphatidic acid-mediated cell signaling
.
Free Radic Biol Med
2014
;
71
:
49
60
.
41.
Okoh
VO
,
Garba
NA
,
Penney
RB
,
Das
J
,
Deoraj
A
,
Sigh
KP
, et al
Redox signalling to nuclear regulatory proteins by reactive oxygen species contributes to oestrogen-induced growth of breast cancer cells
.
Br J Cancer
2015
;
112
:
1687
702
.
42.
Kwon
J
,
Lee
SR
,
Yang
KS
,
Ahn
Y
,
Kim
YJ
,
Stadtman
ER
, et al
Reversible oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors
.
Proc Natl Acad Sci USA
2004
;
101
:
16419
24
.
43.
Lim
KH
,
Ancrile
BB
,
Kashatus
DF
,
Counter
CM
. 
Tumour maintenance is mediated by eNOS
.
Nature
2008
;
452
:
646
9
.
44.
Park
MT
,
Kim
MJ
,
Suh
Y
,
Kim
RK
,
Kim
H
,
Lim
EJ
, et al
Novel signaling axis for ROS generation during K-Ras-induced cellular transformation
.
Cell Death Differ
2014
;
21
:
1185
97
.
45.
Dhar
A
,
Young
MR
,
Colburn
NH
. 
The role of AP-1, NF-kappaB and ROS/NOS in skin carcinogenesis: the JB6 model is predictive
.
Mol Cell Biochem
2002
;
234–235
:
185
93
.
46.
Reynaert
NL
,
van der Vliet
A
,
Guala
AS
,
McGovern
T
,
Hristova
M
,
Pantano
C
, et al
Dynamic redox control of NF-kappaB through glutaredoxin-regulated S-glutathionylation of inhibitory kappaB kinase beta
.
Proc Natl Acad Sci USA
2006
;
103
:
13086
91
.
47.
Yakovlev
VA
,
Barani
IJ
,
Rabender
CS
,
Black
SM
,
Leach
JK
,
Graves
PR
, et al
Tyrosine nitration of IkappaBalpha: a novel mechanism for NF-kappaB activation
.
Biochemistry
2007
;
46
:
11671
83
.
48.
Fan
C
,
Li
Q
,
Ross
D
,
Engelhardt
JF
. 
Tyrosine phosphorylation of I kappa B alpha activates NF kappa B through a redox-regulated and c-Src-dependent mechanism following hypoxia/reoxygenation
.
J Biol Chem
2003
;
278
:
2073
80
.
49.
Lluis
JM
,
Buricchi
F
,
Chiaguri
P
,
Morales
A
,
Fernandez-Checa
JC
. 
Dual role of mitochondrial reactive oxygen species in hypoxia signaling: activation of nuclear factor–{kappa}B via c-Src and oxidant-dependent cell death
.
Cancer Res
2007
;
67
:
7368
77
.
50.
Talbott
SJ
,
Luanpitpong
S
,
Stehlik
C
,
Azad
N
,
Iyer
AK
,
Wang
L
, et al
S-nitrosylation of FLICE inhibitory protein determines its interaction with RIP1 and activation of NF-κB
.
Cell Cycle
2014
;
13
:
1948
57
.
51.
Elsasser
TH
,
Li
CJ
,
Caperna
TJ
,
Kahl
S
,
Schmidt
WF
. 
Growth hormone (GH)-associated nitration of Janus kinase-2 at the 1007Y-1008Y epitope impedes phosphorylation at this site: mechanism for and impact of a GH, AKT, and nitric oxide synthase axis on GH signal transduction
.
Endocrinology
2007
;
148
:
3792
802
.
52.
Yoon
S
,
Woo
SU
,
Kang
JH
,
Kim
K
,
Kwon
MH
,
Park
S
. 
et al. STAT3 transcriptional factor activated by reactive oxygen species induces IL6 in starvation-induced autophagy of cancer cells
.
Autophagy
2010
;
6
:
1125
38
.
53.
Kasiappan
R
,
Jutooru
I
,
Karki
K
,
Hedrick
E
,
Safe
S
. 
Benzyl isothiocyanate (BITC) induces reactive oxygen species-dependent repression of STAT3 protein by down-regulation of specificity proteins in pancreatic cancer
.
J Biol Chem
2016
;
291
:
27122
33
.
54.
Cobbs
CS
,
Whisenhunt
TR
,
Wesemann
DR
,
Harkins
LE
,
Van Meir
EG
,
Samanta
M
. 
Inactivation of wild-type p53 protein function by reactive oxygen and nitrogen species in malignant glioma cells
.
Cancer Res
2003
;
63
:
8670
3
.
55.
Harder
B
,
Jiang
T
,
Wu
T
,
Tao
S
,
Rojo de la Vega
M
,
Tian
W
, et al
Molecular mechanisms of Nrf2 regulation and how these influence chemical modulation for disease intervention
.
Biochem Soc Trans
2015
;
43
:
680
6
.
56.
Kobayashi
M
,
Li
L
,
Iwamoto
N
,
Nakajima-Takagi
Y
,
Kaneko
H
,
Nakayama
Y
, et al
The antioxidant defense system Keap1-Nrf2 comprises a multiple sensing mechanism for responding to a wide range of chemical compounds
.
Mol Cell Biol
2009
;
29
:
493
502
.
57.
Fourquet
S
,
Guerois
R
,
Biard
D
,
Toledano
MB
. 
Activation of NRF2 by nitrosative agents and H2O2 involves Keap1 disulfide formation
.
J. Biol Chem
2010
;
285
:
8463
71
.
58.
He
X
,
Ma
Q
. 
NRF2 cysteine residues are critical for oxidant/electrophile sensing, Kelch-like ECH-associated protein-1-dependent ubiquitination-proteasomal degradation, and transcription activation
.
Mol Pharmacol
2009
;
76
:
1265
78
.
59.
Refaat
A
,
Pararasa
C
,
Arif
M
,
Brown
JE
,
Carmichael
A
,
Ali
SS
, et al
Bardoxolone-methyl inhibits migration and metabolism in MCF7 cells
.
Free Radic Res
2017
;
51
:
211
21
.
60.
Singh
B
,
Shoulson
R
,
Chatterjee
A
,
Ronghe
A
,
Bhat
NK
,
Dim
DC
, et al
Resveratrol inhibits estrogen-induced breast carcinogenesis through induction of NRF2-mediated protective pathways
.
Carcinogenesis
2014
;
35
:
1872
80
.
61.
Joo
JH
,
Oh
H
,
Kim
M
,
An
EJ
,
Kim
RK
,
Lee
SY
, et al
NADPH oxidase 1 activity and ROS generation are regulated by Grb2/Cbl-mediated proteasomal degradation of NoxO1 in colon cancer cells
.
Cancer Res
2016
;
76
:
855
65
.
62.
Chan
DW
,
Liu
VW
,
Tsao
GS
,
Yao
KM
,
Furukawa
T
,
Chan
KK
, et al
Loss of MKP3 mediated by oxidative stress enhances tumorigenicity and chemoresistance of ovarian cancer cells
.
Carcinogenesis
2008
;
29
:
1742
50
.
63.
Choi
BH
,
Hur
EM
,
Lee
JH
,
Jun
DJ
,
Kim
KT
. 
Protein kinase Cdelta-mediated proteasomal degradation of MAP kinase phosphatase-1 contributes to glutamate-induced neuronal cell death
.
J Cell Sci
2006
;
119
:
1329
40
.
64.
Brnjic
S
,
Mazurkiewicz
M
,
Fryknäs
M
,
Sun
C
,
Zhang
Z
,
Larsson
R
, et al
Induction of tumor cell apoptosis by a proteasome deubiquitinase inhibitor is associated with oxidative stress
.
Antioxid Redox Signal
2014
;
21
:
2271
85
.
65.
Bellot
GL
,
Liu
D
,
Pervaiz
S
. 
ROS, autophagy, mitochondria and cancer: RAS, the hidden master?
Mitochondrion
2013
;
13
:
155
62
.
66.
Azad
N
,
Iyer
A
,
Vallyathan
V
,
Wang
L
,
Castranova
V
,
Stehlik
C
, et al
Role of oxidative/nitrosative stress-mediated Bcl-2 regulation in apoptosis and malignant transformation
.
Ann N Y Acad Sci
2010
;
1203
:
1
6
.
67.
Yang
ND
,
Tan
SH
,
Ng
S
,
Shi
Y
,
Zhou
J
,
Tan
KS
, et al
Artesunate induces cell death in human cancer cells via enhancing lysosomal function and lysosomal degradation of ferritin
.
J Biol Chem
2014
;
289
:
33425
41
.
68.
Wang
S
,
Hannafon
BN
,
Lind
SE
,
Ding
WQ
. 
Zinc protoporphyrin suppresses β-catenin protein expression in human cancer cells: the potential involvement of lysosome-mediated degradation
.
PLoS One
2015
;
10
:
e0127413
.
69.
Lin
CY
,
Hu
CT
,
Cheng
CC
,
Lee
MC
,
Pan
SM
,
Lin
TY
, et al
Oxidation of heat shock protein 60 and protein disulfide isomerase activates ERK and migration of human hepatocellular carcinoma HepG2
.
Oncotarget
2016
;
7
:
11067
82
.
70.
Leu
JI
,
Barnoud
T
,
Zhang
G
,
Tian
T
,
Wei
Z
,
Herlyn
M
, et al
Inhibition of stress-inducible HSP70 impairs mitochondrial proteostasis and function
.
Oncotarget
2017
;
8
:
45656
69
.
71.
Caino
MC
,
Chae
YC
,
Vaira
V
,
Ferrero
S
,
Nosotti
M
,
Martin
NM
, et al
Metabolic stress regulates cytoskeletal dynamics and metastasis in cancer cells
.
J Clin Invest
2013
;
123
:
2907
20
.
72.
Beck
R
,
Pedrosa
RC
,
Dejeans
N
,
Glorieux
C
,
Leveque
P
,
Gallez
B
, et al
Ascorbate/menadione-induced oxidative stress kills cancer cells that express normal or mutated forms of the oncogenic protein Bcr-Abl. An in vitro and in vivo mechanistic study
.
Invest New Drugs
2011
;
29
:
891
900
.
73.
Beck
R
,
Verrax
J
,
Gonze
T
,
Zappone
M
,
Pedrosa
RC
,
Taper
H
, et al
Hsp90 cleavage by an oxidative stress leads to its client proteins degradation and cancer cell death
.
Biochem Pharmacol
2009
;
77
:
375
83
.
74.
Hu
WG
,
Lu
QP
. 
Impact of oxidative stress on the cytoskeleton of pancreatic epithelial cells
.
Exp Ther Med
2014
;
8
:
1438
42
.
75.
Sakai
J
,
Li
J
,
Subramanian
KK
,
Mondal
S
,
Bajrami
B
,
Hattori
H
, et al
Reactive oxygen species-induced actin glutathionylation controls actin dynamics in neutrophils
.
Immunity
2012
;
37
:
1037
49
.
76.
Zhou
L
,
Li
Y
,
Yue
BY
. 
Oxidative stress affects cytoskeletal structure and cell-matrix interactions in cells from an ocular tissue: the trabecular meshwork
.
J Cell Physiol
1999
;
180
:
182
9
.
77.
Girouard
MP
,
Pool
M
,
Alchini
R
,
Rambaldi
I
,
Fournier
AE
. 
RhoA proteolysis regulates the actin cytoskeleton in response to oxidative stress
.
PLoS One
2016
;
11
:
e0168641
.