Category: Family

Oxidative stress and cancer

oxidative stress and cancer

Furthermore, in contrast to the mechanism of sister ane, redox signaling is based on migrating strese, and therefore, the Natural immune system boosters in this pathway is much more diffuse. Dtress oxidative stress and cancer PS strwss apoptosis srtess induction of endoplasmic reticulum stress-reactive oxygen species in head and neck squamous cell carcinoma cells. J Clin Oncol — PubMed PubMed Central CAS Google Scholar Kinoshita H, Hirata Y, Nakagawa H, Sakamoto K, Hayakawa Y, Takahashi R, Nakata W, Sakitani K, Serizawa T, Hikiba Y Interleukin-6 mediates epithelial—stromal interactions and promotes gastric tumorigenesis. In this way, p53 has a complementary function to that of FOXO transcription factors that induce the expression of sestrin 3

Oxidative stress and cancer -

Read more about how to correctly acknowledge RSC content. Fetching data from CrossRef. This may take some time to load. Loading related content. Jump to main content.

Jump to site search. You do not have JavaScript enabled. Please enable JavaScript to access the full features of the site or access our non-JavaScript page.

Issue 12, From the journal: Chemical Science. This article is Open Access. Please wait while we load your content Something went wrong. Try again? Cited by. Download options Please wait Supplementary information PDF K. Article type Edge Article. Submitted 27 Nov Accepted 24 Feb First published 25 Feb Download Citation.

Request permissions. Combination of chemotherapy and oxidative stress to enhance cancer cell apoptosis X. Social activity. Search articles by author Xinming Li. Yanan Hou. Approximately 90 percent of all human cancers have an environmental cause non-genetic inheritance predominantly through lifestyle choices smoking, diet, UV radiation while the remaining due to infections and chemical exposure.

Cancer is a multistage process that involves mutational changes and uncontrolled cell proliferation. Research has firmly established a causal and contributory role of oxidative stress and oxidative damage in cancer initiation and progression.

Methods: The purpose of this article is to review the role that oxidative stress and reactive oxygen species play in the development of cancer. Both endogenous and exogenous sources of reactive oxygen species result in increased oxidative stress in the cell. Excess reactive oxygen fumed can result in damage to and modification of cellular macromolecules most importantly genomic DNA that can produce mutations.

In addition, oxidative stress modulates gene expression of downstream targets involved in DNA repair, cell proliferation and antioxidants.

The modulation of gene expression by oxidative stress occurs in part through activation or inhibition of transcription factors and second messengers.

The role of single nuclear polymorphism for oxidative DNA repair and enzymatic antioxidants is important in determining the potential human cancer risk. Conclusion: oxidative stress and the resulting oxidative damage are important contributors to the formation and progression of cancer.

Keywords: Cancer , oxidative stress , ROS , etiology , genetic inheritance , chemical exposure. Volume: 24 Issue: Affiliation: School of Public Health, Indiana University, Bloomington,Indiana. Abstract: Background: Cancer is considered a major cause of death worldwide. Klaunig E. Purchase PDF.

Mark Item. Current Pharmaceutical Design. Title: Oxidative Stress and Cancer Volume: 24 Issue: 40 Author s : James E. Close Print this page.

Export Options ×. Export File: RIS for EndNote, Reference Manager, ProCite. Content: Citation Only. Citation and Abstract. About this article ×. Cite this article as: Klaunig E.

Close About this journal. Related Journals Anti-Cancer Agents in Medicinal Chemistry. Current Bioactive Compounds. Current Cancer Drug Targets. Current Cancer Therapy Reviews.

Current Diabetes Reviews. Current Drug Safety.

Editor-in-Chief: Alessandro Oxidative stress and cancer Department of Clinical and Experimental Srress Oxidative stress and cancer of Primary Oxdative Cells Cancef of Pisa Pisa Italy. ISSN Print snd ISSN Online : DOI: streas Background: Hormone balance and sleep is considered a major cause of death worldwide. The etiology of cancer is linked to environmental and genetic inheritance causes. Approximately 90 percent of all human cancers have an environmental cause non-genetic inheritance predominantly through lifestyle choices smoking, diet, UV radiation while the remaining due to infections and chemical exposure. Cancer is a multistage process that involves mutational changes and uncontrolled cell proliferation.

Barry Stess Oxidative stress and cancer: have we moved forward?, oxidative stress and cancer. Biochem J 1 January ; 1 : 1— Cance, knockout of various antioxidant defence enzymes canceer oxidative Digestive system cleanse levels oxixative promotes age-related cancer oxidative stress and cancer in animals.

Oxicative additional actions of RS oxidatige be important, possibly their effects on p53, oxidativw proliferation, invasiveness and metastasis. Ozidative inflammation predisposes to malignancy, but the role of Ccancer in this is likely oxidativee be complex because RS oxidative stress and cancer sometimes act as anti-inflammatory agents.

Sign In or Create an Account. Search Dropdown Menu. header search search oxidatibe Search input auto suggest. filter your search Oxjdative Content All Journals Ad Oxidative stress and cancer. Advanced Oxirative. Sign In. Toggle Menu Menu Oxodative Issues Issue oxkdative Accepted Manuscripts Collections Oxidativw themed collections Open Blood circulation and healing collections Curated content Anti-viral immunity boost mitochondria Authors Submission checklist Cancrr to authors Oxidative stress and cancer services Why publish with us?

Publishing oxidayive cycle Submit your work Open access options and prices China-based researchers Cancdr and Xtress Transition to open oxiddative Open oxjdative policy Editorial policy Information for reviewers Preprints policy Data policy Immune-boosting gut flora Scope Editorial board Impact and information Sponsored award winners Oxidative stress and cancer Press Biochemical Society Accessibility.

Skip Nav Destination Close navigation menu Article navigation. VolumeIssue 1. Next Article. All Issues. Cover Image Cover Image. EVIDENCE THAT REACTIVE SPECIES ARE IMPORTANT IN CANCER. DIRECT DAMAGE Oxicative DNA BY REACTIVE SPECIES.

ARE MALIGNANT CELLS IN A PRO-OXIDANT STATE? CACHEXIA AND REACTIVE SPECIES. Article Navigation. Review Article December 11 Oxidative stress and cancer: have we moved forward?

Barry Halliwell Barry Halliwell 1. This Site. Google Scholar. Author and article information. Publisher: Portland Press Ltd. Received: July 26 Revision Received: October 16 Accepted: October 18 Online ISSN: The Biochemical Oxidatove, London.

Biochem J 1 : 1— Article history Received:. Revision Received:. Get Permissions. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest. View full article. Sign in Don't already have an account?

Sign in to your personal account. You could not be signed in. Please try again. Sign In Reset password. Biochemical Society Member Sign in Sign In. Sign in via your Institution Sign in via your Institution. Get Access To This Article Buy This Article. View Metrics.

Cited By Web Of Science CrossRef Get Email Alerts Article Activity Alert. Accepted Manuscript Alert. Latest Completed Issue Alert. Submit your work. Latest Most Read Most Cited Beyond the tail: the consequence of context in histone post-translational modification and chromatin research.

Identification of a mer peptide from the death domain of MyD88 which attenuates inflammation and insulin resistance and improves glucose metabolism. IL-4 activates futile triacylglyceride cycle for glucose utilization in white adipocytes. AMPK activation suppresses leptin expression independently of adipogenesis in primary murine adipocytes.

Online Oxidwtive Print ISSN Submit Your Work Language-editing snd Recommend to Your Librarian Request a free trial Accessibility. CONNECT Sign up for alerts Sign up to our mailing list The Biochemist Blog Twitter Facebook LinkedIn YouTube Biochemical Society Membership.

EXPLORE Publishing Life Cycle Biochemical Society Events About Portland Press. com Biochemical Society Company no. GB Facebook Twitter LinkedIn YouTube. Privacy and cookies Accessibility © Copyright Portland Press. This Feature Is Available To Subscribers Only Sign In or Stresz an Account.

Close Modal. This site uses cookies. By continuing to use our website, you are agreeing to our privacy policy.

: Oxidative stress and cancer

Supplementary files Ferroportin and iron regulation in breast cancer progression and prognosis. Exploring novel strategies to improve anti-tumour efficiency: the potential for targeting reactive oxygen species. Article CAS Google Scholar Finkel, T. Similar to ROS, RNS can modulate cell survival, induce DNA damage, and affect mitochondrial functions [ 6 ]. Cellular and tissue techniques like Immunohistochemistry IHC for oxidative stress markers and Cytofluorometric Analysis using fluorescent probes are used for quantifying intracellular levels of ROS or antioxidants [ 75 ]. Squier TC, Bigelow DJ Protein oxidation and age-dependent alterations in calcium homeostasis.
Background Garg M, Moloney JN, Cotter TG. Article PubMed CAS Google Scholar Yagoda, N. Antioxidants accelerate lung cancer progression in mice. Several studies disclosed the chaperone activity of mitochondrial Lon and showed upregulation of Lon induced ROS generation playing a role in stress signaling [ 48 , 81 , 82 , 83 ]. cerevisiae strains.
Oxidative Stress and Cancer Article PubMed PubMed Central CAS Google Scholar Oxidativd, D. On the other hand, high Best ways to increase metabolism oxidative stress and cancer T stresss responses by ans the sterss of Oxidative stress and cancer and MHC antigen complex, which promotes cancer progression through evading immune response [ ]. In fact, it has been found that the breast cancer susceptibility 1 BRCA1 gene interacts with and induces Nrf2 expression with positive outcomes on cancer cell survival Sign In Reset password. Klein EA, Thompson IM Jr, Tangen CM, Crowley JJ, Lucia MS, Goodman PJ, et al. Tosatto A, Sommaggio R, Kummerow C, Bentham RB, Blacker TS, Berecz T, et al. J Nucl Med.
ROS in cancer therapy: the bright side of the moon | Experimental & Molecular Medicine

Current Drug Therapy. View More. Related Books Advanced Pharmacy. Plant-derived Hepatoprotective Drugs. The Role of Chromenes in Drug Discovery and Development. New Avenues in Drug Discovery and Bioactive Natural Products.

Practice and Re-Emergence of Herbal Medicine. Methods for Preclinical Evaluation of Bioactive Natural Products. Nanopharmacology and Nanotoxicology: Clinical Implications and Methods. Natural Immunomodulators: Promising Therapy for Disease Management. Bioactive Phytochemicals from Himalayas: A Phytotherapeutic Approach.

Indopathy for Neuroprotection: Recent Advances. Article Metrics. Journal Information. For Authors. Author Guidelines Graphical Abstracts Fabricating and Stating False Information Research Misconduct Post Publication Discussions and Corrections Publishing Ethics and Rectitude Increase Visibility of Your Article Archiving Policies Peer Review Workflow Order Your Article Before Print Promote Your Article Manuscript Transfer Facility Editorial Policies Allegations from Whistleblowers Announcements Forthcoming Thematic Issues.

For Editors. Guest Editor Guidelines Editorial Management Fabricating and Stating False Information Publishing Ethics and Rectitude Ethical Guidelines for New Editors Peer Review Workflow.

For Reviewers. Reviewer Guidelines Peer Review Workflow Fabricating and Stating False Information Publishing Ethics and Rectitude. Explore Articles. Abstract Ahead of Print 1 Article s in Press 10 Free Online Copy Most Cited Articles Most Accessed Articles Editor's Choice Thematic Issues.

The production of ROS is elevated in tumor cells as a consequence of increased metabolic rate, gene mutation and relative hypoxia, and excess ROS are quenched by increased antioxidant enzymatic and nonenzymatic pathways in the same cells.

Moderate increases of ROS contribute to several pathologic conditions, among which are tumor promotion and progression, as they are involved in different signaling pathways and induce DNA mutation.

However, ROS are also able to trigger programmed cell death PCD. Our review will emphasize the molecular mechanisms useful for the development of therapeutic strategies that are based on modulating ROS levels to treat cancer.

Specifically, we will report on the growing data that highlight the role of ROS generated by different metabolic pathways as Trojan horses to eliminate cancer cells. Reactive oxygen species ROS produced in eukaryotic cells through aerobic metabolism have evolved as regulators of important signaling pathways.

ROS, previously considered mere byproducts of cellular respiration, are oxygen-containing molecules with high reactivity. In fact, H 2 O 2 plays the role of a second messenger in some pathways that involve the transduction of extracellular signals and the control of gene expression, contributing to what is currently defined as redox signaling 1.

Oxidative phosphorylation in mitochondria involves four electron-transporting complexes and a proton-translocating ATP synthase that direct electrons derived from the initial oxidation of NADPH and FADH2 along a multistep pathway that culminates in protons being pumped outside of mitochondria.

ROS are also continuously generated by enzymatic reactions involving cyclooxygenases, NADPH oxidases NOXs , xanthine oxidases and lipoxygenases and through the iron-catalyzed Fenton reaction; indeed, it should be noted that NOXs have primarily evolved to produce ROS 2.

Finally, ROS are generated after exposure to physical agents ultraviolet rays and heat and after chemotherapy and radiotherapy in cancer. Tight regulation of ROS levels is crucial for cellular life; in fact, moderate ROS contribute to the control of cell proliferation and differentiation.

Therefore, eukaryotic cells benefit from a complex scavenging system based on superoxide dismutases SODs , located in the cytoplasm, mitochondria and the extracellular matrix; glutathione peroxidase GPX ; glutathione reductase GR ; peroxiredoxin; thioredoxin; and catalase, which convert superoxide anions into water and recycle the antioxidants in the reduced state.

Here, we focus on the molecular mechanisms that support the elaboration of anticancer therapies that modulate the production and scavenging of ROS and, in particular, on the opportunities raised by their ability to induce cell death upon exceeding a threshold level.

It has been determined that each cell is exposed to ~1. If, for any reason, ROS production increases or the number of scavenged ROS decreases, then cells experience a condition known as oxidative stress.

Oxidative stress has been implicated in the pathophysiology of cancer: in fact, high levels of ROS generated by ongoing aerobic glycolysis followed by pyruvate oxidation in mitochondria the Warburg effect , increase receptor and oncogene activity, and the stimulation of growth factor-dependent pathways or oxidizing enzymes induce genetic instability 3 , 4.

Moreover, excessive intracellular levels of ROS may damage lipids, proteins and DNA, and this ability has been exploited in a series of anticancer strategies, as detailed below. By interacting with lipids, ROS can induce oxidative stress through a feedback loop initiated by the peroxidation of fatty acids, which alters the lipid bilayer of cell membranes and generates free radicals.

This process is potentially dangerous to cells, as peroxidation of mitochondrial phospholipids may affect the integrity of permeability transition pores PTPs and disaggregate complexes I and III of the respiratory chain, thereby enhancing electron leakage within the mitochondrial intermembrane space 5 , 6.

However, free radicals produced by lipid peroxidation are short-lived 7. By interacting with proteins, ROS have an impact on several signaling pathways involved in the control of cell proliferation and apoptosis.

These modifications are reversible and generate a wide array of cellular responses In general, phosphatases are inhibited by ROS 11 , whereas kinases may be inhibited or activated In particular, ROS activate nonreceptor protein kinases belonging to the Src family; small G proteins, such as Ras; and the tyrosine kinase receptors of growth factors 13 , 14 , as well as components of the c-Jun N-terminal kinase JNK and p38 kinase p38MAPK pathways that induce apoptosis Specifically, through the formation of disulfide bonds between catalytic cysteines, H 2 O 2 inactivates phosphatase and tensin homolog phosphatase PTEN and unlocks the phosphoinositide 3-kinase PI3-K -dependent recruitment of its downstream kinases, such as protein kinase B Akt 16 , or oxidizes the redox protein thioredoxin and thus suppresses its inhibitory effect on the p38MAPK signaling cascade ROS also influence the activity of calcium channels; in fact, they induce the release of calcium from cellular stores with the consequent activation of kinases, such as protein kinase C PKC , thereby playing important roles in the proliferation of cancer cells Most ROS-sensitive pathways transduce cytoplasmic signals to the nucleus, where they influence the activity of transcription factors that control the expression of a wide array of genes.

In this regard, to prevent excessive intracellular ROS, cancer cells respond to oxidative stress by inducing the transcription of antioxidant enzymes, highlighting the relevance of an in-depth knowledge of these pathways for use in elaborating therapies that alter ROS levels.

The pivotal redox-sensitive transcription factor is nuclear factor erythroid 2-related factor 2 Nrf2 19 , recognized as the leading transcription factor driving the antioxidant response in cancer cells.

Under normal conditions, Nrf2 is degraded through its interaction with Kelch-like ECH-associated protein 1 Keap1 , whereas under oxidative stress conditions, Keap1 is oxidized and Nrf2 is translocated to the nucleus, where it induces the expression of several genes Nrf2 controls the production of glutathione GSH , the leading antioxidant molecule within cells, through the expression of the enzyme that catalyzes the rate-limiting reaction of GSH synthesis, glutamate-cysteine ligase GCL , and GSH utilization and regeneration 20 , It also controls free Fe II homeostasis, upregulating the expression of heme oxygenase HMOX1, which generates free Fe II via the breakdown of heme molecules.

Notably, high serum concentrations of ferritin have been described in several cancers with a poor prognosis The forkhead box O FOXO family of transcription factors is activated by JNK after ROS levels are increased and induces the expression of SODs and catalase Another important transcription factor that plays a major role in the control of antioxidant gene expression is p In fact, the role of p53 in the control of ROS levels is controversial, as it may promote both oxidant and antioxidant gene expression.

Indeed, moderately elevated ROS levels inhibit p53, while higher levels promote its expression. Among the targets of p53 activity are sestrins sestrin 1 and 2 that induce the activity of peroxiredoxins, increasing the impact of the cellular antioxidant array In this way, p53 has a complementary function to that of FOXO transcription factors that induce the expression of sestrin 3 Interestingly, both p53 and FOXO control a distinct set of genes that are not targets of Nrf2 activity, even though all three factors induce HMOX1 expression and, therefore, Fe II storage and secretion, that plays a role in breast tumorigenesis, highlighting the role of antioxidants in cancer promotion As a consequence of HIF1α activation, several genes important for cancer progression, such as VEGF and VEGF receptors, are induced Finally, the DNA-binding ability of some transcription factors is directly influenced by ROS.

The latter factor is able to interact with thioredoxin to reduce a specific cysteine Cys in the Rel-homology domain RHD of the NF-kB subunit p50 that had been previously oxidized by ROS 34 , restoring its ability to interact with specific responsive DNA sequences 35 , These data show that ROS can either activate or suppress the NF-kB signaling involved in the control of several important cellular processes, such as embryogenesis and cell proliferation and death, and the responses to a variety of stress stimuli ROS influence the activity of epigenetic modulators, such as histone deacetylases HDACs or DNA methyltransferases DNMTs with consequences that are evident in the expression of the target genes 38 , They also oxidize DNA, especially adenine and guanine 8-oxo-A and 8-oxo-G.

Oxidized Gs also impact the methylation of DNA, as indicated by reports showing that damaged bases on the DNA nascent strand can suppress the methylation of a cytosine within a distance of one or two base pairs However, ROS are able to induce DNA hypermethylation as well, with potential consequences on tumor phenotype when promoter regions of tumor suppressor genes are involved 43 , 44 , In addition, 8-oxo-Gs accumulate at telomeres, where they inhibit telomerase and decrease the binding of telomeric proteins, leading to the disruption of telomere length and precluding the maintenance of chromosomal-end capping Finally, ROS also induce mutations in mitochondrial DNA with the potential generation of a feedback loop in which mutations in genes encoding complexes of the ETC may directly affect the efficiency of electron transport.

The major sensitivity of mitochondrial DNA to ROS-induced mutagenesis is intuitive, as this DNA is not protected by histones, and mitochondria lack the nucleotide excision repair NER enzymatic system.

The main consequences of redox signaling and oxidative stress in normal and cancer cells are presented in Fig. The major signaling cascades induced by growth factor-stimulated ROS are highlighted on the left.

The same pathways influence the cell cycle and affect the activity of transcription factors and genes that play roles in the cellular response to the hypoxic microenvironment. The main consequences of oxidative stress in cancer cells are illustrated on the right. Moderately elevated ROS induce oncogenes and inhibit tumor suppressor genes that, in turn, increase ROS levels.

ROS also activate HDACs and have a dual effect on DNMTs with important outcomes for the expression of oncogenes and tumor suppressor genes. Oxidized bases trigger mutations and engage DNA repair enzymes. Cancer is the second cause of death worldwide and is characterized by several hallmarks 47 ; cell transformation, genome instability, hyperproliferation, immortalization, angiogenesis, epithelial-mesenchymal transition EMT and metastasis, which are all influenced in several ways by intracellular ROS 48 , Several noncancer cells associate with tumors: among these, cancer-associated fibroblasts CAFs , particularly represented in the tumor microenvironment TME , actively contribute to the regulation of tumor homeostasis, promoting tumor progression and the invasion of cancer cells.

CAFs and ROS engage in two-way cross-talk: on the one hand, fibroblasts are targeted by ROS, particularly H 2 O 2 , which is able to convert them into active CAFs through the upregulation of HIF1α; on the other hand, CAFs are critical for the increase in ROS levels observed in cancer 50 , CAFs can also promote cancer growth and invasiveness, and both CAFS and ROS are linked through the increases in ROS-generated CAFs to which most cancers respond by increasing the expression of antioxidant genes 52 , 53 , 54 Fig.

ROS, in response to death-inducing ligands TNFα and Fas , enhance the assembly of DISCs and the activation of effector caspases and reduce Bcl-2 activity or, as a consequence of increased permeability of mitochondrial PTPs, stimulate the intracytoplasmic release of cytochrome c, which interacts with Apaf-1 and procaspases and forms the apoptosome apoptosis.

ROS can also inhibit the negative regulators of autophagy TORC1 and increase the formation of LC3-dependent autophagosomes autophagy. Finally, high levels of ROS, induced by several receptor-interacting protein kinases RIPs , increase p53 expression, which increases ROS levels via a mechanism that depends on intracellular iron ferroptosis.

However, a growing body of evidence supports the view that antioxidant activities are essential for tumorigenesis. It has been recently reported that targets of the Nrf2 gene, such as HMOX1, facilitate cancer development because they counteract the effect of oxidative stress in transformed cells Moreover, established oncogenes such as K-RAS and c-MYC , which had been previously demonstrated to induce intracellular ROS 56 , 57 , have been recently shown to stabilize Nrf2 In this regard, mutations to NRF2 and its regulator KEAP1 have been found in cancer cells, supporting the supposition that antioxidant genes are pivotal in tumor progression 59 , 60 , 61 , In fact, it has been found that the breast cancer susceptibility 1 BRCA1 gene interacts with and induces Nrf2 expression with positive outcomes on cancer cell survival Interestingly, estrogen stimulation of breast cancer cells that do not express BRCA1 and, as a result, suffer from high intracellular ROS levels rescues NRF2 transcription, enhancing the survival of these cancer cells Additionally, FOXO transcription factors have recently been implicated in tumorigenesis: in fact, rhabdomyosarcomas present FOXO genes with a high percentage of mutations that render them insensitive to inhibition by AKT signaling In addition, inhibitors of the enzyme glutaminase GLS that converts glutamine to glutamate, which is subsequently transformed to GSH via the glutamate—cysteine ligase complex, efficiently induce cancer cell death through dysregulation of their antioxidant system As mentioned above, another central player in these redox systems is thioredoxin, which is reduced by NADPH to induce the transfer of electrons for use in DNA synthesis, signal transduction and redox regulation.

Interestingly, auranofin, which functions as a thioredoxin inhibitor, has been used with beneficial effects in the treatment of head and neck carcinoma cell lines; prevention of this effect by the ROS scavenger N-acetylcysteine NAC confirms the role of ROS in these cancers Triggered by an extrinsic or an intrinsic pathway, caspase-induced PCD culminates with the formation of apoptotic bodies that are eliminated by adjacent phagocytes The extrinsic pathway is mediated by binding of death-inducing ligands such as TNFα and Fas ligand that bind to cognate receptors that, in turn, recruit adaptor proteins and pro-caspases, leading to the assembly of the death-inducing signaling complex DISC and the activation of effector caspases This interaction is competed by the cellular FLICE-inhibitory protein c-FLIP : ROS have been shown to downregulate the c-FLIP half-life by inducing its ubiquitin-proteasomal degradation, thus enhancing this extrinsic pathway However, compelling evidence suggests that, for the majority of ROS-related anticancer drugs, apoptosis depends on the activation of the intrinsic pathway that involves mitochondrial PTPs, the permeability of which is increased with the cytoplasmic release of pro-apoptotic factors such as cytochrome c that forms a complex with apoptotic protease activating factor 1 Apaf-1 and pro-caspase 9 to build the apoptosome, activating, in turn, effector caspases 77 , 78 , 79 , 80 Fig.

In fact, ROS induce the three major components critical for the opening of the PTPs, the voltage-dependent anion-selective channel VDAC , adenine nucleotide translocase ANT and cyclophilin D, via the oxidation of specific cysteines in their active sites 81 , ROS also trigger apoptosis by inactivating or increasing the ubiquitination of the pivotal anti-apoptotic protein Bcl-2 and by decreasing the intracellular levels of Bax and Bad 83 , 84 Fig.

The induction of apoptosis by elevated ROS levels has been highlighted as the central mechanism responsible for the positive effects of monoclonal antibodies 85 and tyrosine kinase inhibitors 86 , which represent the core of targeted cancer therapy Among tyrosine kinase inhibitors, imatinib a PDGFR inhibitor and erlotinib an EGFR inhibitor induce ROS-dependent apoptosis in melanoma and non-small-cell lung cancer cells, respectively, through disruption of mitochondrial membrane potential upon the stimulation of JNK and p38 phosphorylation 88 , 89 , while vemurafenib a BRAF inhibitor increases the production of superoxide anions with the commensurate depolarization of the mitochondrial membranes in melanoma cells Among the monoclonal antibodies, rituximab specific to the calcium-channel protein CD20 on the surface of B cells and mature plasma cells increases ROS and induces apoptosis via the inhibition of Bcl-2 and p38MAPK signaling and is used in the treatment of B cell lymphomas As noted above, chemotherapy and radiotherapy cause an increase in intracellular ROS that can lead to apoptosis 92 , 93 via extrinsic or intrinsic pathways 94 , Many drugs used in anticancer therapy induce oxidative stress.

A course of treatment with arabinocytosine, which hampers DNA replication, followed by anthracyclines to increase ROS, has been shown to drive PCD, with beneficial effects for patients with acute myeloid leukemia AML Arsenic trioxide has recently carved out a role in cancer therapy because it can induce electron leakage along the respiratory chain It triggers apoptosis in different cancer cells, including those of myeloma, lung cancer, and leukemia , Moreover, 5-fluorouracil, a pyrimidine analog, produces ROS through pdependent pathways and induces apoptosis in colon and rectal cancer cells , ROS-induced apoptosis also explains the beneficial effect of two analogs of nuclear receptor ligands in several types of cancer: 2-methoxyestradiol, a 17β-estradiol metabolite, and N- 4-hydroxyphenyl retinamide, a synthetic analog of retinoic acid, have been shown to induce PCD in neuroblastoma and lung cancer cells, respectively , Furthermore, platinum-based drugs elevate ROS levels that promote PCD; protocols for the administration of these compounds in combination with inhibitors of poly ADP-ribose polymerase PARP , which is involved in the maintenance of DNA integrity, have been shown to arrest the growth of breast cancer cells, even in BRCA -deficient models , Intuitively, the inhibition of DNA damage repair by PARP may sensitize cancer cells to the oxidative stress induced by platinum-containing drugs.

Programmed cell death may also be mediated by the effect of elevated ROS on sphingomyelinase, which generates ceramide from sphingomyelin and binds to death receptors on the cell membrane of cancer cells. Activation of this pathway has been observed after UV irradiation of lymphoma cells Moreover, the use of drugs affecting mitochondria, where more than one-half of all ROS are generated, represents a suitable approach to induce oxidative stress and PCD in cancer cells : gamitrinib, an inhibitor of heat shock protein 90 HSP90 , induces a dramatic collapse of mitochondria in prostate cancer cells , while ARQ a quinone derivative and STA a copper chelator increase ROS through leakage in the electron transport chain and have beneficial effects in patients with solid tumors and pancreatic adenocarcinoma Apoptosis is triggered in cells with excessive endoplasmic reticulum ER stress that is induced when the protein folding ability of the ER is overwhelmed or impaired.

Recently, several drugs have been designed on the basis of their ability to aggravate ER stress in cancer cells via the induction of oxidative stress. Recently, an important therapeutic approach to kill cancer cells has been presented by ROS-induced autophagy Specifically, it has been reported that H 2 O 2 -dependent inactivation of autophagy-related gene-4 ATG4 increases LC3-associated autophagosomes and that ATM-mediated oxidation of AMP-activated protein kinase AMPK inhibits mammalian target of rapamycin 1 TORC1 , a pivotal negative regulator of autophagy , , Fig.

Indeed, autophagy, also known as type II programmed cell death, is now considered not only as a cell survival mechanism but also a tumor suppressor mechanism that induces the death of transformed cells In this regard, it has been reported that H 2 O 2 induces autophagic cell death in glioma cells after treatment with the polycyclic ammonium ion sanguinarine, which increases electron leakage from mitochondria and induces NOXs Rapamycin, administered in combination with inhibitors of HSP90, causes mitochondrial damage with accompanying oxidative stress and autophagy and reduces tumor growth in RAS -dependent tumors ROS are also able to induce necrosis, which was originally considered an unregulated form of cell death but is now recognized as type III programmed cell death necroptosis , Ferroptosis depends on the presence of intracellular iron and is induced by ROS Fig.

Therefore, the role of p53 in this context appears to be different from that reported in several studies showing that it decreases the levels of ROS. A plausible explanation of this apparent dichotomy is that p53 promotes cell survival by preventing excessive increases in ROS under moderate oxidative stress, whereas when the oxygen species increase over a threshold level, it switches to becoming a ROS inducer, triggering cell death.

On the basis that ferroptosis is considered an oxidation-induced cell death mechanism, several trials with different drugs that elicit this pathway have been conducted , Erastin is a synthetic drug that induces cell death through ferroptosis in tumor cells bearing mutant RAS by increasing intracellular ROS levels and altering the permeability of the outer mitochondrial membrane , Increased ROS levels are thought to impair the multidrug resistance of cancer cells, which causes cancer development and metastasis during or after chemotherapy , It has been recently shown that efflux pumps in the plasma membrane of cancer cells are crucial for the extracellular efflux of anticancer drugs These pumps belong to the adenosine triphosphate ATP -binding cassette ABC transporter superfamily and are dependent on intracellular ATP stores ATP is accumulated by a synthase driven by a proton gradient generated in mitochondria by the NADH-dependent electron transport chain , ; therefore, one possible way to overcome efficient efflux in cancer drugs is to inhibit ATP synthesis by promoting NADH conversion to NAD through lipid membrane-coated silica carbon nanoparticles that, under near-infrared laser irradiation, target mitochondria and produce ROS with simultaneous consumption of NADH A new role of ROS related to transcriptional output has been recently highlighted.

It is well known that cells follow a strictly scheduled program for differentiation that is based on an orchestrated sequence of gene expression. Because of spatial constraints, genes must engage in a complex unfolding process to become accessible to the transcriptional machinery, which is triggered through posttranslational modifications at the N-terminal tails of core histones.

Together, these modifications, induced by coordinated targeting of transcription factors that is currently referred to as epigenetic marks, conform to a precise code with specific time requirements to control whole gene expression We have previously shown that estrogen-induced transcription is triggered by LSD1-catalyzed demethylation of lysine 9 in histone H3 H3K9 , which is activated by the binding of liganded estrogen receptor to the enhancers of target genes Intuitively, generation of ROS in this process must be timely and spatially controlled to prevent excessive damage to the DNA: a recent report, in fact, describes a new role for the originally discovered superoxide dismutase, SOD1, that is recruited to the nucleus in response to specific stimuli However, it has also been observed that hormone-induced phosphorylation of serine 10 in H3 histone H3S10 prevents the rapid remethylation of the preceding lysine, serving as the metronome of the process and giving the DNA damage repair system enough time to eliminate the oxidized nucleotides from nearby DNA It has been reported that by inhibiting phosphorylation of serine 10 in this pathway, breast cancer cells simultaneously challenged with estradiol show an overproduction of ROS, with increased oxidation of the DNA that overwhelms the repair apparatus and triggers PCD in a great percentage of these cells Fig.

ROS generated during nuclear receptor-induced transcription of target genes by the activity of lysine demethylases on lysine 9 in histone H3 must be controlled to prevent their accumulation. To this end, SOD1 reaches the nuclear space, while phosphorylation of H3S10 inhibits the rapid remethylation of the same lysine.

If inhibitors of the H3S10 kinases are introduced as a Trojan horse together with nuclear receptor ligands, remethylation of H3K9 is quick, nuclear ROS accumulate, and unrepaired DNA damage triggers PCD.

The complex interconnection between ROS levels and cancer is essentially based on accurate fine-tuning between ROS production and scavenging. Cancer initiation and progression leverage slight increases in ROS levels.

Therefore, cancer cells thrive on levels of ROS that are moderately higher than those in their normal counterparts, as they have developed increased antioxidant systems. This feature renders cancer cells more sensitive to external stimuli that further increase the production of ROS , , , and, as schematically summarized in Table 1 , an increasing number of therapeutic strategies are being developed to elevate ROS levels to overwhelm the redox adaptation of the same cells, inducing oxidative stress incompatible with cellular life , , , Fig.

The first approach is based on lowering ROS levels to counteract their role in cellular transformation; it is aimed at reducing the number of transformed cells by depriving them of fuel represented in the upper right side of the figure as a lower proportion of transformed cells with respect to that of normal cells.

The second approach is based on the consideration that cancer cells, with an antioxidant system already triggered, are more sensitive than their normal counterparts to further increases in ROS and are unable to achieve redox balance. Therefore, by inducing ROS under these metabolic conditions, a high percentage of the cells undergo death represented in the lower right side of the figure, where transformed cells are depicted as apoptotic.

Specifically, cellular responses to ROS must be imagined as the integration of multiple levels in which, in addition to their nature and relative concentration, their location plays an important role.

In fact, mitochondrial ROS have been reported to essentially promote cell death, while NOX-generated ROS have been associated with the promotion of cell proliferation and migration Furthermore, in contrast to the mechanism of sister pathways, redox signaling is based on migrating electrons, and therefore, the signaling in this pathway is much more diffuse.

In reference to the nature of ROS behavior as a double-edged sword, even though several studies have documented the benefits of antioxidant drugs for cancer therapies, none has been supported by solid trials performed on a large scale , In contrast, the most recent studies have shown an increase in tumor development and metastasis in mouse models treated with vitamin E an opposite result of that in which high doses of vitamin C increase ROS levels to induce the death of colon cancer cells bearing KRAS and BRAF mutations In addition, it has been shown that the administration of antioxidants, such as N-acetylcysteine, accelerates the progression of lung cancers and melanomas and that increasing the expression of the antioxidant-encoding Nrf2 gene enhances the growth of lung tumors , , , In fact, and in contrast to the previous view, the results of many studies support a scenario in which the inhibition of antioxidant enzymes ensures the death of cancer cells, especially when this approach is used in combination with treatments that increase ROS.

This approach is an alternative to the traditional strategy of targeting oncogenes and tumor suppressor genes, a strategy that appears complicated because of the high number of genes involved and their ability to drive compensatory pathways Interestingly, increased ROS-induced apoptosis has been reported in cancer cells after depletion of ATP derived from the manipulation of glycolytic enzymes, chemotherapy or radiation therapy; these data highlight the potential eminent role of ROS modulation in anticancer combinatorial therapies , Finally, the most recent ROS-inducing drugs have addressed the pivotal goal of therapists: cancer selectivity.

In this regard, good results have been reached through photodynamic therapy, which is based on the generation of ROS after stimulation of a photosensitizer by light: cancer cells under treatment internalize porphyrin precursor molecules to induce the formation of ROS that lead to photooxidative stress and cancer-specific cell death , In fact, although more studies are required to increase the selectivity of these anticancer ROS-related drugs, the common mechanisms elicited by oncogenes to promote the adaptation to a large set of stress conditions are being revealed in more depth every day, and in a high percentage, they concern the redox balance.

In conclusion, we expect that targeting ROS will represent fruitful ground for future molecular anticancer strategies. Forman, H. An overview of mechanisms of redox signaling.

Article PubMed CAS Google Scholar. Bedard, K. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Vander Heiden, M. Understanding the Warburg effect: the metabolic requirements of cell proliferation.

Science , — Article CAS Google Scholar. Finkel, T. Signal transduction by reactive oxygen species. Cell Biol. Article PubMed PubMed Central CAS Google Scholar. Paradies, G. et al. Circulation Res. Petrosillo, G. Decreased complex III activity in mitochondria isolated from rat heart subjected to ischemia and reperfusion: role of reactive oxygen species and cardiolipin.

FASEB J. Valko, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Ray, P. Reactive oxygen species ROS homeostasis and redox regulation in cellular signaling. Houee-Levin, C. Exploring oxidative modifications of tyrosine: an update on mechanisms of formation, advances in analysis and biological consequences.

Free Radic. Thannickal, V. Activation of an H 2 O 2 -generating NADH oxidase in human lung fibroblasts by transforming growth factor beta 1.

Meng, T. Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo. Cell 9 , — Kamata, H. Reactive oxygen species promote TNF alpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases.

Cell , — Esposito, F. Protein kinase B activation by reactive oxygen species is independent on tyrosine kinase receptor phosphorylation and required Src activity. Sundaresan, M. Requirement for generation of H 2 O 2 for platelet-derived growth factor signal transduction.

Corcoran, A. Redox regulation of protein kinases. FEBS J. Kwon, J. Reversible oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors. Natl Acad. USA , — Article PubMed CAS PubMed Central Google Scholar.

Latimer, H. Peroxiredoxins in regulation of MAPK signalling pathways; sensors and barriers to signal transduction. Gopalakrishna, R. Protein kinase C signaling and oxidative stress. Free Rad. Taguchi, K. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution.

Genes Cells 16 , — Meister, A. Selective modification of glutathione metabolism. Thimmulappa, R. Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray.

Cancer Res. PubMed CAS Google Scholar. Orino, K. Ferritin and the response to oxidative stress. Weinberg, E. The role of iron in cancer. Cancer Prev. Essers, M. FOXO transcription factor activation by oxidative stress mediated by the small GTPase Ral and JNK. EMBO J. Putker, M. Cell 49 , — Melnik, B.

p key conductor of all anti-acne therapies. Article PubMed PubMed Central Google Scholar. Rhee, S. The antioxidant function of sestrins is mediated by promotion of autophagic degradation of keap1 and Nrf2 activation of mTORC1.

Pinnix, Z. Ferroportin and iron regulation in breast cancer progression and prognosis. Huang, L. Regulation of hypoxia-inducible factor 1α is mediated by an O 2 -dependent degradation domain via the ubiquitin-proteasome pathway. USA 95 , — Abid, M.

NADPH oxidase activity selectively modulates vascular endothelial growth factor signaling pathways. Guo, Z. ATM activation by oxidative stress. Svegliati, S. Oxidative DNA damge induces the ATM-mediated transcriptional suppression of the Wnt inhibitor WIF-1 in systemic sclerosis and fibrosis.

Tell, G. Nucleic Acids Res. Ando, K. Toledano, M. Modulation of transcription factor NF-kappa B binding activity by oxidation-reduction in vitro. USA 88 , — Matthews, J. Role of cysteine62 in DNA recognition by the p50 subunit of NF-kappa B.

Lingappan, K. NF-kB in oxidative stress. Article PubMed Google Scholar. Matsushima, S. Increased oxidative stress in the nucleus caused by Nox4 mediates oxidation of HDAC4 and cardiac hypertrophy. Ushijima, T.

Detection and interpretation of altered methylation patterns in cancer cells. Cancer 5 , — Grollman, A. Mutagenesis by 8-oxoguanine: an enemy within. Trends Genet. Egger, G. J Natl Cancer Inst — Murakami T, Cardones AR, Finkelstein SE, Restifo NP, Klaunberg BA, Nestle FO, Castillo SS, Dennis PA, Hwang ST Immune evasion by murine melanoma mediated through CC chemokine receptor Kwong J, Kulbe H, Wong D, Chakravarty P, Balkwill F An antagonist of the chemokine receptor CXCR4 induces mitotic catastrophe in ovarian cancer cells.

Mol Cancer Ther — Kalluri R, Zeisberg M Fibroblasts in cancer. CAS PubMed Google Scholar. Gialeli C, Theocharis AD, Karamanos NK Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting.

Van Lint P, Libert C Chemokine and cytokine processing by matrix metalloproteinases and its effect on leukocyte migration and inflammation. J Leukoc Biol — Deryugina EI, Quigley JP Tumor angiogenesis: MMP-mediated induction of intravasation-and metastasis-sustaining neovasculature.

Matrix Biol — PLoS One 4:e de Souza Junior DA, Santana AC, da Silva EZM, Oliver C, Jamur MC The role of mast cell specific chymases and tryptases in tumor angiogenesis. Biomed Res Int. Mori K, Shibanuma M, Nose K Invasive potential induced under long-term oxidative stress in mammary epithelial cells.

Westermarck J, KÄHÄRI V-M Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J — Swanson KV, Deng M, Ting JP-Y The NLRP3 inflammasome: molecular activation and regulation to therapeutics.

Lin C, Zhang J Inflammasomes in inflammation-induced cancer. Front Immunol Abais JM, Xia M, Zhang Y, Boini KM, Li P-L Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica.

Szabo G, Petrasek J Inflammasome activation and function in liver disease. Nat Rev Gastroenterol Hepatol Zhong Z, Zhai Y, Liang S, Mori Y, Han R, Sutterwala FS, Qiao L TRPM2 links oxidative stress to NLRP3 inflammasome activation. Nat Commun — Download references. Cell Biology Laboratory, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India.

Foram U. Clinical Genomics Center Mount Sinai Hospital, Toronto, ON, Canada. You can also search for this author in PubMed Google Scholar. Correspondence to Chandramani Pathak. Department of Biochemistry, Central University of Haryana, Mahendergarh, Haryana, India.

Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia. Reprints and permissions. Vaidya, F. Oxidative Stress and Inflammation Can Fuel Cancer.

In: Maurya, P. eds Role of Oxidative Stress in Pathophysiology of Diseases. Springer, Singapore. Published : 16 June Publisher Name : Springer, Singapore. Print ISBN : Online ISBN : eBook Packages : Biomedical and Life Sciences Biomedical and Life Sciences R0. Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Policies and ethics. Skip to main content. Abstract Both oxidative stress and inflammation are interdependent cellular consequences of a biological defense system, which can fuel cancer and other pathophysiological provenience.

Keywords Cancer Oxidative stress Inflammation Cancer metabolism Oxidative stress Cell proliferation. Buying options Chapter EUR eBook EUR Softcover Book EUR Hardcover Book EUR Tax calculation will be finalised at checkout Purchases are for personal use only Learn about institutional subscriptions.

References Alfadda AA, Sallam RM Reactive oxygen species in health and disease. Oxid Med Cell Longev PubMed PubMed Central Google Scholar Panieri E, Santoro M ROS homeostasis and metabolism: a dangerous liason in cancer cells.

Cell Death Dis 7:e PubMed PubMed Central CAS Google Scholar Zini R, Berdeaux A, Morin D The differential effects of superoxide anion, hydrogen peroxide and hydroxyl radical on cardiac mitochondrial oxidative phosphorylation. Free Radic Res — PubMed CAS Google Scholar Holmström KM, Finkel T Cellular mechanisms and physiological consequences of redox-dependent signalling.

Nat Rev Mol Cell Biol — PubMed Google Scholar Sumimoto H Structure, regulation and evolution of Nox-family NADPH oxidases that produce reactive oxygen species.

FEBS J — PubMed CAS Google Scholar Finkel T Signal transduction by reactive oxygen species. J Cell Biol —15 PubMed PubMed Central CAS Google Scholar Trachootham D, Lu W, Ogasawara MA, Valle NR-D, Huang P Redox regulation of cell survival.

Antioxid Redox Signal — PubMed PubMed Central CAS Google Scholar Liou G-Y, Storz P Reactive oxygen species in cancer. Free Radic Res — PubMed CAS Google Scholar Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O Oxidative stress and antioxidant defense.

World Allergy Organ J PubMed PubMed Central CAS Google Scholar McCord JM The evolution of free radicals and oxidative stress. Am J Med — PubMed CAS Google Scholar Oberley T, Oberley L Antioxidant enzyme levels in cancer.

Histol Histopathol — PubMed CAS Google Scholar Reiter RJ, Acuña-Castroviejo D, Tan DX, Burkhardt S Free radical-mediated molecular damage. Ann N Y Acad Sci — PubMed CAS Google Scholar Pollack M, Leeuwenburgh C Molecular mechanisms of oxidative stress in aging: free radicals, aging, antioxidants and disease.

Elsevier Science BV, Amsterdam, pp — Google Scholar Wiseman H, Halliwell B Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer.

Biochem J PubMed PubMed Central CAS Google Scholar Matés JM, Sánchez-Jiménez F Antioxidant enzymes and their implications in pathophysiologic processes. Front Biosci 4:D—D PubMed Google Scholar Breitenbach M, Eckl P Introduction to oxidative stress in biomedical and biological research.

Biomol Ther — CAS Google Scholar Burton GJ, Jauniaux EJ Oxidative stress. Best Pract Res Clin Obstet Gynaecol — PubMed PubMed Central Google Scholar Li R, Jia Z, Trush MA Defining ROS in biology and medicine. React Oxyg Species Google Scholar Fukai T, Ushio-Fukai MJA Superoxide dismutases: role in redox signaling, vascular function, and diseases.

Antioxid Redox Signal — PubMed PubMed Central CAS Google Scholar Ushio-Fukai M, Nakamura Y Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy.

Cancer Lett —52 PubMed PubMed Central CAS Google Scholar Aggarwal V, Tuli HS, Varol A, Thakral F, Yerer MB, Sak K, Varol M, Jain A, Khan M, Sethi GJB Role of reactive oxygen species in cancer progression: molecular mechanisms and recent advancements. Biomolecules PubMed Central CAS Google Scholar Lodish H, Berk A, Zipursky SL, Matsudaira P, Baltimore D, Darnell J Electron transport and oxidative phosphorylation.

WH Freeman, New York Google Scholar Phaniendra A, Jestadi DB, Periyasamy L Free radicals: properties, sources, targets, and their implication in various diseases.

Indian J Clin Biochem —26 PubMed CAS Google Scholar Bibov MY, Kuzmin AV, Alexandrova AA, Chistyakov VA, Dobaeva NM, Kundupyan OL Role of the reactive oxygen species induced DNA damage in human spermatozoa dysfunction.

AME Med J —12 Google Scholar Waris G, Ahsan H Reactive oxygen species: role in the development of cancer and various chronic conditions. J Carcinog PubMed PubMed Central Google Scholar Yang H, Villani RM, Wang H, Simpson MJ, Roberts MS, Tang M, Liang X The role of cellular reactive oxygen species in cancer chemotherapy.

J Exp Clin Cancer Res PubMed PubMed Central CAS Google Scholar Sosa V, Moliné T, Somoza R, Paciucci R, Kondoh H, LLeonart ME Oxidative stress and cancer: an overview. Ageing Res Rev — PubMed CAS Google Scholar Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB Oxidative stress, inflammation, and cancer: how are they linked?

Free Radic Biol Med — PubMed PubMed Central CAS Google Scholar Acharya A, Das I, Chandhok D, Saha T Redox regulation in cancer: a double-edged sword with therapeutic potential.

Oxid Med Cell Longev —34 PubMed PubMed Central Google Scholar Reczek CR, Chandel NS The two faces of reactive oxygen species in cancer. Annu Rev Cancer Biol —98 Google Scholar Ward PS, Thompson CB Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell — PubMed PubMed Central CAS Google Scholar Liberti MV, Locasale JW The Warburg effect: how does it benefit cancer cells?

Trends Biochem Sci — PubMed PubMed Central CAS Google Scholar Dhup S, Dadhich RK, Porporato PE, Sonveaux P Multiple biological activities of lactic acid in cancer: influences on tumor growth, angiogenesis and metastasis. Ann Transl Med — PubMed PubMed Central CAS Google Scholar Pathak C, Vinayak M Modulation of lactate dehydrogenase isozymes by modified base queuine.

Mol Biol Rep — PubMed CAS Google Scholar Mohammad GH, Damink SO, Malago M, Dhar DK, Pereira SP Pyruvate kinase M2 and lactate dehydrogenase A are overexpressed in pancreatic cancer and correlate with poor outcome. PLoS One e PubMed PubMed Central Google Scholar Hsu M-C, Hung W-C Pyruvate kinase M2 fuels multiple aspects of cancer cells: from cellular metabolism, transcriptional regulation to extracellular signaling.

Mol Cancer PubMed PubMed Central Google Scholar Gui DY, Lewis CA, Vander Heiden MG Allosteric regulation of PKM2 allows cellular adaptation to different physiological states. Sci Signal 6:pe7 PubMed Google Scholar Anastasiou D, Yu Y, Israelsen WJ, Jiang J-K, Boxer MB, Hong BS, Tempel W, Dimov S, Shen M, Jha A Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis.

Nat Chem Biol PubMed PubMed Central CAS Google Scholar Anastasiou D, Poulogiannis G, Asara JM, Boxer MB, Jiang J-K, Shen M, Bellinger G, Sasaki AT, Locasale JW, Auld DSJS Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses.

Science — PubMed PubMed Central CAS Google Scholar Finley LW, Carracedo A, Lee J, Souza A, Egia A, Zhang J, Teruya-Feldstein J, Moreira PI, Cardoso SM, Clish CB SIRT3 opposes reprogramming of cancer cell metabolism through HIF1α destabilization.

Cancer Cell — PubMed PubMed Central CAS Google Scholar Fiaschi T, Chiarugi P Oxidative stress, tumor microenvironment, and metabolic reprogramming: a diabolic liaison. Int J Cell Biol PubMed PubMed Central Google Scholar Burdon RH, Gill V, Rice-Evans C Oxidative stress and tumour cell proliferation.

Free Radic Res Commun —76 PubMed CAS Google Scholar Kumari S, Badana AK, Malla R Reactive oxygen species: a key constituent in cancer survival.

Biomark Insights PubMed PubMed Central Google Scholar Storz P Reactive oxygen species in tumor progression. Front Biosci — PubMed CAS Google Scholar Yang Y, Fang E, Luo J, Wu H, Jiang Y, Liu Y, Tong S, Wang Z, Zhou R, Tong Q The antioxidant alpha-lipoic acid inhibits proliferation and invasion of human gastric cancer cells via suppression of STAT3-mediated MUC4 gene expression.

Oxid Med Cell Longev PubMed PubMed Central Google Scholar Hamanaka RB, Chandel NS Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem Sci — PubMed PubMed Central CAS Google Scholar Wang M, Kirk JS, Venkataraman S, Domann FE, Zhang HJ, Schafer FQ, Flanagan SW, Weydert CJ, Spitz DR, Buettner GR Manganese superoxide dismutase suppresses hypoxic induction of hypoxia-inducible factor-1 α and vascular endothelial growth factor.

Oncogene — PubMed CAS Google Scholar Patterson JC, Joughin BA, van de Kooij B, Lim DC, Lauffenburger DA, Yaffe MB ROS and oxidative stress are elevated in mitosis during asynchronous cell cycle progression and are exacerbated by mitotic arrest.

e PubMed PubMed Central CAS Google Scholar Ranjan P, Anathy V, Burch PM, Weirather K, Lambeth JD, Heintz NH Redox-dependent expression of cyclin D1 and cell proliferation by Nox1 in mouse lung epithelial cells.

Exp Cell Res —71 PubMed Google Scholar Verbon EH, Post JA, Boonstra J The influence of reactive oxygen species on cell cycle progression in mammalian cells.

Gene —6 PubMed CAS Google Scholar Kelkel M, Schumacher M, Dicato M, Diederich M Antioxidant and anti-proliferative properties of lycopene. Free Radic Res — PubMed CAS Google Scholar Laphanuwat P, Likasitwatanakul P, Sittithumcharee G, Thaphaengphan A, Chomanee N, Suppramote O, Ketaroonrut N, Charngkaew K, Lam EW-F, Okada S Cyclin D1 depletion interferes with oxidative balance and promotes cancer cell senescence.

Nutr J Google Scholar Kozlov SV, Waardenberg AJ, Engholm-Keller K, Arthur JW, Graham ME, Lavin M Reactive oxygen species ROS -activated ATM-dependent phosphorylation of cytoplasmic substrates identified by large-scale phosphoproteomics screen.

Mol Cell Proteomics — PubMed CAS Google Scholar Mantovani F, Collavin L, Del Sal G Mutant p53 as a guardian of the cancer cell. Cell Death Differ — PubMed Google Scholar Ma Q Role of nrf2 in oxidative stress and toxicity.

Annu Rev Pharmacol Toxicol — PubMed PubMed Central CAS Google Scholar Haupt S, Raghu D, Haupt Y Mutant p53 drives cancer by subverting multiple tumor suppression pathways.

Front Oncol PubMed PubMed Central Google Scholar Lisek K, Campaner E, Ciani Y, Walerych D, Del Sal G Mutant p53 tunes the NRF2-dependent antioxidant response to support survival of cancer cells. Oncotarget PubMed PubMed Central Google Scholar Du X, Fu X, Yao K, Lan Z, Xu H, Cui Q, Yang E Bcl-2 delays cell cycle through mitochondrial ATP and ROS.

Cell Cycle — PubMed PubMed Central CAS Google Scholar Liang J, Cao R, Wang X, Zhang Y, Wang P, Gao H, Li C, Yang F, Zeng R, Wei P Mitochondrial PKM2 regulates oxidative stress-induced apoptosis by stabilizing Bcl2. Cell Res — PubMed CAS Google Scholar Martin TA, Ye L, Sanders AJ, Lane J, Jiang WG Cancer invasion and metastasis: molecular and cellular perspective.

Landes Bioscience, Austin Google Scholar Seyfried TN, Huysentruyt LC On the origin of cancer metastasis. Crit Rev Oncog PubMed PubMed Central Google Scholar Lambert AW, Pattabiraman DR, Weinberg RA Emerging biological principles of metastasis.

Cell — PubMed PubMed Central CAS Google Scholar Baba AI, Câtoi C Tumor cell morphology. The Publishing House of the Romanian Academy, Bucharest Google Scholar Heerboth S, Housman G, Leary M, Longacre M, Byler S, Lapinska K, Willbanks A, Sarkar S EMT and tumor metastasis.

Clin Transl Med PubMed PubMed Central Google Scholar Mittal V Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol — PubMed CAS Google Scholar Wang H, Unternaehrer JJ Epithelial-mesenchymal transition and cancer stem cells: at the crossroads of differentiation and dedifferentiation.

Dev Dyn —20 PubMed Google Scholar Nagarsheth N, Wicha MS, Zou W Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Mol Med Rep — PubMed Google Scholar Liao Z, Chua D, Tan NS Reactive oxygen species: a volatile driver of field cancerization and metastasis.

Mol Cancer PubMed PubMed Central Google Scholar Miyazaki Y, Hara A, Kato K, Oyama T, Yamada Y, Mori H, Shibata T The effect of hypoxic microenvironment on matrix metalloproteinase expression in xenografts of human oral squamous cell carcinoma. Int J Oncol — PubMed CAS Google Scholar Zhang L, Huang G, Li X, Zhang Y, Jiang Y, Shen J, Liu J, Wang Q, Zhu J, Feng X Hypoxia induces epithelial-mesenchymal transition via activation of SNAI1 by hypoxia-inducible factor-1α in hepatocellular carcinoma.

BMC Cancer PubMed PubMed Central CAS Google Scholar Nishida N, Kudo M Oxidative stress and epigenetic instability in human hepatocarcinogenesis.

Dig Dis — PubMed Google Scholar Nishida N, Yano H, Nishida T, Kamura T, Kojiro MJ Angiogenesis in cancer.

Vasc Health Risk Manag PubMed PubMed Central CAS Google Scholar Kim Y-W, Byzova TVJB Oxidative stress in angiogenesis and vascular disease. Blood — PubMed PubMed Central CAS Google Scholar Carmeliet PJ VEGF as a key mediator of angiogenesis in cancer. Oncology —10 PubMed CAS Google Scholar Babior BM NADPH oxidase.

Curr Opin Immunol —47 PubMed CAS Google Scholar Xia C, Meng Q, Liu LZ, Rojanasakul Y, Wang XR, Jiang BH Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor.

Cancer Res — PubMed CAS Google Scholar Sun X, Wei J, Tang Y, Wang B, Zhang Y, Shi L, Guo J, Hu F, Li X Leptin-induced migration and angiogenesis in rheumatoid arthritis is mediated by reactive oxygen species.

FEBS Open Bio — PubMed PubMed Central CAS Google Scholar Coussens LM, Werb Z Inflammation and cancer. Nature PubMed PubMed Central CAS Google Scholar Blaylock RL Cancer microenvironment, inflammation and cancer stem cells: a hypothesis for a paradigm change and new targets in cancer control.

Surg Neurol Int PubMed PubMed Central Google Scholar Landskron G, De la Fuente M, Thuwajit P, Thuwajit C, Hermoso MA Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res PubMed PubMed Central Google Scholar Hanahan D, Weinberg RA Hallmarks of cancer: the next generation.

Cell — PubMed CAS Google Scholar Kiraly O, Gong G, Olipitz W, Muthupalani S, Engelward BP Inflammation-induced cell proliferation potentiates DNA damage-induced mutations in vivo. PLoS Genet e PubMed PubMed Central Google Scholar Mantovani A, Allavena P, Sica A, Balkwill F Cancer-related inflammation.

Nature PubMed CAS Google Scholar Grivennikov SI, Greten FR, Karin M Immunity, inflammation, and cancer. Cell — PubMed PubMed Central CAS Google Scholar Kim SS, Ruiz VE, Carroll JD, Moss SF Helicobacter pylori in the pathogenesis of gastric cancer and gastric lymphoma. Cancer Lett — PubMed CAS Google Scholar Wroblewski LE, Peek RM, Wilson KT Helicobacter pylori and gastric cancer: factors that modulate disease risk.

Clin Microbiol Rev — PubMed PubMed Central CAS Google Scholar Arbuthnot P, Kew M Hepatitis B virus and hepatocellular carcinoma. Int J Exp Pathol — PubMed PubMed Central CAS Google Scholar El-Serag HB Hepatocellular carcinoma and hepatitis C in the United States.

Hepatology — Google Scholar de Martel C, Franceschi S Infections and cancer: established associations and new hypotheses. Clin Microbiol Rev — PubMed PubMed Central CAS Google Scholar Balkwill F, Mantovani A Inflammation and cancer: back to Virchow?

Lancet — PubMed CAS Google Scholar Stark T, Livas L, Kyprianou N Inflammation in prostate cancer progression and therapeutic targeting. Transl Androl Urol PubMed PubMed Central Google Scholar Shrihari T Dual role of inflammatory mediators in cancer.

Ecancermedicalscience PubMed PubMed Central CAS Google Scholar Hoesel B, Schmid JA The complexity of NF-κB signaling in inflammation and cancer.

Mol Cancer PubMed PubMed Central CAS Google Scholar Mogensen TH Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev — PubMed PubMed Central CAS Google Scholar Franchi L, Eigenbrod T, Muñoz-Planillo R, Nuñez G The inflammasome: a caspaseactivation platform that regulates immune responses and disease pathogenesis.

Nat Immunol — PubMed PubMed Central CAS Google Scholar Fialkow L, Wang Y, Downey GP Reactive oxygen and nitrogen species as signaling molecules regulating neutrophil function.

Free Radic Biol Med — PubMed CAS Google Scholar Salzano S, Checconi P, Hanschmann E-M, Lillig CH, Bowler LD, Chan P, Vaudry D, Mengozzi M, Coppo L, Sacre S Linkage of inflammation and oxidative stress via release of glutathionylated peroxiredoxin-2, which acts as a danger signal.

Proc Natl Acad Sci — PubMed CAS PubMed Central Google Scholar Martinon F Signaling by ROS drives inflammasome activation. Eur J Immunol — PubMed CAS Google Scholar Latz E, Xiao TS, Stutz A Activation and regulation of the inflammasomes.

Nat Rev Immunol — PubMed CAS Google Scholar Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J Thioredoxin-interacting protein links oxidative stress to inflammasome activation.

Nat Immunol PubMed CAS Google Scholar Zhou R, Yazdi AS, Menu P, Tschopp J A role for mitochondria in NLRP3 inflammasome activation. Nature — PubMed CAS Google Scholar Ghezzi P Role of glutathione in immunity and inflammation in the lung.

Int J Gen Med PubMed PubMed Central CAS Google Scholar Morgan MJ, Liu Z-G Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res — PubMed CAS Google Scholar Blanco P, Palucka AK, Pascual V, Banchereau J Dendritic cells and cytokines in human inflammatory and autoimmune diseases.

Cytokine Growth Factor Rev —52 PubMed PubMed Central CAS Google Scholar Klampfer L Cytokines, inflammation and colon cancer. Curr Cancer Drug Targets — PubMed PubMed Central CAS Google Scholar Liang M, Zhang P, Fu J Up-regulation of LOX-1 expression by TNF-α promotes trans-endothelial migration of MDA-MB breast cancer cells.

Cancer Lett —37 PubMed CAS Google Scholar Zamarron BF, Chen W Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci PubMed PubMed Central CAS Google Scholar Lin W-W, Karin M A cytokine-mediated link between innate immunity, inflammation, and cancer.

J Clin Investig PubMed CAS PubMed Central Google Scholar Knüpfer H, Preiss R Serum interleukin-6 levels in colorectal cancer patients—a summary of published results. Int J Colorectal Dis — PubMed Google Scholar Tanaka T, Bai Z, Srinoulprasert Y, Yang B, Hayasaka H, Miyasaka M Chemokines in tumor progression and metastasis.

Cancer Sci — PubMed CAS Google Scholar Yu JH, Kim H Oxidative stress and cytokines in the pathogenesis of pancreatic cancer. J Cancer Prev PubMed PubMed Central Google Scholar Prawan A, Buranrat B, Kukongviriyapan U, Sripa B, Kukongviriyapan V Inflammatory cytokines suppress NAD P H: quinone oxidoreductase-1 and induce oxidative stress in cholangiocarcinoma cells.

Oncol Rep — PubMed CAS Google Scholar Esquivel-Velázquez M, Ostoa-Saloma P, Palacios-Arreola MI, Nava-Castro KE, Castro JI, Morales-Montor J The role of cytokines in breast cancer development and progression.

J Interferon Cytokine Res —16 PubMed PubMed Central Google Scholar Wojdasiewicz P, Poniatowski ŁA, Szukiewicz D The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm PubMed PubMed Central Google Scholar Ravichandran K, Tyagi A, Deep G, Agarwal C, Agarwal R Interleukininduced iNOS expression in human lung carcinoma A cells: involvement of STAT and MAPK pathways Google Scholar Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S The pro-and anti-inflammatory properties of the cytokine interleukin Biochim Biophys Acta Mol Cell Res — CAS Google Scholar Brizzi MF, Formato L, Dentelli P, Rosso A, Pavan M, Garbarino G, Pegoraro M, Camussi G, Pegoraro L Interleukin-3 stimulates migration and proliferation of vascular smooth muscle cells.

Circulation — PubMed CAS Google Scholar Ehrlich LA, Chung HY, Ghobrial I, Choi SJ, Morandi F, Colla S, Rizzoli V, Roodman GD, Giuliani N IL-3 is a potential inhibitor of osteoblast differentiation in multiple myeloma. Blood — PubMed CAS Google Scholar Gupta N, Barhanpurkar AP, Tomar GB, Srivastava RK, Kour S, Pote ST, Mishra GC, Wani MR IL-3 inhibits human osteoclastogenesis and bone resorption through downregulation of c-Fms and diverts the cells to dendritic cell lineage.

J Immunol — PubMed CAS Google Scholar Heikkilä K, Ebrahim S, Lawlor DA Systematic review of the association between circulating interleukin-6 IL-6 and cancer.

Eur J Cancer — PubMed Google Scholar Hodge DR, Hurt EM, Farrar WL The role of IL-6 and STAT3 in inflammation and canicer. Eur J Cancer — PubMed CAS Google Scholar Aggarwal BB, Sethi G, Ahn KS, Sandur SK, Pandey MK, Kunnumakkara AB, Sung B, Ichikawa H Targeting signal-transducer-and-activator-of-transcription-3 for prevention and therapy of cancer.

Ann N Y Acad Sci — PubMed CAS Google Scholar Rao V, Dyer C, Jameel J, Drew P, Greenman J Potential prognostic and therapeutic roles for cytokines in breast cancer. J Clin Oncol — PubMed PubMed Central CAS Google Scholar Kinoshita H, Hirata Y, Nakagawa H, Sakamoto K, Hayakawa Y, Takahashi R, Nakata W, Sakitani K, Serizawa T, Hikiba Y Interleukin-6 mediates epithelial—stromal interactions and promotes gastric tumorigenesis.

PLoS One 8:e PubMed PubMed Central CAS Google Scholar Gasche JA, Hoffmann J, Boland CR, Goel A Interleukin-6 promotes tumorigenesis by altering DNA methylation in oral cancer cells.

Int J Cancer — PubMed PubMed Central CAS Google Scholar Baffet G, Braciak T, Fletcher R, Gauldie J, Fey G, Northemann W Autocrine activity of interleukin 6 secreted by hepatocarcinoma cell lines.

Mol Biol Med — PubMed CAS Google Scholar Atreya R, Mudter J, Finotto S, Müllberg J, Jostock T, Wirtz S, Schütz M, Bartsch B, Holtmann M, Becker C Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo.

Nat Med PubMed CAS Google Scholar Becker C, Fantini MC, Schramm C, Lehr HA, Wirtz S, Nikolaev A, Burg J, Strand S, Kiesslich R, Huber S TGF-β suppresses tumor progression in colon cancer by inhibition of IL-6 trans-signaling.

Immunity — PubMed CAS Google Scholar McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, Cua DJ TGF-[beta] and IL-6 drive the production of IL and IL by T cells and restrain TH cell-mediated pathology. Nat Immunol PubMed CAS Google Scholar Onishi RM, Gaffen SL Interleukin and its target genes: mechanisms of interleukin function in disease.

Drug Des Devel Ther PubMed PubMed Central Google Scholar Maniati E, Soper R, Hagemann T Up for Mischief? Infect Agent Cancer PubMed PubMed Central Google Scholar Numasaki M, Fukushi J-I, Ono M, Narula SK, Zavodny PJ, Kudo T, Robbins PD, Tahara H, Lotze MT Interleukin promotes angiogenesis and tumor growth.

Blood — PubMed CAS Google Scholar Kolls JK, Lindén A Interleukin family members and inflammation. Immunity — PubMed CAS Google Scholar Kryczek I, Wei S, Zou L, Altuwaijri S, Szeliga W, Kolls J, Chang A, Zou W Cutting edge: Th17 and regulatory T cell dynamics and the regulation by IL-2 in the tumor microenvironment.

J Immunol — PubMed CAS Google Scholar Swann JB, Smyth MJ Immune surveillance of tumors. J Clin Investig PubMed CAS PubMed Central Google Scholar Meier B, Radeke H, Selle S, Younes M, Sies H, Resch K, Habermehl G Human fibroblasts release reactive oxygen species in response to interleukin-1 or tumour necrosis factor-α.

Biochem J — PubMed PubMed Central CAS Google Scholar Li Q, Engelhardt JF Interleukin-1β induction of NFκB is partially regulated by H2O2-mediated activation of NFκB-inducing kinase. J Biol Chem — PubMed CAS Google Scholar Hsu S, Hsu P Autocrine and paracrine functions of cytokines in malignant lymphomas.

Endocrinology — PubMed CAS Google Scholar Yuzhalin A, Kutikhin A Interleukins in cancer biology: their heterogeneous role. Academic, Cambridge Google Scholar Szlosarek P, Charles KA, Balkwill FR Tumour necrosis factor-α as a tumour promoter.

Eur J Cancer — PubMed CAS Google Scholar Szlosarek PW, Balkwill FR Tumour necrosis factor α: a potential target for the therapy of solid tumours. Lancet Oncol — PubMed CAS Google Scholar Leek R, Landers R, Fox S, Ng F, Harris A, Lewis C Association of tumour necrosis factor alpha and its receptors with thymidine phosphorylase expression in invasive breast carcinoma.

Br J Cancer PubMed PubMed Central CAS Google Scholar Sethi G, Sung B, Aggarwal BB TNF: a master switch for inflammation to cancer. Front Biosci — PubMed CAS Google Scholar Dempsey PW, Doyle SE, He JQ, Cheng G The signaling adaptors and pathways activated by TNF superfamily.

Cytokine Growth Factor Rev — PubMed CAS Google Scholar Aggarwal BB Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol PubMed CAS Google Scholar Aggarwal BB, Shishodia S, Ashikawa K, Bharti AC The role of TNF and its family members in inflammation and cancer: lessons from gene deletion.

Curr Drug Targets Inflamm Allergy — PubMed CAS Google Scholar Wajant H, Pfizenmaier K, Scheurich P Tumor necrosis factor signaling. Cell Death Differ PubMed CAS Google Scholar Wang X, Lin Y Tumor necrosis factor and cancer, buddies or foes?

Acta Pharmacol Sin — PubMed Google Scholar Devin A, Cook A, Lin Y, Rodriguez Y, Kelliher M, Liu Z-G The distinct roles of TRAF2 and RIP in IKK activation by TNF-R1: TRAF2 recruits IKK to TNF-R1 while RIP mediates IKK activation. Immunity — PubMed CAS Google Scholar Yang J, Lin Y, Guo Z, Cheng J, Huang J, Deng L, Liao W, Chen Z, Zheng-gang L, Su B The essential role of MEKK3 in TNF-induced NF-[kappa] B activation.

Nat Immunol PubMed CAS Google Scholar Karin M, Yamamoto Y, Wang QM The IKK NF-[kappa] B system: a treasure trove for drug development. Nat Rev Drug Discov PubMed CAS Google Scholar Kamata H, Honda S-I, Maeda S, Chang L, Hirata H, Karin M Reactive oxygen species promote TNFα-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases.

Cell — PubMed CAS Google Scholar Reinhard C, Shamoon B, Shyamala V, Williams LT Tumor necrosis factor α-induced activation of c-jun N-terminal kinase is mediated by TRAF2.

Proc Natl Acad Sci — PubMed CAS PubMed Central Google Scholar Liu J, Lin A Role of JNK activation in apoptosis: a double-edged sword. Cell Res PubMed Google Scholar Shih R-H, Wang C-Y, Yang C-M NF-kappaB signaling pathways in neurological inflammation: a mini review.

Front Mol Neurosci PubMed PubMed Central Google Scholar Alkhamesi NA, Ziprin P, Pfistermuller K, Peck DH, Darzi AW ICAM-1 mediated peritoneal carcinomatosis, a target for therapeutic intervention. Clin Exp Metastasis — PubMed CAS Google Scholar Choo MK, Sakurai H, Koizumi K, Saiki I TAK1-mediated stress signaling pathways are essential for TNF-α-promoted pulmonary metastasis of murine colon cancer cells.

Int J Cancer — PubMed CAS Google Scholar Kitakata H, Nemoto-Sasaki Y, Takahashi Y, Kondo T, Mai M, Mukaida N Essential roles of tumor necrosis factor receptor p55 in liver metastasis of intrasplenic administration of colon 26 cells.

Cancer Res — PubMed CAS Google Scholar Kulbe H, Hagemann T, Szlosarek PW, Balkwill FR, Wilson JL The inflammatory cytokine tumor necrosis factor-α regulates chemokine receptor expression on ovarian cancer cells. Cancer Res — PubMed CAS Google Scholar Mochizuki Y, Nakanishi H, Kodera Y, Ito S, Yamamura Y, Kato T, Hibi K, Akiyama S, Nakao A, Tatematsu M TNF-α promotes progression of peritoneal metastasis as demonstrated using a green fluorescence protein GFP -tagged human gastric cancer cell line.

Clin Exp Metastasis —47 PubMed CAS Google Scholar Van Grevenstein W, Hofland L, Van Rossen M, Van Koetsveld P, Jeekel J, Van Eijck C Inflammatory cytokines stimulate the adhesion of colon carcinoma cells to mesothelial monolayers. Dig Dis Sci — PubMed Google Scholar Ziprin P, Ridgway PF, Pfistermüller KL, Peck DH, Darzi AW ICAM-1 mediated tumor-mesothelial cell adhesion is modulated by IL-6 and TNF-α: a potential mechanism by which surgical trauma increases peritoneal metastases.

Cell Commun Adhes — PubMed CAS Google Scholar Balkwill F Tumour necrosis factor and cancer. Nat Rev Cancer PubMed CAS Google Scholar Kehrl JH, Wakefield LM, Roberts AB, Jakowlew S, Alvarez-Mon M, Derynck R, Sporn MB, Fauci AS Production of transforming growth factor beta by human T lymphocytes and its potential role in the regulation of T cell growth.

J Exp Med — PubMed CAS Google Scholar Roberts AB, Sporn MB, Assoian RK, Smith JM, Roche NS, Wakefield LM, Heine UI, Liotta LA, Falanga V, Kehrl JH Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro.

Buying options

The latter factor is able to interact with thioredoxin to reduce a specific cysteine Cys in the Rel-homology domain RHD of the NF-kB subunit p50 that had been previously oxidized by ROS 34 , restoring its ability to interact with specific responsive DNA sequences 35 , These data show that ROS can either activate or suppress the NF-kB signaling involved in the control of several important cellular processes, such as embryogenesis and cell proliferation and death, and the responses to a variety of stress stimuli ROS influence the activity of epigenetic modulators, such as histone deacetylases HDACs or DNA methyltransferases DNMTs with consequences that are evident in the expression of the target genes 38 , They also oxidize DNA, especially adenine and guanine 8-oxo-A and 8-oxo-G.

Oxidized Gs also impact the methylation of DNA, as indicated by reports showing that damaged bases on the DNA nascent strand can suppress the methylation of a cytosine within a distance of one or two base pairs However, ROS are able to induce DNA hypermethylation as well, with potential consequences on tumor phenotype when promoter regions of tumor suppressor genes are involved 43 , 44 , In addition, 8-oxo-Gs accumulate at telomeres, where they inhibit telomerase and decrease the binding of telomeric proteins, leading to the disruption of telomere length and precluding the maintenance of chromosomal-end capping Finally, ROS also induce mutations in mitochondrial DNA with the potential generation of a feedback loop in which mutations in genes encoding complexes of the ETC may directly affect the efficiency of electron transport.

The major sensitivity of mitochondrial DNA to ROS-induced mutagenesis is intuitive, as this DNA is not protected by histones, and mitochondria lack the nucleotide excision repair NER enzymatic system. The main consequences of redox signaling and oxidative stress in normal and cancer cells are presented in Fig.

The major signaling cascades induced by growth factor-stimulated ROS are highlighted on the left. The same pathways influence the cell cycle and affect the activity of transcription factors and genes that play roles in the cellular response to the hypoxic microenvironment.

The main consequences of oxidative stress in cancer cells are illustrated on the right. Moderately elevated ROS induce oncogenes and inhibit tumor suppressor genes that, in turn, increase ROS levels. ROS also activate HDACs and have a dual effect on DNMTs with important outcomes for the expression of oncogenes and tumor suppressor genes.

Oxidized bases trigger mutations and engage DNA repair enzymes. Cancer is the second cause of death worldwide and is characterized by several hallmarks 47 ; cell transformation, genome instability, hyperproliferation, immortalization, angiogenesis, epithelial-mesenchymal transition EMT and metastasis, which are all influenced in several ways by intracellular ROS 48 , Several noncancer cells associate with tumors: among these, cancer-associated fibroblasts CAFs , particularly represented in the tumor microenvironment TME , actively contribute to the regulation of tumor homeostasis, promoting tumor progression and the invasion of cancer cells.

CAFs and ROS engage in two-way cross-talk: on the one hand, fibroblasts are targeted by ROS, particularly H 2 O 2 , which is able to convert them into active CAFs through the upregulation of HIF1α; on the other hand, CAFs are critical for the increase in ROS levels observed in cancer 50 , CAFs can also promote cancer growth and invasiveness, and both CAFS and ROS are linked through the increases in ROS-generated CAFs to which most cancers respond by increasing the expression of antioxidant genes 52 , 53 , 54 Fig.

ROS, in response to death-inducing ligands TNFα and Fas , enhance the assembly of DISCs and the activation of effector caspases and reduce Bcl-2 activity or, as a consequence of increased permeability of mitochondrial PTPs, stimulate the intracytoplasmic release of cytochrome c, which interacts with Apaf-1 and procaspases and forms the apoptosome apoptosis.

ROS can also inhibit the negative regulators of autophagy TORC1 and increase the formation of LC3-dependent autophagosomes autophagy. Finally, high levels of ROS, induced by several receptor-interacting protein kinases RIPs , increase p53 expression, which increases ROS levels via a mechanism that depends on intracellular iron ferroptosis.

However, a growing body of evidence supports the view that antioxidant activities are essential for tumorigenesis. It has been recently reported that targets of the Nrf2 gene, such as HMOX1, facilitate cancer development because they counteract the effect of oxidative stress in transformed cells Moreover, established oncogenes such as K-RAS and c-MYC , which had been previously demonstrated to induce intracellular ROS 56 , 57 , have been recently shown to stabilize Nrf2 In this regard, mutations to NRF2 and its regulator KEAP1 have been found in cancer cells, supporting the supposition that antioxidant genes are pivotal in tumor progression 59 , 60 , 61 , In fact, it has been found that the breast cancer susceptibility 1 BRCA1 gene interacts with and induces Nrf2 expression with positive outcomes on cancer cell survival Interestingly, estrogen stimulation of breast cancer cells that do not express BRCA1 and, as a result, suffer from high intracellular ROS levels rescues NRF2 transcription, enhancing the survival of these cancer cells Additionally, FOXO transcription factors have recently been implicated in tumorigenesis: in fact, rhabdomyosarcomas present FOXO genes with a high percentage of mutations that render them insensitive to inhibition by AKT signaling In addition, inhibitors of the enzyme glutaminase GLS that converts glutamine to glutamate, which is subsequently transformed to GSH via the glutamate—cysteine ligase complex, efficiently induce cancer cell death through dysregulation of their antioxidant system As mentioned above, another central player in these redox systems is thioredoxin, which is reduced by NADPH to induce the transfer of electrons for use in DNA synthesis, signal transduction and redox regulation.

Interestingly, auranofin, which functions as a thioredoxin inhibitor, has been used with beneficial effects in the treatment of head and neck carcinoma cell lines; prevention of this effect by the ROS scavenger N-acetylcysteine NAC confirms the role of ROS in these cancers Triggered by an extrinsic or an intrinsic pathway, caspase-induced PCD culminates with the formation of apoptotic bodies that are eliminated by adjacent phagocytes The extrinsic pathway is mediated by binding of death-inducing ligands such as TNFα and Fas ligand that bind to cognate receptors that, in turn, recruit adaptor proteins and pro-caspases, leading to the assembly of the death-inducing signaling complex DISC and the activation of effector caspases This interaction is competed by the cellular FLICE-inhibitory protein c-FLIP : ROS have been shown to downregulate the c-FLIP half-life by inducing its ubiquitin-proteasomal degradation, thus enhancing this extrinsic pathway However, compelling evidence suggests that, for the majority of ROS-related anticancer drugs, apoptosis depends on the activation of the intrinsic pathway that involves mitochondrial PTPs, the permeability of which is increased with the cytoplasmic release of pro-apoptotic factors such as cytochrome c that forms a complex with apoptotic protease activating factor 1 Apaf-1 and pro-caspase 9 to build the apoptosome, activating, in turn, effector caspases 77 , 78 , 79 , 80 Fig.

In fact, ROS induce the three major components critical for the opening of the PTPs, the voltage-dependent anion-selective channel VDAC , adenine nucleotide translocase ANT and cyclophilin D, via the oxidation of specific cysteines in their active sites 81 , ROS also trigger apoptosis by inactivating or increasing the ubiquitination of the pivotal anti-apoptotic protein Bcl-2 and by decreasing the intracellular levels of Bax and Bad 83 , 84 Fig.

The induction of apoptosis by elevated ROS levels has been highlighted as the central mechanism responsible for the positive effects of monoclonal antibodies 85 and tyrosine kinase inhibitors 86 , which represent the core of targeted cancer therapy Among tyrosine kinase inhibitors, imatinib a PDGFR inhibitor and erlotinib an EGFR inhibitor induce ROS-dependent apoptosis in melanoma and non-small-cell lung cancer cells, respectively, through disruption of mitochondrial membrane potential upon the stimulation of JNK and p38 phosphorylation 88 , 89 , while vemurafenib a BRAF inhibitor increases the production of superoxide anions with the commensurate depolarization of the mitochondrial membranes in melanoma cells Among the monoclonal antibodies, rituximab specific to the calcium-channel protein CD20 on the surface of B cells and mature plasma cells increases ROS and induces apoptosis via the inhibition of Bcl-2 and p38MAPK signaling and is used in the treatment of B cell lymphomas As noted above, chemotherapy and radiotherapy cause an increase in intracellular ROS that can lead to apoptosis 92 , 93 via extrinsic or intrinsic pathways 94 , Many drugs used in anticancer therapy induce oxidative stress.

A course of treatment with arabinocytosine, which hampers DNA replication, followed by anthracyclines to increase ROS, has been shown to drive PCD, with beneficial effects for patients with acute myeloid leukemia AML Arsenic trioxide has recently carved out a role in cancer therapy because it can induce electron leakage along the respiratory chain It triggers apoptosis in different cancer cells, including those of myeloma, lung cancer, and leukemia , Moreover, 5-fluorouracil, a pyrimidine analog, produces ROS through pdependent pathways and induces apoptosis in colon and rectal cancer cells , ROS-induced apoptosis also explains the beneficial effect of two analogs of nuclear receptor ligands in several types of cancer: 2-methoxyestradiol, a 17β-estradiol metabolite, and N- 4-hydroxyphenyl retinamide, a synthetic analog of retinoic acid, have been shown to induce PCD in neuroblastoma and lung cancer cells, respectively , Furthermore, platinum-based drugs elevate ROS levels that promote PCD; protocols for the administration of these compounds in combination with inhibitors of poly ADP-ribose polymerase PARP , which is involved in the maintenance of DNA integrity, have been shown to arrest the growth of breast cancer cells, even in BRCA -deficient models , Intuitively, the inhibition of DNA damage repair by PARP may sensitize cancer cells to the oxidative stress induced by platinum-containing drugs.

Programmed cell death may also be mediated by the effect of elevated ROS on sphingomyelinase, which generates ceramide from sphingomyelin and binds to death receptors on the cell membrane of cancer cells. Activation of this pathway has been observed after UV irradiation of lymphoma cells Moreover, the use of drugs affecting mitochondria, where more than one-half of all ROS are generated, represents a suitable approach to induce oxidative stress and PCD in cancer cells : gamitrinib, an inhibitor of heat shock protein 90 HSP90 , induces a dramatic collapse of mitochondria in prostate cancer cells , while ARQ a quinone derivative and STA a copper chelator increase ROS through leakage in the electron transport chain and have beneficial effects in patients with solid tumors and pancreatic adenocarcinoma Apoptosis is triggered in cells with excessive endoplasmic reticulum ER stress that is induced when the protein folding ability of the ER is overwhelmed or impaired.

Recently, several drugs have been designed on the basis of their ability to aggravate ER stress in cancer cells via the induction of oxidative stress. Recently, an important therapeutic approach to kill cancer cells has been presented by ROS-induced autophagy Specifically, it has been reported that H 2 O 2 -dependent inactivation of autophagy-related gene-4 ATG4 increases LC3-associated autophagosomes and that ATM-mediated oxidation of AMP-activated protein kinase AMPK inhibits mammalian target of rapamycin 1 TORC1 , a pivotal negative regulator of autophagy , , Fig.

Indeed, autophagy, also known as type II programmed cell death, is now considered not only as a cell survival mechanism but also a tumor suppressor mechanism that induces the death of transformed cells In this regard, it has been reported that H 2 O 2 induces autophagic cell death in glioma cells after treatment with the polycyclic ammonium ion sanguinarine, which increases electron leakage from mitochondria and induces NOXs Rapamycin, administered in combination with inhibitors of HSP90, causes mitochondrial damage with accompanying oxidative stress and autophagy and reduces tumor growth in RAS -dependent tumors ROS are also able to induce necrosis, which was originally considered an unregulated form of cell death but is now recognized as type III programmed cell death necroptosis , Ferroptosis depends on the presence of intracellular iron and is induced by ROS Fig.

Therefore, the role of p53 in this context appears to be different from that reported in several studies showing that it decreases the levels of ROS.

A plausible explanation of this apparent dichotomy is that p53 promotes cell survival by preventing excessive increases in ROS under moderate oxidative stress, whereas when the oxygen species increase over a threshold level, it switches to becoming a ROS inducer, triggering cell death.

On the basis that ferroptosis is considered an oxidation-induced cell death mechanism, several trials with different drugs that elicit this pathway have been conducted , Erastin is a synthetic drug that induces cell death through ferroptosis in tumor cells bearing mutant RAS by increasing intracellular ROS levels and altering the permeability of the outer mitochondrial membrane , Increased ROS levels are thought to impair the multidrug resistance of cancer cells, which causes cancer development and metastasis during or after chemotherapy , It has been recently shown that efflux pumps in the plasma membrane of cancer cells are crucial for the extracellular efflux of anticancer drugs These pumps belong to the adenosine triphosphate ATP -binding cassette ABC transporter superfamily and are dependent on intracellular ATP stores ATP is accumulated by a synthase driven by a proton gradient generated in mitochondria by the NADH-dependent electron transport chain , ; therefore, one possible way to overcome efficient efflux in cancer drugs is to inhibit ATP synthesis by promoting NADH conversion to NAD through lipid membrane-coated silica carbon nanoparticles that, under near-infrared laser irradiation, target mitochondria and produce ROS with simultaneous consumption of NADH A new role of ROS related to transcriptional output has been recently highlighted.

It is well known that cells follow a strictly scheduled program for differentiation that is based on an orchestrated sequence of gene expression.

Because of spatial constraints, genes must engage in a complex unfolding process to become accessible to the transcriptional machinery, which is triggered through posttranslational modifications at the N-terminal tails of core histones.

Together, these modifications, induced by coordinated targeting of transcription factors that is currently referred to as epigenetic marks, conform to a precise code with specific time requirements to control whole gene expression We have previously shown that estrogen-induced transcription is triggered by LSD1-catalyzed demethylation of lysine 9 in histone H3 H3K9 , which is activated by the binding of liganded estrogen receptor to the enhancers of target genes Intuitively, generation of ROS in this process must be timely and spatially controlled to prevent excessive damage to the DNA: a recent report, in fact, describes a new role for the originally discovered superoxide dismutase, SOD1, that is recruited to the nucleus in response to specific stimuli However, it has also been observed that hormone-induced phosphorylation of serine 10 in H3 histone H3S10 prevents the rapid remethylation of the preceding lysine, serving as the metronome of the process and giving the DNA damage repair system enough time to eliminate the oxidized nucleotides from nearby DNA It has been reported that by inhibiting phosphorylation of serine 10 in this pathway, breast cancer cells simultaneously challenged with estradiol show an overproduction of ROS, with increased oxidation of the DNA that overwhelms the repair apparatus and triggers PCD in a great percentage of these cells Fig.

ROS generated during nuclear receptor-induced transcription of target genes by the activity of lysine demethylases on lysine 9 in histone H3 must be controlled to prevent their accumulation.

To this end, SOD1 reaches the nuclear space, while phosphorylation of H3S10 inhibits the rapid remethylation of the same lysine.

If inhibitors of the H3S10 kinases are introduced as a Trojan horse together with nuclear receptor ligands, remethylation of H3K9 is quick, nuclear ROS accumulate, and unrepaired DNA damage triggers PCD.

The complex interconnection between ROS levels and cancer is essentially based on accurate fine-tuning between ROS production and scavenging. Cancer initiation and progression leverage slight increases in ROS levels. Therefore, cancer cells thrive on levels of ROS that are moderately higher than those in their normal counterparts, as they have developed increased antioxidant systems.

This feature renders cancer cells more sensitive to external stimuli that further increase the production of ROS , , , and, as schematically summarized in Table 1 , an increasing number of therapeutic strategies are being developed to elevate ROS levels to overwhelm the redox adaptation of the same cells, inducing oxidative stress incompatible with cellular life , , , Fig.

The first approach is based on lowering ROS levels to counteract their role in cellular transformation; it is aimed at reducing the number of transformed cells by depriving them of fuel represented in the upper right side of the figure as a lower proportion of transformed cells with respect to that of normal cells.

The second approach is based on the consideration that cancer cells, with an antioxidant system already triggered, are more sensitive than their normal counterparts to further increases in ROS and are unable to achieve redox balance. Therefore, by inducing ROS under these metabolic conditions, a high percentage of the cells undergo death represented in the lower right side of the figure, where transformed cells are depicted as apoptotic.

Specifically, cellular responses to ROS must be imagined as the integration of multiple levels in which, in addition to their nature and relative concentration, their location plays an important role.

In fact, mitochondrial ROS have been reported to essentially promote cell death, while NOX-generated ROS have been associated with the promotion of cell proliferation and migration Furthermore, in contrast to the mechanism of sister pathways, redox signaling is based on migrating electrons, and therefore, the signaling in this pathway is much more diffuse.

In reference to the nature of ROS behavior as a double-edged sword, even though several studies have documented the benefits of antioxidant drugs for cancer therapies, none has been supported by solid trials performed on a large scale , In contrast, the most recent studies have shown an increase in tumor development and metastasis in mouse models treated with vitamin E an opposite result of that in which high doses of vitamin C increase ROS levels to induce the death of colon cancer cells bearing KRAS and BRAF mutations In addition, it has been shown that the administration of antioxidants, such as N-acetylcysteine, accelerates the progression of lung cancers and melanomas and that increasing the expression of the antioxidant-encoding Nrf2 gene enhances the growth of lung tumors , , , In fact, and in contrast to the previous view, the results of many studies support a scenario in which the inhibition of antioxidant enzymes ensures the death of cancer cells, especially when this approach is used in combination with treatments that increase ROS.

This approach is an alternative to the traditional strategy of targeting oncogenes and tumor suppressor genes, a strategy that appears complicated because of the high number of genes involved and their ability to drive compensatory pathways Interestingly, increased ROS-induced apoptosis has been reported in cancer cells after depletion of ATP derived from the manipulation of glycolytic enzymes, chemotherapy or radiation therapy; these data highlight the potential eminent role of ROS modulation in anticancer combinatorial therapies , Finally, the most recent ROS-inducing drugs have addressed the pivotal goal of therapists: cancer selectivity.

In this regard, good results have been reached through photodynamic therapy, which is based on the generation of ROS after stimulation of a photosensitizer by light: cancer cells under treatment internalize porphyrin precursor molecules to induce the formation of ROS that lead to photooxidative stress and cancer-specific cell death , In fact, although more studies are required to increase the selectivity of these anticancer ROS-related drugs, the common mechanisms elicited by oncogenes to promote the adaptation to a large set of stress conditions are being revealed in more depth every day, and in a high percentage, they concern the redox balance.

In conclusion, we expect that targeting ROS will represent fruitful ground for future molecular anticancer strategies. Forman, H. An overview of mechanisms of redox signaling. Article PubMed CAS Google Scholar.

Bedard, K. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Vander Heiden, M. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science , — Article CAS Google Scholar. Finkel, T. Signal transduction by reactive oxygen species.

Cell Biol. Article PubMed PubMed Central CAS Google Scholar. Paradies, G. et al. Circulation Res. Petrosillo, G. Decreased complex III activity in mitochondria isolated from rat heart subjected to ischemia and reperfusion: role of reactive oxygen species and cardiolipin.

FASEB J. Valko, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Ray, P. Reactive oxygen species ROS homeostasis and redox regulation in cellular signaling. Houee-Levin, C. Exploring oxidative modifications of tyrosine: an update on mechanisms of formation, advances in analysis and biological consequences.

Free Radic. Thannickal, V. Activation of an H 2 O 2 -generating NADH oxidase in human lung fibroblasts by transforming growth factor beta 1.

Meng, T. Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo. Cell 9 , — Kamata, H. Reactive oxygen species promote TNF alpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases.

Cell , — Esposito, F. Protein kinase B activation by reactive oxygen species is independent on tyrosine kinase receptor phosphorylation and required Src activity. Sundaresan, M. Requirement for generation of H 2 O 2 for platelet-derived growth factor signal transduction. Corcoran, A.

Redox regulation of protein kinases. FEBS J. Kwon, J. Reversible oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors.

Natl Acad. USA , — Article PubMed CAS PubMed Central Google Scholar. Latimer, H. Peroxiredoxins in regulation of MAPK signalling pathways; sensors and barriers to signal transduction.

Gopalakrishna, R. Protein kinase C signaling and oxidative stress. Free Rad. Taguchi, K. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution.

Genes Cells 16 , — Meister, A. Selective modification of glutathione metabolism. Thimmulappa, R. Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray.

Cancer Res. PubMed CAS Google Scholar. Orino, K. Ferritin and the response to oxidative stress. Weinberg, E. The role of iron in cancer. Cancer Prev. Essers, M. FOXO transcription factor activation by oxidative stress mediated by the small GTPase Ral and JNK.

EMBO J. Putker, M. Cell 49 , — Melnik, B. p key conductor of all anti-acne therapies. Article PubMed PubMed Central Google Scholar. Rhee, S. The antioxidant function of sestrins is mediated by promotion of autophagic degradation of keap1 and Nrf2 activation of mTORC1. Pinnix, Z. Ferroportin and iron regulation in breast cancer progression and prognosis.

Huang, L. Regulation of hypoxia-inducible factor 1α is mediated by an O 2 -dependent degradation domain via the ubiquitin-proteasome pathway. USA 95 , — Abid, M. NADPH oxidase activity selectively modulates vascular endothelial growth factor signaling pathways. Guo, Z. ATM activation by oxidative stress.

Svegliati, S. Oxidative DNA damge induces the ATM-mediated transcriptional suppression of the Wnt inhibitor WIF-1 in systemic sclerosis and fibrosis.

Tell, G. Nucleic Acids Res. Ando, K. Toledano, M. Modulation of transcription factor NF-kappa B binding activity by oxidation-reduction in vitro.

USA 88 , — Matthews, J. Role of cysteine62 in DNA recognition by the p50 subunit of NF-kappa B. Lingappan, K. NF-kB in oxidative stress. Article PubMed Google Scholar. Matsushima, S. Increased oxidative stress in the nucleus caused by Nox4 mediates oxidation of HDAC4 and cardiac hypertrophy.

Ushijima, T. Detection and interpretation of altered methylation patterns in cancer cells. Cancer 5 , — Grollman, A. Mutagenesis by 8-oxoguanine: an enemy within. Trends Genet.

Egger, G. Epigenetics in human disease and prospects for epigenetic therapy. Nature , — Turk, P. Carcinogenesis 16 , — Pezone, A. High-coverage methylation data of a gene model before and after DNA damage and homologous repair.

Russo, G. DNA damage and repair modify DNA methylation and chromatin domain of the targeted locus: mechanism of allele methylation polymorphism. Morano, A. Targeted DNA methylation by homology-directed repair in mammalian cells.

Transcription reshapes methylation on the repair gene. Opresko, P. Oxidative damage in telomeric DNA disrupts recognition by TRF1 and TRF2.

Hanahan, D. Hallmarks of cancer: the next generation. Radisky, D. Rac1b and reactive oxygen species mediate MMPinduced EMT and genomic instability. Nishikawa, M. Reactive oxygen species in tumor metastasis. Cancer Lett. Chan, J. Cancer-associated fibroblasts enact field cancerization by promoting extratumoral oxidative stress.

Cell Death Dis. Toullec, A. Oxidative stress promotes myofibroblast differentiation and tumour spreading. EMBO Mol. Joyce, J. Microenvironmental regulation of metastasis. Cancer 9 , — Accessories to the crime: functions of cells recruited to the tumor microenvironment.

Cancer Cell 21 , — Holmstrom, K. Cellular mechanisms and physiological consequences of redox-dependent signaling. Diehn, M.

Association of reactive oxygen species levels and radioresistance in cancer stem cells. Irani, K. Mitogenic signaling mediated by oxidants in Ras-transformed fibroblasts. Vafa, O. C-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability.

De Nicola, G. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Shibata, T. Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Singh, A. Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer.

PLoS Med. Menegon, S. The dual roles of NRF2 in cancer. Trends Mol. Lignitto, L. Nrf2 activation promotes lung cancer metastasis by inhibiting the degradation of Bach1. Bae, I. BRCA1 induces antioxidant gene expression and resistance to oxidative stress.

Gorrini, C. BRCA1 interacts with Nrf2 to regulate antioxidant signaling and cell survival. Olanich, M. A call to ARMS: targeting the PAX3-FOXO1 gene in alveolar rhabdomyosarcoma.

Expert Opin. Targets 17 , — Harris, I. Glutathione and thioredoxin antioxidant pathways synergize to drive cancer initiation and progression. Cancer Cell 27 , — Sayin, V. Antioxidants accelerate lung cancer progression in mice.

Lo, M. Cell Physiol. Potential use of the anti-inflammatory drug, sulfasalazine, for targets therapy of pancreatic cancer.

PubMed PubMed Central CAS Google Scholar. Guan, J. Cancer Chemoter. Townsend, D. NOV, a glutathione disulfide mimetic, as a modulator of cellular redox balance. Wang, J. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell 18 , — Sobhakumari, A.

Susceptibility of human head and neck cancer cells to combined inhibition of glutathione and thioredoxin metabolism. PLoS ONE 7 , e Chandra, J. Triggering and modulation of apoptosis by oxidative stress. Wang, L. The Fas death signaling pathway connecting reactive oxygen species generation and FLICE inhibitory protein down-regulation.

Wilkie-Grantham, R. Novel phosphorylation and ubiquitination sites regulate reactive oxygen species-dependent degradation of anti-apoptotic c-FLIP protein. Stennicke, H. Caspase 9 can be activated without proteolytic processing. Kaufmann, S.

Induction of apoptosis by cancer chemotherapy. Cell Res. Kagan, V. Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors.

Zuo, Y. Oxidative modification of caspase-9 facilitates its activation via disulfide-mediated interaction with Apaf Madesh, M. VDAC-dependent permeabilization of the outer mitochondrial membrane by superoxide induces rapid and massive cytochrome c release.

McStay, G. Role of critical thiol groups on the matrix surface of the adenine nucleotide translocase in the mechanism of the mitochondrial permeability transition pore. Luanpitpong, S. Regulation of apoptosis by Bcl-2 cysteine oxidation in human lung epithelial cells. Cell 24 , — Li, D.

Reactive oxygen species ROS control the expression of Bcl-2 family proteins by regulating their phosphorylation and ubiquitination. Cancer Sci. Dimitrov, D. Therapeutic antibodies current state and future trends-is a paradigm change coming soon? Methods Mol. Hartmann, J. Tyrosine kinase inhibitors- a review on pharmacology, metabolism and side effects.

Drug Metab. Teppo, H. Reactive oxygen species-mediated mechanisms of action of targeted cancer therapy. Cell Longev. Chang, S. Imatinib mesylate induction of ROS-dependent apoptosis in melanoma B16F0 cells.

Shan, F. Erlotinib induces the human non-small-cell lung cancer cells apoptosis via activating ROS-dependent JNK pathways. Cancer Med.

Bauer, D. Critical role of reactive oxygen species ROS for synergistic enhancement of apoptosis by vemurafenib and the potassium channel inhibitor TRAM in melanoma cells. Alas, S. Shen, B. Norcantharidin induced DU cell apoptosis through ROS-mediated mitochondrial dysfunction and energy depletion.

PLoS One 8 , e Brenneisen, P. Nanotherapy and reactive oxygen species ROS in cancer: a novel perspective. Antioxidants 7 , E31 Qu, W. Life Sci. Oliveira, M.

Redox Biol. Renschler, M. The emerging role of reactive oxygen species in cancer therapy. Cancer 40 , — Kotamraju, S. Transferrin receptor-dependent iron uptake is responsible for doxorubicin-mediated apoptosis in endothelial cells: role of oxidant-induced iron signaling in apoptosis.

Prieto-Bermejo, R. Reactive oxygen species in hematopoiesis: leukemic cells take a walk on the wild side. Pelicano, H.

Inhibition of mitochondrial respiration: a novel strategy to enhance drug-induced apoptosis in human leukemia cells by a reactive oxygen species-mediated mechanism. Miller, W. Mechanisms of action of arsenic trioxide. Yi, J. The inherent cellular level of reactive oxygen species: one of the mechanisms determining the apoptotic susceptibility of leukemic cells to arsenic trioxide.

Apoptosis 7 , — Hwang, P. Ferrodoxin reductase affects pdependent, 5-fluorouracil-induced apoptosis in colorectal cancer cells. Longley, D. Cancer 3 , — Zhang, Q. Involvement of reactive oxygen species in 2-methoxyestradiol-induced apoptosis in human neuroblastoma cells.

Lai, W. ROS mediates 4-HPR-induced posttranscriptional expression of the Gadd gene. Berndtsson, M. Acute apoptosis by cisplatin requires induction of reactive oxygen species but is not associated with damage to nuclear DNA. Cancer , — Rottenberg, S. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD alone and in combination with platinum drugs.

Charruyer, A. UV-C light induces raft-associated acid sphingomyelinase and JNK activation and translocation independently on a nuclear signal. Caino, M. Molecular pathways: mitochondrial reprogramming in tumor progression and therapy. Kang, B. Preclinical characterization of mitochondria-targeted small molecule hsp90 inhibitors, gamitrinibs, in advanced prostate cancer.

Sborov, D. Investigational cancer drugs targeting cell metabolism in clinical development. Drugs 24 , 79—94 Fribley, A. Proteasome inhibitor PS induces apoptosis through induction of endoplasmic reticulum stress-reactive oxygen species in head and neck squamous cell carcinoma cells. Zhu, J. Affiliation: School of Public Health, Indiana University, Bloomington,Indiana.

Abstract: Background: Cancer is considered a major cause of death worldwide. Klaunig E. Purchase PDF. Mark Item. Current Pharmaceutical Design. Title: Oxidative Stress and Cancer Volume: 24 Issue: 40 Author s : James E.

Close Print this page. Export Options ×. Export File: RIS for EndNote, Reference Manager, ProCite. Content: Citation Only. Citation and Abstract. About this article ×. Cite this article as: Klaunig E. Close About this journal.

Related Journals Anti-Cancer Agents in Medicinal Chemistry. Current Bioactive Compounds. Current Cancer Drug Targets. Current Cancer Therapy Reviews. Current Diabetes Reviews. Current Drug Safety. Current Drug Targets. Current Drug Therapy.

View More. Related Books Advanced Pharmacy. Plant-derived Hepatoprotective Drugs. The Role of Chromenes in Drug Discovery and Development. New Avenues in Drug Discovery and Bioactive Natural Products.

Practice and Re-Emergence of Herbal Medicine. Methods for Preclinical Evaluation of Bioactive Natural Products. Nanopharmacology and Nanotoxicology: Clinical Implications and Methods. Natural Immunomodulators: Promising Therapy for Disease Management. Bioactive Phytochemicals from Himalayas: A Phytotherapeutic Approach.

Video

Inflammation, Oxidative Stress \u0026 Free Radical Damage to Health

Author: Bralmaran

1 thoughts on “Oxidative stress and cancer

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com