Category: Diet

Free radicals and cancer

Free radicals and cancer

Hearty Mediterranean Probiotics and Mental Clarity. IntechOpen Free Radicals and Diseases Radicalz Free radicals and cancer Rizwan Ahmad. The radiccals certified registered dietitians at cancet Stanford Radicqls Center are here to Free radicals and cancer you make informed choices about nutrition, answer your nutrition-related questions, and help you to achieve and maintain good health. Cajun Salmon over Polenta. Nutrition Burbank, Los Angeles County, Calif29 1 Miguel, eds. Google Scholar Jomova K, Valko M: Importance of iron chelation in free radical-induced oxidative stress and human disease.

The antioxidants found xancer fruits and vegetables radicaos Free radicals and cancer free radicals, which Free radicals and cancer lead to breast cancer. Free radicals are Free radicals and cancer mentioned as a cause of breast Balanced snacks for cravingsbut what exactly are free radicals?

Free radicals are highly reactive and unstable molecules, dancer oxygen molecules, but not raddicals. Their unstable cancef is caused by radicls an unpaired electron. As a andd of this unpaired electron, free radicals seek out and take electrons radiczls other molecules, which oftentimes causes adn to the second molecule.

Like many things that Free radicals and cancer in nature, Free radicals and cancer radicals are not Fred impossible to avoid, Free radicals and cancer necessary for life. Free radicals help us fight infection, Reduce cholesterol intake the inflammation process that helps repair tissue injury, and short-term oxidative stress may inhibit aging.

At the same time, excessive amounts are harmful to humans. Free radicals are thought to be one cause of cancer, as well as the cause of some auto-immune diseases, heart diseases, neurological diseases, and the aging process in general. can cause cell mutations which increases breast cancer risk.

of cancer cells. Antioxidants are molecules that can safely interact with free radicals. Unlike other molecules, antioxidants can donate an electron to a free radical and still remain stable, therefore stopping the cycle A persistent and recurring way.

of cell damage. Examples of antioxidants are:. One of the best ways to reduce free radical damage in the body, and therefore decrease your risk of breast cancer, is to increase your intake of these vitamins.

One of the best sources? Fruits and vegetables! Consider having one vegetarian day per week or even going completely vegetarian. Make your goal servings of fruits and vegetables per day. If you are unsure what a serving is, check out our article Confused About the Serving Sizes of Vegetables?

Have you increased antioxidants in your diet? Maurer Foundation. July 11, Read More About the Author April Zubko Categories: Breast Cancer Prevention 0 comments.

This field is for validation purposes and should be left unchanged. Search for:.

: Free radicals and cancer

The Role of Free Radicals in Cancer and Aging

The researchers found that different cancer-associated genes affect the heart in different ways — a sign that genetic information could one day guide heart-protective treatment decisions in cancer patients. The study is published in the journal Antioxidants. However, emerging research has suggested that heart problems can surface before cancer treatment or muscle wasting occur.

The Ohio State team noted that a study published recently in the Journal of the America Heart Association reported the detection of abnormalities in heart tissue and cardiac function in human cancer patients before cancer treatment had begun.

The scientists observed significantly lower ejection fraction and fractional shortening — similar to what was seen in mice with tumors — as well as an increase in heart rate in flies with tumors.

The researchers found a total-body increase in the rate of production as well as a higher total number of free radicals — also known as reactive oxygen species — in fruit flies with tumors compared to controls.

The rate of reactive oxygen species production was also significantly higher in mice with tumors compared to controls. She and Singh also emphasized that reactive oxygen species are just one identified mechanism of tumor-related heart damage, and that there is still a lot to learn about how antioxidants might fit into a treatment regimen.

Though this research zeroed in on one cancer-causing gene to study the mechanism of heart damage in fruit flies, the researchers initially tested the effects of several cancer-causing genes in the flies.

The heart function affected and the intensity of the effects on the heart varied, depending on the gene. Singh is collaborating with clinicians at Ohio State and other institutions to collect blood samples from cancer patients who have heart failure.

Second, we want to see what the message is and whether we can prescribe antioxidants. Additional co-authors include Priyanka Karekar, Haley Jensen, Kathryn Russart, Devasena Ponnalagu, Shridhar Sanghvi, Sakima Smith and Leah Pyter from Ohio State and Sarah Seeley from Ohio Northern University ONU.

The work was supported by Commonwealth Universal Research Enhancement Program grants, the W. Int J Radiat Biol , 85 12 Jian Liu K, Rosenberg GA: Matrix metalloproteinases and free radicals in cerebral ischemia.

Free Radical Biol Med , 39 1 Jomova K, Valko M: Importance of iron chelation in free radical-induced oxidative stress and human disease. Curr Pharm Des , 17 31 Jones DP, Carlson JL, Mody VC Jr, Cai J, Lynn MJ, Sternberg P Jr: Redox state of glutathione in human plasma.

Free Radical Biol Med , 28 4 J-w Z, Rubio V, Zheng S, Z-z S: Knockdown of OLA1, a regulator of oxidative stress response, inhibits motility and invasion of breast cancer cells.

Journal of Zhejiang University SCIENCE B , 10 11 Kempner ES: Direct effects of ionizing radiation on macromolecules. J Polym Sci B , 49 12 Kheradmand F, Werner E, Tremble P, Symons M, Werb Z: Role of Rac1 and oxygen radicals in collagenase-1 expression induced by cell shape change.

Science , Kim J, Gherasim C, Banerjee R: Decyanation of vitamin B12 by a trafficking chaperone. Proc Natl Acad Sci , 38 Kim H-S, Patel K, Muldoon-Jacobs K, Bisht KS, Aykin-Burns N, Pennington JD, van der Meer R, Nguyen P, Savage J, Owens KM: SIRT3 is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress.

Cancer cell , 17 1 Klaunig JE, Kamendulis LM: The role of oxidative stress in carcinogenesis. Annu Rev Pharmacol Toxicol , Klaunig JE, Kamendulis LM, Hocevar BA: Oxidative stress and oxidative damage in carcinogenesis. Toxicologic pathology , 38 1 Kumar B, Koul S, Khandrika L, Meacham RB, Koul HK: Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype.

Cancer research , 68 6 Kumar A, Pant M, Singh H, Khandelwal S: Assessment of the redox profile and oxidative DNA damage 8-OHdG in squamous cell carcinoma of head and neck. J Cancer Res Ther , 8 2 Lee JJ, Lee JH, Ko YG, Hong SI, Lee JS: Prevention of premature senescence requires JNK regulation of Bcl-2 and reactive oxygen species.

Oncogene , 29 4 Leonardo CC, Pennypacker KR: Neuroinflammation and MMPs: potential therapeutic targets in neonatal hypoxic-ischemic injury. J Neuroinflammation , 6: Differentiation , 70 2—3 Li H, Sekine M, Seng S, Avraham S, Avraham HK: BRCA1 Interacts with Smad3 and Regulates Smad3-Mediated TGF-b Signaling during Oxidative Stress Responses.

PloS one , 4 9 :e Li L, Ishdorj G, Gibson SB: Reactive oxygen species regulation of autophagy in cancer: Implications for cancer treatment. Free Radical Biol Med , 53 7 Luo J, Solimini NL, Elledge SJ: Principles of cancer therapy: oncogene and non-oncogene addiction.

Cell , 5 Mahelkova G, Korynta J, Moravova A, Novotna J, Vytasek R, Wilhelm J: Changes of extracellular matrix of rat cornea after exposure to hypoxia. Physiol Res , 57 1 Marnett LJ: Oxyradicals and DNA damage. Carcinogenesis , 21 3 Martínez Sarrasague M, Barrado DA, Zubillaga M, Hager A, De Paoli T, Boccio J: Conceptos actuales del metabolismo del glutatión Utilización de los isótopos estables para la evaluación de su homeostasis.

Acta bioquímica clínica latinoamericana , 40 1 Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen H-Y, Bray K, Reddy A, Bhanot G, Gelinas C: Autophagy suppresses tumorigenesis through elimination of p Meley D, Bauvy C, Houben-Weerts JH, Dubbelhuis PF, Helmond MT, Codogno P, Meijer AJ: AMP-activated protein kinase and the regulation of autophagic proteolysis.

J Biol Chem , 46 Mena S, Ortega A, Estrela JM: Oxidative stress in environmental-induced carcinogenesis. Mori K, Shibanuma M, Nose K: Invasive potential induced under long-term oxidative stress in mammary epithelial cells. Cancer research , 64 20 Muriel P: Role of free radicals in liver diseases.

Hepatology international , 3 4 Murley JS, Kataoka Y, Miller RC, Li JJ, Woloschak G, Grdina DJ: SOD2-mediated effects induced by WR and low-dose ionizing radiation on micronucleus formation in RKO human colon carcinoma cells.

Radiation research , 1 Nathan C: Nitric oxide as a secretory product of mammalian cells. FASEB J , 6 12 Nelson KK, Ranganathan AC, Mansouri J, Rodriguez AM, Providence KM, Rutter JL, Pumiglia K, Bennett JA, Melendez JA: Elevated Sod2 Activity Augments Matrix Metalloproteinase Expression Evidence for the Involvement of Endogenous Hydrogen Peroxide in Regulating Metastasis.

Clin Cancer Res , 9 1 Okezie I, Harparkash K, Haliwell B: Oxygen free radicals and human diseases. J Roy Soc Health , Oronsky BT, Knox SJ, Scicinski JJ: Is nitric oxide NO the last word in radiosensitization? A review. Translational Oncology , 5 2 Pande D, Negi R, Khanna S, Khanna R, Khanna HD: Vascular endothelial growth factor levels in relation to oxidative damage and antioxidant status in patients with breast cancer.

Journal of breast cancer , 14 3 Pande D, Negi R, Karki K, Khanna S, Khanna RS, Khanna HD: Oxidative damage markers as possible discriminatory biomarkers in breast carcinoma.

Transl Res , 6 Pani G, Galeotti T, Chiarugi P: Metastasis: cancer cell's escape from oxidative stress. Cancer Metastasis Rev , 29 2 Panis C, Herrera AC, Victorino VJ, Campos FC, Freitas LF, De Rossi T, Colado Simão AN, Cecchini AL, Cecchini R: Oxidative stress and hematological profiles of advanced breast cancer patients subjected to paclitaxel or doxorubicin chemotherapy.

Breast Cancer Res Treat , 1 Paquette B, Baptiste C, Therriault H, Arguin G, Plouffe B, Lemay R: In vitro irradiation of basement membrane enhances the invasiveness of breast cancer cells.

Brit J Cancer , 97 11 Pervin S, Tran L, Urman R, Braga M, Parveen M, Li S, Chaudhuri G, Singh R: Oxidative stress specifically downregulates survivin to promote breast tumour formation. Brit J Cancer , 4 Polidori MC, Stahl W, Eichler O, Niestroj I, Sies H: Profiles of antioxidants in human plasma.

Free Radical Biol Med , 30 5 Polyak K, Weinberg RA: Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer , 9 4 Pourahmad J, O'Brien PJ, Jokar F, Daraei B: Carcinogenic metal induced sites of reactive oxygen species formation in hepatocytes.

Toxicol In Vitro , 17 5—6 Prousek J: Fenton chemistry in biology and medicine. Pure Appl Chem , 79 12 Purdom S, Chen QM: Epidermal growth factor receptor-dependent and-independent pathways in hydrogen peroxide-induced mitogen-activated protein kinase activation in cardiomyocytes and heart fibroblasts.

J Pharmacol Exp Ther , 3 Quievryn G, Messer J, Zhitkovich A: Carcinogenic chromium VI induces cross-linking of vitamin C to DNA in vitro and in human lung A cells. Biochemistry , 41 9 Rahman MA, Senga T, Ito S, Hyodo T, Hasegawa H, Hamaguchi M: S-nitrosylation at cysteine of c-Src tyrosine kinase regulates nitric oxide-mediated cell invasion.

J Biol Chem , 6 Raj L, Ide T, Gurkar AU, Foley M, Schenone M, Li X, Tolliday NJ, Golub TR, Carr SA, Shamji AF: Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S: Regulation of mammalian autophagy in physiology and pathophysiology.

Physiol Rev , 90 4 Reynolds M, Stoddard L, Bespalov I, Zhitkovich A: Ascorbate acts as a highly potent inducer of chromate mutagenesis and clastogenesis: linkage to DNA breaks in G2 phase by mismatch repair.

Nucleic Acids Res , 35 2 Ridnour LA, Thomas DD, Mancardi D, Espey MG, Miranda KM, Paolocci N, Feelisch M, Fukuto J, Wink DA: The chemistry of nitrosative stress induced by nitric oxide and reactive nitrogen oxide species.

Putting perspective on stressful biological situations. Biol Chem , 1 Roberts RA, Laskin DL, Smith CV, Robertson FM, Allen EM, Doorn JA, Slikker W: Nitrative and oxidative stress in toxicology and disease. Toxicological sciences , 1 Rosenberg GA: Matrix metalloproteinases in neuroinflammation.

Glia , 39 3 Sablina AA, Budanov AV, Ilyinskaya GV, Agapova LS, Kravchenko JE, Chumakov PM: The antioxidant function of the p53 tumor suppressor. Nature medicine , 11 12 Free Radical Biol Med , 30 11 Schafer ZT, Grassian AR, Song L, Jiang Z, Gerhart-Hines Z, Irie HY, Gao S, Puigserver P, Brugge JS: Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment.

Scherz-Shouval R, Elazar Z: ROS, mitochondria and the regulation of autophagy. Trends in cell Biol , 17 9 Schetter AJ, Heegaard NH, Harris CC: Inflammation and cancer: interweaving microRNA, free radical, cytokine and p53 pathways. Carcinogenesis , 31 1 Schramek D, Kotsinas A, Meixner A, Wada T, Elling U, Pospisilik JA, Neely GG, Zwick R-H, Sigl V, Forni G: The stress kinase MKK7 couples oncogenic stress to p53 stability and tumor suppression.

Nature genetics , 43 3 Sethy-Coraci I, Crock LW, Silverstein SC: PAF-receptor antagonists, lovastatin, and the PTK inhibitor genistein inhibit H2O2 secretion by macrophages cultured on oxidized-LDL matrices. Journal of leukocyte biology , 78 5 Shen K, Ji L, Chen Y, Yu Q, Wang Z: Influence of glutathione levels and activity of glutathione-related enzymes in the brains of tumor-bearing mice.

Bioscience trends , 5 1 Cancer chemotherapy and pharmacology , 67 6 Singh S, Sreenath K, Pavithra L, Roy S, Chattopadhyay S: SMAR1 regulates free radical stress through modulation of AKR1a4 enzyme activity. Int J Biochem Cell Biol , 42 7 Skrzydlewska E, Sulkowski S, Koda M, Zalewski B, Kanczuga-Koda L, Sulkowska M: Lipid peroxidation and antioxidant status in colorectal cancer.

World J Gastroenterol , 11 3 Speisky H, Gómez M, Burgos-Bravo F, López-Alarcón C, Jullian C, Olea-Azar C, Aliaga ME: Generation of superoxide radicals by copper-glutathione complexes: Redox-consequences associated with their interaction with reduced glutathione.

Bioorg Med Chem , 17 5 Sung HJ, Ma W, P-y W, Hynes J, O'Riordan TC, Combs CA, McCoy JP, Bunz F, Kang J-G, Hwang PM: Mitochondrial respiration protects against oxygen-associated DNA damage. Nat Commun , 1: 5. Tao R, Coleman MC, Pennington JD, Ozden O, Park S-H, Jiang H, Kim H-S, Flynn CR, Hill S, Hayes McDonald W: Sirt3-mediated deacetylation of evolutionarily conserved lysine regulates MnSOD activity in response to stress.

Molecular cell , 40 6 Teixeira V, Valente H, Casal S, Marques F, Moreira P: Blood antioxidant and oxidative stress biomarkers acute responses to a m kayak sprint in elite male kayakers. J Sports Med physical fitness , 53 1 Ten Kate M, van der Wal J, Sluiter W, Hofland L, Jeekel J, Sonneveld P, van Eijck C: The role of superoxide anions in the development of distant tumour recurrence.

Brit J Cancer , 95 11 Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer , 2 6 Turi JL, Jaspers I, Dailey LA, Madden MC, Brighton LE, Carter JD, Nozik-Grayck E, Piantadosi CA, Ghio AJ: Oxidative stress activates anion exchange protein 2 and AP-1 in airway epithelial cells.

Am J Physiol Lung Cell Mol Physiol , 4 :LL Valko M, Rhodes C, Moncol J, Izakovic M, Mazur M: Free radicals, metals and antioxidants in oxidative stress-induced cancer.

Chemico-biological interactions , 1 Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J: Free radicals and antioxidants in normal physiological functions and human disease.

Int J Biochem Cell Biol , 39 1 Vera-Ramirez L, Sanchez-Rovira P, Ramirez-Tortosa MC, Ramirez-Tortosa CL, Granados-Principal S, Lorente JA, Quiles JL: Free radicals in breast carcinogenesis, breast cancer progression and cancer stem cells.

Biological bases to develop oxidative-based therapies. Crit Rev Oncol Hematol , 80 3 Vesper BJ, Elseth KM, Tarjan G, Haines GK III, Radosevich JA: Long-term adaptation of breast tumor cell lines to high concentrations of nitric oxide.

Tumor Biology , 31 4 Wang Y, Shang Y: Epigenetic control of epithelial-to-mesenchymal transition and cancer metastasis.

Exp Cell Res , 2 Weinberg F, Chandel NS: Reactive oxygen species-dependent signaling regulates cancer. Cellular and molecular life sciences , 66 23 Whitaker-Menezes D, Martinez-Outschoorn UE, Flomenberg N, Birbe RC, Witkiewicz AK, Howell A, Pavlides S, Tsirigos A, Ertel A, Pestell RG: Hyperactivation of oxidative mitochondrial metabolism in epithelial cancer cells in situ: visualizing the therapeutic effects of metformin in tumor tissue.

Cell Cycle , 10 23 Wolf FI, Torsello A, Tedesco B, Fasanella S, Boninsegna A, D'Ascenzo M, Grassi C, Azzena GB, Cittadini A: Hz extremely low frequency electromagnetic fields enhance cell proliferation and DNA damage: possible involvement of a redox mechanism.

Biochimica et Biophysica Acta BBA -Molecular Cell Research , 1 Wong K-K, Engelman JA, Cantley LC: Targeting the PI3K signaling pathway in cancer. Wood LD, Parsons DW, Sn J, Lin J, Sjoblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J: The genomic landscapes of human breast and colorectal cancers.

Woolston CM, Al-Attar A, Storr SJ, Ellis IO, Morgan DA, Martin SG: Redox protein expression predicts radiotherapeutic response in early-stage invasive breast cancer patients. Int J Radiat Oncol Biol Phys , 79 5 Wright DT, Cohn LA, Li H, Fischer B, Li CM, Adler KB: Interactions of oxygen radicals with airway epithelium.

Environmental health perspectives , Suppl 10 Xiao C, Sharp JA, Kawahara M, Davalos AR, Difilippantonio MJ, Hu Y, Li W, Cao L, Buetow K, Ried T: The XIST Noncoding RNA Functions Independently of BRCA1 in X Inactivation.

Zaremba T, Olinski R: Oxidative DNA damage—analysis and clinical significance. Postepy biochemistry , 56 2 Zhang HJ, Zhao W, Venkataraman S, Robbins ME, Buettner GR, Kregel KC, Oberley LW: Activation of matrix metalloproteinase-2 by overexpression of manganese superoxide dismutase in human breast cancer MCF-7 cells involves reactive oxygen species.

J Biol Chem , 23 Zhou L, Wang Y, Tian D, Yang J, Yang Y: Decreased levels of nitric oxide production and nitric oxide synthase-2 expression are associated with the development and metastasis of hepatocellular carcinoma.

Molecular medicine reports , 6 6 Ziech D, Franco R, Georgakilas AG, Georgakila S, Malamou-Mitsi V, Schoneveld O, Pappa A, Panayiotidis MI: The role of reactive oxygen species and oxidative stress in environmental carcinogenesis and biomarker development.

Chemico-biological interactions , 2 Z-y L, Yang Y, Ming M, Liu B: Mitochondrial ROS generation for regulation of autophagic pathways in cancer. Biochem Biophys Res Commun , 1 Download references. This study was supported by the Fondo de Investigación Sanitaria PI to M.

Artacho-Cordón is supported by the Spanish Ministry of Science and Education AP A grant from the Fundación Benéfica San Francisco Javier y Santa Cándida, University of Granada to S. Ríos-Arrabal greatly aided this work. This research was also funded by the Unidad de Apoyo a la Investigación of San Cecilio University Hospital, Granada.

The authors thank Glenn Harding for improving the English style of the manuscript. It has been confirmed that these organelles are the principal intracellular source of ROS production in most tissues.

Studies have shown that under physiological condition, complex II respiratory chain could also be a main regulator of ROS generation from mitochondria [ 13 ]. In eukaryotic cells, the mitochondrion is an important organelle that plays a main role in several critical processes.

The important role of mitochondria is mentioned in the physiology of cancer, such as in energy metabolism and cell cycle regulation. There is powerful documentary evidence to support the rationale for the expansion of anticancer strategies based on mitochondrial targets.

This organelle is recognizing to play an important role in the apoptosis mechanism and initiate cell death through various mechanisms that comprise disrupting electron transport and energy metabolism in the respiratory chain, releasing agents or proteins such as cytochrome c that mediate apoptosis signaling, and changing the cellular redox status by ROS generation.

The therapeutic targeting of cancer cells based on mitochondria have often depended on the intrinsic various differences between mitochondria in normal and cancer cells, which allow for better options, manipulation different pathways, and destruction of these cancer cells.

These differences are including bioenergetics change, disturbance of the mitochondrial DNA mtDNA , and morphological and physiological changes in the cancer cell Table 1. The mtDNA is one main target of ROS and the lack of sufficient protective histones surrounding the mtDNA in cancer cells makes the mtDNA more easily tending to ROS-caused DNA damage.

In addition, various researches have shown that the mitochondria were different in cancer cells than in normal cells, such as grow faster, fewer and smaller, and also had the morphological transformed [ 13 ]. Some differences between cancer and normal cells such as bioenergetics process, MMP and ROS level, and morphological and physiological differences.

Today, several mitochondria targeted strategies for cancer therapy have been focused on the development of agents that manage increased the ROS generation in mitochondria from the cancer cells without effect on the normal cells. It has been shown that ROS generation in the mitochondria is evaluated through the rates of both mitochondria ROS mtROS disposal and production, and ROS levels in mitochondria are regulated by several agents, including mitochondria O 2 levels, mitochondrial membrane potential MMP or Δψm , the metabolic condition of mitochondria, and other factors Figure 4.

A number of recent researches reveal the fact that mtROS at low to high levels act as several functions. That is it at low levels, involved in the hypoxia adaptation process, at moderate levels, involved in controlling inflammatory response, and at high level involved in regulating apoptosis signaling.

Adjustment of mitochondria ROS mtROS generation. Several agents such as MMP, the metabolic state of mitochondrial, O 2 level and STAT3 adjust the generation of mtROS. As mentioned in the previous section, the increase of ROS level of intracellular by sever agents through activating signaling pathways in cancer cells represents that these cells are more vulnerable than normal cells to ROS-caused cell death.

The results of the studies suggest that ROS such as H 2 O 2 could affect the extrinsic apoptosis pathway through changing the intracellular space. The up-regulation of receptor shows in various systems with increase ROS by exogenous ROS and ROS causing agents.

It has also been found to sensitize cancer cells, but not normal cells to TRAIL-caused apoptosis. Adenine nucleotide translocator ANT , as an inner mitochondrial protein, is also a target of ROS regulation by integrity of its redox-sensitive cysteines, supplying an extra mechanism by which drug-caused ROS generation may activate mitochondrial apoptosis signaling.

On the other hand, ROS also could regulate protein complexes inner place the mitochondrial electron transport chain mETC , activate caspases-3 and initiate apoptosis signaling. Today, agents that are used in the treatment of cancer through the mechanism of ROS generation are known as an important drug class.

Some studies have shown that several mitochondria-targeted drugs have potency useful in selective cancer cell killing and no effect on normal cells in pre-clinical and clinical testing, such as ROS regulators.

It has been shown that cancer cells in comparison with normal cells are more vulnerable to irreversible damage induced by stress oxidative and subsequent apoptosis. Researchers in previous studies have been used of the differences between the mitochondria between cancer and normal cells as a means to kill cancer cells by anti-cancer drugs.

Developing cancer therapies based on increasing further the high ROS level in cancer cells to a toxic level by the several mechanisms such as triggering ROS accumulation directly and inhibiting the antioxidant systems display powerful phenomenon of selectively killing cancer cells [ 6 ].

A number of drugs class have been recognized as increasing ROS production. It is well documented that some of chemotherapeutic agents can induce ROS generation through mitochondrial respiratory chain complexes in patients during cancer therapy.

These compounds can be separated into various categories such as alkylating agents, anthracycline antibiotics, platinum compounds, mitotic inhibitors, antimetabolites, biological response modifiers, and hormone therapies.

Arsenic trioxide ATO is used in the treatment of acute promyelocytic leukemia APL. It was reported that ATO induces apoptosis signaling in several cancer cells such as lung, leukemia, and myeloma cancer through the induction of ROS. The mechanism by which ATO cause increased ROS generation is not completely well known.

The most recent investigations indicated that ATO can impair the function of respiratory chain in the mitochondria, leading to increased generation of superoxide, likely by causing leakage of electrons from the mitochondrial respiratory chain complexes Figure 5.

On the other hand, ATO could be used in mixture with several anticancer drugs, which play a role through increasing ROS production. Mechanism of ROS generation and apoptosis induction by the ATO, doxorubicin, daunorubicin and bleomycin anthracycline antibiotics and cisplatin.

These drugs, leading to increased generation of ROS, likely by causing leakage of electrons from the mitochondrial respiratory chain complexes. The doxorubicin, daunorubicin, and bleomycin are an anthracycline antibiotics, cisplatin is a platinum compound, and amitriptyline as a tricyclic antidepressant are used in the treatment of several types of cancer.

The mechanism of doxorubicin bleomycin, cisplatin, and amitriptyline in the ROS production is the same of ATO. These drugs with impair the function of respiratory chain in the mitochondria, leading to increase generation of superoxide. These compounds due to this mechanism ROS generation are used for the treatment of several types of cancers Figure 5.

Studies have shown that other drugs such as dequalinium chloride preclinical and metformin preclinical and clinical, Phase I through inhibition of mitochondrial complex 1 have the ability to produce ROS.

VDACs, also known as mitochondrial porins, show high similarity between some animals especially mice and humans. VDAC play an important role in the cell, such as regulating mitochondrial shape and structural changes, regulating apoptosis signaling, regulating ATP and calcium transport. Several studies have demonstrated the role of VDAC in the regulation of apoptosis signaling and VDAC is being studied as a cancer-specific target.

As has been mentioned, these drugs promote ROS production through the disturbance of VDAC and cause non-apoptotic form of cell death in KRAS. Several studies have shown that some drugs and agents including, paclitaxel taxol , ionizing radiation, niclosamide, AGX, AG with effect on NOX induces ROS generation.

The recent results from both in vitro and in vivo studies have shown that this drug causes the translocation of Rac1, which favorably regulates the activity of NOX, thereby furthering ROS H 2 O 2 production.

It was reported that taxol can raise the levels of ROS in the extracellular and subsequently induced cancer cell death resulting in the release of cytochrome c from the mitochondria.

P53 act as a transcription factor to regulate the expression of many pro-oxidant genes. The 5-fluorouracil 5-FU is an antimetabolites and pyrimidine analog. It is used for therapies for several types of cancers, such as gastrointestinal, colon, rectal, and head and neck cancer, through inducing intracellular increase in superoxide.

The mechanism by which 5-FU cause increased ROS generation from mitochondria is through a pdependent pathway [ 4 ].

The two pathways GSH through enzymes such as GPX and GST and catalase can act directly on scavenging ROS in cells. For that reason, oxidative stress can be promoted with methods based on the loss of the reduced GSH storage and other antioxidant sources. A number of drugs class have been recognized as increasing oxidative stress process and ROS generation through targeting antioxidant system [ 4 , 5 ].

These drugs are, including buthionine sulfoximine BSO , imexon, phenylethyl isothiocyanate, mangafodipir, 2-methoxyestradiol, tetrathiomolybdate ATN , and auranofin, used in the treatment of various types of cancer.

For example, BSO through inhibition of the antioxidant system especially GSH in cancer cells such as ovarian and breast cancers can induce an accumulation of ROS due to the high basal ROS output in ovarian and breast cancers, and initiate cell death [ 4 , 5 ].

Other studies have shown that imexon through decrease GSH pool and subsequently increase the production of ROS and decrease mitochondria function was induced apoptosis.

Other studies have shown that some other drugs, including ascorbic acid and diethylmaleate are able effects on GSH GSH depletion. One the other hand, mercaptosuccinic acid, aminotriazol, and 2-Methoxyoestradiol were able to inhibit of GPx, catalase, and SOD, respectively, and thereby increase ROS production [ 7 ].

Cancer is a multistage disease including initiation, promotion, and progression. The increased ROS causes DNA damage, which may lead to DNA damage or gene mutation, resulting in the progression of cancer.

Increased generation of ROS and an altered redox status have observed in cancer cells, and investigations suggest that this biochemical property of cancer cells can be exploited for cancer therapy.

For treatment of cancer, since high levels of ROS can induce cell death, treatment of radiation, chemotherapy, and molecule compounds all can increase the level of intracellular ROS to induce cancer cell death and apoptosis.

The increased intracellular ROS levels could make cancer cells more vulnerable than normal cells to oxidative stress-induced cell death. Licensee IntechOpen. This chapter is distributed under the terms of the Creative Commons Attribution 3. Edited by Rizwan Ahmad. Open access peer-reviewed chapter Role of Oxygen Free Radicals in Cancer Development and Treatment Written By Jalal Pourahmad, Ahmad Salimi and Enaytollah Seydi.

DOWNLOAD FOR FREE Share Cite Cite this chapter There are two ways to cite this chapter:. Choose citation style Select style Vancouver APA Harvard IEEE MLA Chicago Copy to clipboard Get citation. Choose citation style Select format Bibtex RIS Download citation.

IntechOpen Free Radicals and Diseases Edited by Rizwan Ahmad. From the Edited Volume Free Radicals and Diseases Edited by Rizwan Ahmad Book Details Order Print. Chapter metrics overview 3, Chapter Downloads View Full Metrics.

Impact of this chapter. Abstract It is well known that species derived from oxygen are cytotoxic and are involved in the etiology of cancer. Keywords cancer reactive oxygen species oxidation therapy mitochondria.

pourahmadjaktaji utoronto. Mechanisms of free radical-induced DNA base modification and mutagenesis It has been appraised that in one human cell is exposed to nearly 10 5 oxidative hits such as hydroxyl radical and other such species in a day.

Role of ROS in genotoxicity and DNA damage ROS-caused DNA lesion may be characterized both structurally and chemically and displays a typical schema of modifications. Lipid peroxidation and DNA damage While major consideration has centralized on direct DNA lesion by oxygen free radicals because of the genetic outcomes of such lesion, reactive radical species may also induce damage to other cellular members.

Functions of ROS in the cancer cells. Antioxidant and oxidation pathways regulate ROS generation. NRF2 nuclear factor, erythroid-derived 2, like 2 NRF2 is an important regulator of the antioxidant system and cellular stress responses in the several cancers.

FOXO Forkhead box O and p53 FOXO, as a transcription factors, are involved in different signaling pathways and play key roles in some physiological and pathological processes such as cancer. Hypoxia and hypoglycemia Hypoxic conditions caused by the imbalance between intake and oxygen consumption [ 4 ].

Mitochondria In recent decades, several studies have shown that mitochondria organelle plays an important role in human health and disease. Cancer cells Normal cell Bioenergetics process Aerobic glycolysis condition "Warburg Effect" Aerobic condition Mitochondrial DNA mtDNA mtDNA is mutated mtDNA is normal Morphological and physiological differences shape and count Size and shape: smaller Size and shape: larger MMP level Higher ~60 mV Lower ROS level Higher Lower Intracellular pH Acidic No acidic Metabolic rates Higher Lower.

Table 1. Targeting mitochondrial respiratory chain Arsenic trioxide ATO is used in the treatment of acute promyelocytic leukemia APL. Targeting VDACs VDACs, also known as mitochondrial porins, show high similarity between some animals especially mice and humans. Targeting NOX Several studies have shown that some drugs and agents including, paclitaxel taxol , ionizing radiation, niclosamide, AGX, AG with effect on NOX induces ROS generation.

Targeting p53 P53 act as a transcription factor to regulate the expression of many pro-oxidant genes. References 1. Dizdaroglu M, Jaruga P, Birincioglu M, Rodriguez H. Free radical-induced damage to DNA: mechanisms and measurement 1, 2.

Free Radical Biology and Medicine. Waris G, Ahsan H. Reactive oxygen species: role in the development of cancer and various chronic conditions. Journal of Carcinogenesis.

Marnett LJ.

Role of Oxygen Free Radicals in Cancer Development and Treatment | IntechOpen These antioxidants play a crucial role in inhibiting free radical production, which is why diets rich in fruits and vegetables are linked with better health and a reduced risk of many diseases, including cancer. This vegetable family contains powerful phytochemicals, including carotenoids, indoles and glucosinolates and isothiocyanates, which have been studied and shown to slow the growth of many cancers. Free radical levels increased, and the cells became long and spindly, losing their ability to move around as much. Broccoli Sunflower Salad. Aspects Med.
On Nutrition Kumar B, An S, Khandrika L, Meacham RB, Raducals HK: Oxidative stress Free radicals and cancer qnd in prostate cancer cells and is required for aggressive phenotype. Apple Crisp. Doll, J. Braised Kale with Black Beans and Tomatoes. Oliver, M. The activity of MMP-2 can be modulated according to intracellular levels of RONS. Stauber, and T.
Antioxidants and Cancer Prevention

Oxidative stress is a serious health concern. It damages fatty tissue, DNA and proteins that the body needs to support essential, everyday processes. As the damage compounds, it can negatively impact your immune system.

Scientists believe it can event contribute to the development of serious diseases including diabetes, heart disease and cancer.

As free radicals build up and create progressively more oxidative stress, they do major damage to genes in our DNA. Over time, the damage can cause DNA to produce ineffective proteins, which affects DNA integrity. When DNA suffers, mutations occur.

The most common form of mutation occurs in genes known as tumor suppressor genes: the genes that function to repair cell damage and prevent cellular death.

A series of these mutations in the tumor suppressor genes and other genes are usually responsible for the formation of cancer cells. These are the cells that multiply into a malignant tumor. Fruits and vegetables! Consider having one vegetarian day per week or even going completely vegetarian.

Make your goal servings of fruits and vegetables per day. If you are unsure what a serving is, check out our article Confused About the Serving Sizes of Vegetables? Have you increased antioxidants in your diet? Maurer Foundation. July 11, Read More About the Author April Zubko Categories: Breast Cancer Prevention 0 comments.

This field is for validation purposes and should be left unchanged. Antioxidant Supplementation and Breast Cancer Prognosis in Postmenopausal Women Undergoing Chemotherapy and Radiation Therapy.

The American Journal of Clinical Nutrition. Lignitto L, LeBoeuf SE, Hamer H, et al. Nrf2 Activation Promotes Lung Cancer Metastasis by Inhibiting the Degradation of Bach1.

doi: By Lynne Eldridge, MD Lynne Eldrige, MD, is a lung cancer physician, patient advocate, and award-winning author of "Avoiding Cancer One Day at a Time.

Use limited data to select advertising. Create profiles for personalised advertising. Use profiles to select personalised advertising.

Create profiles to personalise content. Use profiles to select personalised content. Measure advertising performance. Measure content performance. Understand audiences through statistics or combinations of data from different sources.

Develop and improve services. Use limited data to select content. List of Partners vendors. By Lynne Eldridge, MD. Medically reviewed by Doru Paul, MD. Table of Contents View All. Table of Contents. What Are Free Radicals?

Causes and Sources. Free Radicals and Cancer. Reducing Free Radicals. Free Radicals and Oxidized Cholesterol in Your Body. How Free Radicals and Carcinogens Are Linked.

The Free Radical Theory of Aging There are several theories about why our bodies age and free radicals are a key player in many of them. Free Radicals and Aging. Do Free Radicals Cause Cancer Cells to Form? Anthocyanins and Free Radicals.

What Causes Cancer? Can I Take Supplements During Cancer Treatment? Does Glutathione Fight Free Radicals? Verywell Health uses only high-quality sources, including peer-reviewed studies, to support the facts within our articles.

Read our editorial process to learn more about how we fact-check and keep our content accurate, reliable, and trustworthy. See Our Editorial Process. Meet Our Medical Expert Board. Share Feedback. Was this page helpful?

Thanks for your feedback! What is your feedback? Related Articles.

The Link Between Free Radicals and Cancer

Harman, and J. Miguel, eds. Oxygen radicals: A commonsense look at their nature and medical importance. Free radicals and metal ions in health and disease.

Halliwell and J. Oxygen toxicity, oxygen radicals, transition metals, and disease. Halliwell, and J. Free Radicals in Biology and Medicine. Clarendon Press, Oxford, The importance of free radicals and catalytic metal ions in human diseases. Aspects Med.

Oxygen radicals and the nervous system. Trends in Neurosci. Halliwell, J. Gutteridge, and D. Metal ions and oxygen radical reactions in human inflammatory joint disease. Royal Soc. B, — Oxygen free radicals and iron in relation to biology and medicine: some problems and concepts. Iron and free radical reactions: two aspects of antioxidant protection.

TIBS 11, — The aging process. USA 78, — Free radicals and the origination, evolution, and present status of the free radical theory of aging. In Free Radicals in Molecular Biology , Ageing , and Disease , D. Armstrong, R. Sohal, R. Cutler, and T. Slater, eds.

Age 7, — Free radical theory of aging: Role of free radicals in the origination and evolution of life, aging, and disease processes. In Free Radicals, Ageing, and Degenerative Diseases, J.

Theories of biological aging. Micronutrients and cancer prevention. Chemical carcinogens in tobacco. In Cancer Risks , P. Bannasch, ed.

The ageing process is a key problem in biomedical research. Lancet ii , — Hughes, F. McDermott, A. Polglase, W. Johnson, and E. Large bowel cancer-the next move? Australia 1, 36—37 Brave New World. Chatto and Windus, London, Selenium inhibition of chemical carcinogenesis.

Kensler, and M. Role of oxygen radicals in tumour promotion. Germanium Ge : Homeostatic normalizer and immunostimulant. A review of its preventive and therapeutic efficacy.

Lathia, A. Braasch, and V. Inhibitory effects of vitamin C and E on in vitro formation of N-nitrosamine under physiological conditions. Loeb, V. Emster, K. Wamer, J. Abbots, and J. Smoking and lung cancer: An overview.

Marcus, N. Ibrahim, and M. Age-related decline in the biosynthesis of mitochondrial inner membrane proteins. Oxygen free radicals linked to many diseases.

Massie, V. Aiello, and T. Dietary vitamin C improves the survival of mice. Gerontology 30, — Oxygen-derived free radicals in postischemic tissue injury. Miller, and J. Searches for ultimate chemical carcinogens and their reactions with cellular macromolecules.

Cancer 47, — Miguel, and J. Muir, and D. The world cancer burden: prevent or perish. Naqui, and B. Reactive oxygen intermediates in biochemistry. Oberley, and T. Free radicals, cancer, and aging. In Free Radicals , Ageing and Degenerative Diseases , J.

Oberley, T. Oberley, and G. Cell differentiation, aging, and cancer. The possible roles of superoxide and superoxide dis-mutases. Hypotheses 6, — Oliver, R. Fulks, R. Levine, L. Fucci, A.

Rivett, J. Roseman, and E. Oxidative inactivation of key metabolic enzymes. In Molecular Basis of Ageing , A. Ono, and S. Unique increase of superoxide dismutase level in brains of long living mammals. Evolution and the need for ascorbic acid.

USA 67, — How to Live Longer and Feel Better. Avon Books, New York, Peto, R. Doll, J. Buckley, and M. Can dietary beta-carotene materially reduce human cancer rates? Nature — Fundamentals of Oncology.

Marcel Dekker, New York, Proctor, and E. Free radicals and disease in man. NMR 16, — Cancer and free radicals.

Oxyradicals and related species: Their formation, lifetimes, and reactions. The New Zealand selenium experience. The alteration of enzymes in aging animals.

Salonen, G. Alfthan, J. Huttunen, and P. Association between serum selenium and the risk of cancer. Salonen, R. Lappetelainen, P. Maenpaa, G. Alfthan, and P. Risk of cancer in relation to serum concentrations of selenium and vitamins A and E: matched case-control analysis of prospective data.

Nutrition and Cancer , Plenum, New York, In Biochemistry of the Essential Ultratrace Elements , E. Frieden, ed. Shankel, P. Hartman, T. Kada, and A. Synopsis of the first international conference on mutagenesis and anticarcinogenesis.

Hypoxia, on the one hand, as a ROS inducer, can also directly induce the raised expression of oncogenes such as MYC and RAS through HIF-2α. On the other hand, MYC increases mitochondrial biogenesis, which adds to the raised ROS generation under hypoxic conditions and the raised mitochondrial ROS generation elevate the oxidative stress process [ 12 ].

It has been suggested that RAS, p53, and c-MYC through the mitochondria to regulate ROS generation, thereby affecting apoptosis. RAS in K-Ras-transformed fibroblast cell under the condition that glucose deprivation induces apoptosis through changes in mitochondrial complex gene expression.

It has also been reported that RAS in p66SHC overexpression condition in the transformed cells increase ROS generation through mitochondria and p53, MPTP opening and mitochondrial swelling could induce apoptosis Figure 3 [ 12 ]. In recent decades, several studies have shown that mitochondria organelle plays an important role in human health and disease.

These studies led to the emergence of a new field of study named "mitochondrial medicine. It has been confirmed that these organelles are the principal intracellular source of ROS production in most tissues.

Studies have shown that under physiological condition, complex II respiratory chain could also be a main regulator of ROS generation from mitochondria [ 13 ]. In eukaryotic cells, the mitochondrion is an important organelle that plays a main role in several critical processes.

The important role of mitochondria is mentioned in the physiology of cancer, such as in energy metabolism and cell cycle regulation. There is powerful documentary evidence to support the rationale for the expansion of anticancer strategies based on mitochondrial targets.

This organelle is recognizing to play an important role in the apoptosis mechanism and initiate cell death through various mechanisms that comprise disrupting electron transport and energy metabolism in the respiratory chain, releasing agents or proteins such as cytochrome c that mediate apoptosis signaling, and changing the cellular redox status by ROS generation.

The therapeutic targeting of cancer cells based on mitochondria have often depended on the intrinsic various differences between mitochondria in normal and cancer cells, which allow for better options, manipulation different pathways, and destruction of these cancer cells.

These differences are including bioenergetics change, disturbance of the mitochondrial DNA mtDNA , and morphological and physiological changes in the cancer cell Table 1. The mtDNA is one main target of ROS and the lack of sufficient protective histones surrounding the mtDNA in cancer cells makes the mtDNA more easily tending to ROS-caused DNA damage.

In addition, various researches have shown that the mitochondria were different in cancer cells than in normal cells, such as grow faster, fewer and smaller, and also had the morphological transformed [ 13 ]. Some differences between cancer and normal cells such as bioenergetics process, MMP and ROS level, and morphological and physiological differences.

Today, several mitochondria targeted strategies for cancer therapy have been focused on the development of agents that manage increased the ROS generation in mitochondria from the cancer cells without effect on the normal cells.

It has been shown that ROS generation in the mitochondria is evaluated through the rates of both mitochondria ROS mtROS disposal and production, and ROS levels in mitochondria are regulated by several agents, including mitochondria O 2 levels, mitochondrial membrane potential MMP or Δψm , the metabolic condition of mitochondria, and other factors Figure 4.

A number of recent researches reveal the fact that mtROS at low to high levels act as several functions. That is it at low levels, involved in the hypoxia adaptation process, at moderate levels, involved in controlling inflammatory response, and at high level involved in regulating apoptosis signaling.

Adjustment of mitochondria ROS mtROS generation. Several agents such as MMP, the metabolic state of mitochondrial, O 2 level and STAT3 adjust the generation of mtROS. As mentioned in the previous section, the increase of ROS level of intracellular by sever agents through activating signaling pathways in cancer cells represents that these cells are more vulnerable than normal cells to ROS-caused cell death.

The results of the studies suggest that ROS such as H 2 O 2 could affect the extrinsic apoptosis pathway through changing the intracellular space. The up-regulation of receptor shows in various systems with increase ROS by exogenous ROS and ROS causing agents.

It has also been found to sensitize cancer cells, but not normal cells to TRAIL-caused apoptosis. Adenine nucleotide translocator ANT , as an inner mitochondrial protein, is also a target of ROS regulation by integrity of its redox-sensitive cysteines, supplying an extra mechanism by which drug-caused ROS generation may activate mitochondrial apoptosis signaling.

On the other hand, ROS also could regulate protein complexes inner place the mitochondrial electron transport chain mETC , activate caspases-3 and initiate apoptosis signaling. Today, agents that are used in the treatment of cancer through the mechanism of ROS generation are known as an important drug class.

Some studies have shown that several mitochondria-targeted drugs have potency useful in selective cancer cell killing and no effect on normal cells in pre-clinical and clinical testing, such as ROS regulators.

It has been shown that cancer cells in comparison with normal cells are more vulnerable to irreversible damage induced by stress oxidative and subsequent apoptosis.

Researchers in previous studies have been used of the differences between the mitochondria between cancer and normal cells as a means to kill cancer cells by anti-cancer drugs.

Developing cancer therapies based on increasing further the high ROS level in cancer cells to a toxic level by the several mechanisms such as triggering ROS accumulation directly and inhibiting the antioxidant systems display powerful phenomenon of selectively killing cancer cells [ 6 ].

A number of drugs class have been recognized as increasing ROS production. It is well documented that some of chemotherapeutic agents can induce ROS generation through mitochondrial respiratory chain complexes in patients during cancer therapy. These compounds can be separated into various categories such as alkylating agents, anthracycline antibiotics, platinum compounds, mitotic inhibitors, antimetabolites, biological response modifiers, and hormone therapies.

Arsenic trioxide ATO is used in the treatment of acute promyelocytic leukemia APL. It was reported that ATO induces apoptosis signaling in several cancer cells such as lung, leukemia, and myeloma cancer through the induction of ROS.

The mechanism by which ATO cause increased ROS generation is not completely well known. The most recent investigations indicated that ATO can impair the function of respiratory chain in the mitochondria, leading to increased generation of superoxide, likely by causing leakage of electrons from the mitochondrial respiratory chain complexes Figure 5.

On the other hand, ATO could be used in mixture with several anticancer drugs, which play a role through increasing ROS production. Mechanism of ROS generation and apoptosis induction by the ATO, doxorubicin, daunorubicin and bleomycin anthracycline antibiotics and cisplatin.

These drugs, leading to increased generation of ROS, likely by causing leakage of electrons from the mitochondrial respiratory chain complexes. The doxorubicin, daunorubicin, and bleomycin are an anthracycline antibiotics, cisplatin is a platinum compound, and amitriptyline as a tricyclic antidepressant are used in the treatment of several types of cancer.

The mechanism of doxorubicin bleomycin, cisplatin, and amitriptyline in the ROS production is the same of ATO. These drugs with impair the function of respiratory chain in the mitochondria, leading to increase generation of superoxide. These compounds due to this mechanism ROS generation are used for the treatment of several types of cancers Figure 5.

Studies have shown that other drugs such as dequalinium chloride preclinical and metformin preclinical and clinical, Phase I through inhibition of mitochondrial complex 1 have the ability to produce ROS. VDACs, also known as mitochondrial porins, show high similarity between some animals especially mice and humans.

VDAC play an important role in the cell, such as regulating mitochondrial shape and structural changes, regulating apoptosis signaling, regulating ATP and calcium transport. Several studies have demonstrated the role of VDAC in the regulation of apoptosis signaling and VDAC is being studied as a cancer-specific target.

As has been mentioned, these drugs promote ROS production through the disturbance of VDAC and cause non-apoptotic form of cell death in KRAS.

Several studies have shown that some drugs and agents including, paclitaxel taxol , ionizing radiation, niclosamide, AGX, AG with effect on NOX induces ROS generation. The recent results from both in vitro and in vivo studies have shown that this drug causes the translocation of Rac1, which favorably regulates the activity of NOX, thereby furthering ROS H 2 O 2 production.

It was reported that taxol can raise the levels of ROS in the extracellular and subsequently induced cancer cell death resulting in the release of cytochrome c from the mitochondria. P53 act as a transcription factor to regulate the expression of many pro-oxidant genes. The 5-fluorouracil 5-FU is an antimetabolites and pyrimidine analog.

It is used for therapies for several types of cancers, such as gastrointestinal, colon, rectal, and head and neck cancer, through inducing intracellular increase in superoxide. The mechanism by which 5-FU cause increased ROS generation from mitochondria is through a pdependent pathway [ 4 ].

The two pathways GSH through enzymes such as GPX and GST and catalase can act directly on scavenging ROS in cells. For that reason, oxidative stress can be promoted with methods based on the loss of the reduced GSH storage and other antioxidant sources. A number of drugs class have been recognized as increasing oxidative stress process and ROS generation through targeting antioxidant system [ 4 , 5 ].

These drugs are, including buthionine sulfoximine BSO , imexon, phenylethyl isothiocyanate, mangafodipir, 2-methoxyestradiol, tetrathiomolybdate ATN , and auranofin, used in the treatment of various types of cancer.

For example, BSO through inhibition of the antioxidant system especially GSH in cancer cells such as ovarian and breast cancers can induce an accumulation of ROS due to the high basal ROS output in ovarian and breast cancers, and initiate cell death [ 4 , 5 ]. Other studies have shown that imexon through decrease GSH pool and subsequently increase the production of ROS and decrease mitochondria function was induced apoptosis.

Other studies have shown that some other drugs, including ascorbic acid and diethylmaleate are able effects on GSH GSH depletion.

One the other hand, mercaptosuccinic acid, aminotriazol, and 2-Methoxyoestradiol were able to inhibit of GPx, catalase, and SOD, respectively, and thereby increase ROS production [ 7 ].

Cancer is a multistage disease including initiation, promotion, and progression. The increased ROS causes DNA damage, which may lead to DNA damage or gene mutation, resulting in the progression of cancer.

Increased generation of ROS and an altered redox status have observed in cancer cells, and investigations suggest that this biochemical property of cancer cells can be exploited for cancer therapy. For treatment of cancer, since high levels of ROS can induce cell death, treatment of radiation, chemotherapy, and molecule compounds all can increase the level of intracellular ROS to induce cancer cell death and apoptosis.

The increased intracellular ROS levels could make cancer cells more vulnerable than normal cells to oxidative stress-induced cell death. Licensee IntechOpen. This chapter is distributed under the terms of the Creative Commons Attribution 3.

Edited by Rizwan Ahmad. Open access peer-reviewed chapter Role of Oxygen Free Radicals in Cancer Development and Treatment Written By Jalal Pourahmad, Ahmad Salimi and Enaytollah Seydi. DOWNLOAD FOR FREE Share Cite Cite this chapter There are two ways to cite this chapter:.

Choose citation style Select style Vancouver APA Harvard IEEE MLA Chicago Copy to clipboard Get citation. Choose citation style Select format Bibtex RIS Download citation. IntechOpen Free Radicals and Diseases Edited by Rizwan Ahmad. From the Edited Volume Free Radicals and Diseases Edited by Rizwan Ahmad Book Details Order Print.

Chapter metrics overview 3, Chapter Downloads View Full Metrics. Impact of this chapter. Abstract It is well known that species derived from oxygen are cytotoxic and are involved in the etiology of cancer.

Keywords cancer reactive oxygen species oxidation therapy mitochondria. pourahmadjaktaji utoronto. Mechanisms of free radical-induced DNA base modification and mutagenesis It has been appraised that in one human cell is exposed to nearly 10 5 oxidative hits such as hydroxyl radical and other such species in a day.

Role of ROS in genotoxicity and DNA damage ROS-caused DNA lesion may be characterized both structurally and chemically and displays a typical schema of modifications.

Lipid peroxidation and DNA damage While major consideration has centralized on direct DNA lesion by oxygen free radicals because of the genetic outcomes of such lesion, reactive radical species may also induce damage to other cellular members.

Functions of ROS in the cancer cells. Antioxidant and oxidation pathways regulate ROS generation. NRF2 nuclear factor, erythroid-derived 2, like 2 NRF2 is an important regulator of the antioxidant system and cellular stress responses in the several cancers.

FOXO Forkhead box O and p53 FOXO, as a transcription factors, are involved in different signaling pathways and play key roles in some physiological and pathological processes such as cancer. Hypoxia and hypoglycemia Hypoxic conditions caused by the imbalance between intake and oxygen consumption [ 4 ].

Mitochondria In recent decades, several studies have shown that mitochondria organelle plays an important role in human health and disease. Cancer cells Normal cell Bioenergetics process Aerobic glycolysis condition "Warburg Effect" Aerobic condition Mitochondrial DNA mtDNA mtDNA is mutated mtDNA is normal Morphological and physiological differences shape and count Size and shape: smaller Size and shape: larger MMP level Higher ~60 mV Lower ROS level Higher Lower Intracellular pH Acidic No acidic Metabolic rates Higher Lower.

Table 1. Targeting mitochondrial respiratory chain Arsenic trioxide ATO is used in the treatment of acute promyelocytic leukemia APL. Targeting VDACs VDACs, also known as mitochondrial porins, show high similarity between some animals especially mice and humans.

Targeting NOX Several studies have shown that some drugs and agents including, paclitaxel taxol , ionizing radiation, niclosamide, AGX, AG with effect on NOX induces ROS generation. Targeting p53 P53 act as a transcription factor to regulate the expression of many pro-oxidant genes.

References 1. Antioxidants are chemicals that interact with and neutralize free radicals , thus preventing them from causing damage. The body makes some of the antioxidants that it uses to neutralize free radicals.

These antioxidants are called endogenous antioxidants. However, the body relies on external exogenous sources, primarily the diet, to obtain the rest of the antioxidants it needs. These exogenous antioxidants are commonly called dietary antioxidants.

Fruits, vegetables, and grains are rich sources of dietary antioxidants. Some dietary antioxidants are also available as dietary supplements 1 , 3. Examples of dietary antioxidants include beta-carotene , lycopene , and vitamins A, C, and E alpha-tocopherol.

The mineral element selenium is often thought to be a dietary antioxidant, but the antioxidant effects of selenium are most likely due to the antioxidant activity of proteins that have this element as an essential component i.

In laboratory and animal studies , the presence of increased levels of exogenous antioxidants has been shown to prevent the types of free radical damage that have been associated with cancer development. Therefore, researchers have investigated whether taking dietary antioxidant supplements can help lower the risk of developing or dying from cancer in humans.

Many observational studies , including case—control studies and cohort studies , have been conducted to investigate whether the use of dietary antioxidant supplements is associated with reduced risks of cancer in humans. Overall, these studies have yielded mixed results 5.

Because observational studies cannot adequately control for biases that might influence study outcomes, the results of any individual observational study must be viewed with caution. Randomized controlled clinical trials , however, lack most of the biases that limit the reliability of observational studies.

To date, nine randomized controlled trials of dietary antioxidant supplements for cancer prevention have been conducted worldwide. Many of the trials were sponsored by the National Cancer Institute.

The results of these nine trials are summarized below. Initial: no effect on risk of developing either cancer; decreased risk of dying from gastric cancer only Later: no effect on risk of dying from gastric cancer.

Initial: lower total cancer and prostate cancer incidence and all-cause mortality among men only; increased incidence of skin cancer among women only.

Learn about the Stress reduction exercises for seniors shotCOVID Frew Free radicals and cancer, and our masking policy ». View the changes to our visitor znd ». View Free radicals and cancer for Guest Services ». Access your health information from any device with MyHealth. You can message your clinic, view lab results, schedule an appointment, and pay your bill. Learn More about MyHealth » Learn More about Video Visits ». Get the iPhone MyHealth app » Get the Android MyHealth app ». Free radicals and cancer

Free radicals and cancer -

Chicken Chili. Chicken Enchilada Casserole. Cranberry Salmon. Cranberry-Turkey Salad Sandwiches. Crispy Parmesan Turkey Cutlets. Crunchy Veggie Wrap. Easy Spinach Lasagna.

Eating Well Sloppy Joe. Egg, Spinach, and Bacon Sandwiches. Fish Filet with Squash and Herbs. Greek-Style Scallops. Grilled Ginger Tuna. Grilled Halibut with a Tomato-Herb Sauce. Grilled Portobello Burgers. Grilled Vegetable Polenta with Pan Roasted Red Pepper and Tomato Sauce.

Halibut with Citrus and Garlic. Healthy Jambalaya. Hearty Beef Stew with Winter Vegetables. Hearty Mediterranean Stew. Herbed Polenta with Grilled Portobello Mushrooms.

Indonesian Salmon. Lasagna Rolls. Lemon Dijon Salmon. Mediterranean Grilled Veggie Pockets. Molasses-Cured Pork Loin with Apples. Mushroom Goulash.

New American Plate "Tetrazzini" Casserole. New Tuna Salad. Peppers Stuffed with Barley, Parmesan and Onion. Pizza Meat Loaf. Pumpkin Gnocchi.

Quinoa and Mushroom Pilaf with Dill. Quinoa Stuffed Peppers. Roasted Pork Tenderloin with Maple Mustard Sauce. Scallion Crusted Arctic Char. Seared Scallops with Beet Puree and Arugula Salad.

Soft Tacos with Southwestern Vegetables. Spaghetti alla Carbonara. Speedy Summer Ratatouille. Spicy Broccoli, Cauliflower and Tofu. Steamed Halibut on Spinach with Lemon Sauce. Stuffed Cornish Hens. Summer Tofu Kebab with Peanut Sauce.

Sweet and Sour Chicken. Sweet and Sour Tofu. Tofu Cutlets Marsala. Turkey Reuben Grilled Sandwiches. Udon Noodles with Spicy Peanut Ginger Sauce. Veggie Pita Pizzas. White Wine Coq au Vin. Whole Wheat Pasta with Fennel, Peas and Arugula.

Zesty Roasted Chicken. Asian Green Bean Stir-Fry. Asian Pilaf. Avocado and Mango Salsa. Baked Sweet Potato Wedges. Bok Choy with Sautéed Mushrooms and Shallots. Braised Kale with Black Beans and Tomatoes. Broccoli with Hazelnuts. Brussels Sprouts with Pecans and Dried Cranberries.

Butternut Squash Pilaf. Garlicky Greens. Honey-Roasted Parsnips, Sweet Potatoes and Apples. Lite Hummus Dip. Parmesan Orzo Primavera. Peas-Mushroom Pilaf. Quinoa Salad with Roasted Autumn Vegetables.

Seasoned Spinach with Garlic. Simply Grilled Portobello Mushrooms. Spring Barley. Stir-Fried Kale with Slivered Carrots. Summer Gazpacho. Sweet Potato Power. Tofu Fried Rice.

Winter Caponata. Apple Cranberry Cobbler. Apple Crisp. Apple-Cranberry Crisp. Baked Summer Fruit. Better Brownies. Blueberry Crumble Pie. Cranberry-Orange Fruit Bars. Crunchy Oat Apricot Bars. Fresh Berry Sundaes.

Fudge Brownie Sundaes. Ginger Spice Biscotti. Grilled Fruit with Strawberry Dip. Grilled Peaches with Honey and Yogurt. Harvest Apples. Lemon Cake. Marbled Pumpkin Cheesecake. Melon Sorbet. Pear Crisp. Pumpkin Bread. Pumpkin Mousse. Raspberry Cinnamon Sorbet.

Rhubarb-Strawberry Parfaits. Sliced Oranges with Almonds and Ginger. Summer Fruit Gratin. Warm Chocolate Fantasy. Yogurt Berry Brûlée with Maple Almond Brittle. Apple Pumpkin Shake. Avocado and Melon Smoothie.

Banana Cinnamon Vanilla Shake. Berry Blast Protein Shake. Cinnamint Green Tea. Cinnamon Hot Chocolate. Green Tea Slush. High Calorie Recipe: Cinnamon-Peach Smoothie.

High Calorie Recipe: Super Protein Power Smoothie. Hot and Healthy Winter Teas. Juicing Recipes. Peach Apricot Dessert Smoothie. Sour Citrus Blast Smoothie. Spiced Brazilian Mocha. Tips for Making Smoothies and Shakes. High-Calorie Snack Recipes.

Our dietitians are available for 45 minute consults by appointment only, Monday — Friday from 8 a. Download our nutrition appointment flyer. Billing and insurance. Our dietitians are available for 45 minute consults by appointment only on: Mondays — Friday, 8 a.

Call us at to refer a new patient for a nutrition consultation. If you wish to refer a patient to the Stanford Cancer Center, please call the Physician Helpline. Learn More About PRISM ». Nutrition Services for Cancer Patients at Stanford Cancer Center Palo Alto Nutrition Services for Cancer Patients at Stanford Cancer Center South Bay Share on Facebook.

Notice: Users may be experiencing issues with displaying some pages on stanfordhealthcare. We are working closely with our technical teams to resolve the issue as quickly as possible. Thank you for your patience. Billing Insurance Medical Records Support Groups Help Paying Your Bill COVID Resource Center.

Locations and Parking Visitor Policy Hospital Check-in Video Visits International Patients Contact Us. View the changes to our visitor policy » View information for Guest Services ».

New to MyHealth? Manage Your Care From Anywhere. ALREADY HAVE AN ACCESS CODE? Activate Account. DON'T HAVE AN ACCESS CODE? Create a New Account. NEED MORE DETAILS? MyHealth for Mobile Get the iPhone MyHealth app » Get the Android MyHealth app ».

WELCOME BACK. Forgot Username or Password? Stanford Medicine Cancer Center. Nutrition Services for Cancer Patients Nutrition is an important part of life, cancer treatment, recovery, and prevention. Services Available at These Locations 2.

See All Locations ». Make an appointment. Care and Treatment. Your Dietitians. Reducing Cancer Risk. Before Cancer Treatment. Cancer Diet During Treatment. Food safety during cancer treatment Organic produce Making vegetables taste good High protein foods High calorie snacks Clear liquids and full liquids Enteral and parenteral nutrition for adults Exercise for appetite and digestion Lactose intolerance Increasing calories and protein low to no lactose Nutrition during chemo Nutrition during radiation therapy.

Managing Treatment Side Effects. Antioxidants as Part of Your Cancer Diet. Antioxidant sources. Antioxidants include: Vitamin C ascorbic acid According to the National Cancer Institute NCI , vitamin C may protect against cancer of the oral cavity, stomach, and esophagus and may also reduce the risk of developing cancers of the rectum, pancreas, and cervix.

In addition, the production of ROS can be used therapeutically for cancer therapy. In this chapter, we described the role of ROS in cancer development and treatments. It has been appraised that in one human cell is exposed to nearly 10 5 oxidative hits such as hydroxyl radical and other such species in a day.

Constant change of genetic material resulting from these oxidative damage incidents demonstrates the initial step of carcinogenesis involved in aging and mutagenesis.

The mechanisms such as specific and nonspecific repair play an important role in the removal of DNA changes by free radicals. It has been documented that base-excision is one method of repair DNA base damage.

The alterations, including base deletion and substitution, play a role of in the DNA damage misrepairing and carcinogenesis. Mutagenic potential is directly equal to the number of oxidative DNA changes that flee repair.

It is known that repair mechanisms decline with age and so DNA damages accumulate with age. The subsequence specificity of DNA lesion locates modifies the mutation frequency.

The particular mechanism by OS which helps to the expansion of carcinogenesis is mainly unknown. However, two distinct mechanisms are supposed to act an important role in the expansion of oxidative and carcinogenesis.

The modulation of gene expression by oxidative damage, can affect carcinogenesis. The epigenetic effects on gene expression could lead to the stimulation of proliferation and growth signals. Chromosomal rearrangements are speculated to result from loss of heterozygosity, alterations in gene expression, contributing to genetic amplifications and strand breakage misrepair, which in turn may advance neoplastic progression.

Active oxygen species have been shown to motive poly ADP ribosylation and protein kinase pathways, thus affecting signal transduction pathways. This can lead to modulation of the expression of necessary genes for tumor promotion and proliferation.

One previous study shows that RAS signal transduction pathways play a role in the mediating free radical signaling.

Second, free radicals cause genetic changes, including chromosomal rearrangements and mutations, play a vital role in the beginning of carcinogenesis process.

The oxidative DNA damage leads to a wide range of chromosomal abnormalities, inducing a wide cytotoxicity and stoppage of DNA duplication.

Mutations can happen a failure to arrest in G1, diminishing their capacity to repair damaged DNA. This enhancement in replication errors can begin tumor suppressor gene inactivation and additional oncogene activation, eventually contributing to malignancy.

Free radical-induced cytotoxicity may also help the beginning of carcinogenesis by promoting the clonal expansion of more resistant-initiated cells depleting the normal cell population, then increasing the possibility of mutation through incorrect replication or due to misrepair, while chromosomal rearrangements can end strand breakage misrepair.

The initiation potential of oxidants might help to induce carcinogenesis as a result of their ability to cause DNA base alterations in tumor suppressor genes and certain oncogenes.

Researches have shown that the radicals especially hydroxyl radicals are able to activate some oncogenes, including C-Raf-1 and K-ras. On the one hand, the activation launches through N-terminal deletions in these genes and the induction of DNA point mutations in GC base pairs. On the other hand, the base point mutations in CpG dinucleotides are also mostly found in specific tumor suppressor genes, including retinoblastoma and p53, which leading to their inactivation.

It is shown that cells containing mutant or absent p53 are attacked by hydroxyl radical, which leading to a failure to arrest in G1 stage, diminishing their ability to repair damaged DNA.

This enhancement in replication errors can initiate tumor suppressor gene inactivation and additional oncogene activation, eventually contributing to malignancy.

Free radical-induced cytotoxicity may also contribute to the initiation of carcinogenesis by promoting the clonal expansion of more resistant-initiated cells, depleting the normal cell population, then increasing the likelihood of mutation [ 1 ].

ROS-caused DNA lesion may be characterized both structurally and chemically and displays a typical schema of modifications.

The free radicals-induced DNA lesion was detected in the various cancer tissues. Most of these alterations can be modified in the in vitro situation.

The figures of DNA lesion induced through ROS experimentally include production of base-free sites, modification of all bases, frame shifts, deletions, DNA-protein cross-links, strand breaks, and chromosomal rearrangements.

The Fenton chemistry mechanism is one of the reactions involved in DNA damage through the generation of hydroxyl radical form. It is well known that hydroxyl radical responds with all ingredients of the DNA molecule: the pyrimidine bases and purine.

Regarding oxidative DNA lesion, main concern has centralized on repair to bases of DNA, with over 20 yields known, but only a few have been investigated with more details. Provided OH-adduct radicals of DNA bases are produced through additional reactions, the carbon-centered sugar radicals and allyl radical of thymine are formed from abstraction reactions.

Peroxyl radicals are generated in environments full of oxygen through oxygen addition to OH-adduct radicals and also to carbon-centered radicals at diffusion controlled rates.

Further reactions of base and sugar radicals generate a variety of sites, modified bases and sugars, protein of DNA, strand breaks, and cross-links.

Hydroxyl radical attacks to pyrimidines: to the C5 and C6 site of cytosine and thymine, generating C5-OH- and C6-OH-adduct radicals.

Oxidative reactions of the C5-OH-adduct radicals of thymine and cytosine followed by release of proton deprotonation and addition of OH or water lead to the generation of glycols of cytosine and thymine.

Oxygen adds to C5-OH-adduct radicals to produce 5-hydroxyperoxyl radicals that may remove superoxide followed by reaction with water, giving rise to cytosine glycol and thymine glycol.

Oxidation of the allyl radical of thymine generates 5- hydroxymethyl uracil 5-OHMeUra and 5-formyluracil. In the lack of O 2 , 6-hydroxyhydropyrimidines and 5-hydroxyhydroare generated by reduction of 6-OH- and 5-OH-adduct radicals of pyrimidines, respectively.

Hydroxyl radical is as well as capable to attacks to purines giving rise to C4-OH-, C5-OH-, and C8-OH-adducts. One electron oxidation and one electron reduction of C8-OH-adduct radicals yield formamidopyrimidines and 8-hydroxypurines 7,8-dihydrooxopurines.

The most studied of these oxidized DNA products is 8-oxo-deoxyguanosine 8-oxo-dG , mainly because it is the most detectable. This base ornamentation falls out in nearly one in 10 5 guanidine residues in a healthy human cell. Therefore, 8-OH-G is mostly named 8-oxoG or 8-oxyhydroguanine.

The nucleoside is thereupon named 8-oxohydrodeoxyguanosine or 8-oxo-dG so, 8-OH-dG and 8-oxo-dG are the identical compounds. Several methods for evaluating oxidative DNA damage exist; a favorite method engages enzymatic digestion of DNA, which releases 8-hydroxypurines for analysis by HPLC usually with electrochemical detector.

Another method uses acidic hydrolysis of DNA, which releases the free base, because the glycosidic bond is cleaved by acid.

Measurement is through HPLC or, transformation to volatile compounds, through GC-MS. The 8-oxoG damage is main due to it is relatively simply generated and is mutagenic, thus is a main indicator for the detection of carcinogenesis.

The studies suggested mutagenic potential of 8-oxo-dG is supported by insertion of adenine opposite the lesions, or a loss of base pairing specificity, misreading of adjacent pyrimidines.

Former studies have shown that the mispairing of 8-oxo-dG with adenine appears to be feasible due to the energetically favored syn glycosidic conformation, while coupling with dG assumes the antiform. These data propose that the way of life might remarkably affect the level of oxidative lesion.

The generation of 2-oxy-dA in the nucleotide unite is another mechanism of mutations. While major consideration has centralized on direct DNA lesion by oxygen free radicals because of the genetic outcomes of such lesion, reactive radical species may also induce damage to other cellular members.

Phospholipids in the cell membrane are extremely susceptible to oxidative process and have been discovered to be repeated targets of radical-caused injury that supply them to be involved in free radical chain reactions.

Several of the fatty acids are polyunsaturated, have a methylene group between two double bonds that predisposes the fatty acid more susceptible to oxidation. In addition, it is reported that polyunsaturated fatty acids at high concentration in phospholipids predisposes play a role of in the free radical chain reactions.

Linoleic acid is the most common fatty acid in cell membranes. A set of arachidonic acid oxidation products termed isoprostanes is the best biomarker of lipid peroxidation that generally detected through GC-MS.

The first products of unsaturated fatty acid oxidation are short-lived lipid hydroperoxides. When they react with metals, they produce some of products for example epoxides and aldehydes, which are themselves reactive. Malondialdehyde MDA is one of the important aldehyde products through lipid peroxidation.

This product of lipid peroxidation is mutagenic and carcinogenic in mammalian cells and animals, respectively. MDA can react with DNA bases dA, dC, and dG, to form adducts, M 1 A, M 1 C, and M 1 G. M 1 G has been indicated in the several tissues such as pancreas, liver, and breast.

The M 1 G content corresponds nearly to adducts in cell. Many researches have shown that M 1 G is an electrophile in the genome. N 2 -Oxo-propenyl-dG, as a yield of quantitative and rapid ring-opening of M 1 G, is as well as electrophilic, but aims regions of DNA distinct from M 1 G.

Therefore, the conversion of M 1 G and N 2 -oxo-propenyl-dG may unfold varying reactive groups of DNA that could take part in the production of DNA-DNA inter-strand cross-links or DNA-protein cross-links. It has been shown that hydroxypropanodeox-oguanosines OH-PdGs are exist in rodent and human liver DNA.

It has been proposed that these propano adducts are interceded by the reaction of DNA with crotonaldehyde and acrolein, which in turn are products of lipid peroxidation.

Crotonaldehyde and acrolein are mutagenic in mammalian cells and bacteria. There is a few information associated with the repair of OH-PdGs. Studies show that PdG is a main substrate for the nucleotide cut repair complex of mammalian cells and E.

coli and is identified and repaired through the mismatch repair system. Various exocyclic etheno DNA adducts increasing from lipid peroxidation have been found in DNA from healthy human volunteers. The most important involves etheno-dG, etheno-dC, and etheno-dA.

Etheno-dC and etheno-dA are found to be strongly genotoxic but weakly mutagenic [ 3 ]. The interaction between different ROS levels in cancer cells. In cancer cells, ROS at low to moderate levels induces cell proliferation and cell survival, at high level induce cell damage and at an excessive level induce cell death.

The findings from both in vitro and in vivo studies have shown that endogenous oxidative stress in cancer cells is higher than normal cells. ROS might function as a double-edged sword and as varied ROS levels could cause various biological responses.

A low to moderate raise of ROS may help with the proliferation and survival of cells. But, at a high level, ROS may suppress the antioxidant capacity of the cell and start cell death Figure 1. On the other hand, at the accumulation of ROS, these cells may be more sensitive than normal cells.

The normal cells at under physiological status play an important role in maintaining redox homeostasis with a low level of basal ROS by controlling the balance between pro-oxidants and antioxidant capacity.

The physiological conditions are affected by ROS inducers such as hypoxia, metabolic defects, ER stress, and oncogenes and ROS elimination such as NRF2, glutathione, NADPH, tumor suppressors, and dietary antioxidant agents [ 4 , 5 ].

The increase of ROS level of intracellular by activating signaling pathways in cancer cells represents that these cells very more vulnerable than normal cells to ROS-caused cell death. As a result, these cells in comparison to normal cells very more dependent on the capacity of the antioxidant system and more vulnerable to major oxidative stress induced through exogenous ROS-generating agents or compounds that inhibit the antioxidant system.

This might constitute a biochemical basis to plan therapeutic strategies to selectively death cancer cells using ROS-moderated mechanisms [ 4 — 6 ]. As described above, the increase of ROS in cancer cells was induced several biological responses.

These biological responses including adaptation, increase in cellular proliferation, cell damage, and cell death are likely to be dependent on the cellular genetic background, the types of the specific ROS involved, and the levels of ROS at the duration of the oxidative stress [ 7 ].

The regulation of ROS level by ROS inducers and ROS scavengers. Oxidative stress plays an important role in cell signaling as a sensor and regulator.

It was reported that a lot of regulator agents have a considerable effect on up-expression and down-expression of antioxidant genes. In the following, we explain some of the major factors that act directly in the expression of antioxidant genes Figure 2.

On the other hand, good understanding of the particular pathways that are affected by these regulators is important before designing therapeutic approaches to the adjustment of ROS levels [ 4 ].

NRF2 is an important regulator of the antioxidant system and cellular stress responses in the several cancers. From the support on the function of Nrf2 target genes, one can easily conclude that activation of Nrf2 may protect cells from several stresses imposed through toxic exposure.

Actually, it is recognized that NRF2 regulate various anti-oxidative stress responses and for detoxification reactions, its expression in the tissues increases [ 8 , 9 ].

NRF2 adjust the common various different antioxidant pathways such as GSH production and regeneration, GSH utilization, NADPH production, thioredoxin TXN production, regeneration and utilization, Quinone detoxification and Iron sequestration Figure 2.

It is directly through GSH metabolism and indirectly controlling free Fe II homeostasis involved in ROS detoxification. NRF2 decreases the generation of harmful hydroxyl radicals from ROS by increasing the release of Fe II from haem molecules [ 4 ].

It was suggested that phytochemical compounds such as dietary and medicinal plants through the effect on NRF2 pathway played a key role in cancer therapy [ 8 , 9 ].

FOXO, as a transcription factors, are involved in different signaling pathways and play key roles in some physiological and pathological processes such as cancer. It could play and act as a self-regulatory mechanism, which protects cells from an oxidative damages, via keep in good condition a balance of ROS and antioxidant productions.

FOXO and p53 as a tumor suppressor have a key role in inhibiting oxidative stress process through inducing antioxidant gene expression [ 4 ]. It was reported that an increase of ROS level leads to up-regulation of anti-oxidative proteins, such as MnSOD and catalase through FOXO3a- and FOXO1 [ 10 ].

The p53, as a final transcription factor, has an important role in regulating antioxidant gene expression is p53 and a double-edged as a pro- and antioxidant role in ROS controlling. Hypoxic conditions caused by the imbalance between intake and oxygen consumption [ 4 ].

The production of ROS through the mitochondrial complex I and III, xanthine oxidase, and NADPH oxidase related to hypoxia is recognized as one of the most harmful causes of oxidative process. Some studies have shown that hypoxia condition-caused superoxide generation occurs through the activation of NADPH oxidase placed in the cell membrane and under moderate condition, NO is generated in mitochondria.

Studies suggested that hypoxia-induced loss in mitochondria membrane potential and this and this event is related to raising ROS [ 11 ]. Studies have shown that the mitochondria complex III at the Qo site at during the transfer of electrons from ubisemiquinone to molecular oxygen is the main source for ROS generation under hypoxia.

In addition, it has shown that activation the transcription factor hypoxia-inducible factor HIF dependent on ROS level. HIFs regulate physiological responses to hypoxia, such as pathophysiological processes especially in cancer [ 4 ]. It was reported that, there is a relation between an increase in the level of H 2 O 2 generation of mitochondria and an increase in susceptible cancer cells to apoptosis.

This susceptibility caused cytotoxicity and also to oxidative stress-induced apoptosis when compared to normal cells [ 12 ].

In the cancer cells, oxidative stress due to increase ROS level and decrease antioxidant level is higher than normal cells. At a time when equivalent levels of oxidative stress are added by the administration of exogenous ROS-inducing agents, oxidative stress levels in cancer cells but not normal cells can readily over the threshold of cell death.

Hence, cancer cells in compared normal cells are expected to be more vulnerable to cell damage caused by ROS-inducing agents and this vulnerability can be exploited to selectively kill these cells [ 11 ]. A model for increase ROS and apoptosis signaling by oncogenes such as c-MYC.

The activation of MYC in cancer cells leads to an elevating in intracellular ROS through mechanisms such as changes in scavenging process, metabolic rate, and eventual activation of intracellular oxidases Figure 3.

The previous investigations show that the increased expression of c-MYC and E2F1 induces accumulation of ROS and increases ROS by c-MYC and E2F1 sensitizes host cells to apoptosis. Hypoxia, on the one hand, as a ROS inducer, can also directly induce the raised expression of oncogenes such as MYC and RAS through HIF-2α.

On the other hand, MYC increases mitochondrial biogenesis, which adds to the raised ROS generation under hypoxic conditions and the raised mitochondrial ROS generation elevate the oxidative stress process [ 12 ].

It has been suggested that RAS, p53, and c-MYC through the mitochondria to regulate ROS generation, thereby affecting apoptosis. RAS in K-Ras-transformed fibroblast cell under the condition that glucose deprivation induces apoptosis through changes in mitochondrial complex gene expression.

It has also been reported that RAS in p66SHC overexpression condition in the transformed cells increase ROS generation through mitochondria and p53, MPTP opening and mitochondrial swelling could induce apoptosis Figure 3 [ 12 ].

In recent decades, several studies have shown that mitochondria organelle plays an important role in human health and disease. These studies led to the emergence of a new field of study named "mitochondrial medicine.

It has been confirmed that these organelles are the principal intracellular source of ROS production in most tissues. Studies have shown that under physiological condition, complex II respiratory chain could also be a main regulator of ROS generation from mitochondria [ 13 ].

In eukaryotic cells, the mitochondrion is an important organelle that plays a main role in several critical processes. The important role of mitochondria is mentioned in the physiology of cancer, such as in energy metabolism and cell cycle regulation. There is powerful documentary evidence to support the rationale for the expansion of anticancer strategies based on mitochondrial targets.

This organelle is recognizing to play an important role in the apoptosis mechanism and initiate cell death through various mechanisms that comprise disrupting electron transport and energy metabolism in the respiratory chain, releasing agents or proteins such as cytochrome c that mediate apoptosis signaling, and changing the cellular redox status by ROS generation.

The therapeutic targeting of cancer cells based on mitochondria have often depended on the intrinsic various differences between mitochondria in normal and cancer cells, which allow for better options, manipulation different pathways, and destruction of these cancer cells.

These differences are including bioenergetics change, disturbance of the mitochondrial DNA mtDNA , and morphological and physiological changes in the cancer cell Table 1.

The mtDNA is one main target of ROS and the lack of sufficient protective histones surrounding the mtDNA in cancer cells makes the mtDNA more easily tending to ROS-caused DNA damage. In addition, various researches have shown that the mitochondria were different in cancer cells than in normal cells, such as grow faster, fewer and smaller, and also had the morphological transformed [ 13 ].

Some differences between cancer and normal cells such as bioenergetics process, MMP and ROS level, and morphological and physiological differences.

Today, several mitochondria targeted strategies for cancer therapy have been focused on the development of agents that manage increased the ROS generation in mitochondria from the cancer cells without effect on the normal cells.

It has been shown that ROS generation in the mitochondria is evaluated through the rates of both mitochondria ROS mtROS disposal and production, and ROS levels in mitochondria are regulated by several agents, including mitochondria O 2 levels, mitochondrial membrane potential MMP or Δψm , the metabolic condition of mitochondria, and other factors Figure 4.

A number of recent researches reveal the fact that mtROS at low to high levels act as several functions. That is it at low levels, involved in the hypoxia adaptation process, at moderate levels, involved in controlling inflammatory response, and at high level involved in regulating apoptosis signaling.

Adjustment of mitochondria ROS mtROS generation. Several agents such as MMP, the metabolic state of mitochondrial, O 2 level and STAT3 adjust the generation of mtROS.

As mentioned in the previous section, the increase of ROS level of intracellular by sever agents through activating signaling pathways in cancer cells represents that these cells are more vulnerable than normal cells to ROS-caused cell death.

The results of the studies suggest that ROS such as H 2 O 2 could affect the extrinsic apoptosis pathway through changing the intracellular space.

The antioxidants found in fruits Almond health supplements vegetables help fight free cancerr, which can lead to cancre cancer. Free radicals and cancer radical are often mentioned raddicals a cause of breast cancerbut what exactly are radicalz radicals? Free radicals are highly reactive and unstable molecules, usually oxygen molecules, but not always. Their unstable nature is caused by having an unpaired electron. As a result of this unpaired electron, free radicals seek out and take electrons from other molecules, which oftentimes causes damage to the second molecule. Like many things that occur in nature, free radicals are not only impossible to avoid, but necessary for life.

Author: Nigul

0 thoughts on “Free radicals and cancer

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com