+ All Categories
Home > Documents > Chronic inflammation and oxidative stress in human...

Chronic inflammation and oxidative stress in human...

Date post: 27-Apr-2018
Category:
Upload: trinhdien
View: 217 times
Download: 3 times
Share this document with a friend
22
Chronic inflammation and oxidative stress in human carcinogenesis Federico A, Morgillo F, Tuccillo C, Ciardiello F & Loguercio C. International Journal of Cancer. Volume 121, Issue 11, pages 2381–2386, 1 December 2007. DOI: 10.1002/ijc.23192 © 2007 Wiley-Liss, Inc. A wide array of chronic inflammatory conditions predisposes susceptible cells to neoplastic transformation. In general, the longer the inflammation persists, the higher the risk of cancer. A mutated cell is a sine qua non for carcinogenesis. Inflammatory processes may induce DNA mutations in cells via oxidative/nitrosative stress. This condition occurs when the generation of free radicals and active intermediates in a system exceeds the system's ability to neutralize and eliminate them. Inflammatory cells and cancer cells themselves produce free radicals and soluble mediators such as metabolites of arachidonic acid, cytokines and chemokines, which act by further producing reactive species. These, in turn, strongly recruit inflammatory cells in a vicious circle. Reactive intermediates of oxygen and nitrogen may directly oxidize DNA, or may interfere with mechanisms of DNA repair. These reactive substances may also rapidly react with proteins, carbohydrates and lipids, and the derivative products may induce a high perturbation in the intracellular and intercellular homeostasis, until DNA mutation. The main substances that link inflammation to cancer via oxidative/nitrosative stress are prostaglandins and cytokines. The effectors are represented by an imbalance between pro-oxidant and antioxidant enzyme activities (lipoxygenase, cyclooxygenase and phospholipid hydroperoxide glutathione- peroxidase), hydroperoxides and lipoperoxides, aldehydes and peroxinitrite. This review focalizes some of these intricate events by discussing the relationships occurring among oxidative/nitrosative/metabolic stress, inflammation and cancer. Introduction General concepts on carcinogenesis Carcinogenesis is a long and multistep process that includes initiation (selection of a mutated cell), promotion (selective expansion of the initiated cell) and progression as a consequence of an imbalance between cell proliferation and cell death. More genetic and epigenetic events are required to drive from initiated cells to malignant tumors, each conferring one or another type of growth advantage, and leads to the progressive conversion of normal human cells into cancer cells. The establishment of a mutation is a sine qua non of cancer. Most notable among proneoplastic mutations are those that result in increased expression of oncogenes (e.g., myc, ras, abl, bcl-2) or decreased activity of tumor-suppressor genes1 (e.g., p53, Rb), conferring a selective growth or survival advantage to the cell. Phenotypic changes representative of preneoplastic mutations include a decreased need for metabolites and growth factors, abnormal
Transcript

Chronic  inflammation  and  oxidative  stress  in  human  carcinogenesis  

Federico A, Morgillo F, Tuccillo C, Ciardiello F & Loguercio C. International Journal of Cancer. Volume 121, Issue 11, pages 2381–2386, 1 December 2007. DOI: 10.1002/ijc.23192 © 2007 Wiley-Liss, Inc.

A wide array of chronic inflammatory conditions predisposes susceptible cells to neoplastic transformation. In general, the longer the inflammation persists, the higher the risk of cancer. A mutated cell is a sine qua non for carcinogenesis. Inflammatory processes may induce DNA mutations in cells via oxidative/nitrosative stress. This condition occurs when the generation of free radicals and active intermediates in a system exceeds the system's ability to neutralize and eliminate them. Inflammatory cells and cancer cells themselves produce free radicals and soluble mediators such as metabolites of arachidonic acid, cytokines and chemokines, which act by further producing reactive species. These, in turn, strongly recruit inflammatory cells in a vicious circle. Reactive intermediates of oxygen and nitrogen may directly oxidize DNA, or may interfere with mechanisms of DNA repair. These reactive substances may also rapidly react with proteins, carbohydrates and lipids, and the derivative products may induce a high perturbation in the intracellular and intercellular homeostasis, until DNA mutation. The main substances that link inflammation to cancer via oxidative/nitrosative stress are prostaglandins and cytokines. The effectors are represented by an imbalance between pro-oxidant and antioxidant enzyme activities (lipoxygenase, cyclooxygenase and phospholipid hydroperoxide glutathione-peroxidase), hydroperoxides and lipoperoxides, aldehydes and peroxinitrite. This review focalizes some of these intricate events by discussing the relationships occurring among oxidative/nitrosative/metabolic stress, inflammation and cancer.

Introduction

General concepts on carcinogenesis

Carcinogenesis is a long and multistep process that includes initiation (selection of a mutated cell), promotion (selective expansion of the initiated cell) and progression as a consequence of an imbalance between cell proliferation and cell death. More genetic and epigenetic events are required to drive from initiated cells to malignant tumors, each conferring one or another type of growth advantage, and leads to the progressive conversion of normal human cells into cancer cells. The establishment of a mutation is a sine qua non of cancer. Most notable among proneoplastic mutations are those that result in increased expression of oncogenes (e.g., myc, ras, abl, bcl-2) or decreased activity of tumor-suppressor genes1 (e.g., p53, Rb), conferring a selective growth or survival advantage to the cell. Phenotypic changes representative of preneoplastic mutations include a decreased need for metabolites and growth factors, abnormal

signal transduction, inappropriate expression of receptors for available growth factors (epidermal growth factor receptor, HER2/neu), dysregulation of cell-cycle checkpoints and resistance to apoptosis.1 At this point, any agent that causes increased cell proliferation increases the risk of neoplastic transformation. The damage to the DNA must survive the many DNA-repair processes and must be readable by DNA polymerase, which creates and locks in the mutation. If, by chance, this damage to the DNA results in a selective growth or survival advantage to the cell, it may become a precancerous lesion. On the other side, while normal cells have the ability to turn on and off transiently the genes that help them survive toxic signals (e.g., induction of P450 enzymes for metabolism of toxic chemicals and drugs), under circumstances of prolonged stress, such as during chronic inflammation, a mutation may lock in the growth-advantaged phenotype. Hence, prolonged exposure to stress can result in selection of preneoplastic cells. Some examples of adaptive changes in a cell are increased expression of antioxidant enzymes, matrix metalloproteinases and growth factor receptors; increased anaerobic respiration; and de novo synthesis of angiogenic factors. Studies in experimental tumor models suggest that the inhibition of angiogenesis can impede tumor growth and metastasis and can cause preformed tumors to necrotize and regress.

The role of inflammation on carcinogenesis

There is a large series of reviews that extensively demonstrate the direct relationship between chronic inflammation and cancer.2, 3, 4, 5, 6, 7, 8, 9, 10

In fact, a wide array of chronic inflammatory conditions predispose susceptible cells to neoplastic transformation.11 In general, the longer the inflammation persists, the higher the risk of cancer. Inflammation is a step-by step process that includes injury, repair and resolution. All inflammatory cells (neutrophils, monocytes, macrophages, eosinophils, dendritic cells, mast cells and lymphocytes) are recruited after a damage or an infection, and may contribute to the onset and progression of cancer. Key molecular players that link inflammation to genetic alterations are prostaglandins, cytokines, nuclear factor NFkB, chemokines and angiogenic factors. The main chemical effectors are free radical species derived from oxygen (ROI) and nitrogen (RNI). Free radicals may act as direct or indirect damaging agents through their reaction with other chemical or structural components in cells. ROI and RNI also recruit other inflammatory cells with secondary amplification of the damage. In this review, we describe the role of free radicals and of oxidative stress consequent to a chronic inflammation on the induction and progression of cancer.

Free radicals as a link between inflammation and cancer

General concepts on free radicals and oxidative stress

The main sources of reactive species in all cells are mitochondria, cytochrome P450 and peroxisome. Under physiological conditions, there is a constant endogenous production of reactive intermediates of ROI and RNI that interact as “signaling”

molecules for metabolism, cell cycle and intercellular transduction pathways.12, 13 The balance between beneficial and/or harmful effects of intermediate species is the crucial event in living organisms. In fact, the redox homeostasis is, in vivo, the main protective process from cell death. To control the balance between production and removal of ROI and RNI, there are a series of protective molecules and systems globally defined as “antioxidant defences.”.These include enzymes such as superoxide dismutase, catalase, glutathione peroxidase and glutathione-S-transferase, proteins that sequester transition metals, glutathione (GSH), cysteine, thioredoxin, vitamins, etc.14 Oxidative stress occurs when the generation of free radicals and active intermediates in a system exceeds the system's ability to neutralize and eliminate them.15, 16, 17 At the moment, the concept of oxidative stress originally confined to ROI—such as hydroxyl and superoxide radicals, and hydrogen peroxide and singlet oxygen—has been extended onto reactive nitrogen species (RNI) as nitric oxide (NO), peroxynitrite and, recently, S-nitrosothiols.13 Therefore, the current concept of “oxidative stress” should also include the pathways related to the “nitrosative stress” and, for their implication in cellular and extracellular metabolic events, to the “metabolic stress.” In these conditions, ROI and RNI act as “toxic” substances that may react with proteins, carbohydrates and lipids, with consequent alteration both in the intracellular and intercellular homeostasis, leading to possible cell death and regeneration (see Fig. 1).

Figure 1. Reactions of ROI and RNI with proteins, carbohydrates and lipids, with consequent alteration both in the intracellular and intercellular homeostasis until possible cell death and regeneration.

   

Sources of free radicals during inflammation

Inflammatory cells secrete a large number of cytokines and chemokines that promote the outgrowth of neoplastic cells, in addition to the autocrine growth factor production by the tumor cells themselves. ROI and RNI are produced under the stimulus of

proinflammatory cytokines in phagocytic and nonphagocytic cells through the activation of protein-kinases signaling. For example, TNF-α enhances the formation of ROI by neutrophils and other cells, while interleukin-1-β (IL-1-β), TNF-α and interferon (IFN)-γ stimulate the expression of inducible nitric oxide synthase in inflammatory and epithelial cells. In experimental animals, pristane-induced plasma cell tumors require IL-6 for their growth, and it is provided by macrophages in the chronically inflamed tissue. Production of IL-6 in this system is stimulated by PGE2 derived from cyclooxygenase-2 (COX-2), which is elevated in inflammatory macrophages; this process is inhibited by the administration of indomethacin, a potent antiinflammatory drug.18, 19 Knockout of the TNF-α gene in mice significantly inhibits the development of skin tumors in response to DMBA and phorbol esters.20 IL-8, an inflammatory chemokine derived from monocytes, macrophages and endothelial cells, is implicated in the progression and metastasis of tumors in the colon, bladder, lung and stomach.21

Chronic inflammation is closely associated with angiogenesis.22 Macrophages, platelets, fibroblasts and tumor cells themselves are a major source of angiogenic factors such as basic fibroblast growth factor, vascular endothelial growth factor, prostaglandins-1 and -2 other than inflammatory cytokines, chemokines and NO.21 There is a significant interaction and synergy among these mediators of inflammation until cancer. As an example, it has been demonstrated that prostaglandins, derived from the oxidative metabolism of arachidonic acid in inflammatory cells, contribute to the development of cancer.23, 24 Prostaglandins induce the expression of certain inflammatory cytokines, which can, in turn, enhance the production of ROI and RNI. COX-2 is the main enzyme that responds to inflammation through the synthesis of prostaglandins in monocytes and macrophages; however, COX-2 is also expressed in noninflammatory cells such as fibroblasts, epithelial and endothelial cells. In vitro, the expression of COX-2 is induced by bacterial cell products and inflammatory cytokines.25, 26 COX-2 is known to be involved in cancer biology, and it is upregulated in many cancer types including carcinomas of the colon, breast, lung, pancreas, esophagus, and head and neck.27, 28 Studies from COX-2 transgenic mice and COX-2 knockout mice confirm that COX-2 plays a role in colonic cancer development, both through angiogenesis29, 30 and through the activation of different oncogenes, including v-src, v-Ha-ras, HER-2/neu and Wnt.31 Notably, prostaglandin synthesis can also be stimulated by peroxynitrite,32 a toxic radical derived from NO.

Free radicals may react with phospholipids of membranes generating hydroperoxides, lipoperoxides and toxic aldehydes such as malondialdehyde (MDA), which in turn may alter membrane permeability and microcirculation. These last events may induce a further adhesion of granulocytes to endothelium that may activate the xantine–xantine–oxidase pathway with an enhanced production of hydroperoxide in a vicious circle. ROI, RNI and their derivative products may also activate nuclear factors such as, for example, NFkB, leading to the production of other amount of proinflammatory cytokines, which in turn enhance inflammation and, therefore, the generation of other reactive species.33 The imbalance in redox status and the enhanced production of intermediate reactive species progressively consumes the antioxidant defences, leading

the cells to develop oxidative stress. Cells could respond to these insults by enhancing their antioxidant potential or by activating the system of caspases that induce programmed cell death (apoptosis). It is also possible that the oxidative stress induces cell necrosis through the release of cytochrome c and the depletion of ATP at mitochondrial level.34, 35 Peroxynitrite participates with the apoptotic program through the nitrosylation of proteins, which in turn alters the function of several signaling molecules including NF-KB, AP-1, p53 and caspases.21, 36, 37 When the system fails in 1 or more than 1 step, the possibility to have a mutated cell increases as a consequence of the high perturbation in the redox status that progresses, through ROI and RNI, into the nucleus. Figures 2 and 3 summarize these pathways.

Figure 2. Role of oxidative/nitrosative/metabolic stress in carcinogenesis.

Figure 3. The main steps that link inflammation to cancer via free radical-mediated processes.

It is now known that many different oncogenes act by inhibiting apoptosis, thereby conferring a survival advantage to preneoplastic and malignant cells. On the other

hand, several chemotherapeutic drugs exert their action of cell death promotion by increasing ROI production.38, 39

The start of carcinogenesis mediated by ROI and RNI following chronic inflammation may be direct (oxidation, nitration, halogenation of nuclear DNA, RNA and lipids), or may be mediated by the products of ROI-RNI and proteins, lipids and carbohydrates that are capable of forming DNA adducts. ROI can also increase the expression of transcriptional factors including c-fos and c-jun oncogenes involved in neoplastic transformation.

Direct effect of free radicals on DNA damage and carcinogenesis

To date, more than 100 oxidized DNA products have been identified. ROI-induced DNA damage involves single- or double-stranded DNA breaks; purine, pyrimidine or deoxyribose modifications; DNA intrastrand adducts; and DNA protein crosslinks.40 DNA damage can result in either arrest or induction of transcription, induction of signal transduction pathways, replication errors and genomic instability, all processes associated with carcinogenesis.41, 42 NO reacts with superoxide (O2

−) to form peroxynitrite (ONOO−), a highly reactive species that induce nitrative and oxidative DNA damage. In fact, ONOO− mediates the formation of 8-oxo-7,8-dihydro-2′-deoxyguanosine43 and 8-nitroguanine,44, 45 which has also been suggested as a potential biomarker of inflammation-related carcinogenesis.42, 46 Recent evidences indicate that 8-nitroguanosine is a highly redox-active molecule47, 48 and that 8-nitroguanine is a mutagenic substance, which preferentially leads to G>T transversions.44, 49 Indeed, G>T transversions have been observed in vivo in the ras gene50 and the p53 tumor suppressor gene, in lung and liver cancer.51, 52 These findings indicate that the DNA damage mediated by ROI and RNI may participate to carcinogenesis, both via activation of proto-oncogenes and inactivation of tumor suppressor genes. In the carcinogenesis also, the oxidative damage of mitochondria is strongly involved.42 In fact, hydrogen peroxide and other reactive oxygen species activate nuclear genes that regulate the biogenesis, transcription and replication of the mitochondrial genome. Although the rate of tumor cells that possess mutated mitochondrial DNA and the cancer processes to which mitochondrial DNA alterations participate have not been satisfactorily established, a significant amount of information support the involvement of the mitochondria in carcinogenesis.42 Fragments of mitochondrial DNA have been found to be inserted into nuclear DNA, suggesting a possible mechanism of oncogene activation. Mitochondrial DNA (mtDNA), which is bound to the inner mitochondria membrane, is particularly liable to oxidative damage because of the lack of histone proteins, the presence of incomplete repairing mechanisms53, 54, 55 and its proximity to the mitochondrial electron transport chain. Damage to mtDNA causes mitochondrial respiratory chain dysfunction, which in turn increases the production of hydroxyl radicals which are the major source of oxidative damage for DNA.56, 57, 58 MtDNA also encodes essential proteins involved in the phosphorylation processes,59 and its damage is associated with a cascade of reactions that amplify the oxidative stress.60, 61, 62, 63 Mutations and altered expression in mitochondrialgenes encoding for complexes I, III, IV and V,

and in the hypervariable regions of mitochondrial DNA, have been identified in various human cancers.64, 65

The role of ROI- and RNI-derived products on carcinogenesis

Free radicals immediately react with all components of cells by forming stable products. Proteins are more susceptible to oxidation by free radicals. In the global cellular economy, the oxidation of SH groups of cysteine reduces the activity of various enzymes as well as the synthesis of GSH, which is the main intracellular free radical scavenger.

The oxidation of lipids induces the formation of aldehydes and lipid peroxides. These molecules, at very low and nontoxic concentrations, act as signaling transductors of ROI-mediated metabolic reactions, and therefore, they modulate several cell functions including gene expression and cell proliferation.66 In high concentrations, lipid-derived products are considered the more damaging species because they easily react with proteins, DNA and phospholipids, generating a variety of intra- and intermolecular toxic covalent adducts that lead to the propagation and amplification of oxidative stress.67 This has been demonstrated in human cancerogenesis.68, 69, 70 Lipid hydroperoxides are formed in vivo through the action of ROI on polyunsaturated fatty acids, both directly and as specific products of lipoxygenase (LOX) and COX activities.67 Cell membranes are the main target for lipid peroxidation. Lipid peroxides cause cell damage by its decomposition in breakdown toxic products that are bifunctional aldehydes,71 which are relatively stable and able to diffuse in and outside the cell. The most abundant ROI–lipid-derived product is—in conditions of oxidative stress—the 4-hydroxynonenal (HNE). At low levels it promotes cell proliferation, while at a higher concentration it induces oxidative alterations of DNA72 and apoptosis.73, 74, 75 Therefore, 4-HNE is directly involved in cell cycle control,76 and in human cancer, it causes mutation in the p53 gene expression.68 4-HNE forms etheno adducts with DNA and upregulates COX-2 expression.77 It has previously been stressed that COX2 expression induces an enhanced production of ROI. The breakdown of membrane lipid hydroperoxides also increases the levels of MDA,78 which can react with DNA bases G, A, and C by forming adducts M1G, M1A and M1C, respectively.79 M1G adducts have been found in human tissues at levels as high as 1.2 adducts per 106 bases (which corresponds to ∼6,000 adducts per cell). M1G has been detected in human breast tissue by32phospho-postlabeling as well as in rodent tissues.80 Moreover, M1G has been demonstrated to be mutagenic in Escherichia coli, through the transversions to T and transitions to A.81, 82 There are also other exocyclic DNA adducts that arise from lipid peroxidation. For example, etheno-dA, etheno-dC and etheno-dG have been detected by both32phospho-postlabeling and GC–MS.83 It has been demonstrated that etheno-dA and etheno-dC are strongly genotoxic but weakly mutagenic when introduced on single-stranded vectors in E. coli. In addition, hydroxypropanodeoxyguanosines are present in human DNA in presence of cancer.41 These adducts are most probably derived from the reaction of DNA with acrolein and crotonaldehyde generated by the lipid peroxidation process.

Two enzymes, with opposed activities, control the peroxidation of cell membranes. These are LOXs and the phospholipid hydroperoxide glutathione-peroxidase (PH-GPx). The regulatory relation of these enzymes is of crucial importance to maintain a normal redox status in cells. An imbalance in this regulation leads to the generation of cytotoxic molecules that are involved in carcinogenesis.71 LOXs catalyze the specific dioxygenation of polyenoic fatty acids, forming reactive fatty acid hydroperoxides. PH-GPx is a member of the GPx enzymes family, a selenium-dependent protein family that contains selenocysteine at its active site, and it has the ability to reduce organic and inorganic hydroperoxides by utilizing GSH as a reducing agent.84

Among the three 12-LOX forms that have been identified, both the leukocyte and platelet types have been found in different cancer tissues, including melanoma, prostate and epidermal cancers.85 Inhibition of LOXs induces apoptosis and blocks the proliferation in carcinosarcoma cells.86 The expression of 12-LOX also correlates with tumor cell metastasis. Clinically, the degree of 12-LOX expression in human prostate cancer correlates with the tumor grade and stage, and its expression level is higher in metastasic prostate cancers than in nonmetastatic tumors.87 Some studies have suggested that 15-LOX enhances colonic tumorogenesis by potentiating the cell proliferation and the mitogenic response to EGF.88, 89 12/15 LOX, that is the oxidative counterpart of PH-GPx, is transcriptionally regulated by ILs. In a variety of human cells, the expression of the human 12/15-LOX is upregulated when cells are exposed to IL-4. Other cytokines, such as IFN-γ, IL-6 and IL-10 did not induce the enzyme.90, 91 Experiments with human epidermoid carcinoma A431 cells that were transfected with the cDNA sequence of cytoplasmic PH-GPx revealed that PH-GPx downregulates the 12-LOX activity.92 In animals, platelets 12-LOX, 15-LOX and COX-2 are more sensitive to PH-GPx inhibition than do 5-LOX and COX-1, which are more resistant.93 This inhibition is probably caused by a requirement of fatty acid hydroperoxides for the activation of LOX and COX that are regulated by its own products.94, 95 These results support the hypothesis that cytoplasmic PH-GPx plays a pivotal role in the regulation of arachidonate metabolism. On the other hand, inhibition of PH-GPx activity with the treatment of cells with an antisense oligonucleotide of PH-GPx mRNA increases the enzymatic activity of 12-LOX in A431cells.96 This interplay can be further complicated by stimulation with ILs. When the human lung carcinoma cell line A549 is exposed to IL-4 and 13, an upregulation of the 12/15-LOX and a downregulation of the PH-GPx are observed.90, 91 Mitochondrial PH-GPx protects from cell death induced by phospholipid hydroperoxidation triggered by photodynamic therapy.97 Likewise, 2 arsenic compounds, arsenic trioxide and arsenic disulfide, employed against acute promyelocytic leukemia, induce apoptosis in leukemia cells98 by inhibiting PH-GPx expression.99

In cancer tissues, a great part of the increased lipid peroxidation could be triggered by increased arachidonic acid metabolism, as a result of elevated COXs. On the other hand, MDA can be produced by isomerization of prostaglandins PGH2. Therefore, a high interplay exists among PHGPx, 12/15LOX and COX-2 in cancerogenesis mechanisms. Figure 4 summarizes the role of free radicals on carcinogenesis.

Figure 4. The role of free radicals on carcinogenesis.

Conclusive remarks

ROI and RNI participate in various redox-regulatory mechanisms of cells in order to maintain a normal and constant “redox homeostasis.” When the system fails and the antioxidant control mechanisms are exhausted or overrun, the cellular redox potential shifts toward an oxidative and nitrosative stress, which leads to DNA mutations and genomic instability. A chronic inflammation is the main system capable of inducing an oxidative/nitrosative stress in a self-perpetuating system. The most widely studied and best established of these links are colon carcinoma associated with inflammatory bowel diseases (chronic ulcerative colitis and Crohn's disease), esophageal adenocarcinoma associated with reflux esophagitis (Barrett's esophagus), hepatitis predisposing to liver cancer, schistosomiasis causing an increased risk of bladder and colon carcinomas and chronic Helicobacter infection leading to cancer of the stomach. Inflammatory cytokines also regulate the progression and metastasis of tumors (colon, bladder, lung and stomach). Examples of tumor cell cytokine dependence in human disease are the growth dependence of AIDS- and EBV-associated B-cell lymphomas, B-cell leukemias and multiple myeloma on the inflammatory cytokines IL-6 and IL-15, and the dependence of malignant mesothelioma on platelet-derived growth factor.100, 101

In animals, indomethacin blocks carcinogenesis by reducing the production of inflammatory cytokines. In humans, epidemiological studies documented a lower cancer risk for people regularly taking nonsteroidal antiinflammatory drugs.102 For example, the use of COX-2 inhibitors, like celecoxib, are now being tested as antitumor agents in clinical trials.103 In fact, this drug reduces the number of colorectal polyps in patients with familial adenomatous polyposis.

On the basis of these considerations, future research should be aimed to precociously discover the preneoplastic tranformation of a chronically inflamed tissue. Lipid peroxides and 8-dehoxiguanosine and guanine appear at the moment as promising tools for quantifying the risk of DNA mutations. The evaluation of the levels of these supposed markers should also be useful to monitor therapies aimed to block the carcinogenic potential of the inflammation-related oxidative and nitrosative cascade.

There are studies on these aspects in humans following a diet rich in antioxidants. Depletion of antioxidants, and in particular of GSH, may sensitize tumor cells to some chemotherapy agents, and many of the GSH-depleting agents have a dose-related toxicity. In this context, the development of nontoxic GSH-depleting agent may be strongly relevant in overcoming multidrug resistance. Finally, future researches are needed to better determine the most rational and effective combination of redox-active and antiinflammatory agents to other routine drugs in the treatment of cancer.

References

• 1  

Hanahan  D,Weinberg  RA.  The  hallmarks  of  cancer.  Cell  2000;  100:  57–70.  

• 2  

Bartsch  H,Nair  J.  Chronic  inflammation  and  oxidative  stress  in  the  genesis  and  perpetuation  of  cancer:  role  of  lipid  peroxidation,  DNA  damage,  and  repair.  Langenbecks  Arch  Surg  2006;  391:  499–510.  

• 3  

Aggarwal  BB,Shishodia  S,Sandur  SK,Pandey  MK,Sethi  G.  Inflammation  and  cancer:  how  hot  is  the  link?  Biochem  Pharmacol  2006;  72:  1605–21.  

• 4  

Lu  H,Ouyang  W,Huang  C.  Inflammation,  a  key  event  in  cancer  development.  Mol  Cancer  Res  2006;  4:  221–33.  

• 5  

Moss  SF,Blaser  MJ.  Mechanisms  of  disease:  inflammation  and  the  origins  of  cancer.  Nat  Clin  Pract  Oncol  2005;  2:  90–7.  

• 6  

Hofseth  LJ,Ying  L.  Identifying  and  defusing  weapons  of  mass  inflammation  in  carcinogenesis.  Biochim  Biophys  Acta  2006;  1765:  74–84.  

• 7  

Hussain  SP,Hofseth  LJ,Harris  CC.  Radical  causes  of  cancer.  Nat  Rev  Cancer  2003;  3:  276–85.  

• 8  

Ohshima  H,Tatemichi  M,Sawa  T.  Chemical  basis  of  inflammation-­induced  carcinogenesis.  Arch  Biochem  Biophys  2003;  417:  3–11.  

• 9  

Coussens  LM,Werb  Z.  Inflammation  and  cancer.  Nature  2002;  420:  860–7.  

• 10  

Fitzpatrick  FA.  Inflammation,  carcinogenesis  and  cancer.  Int  Immunopharmacol  2001;  1:  1651–67.  

• 11  

Slaga  TJ,Lichti  U,Hennings  H,Elgjo  K,Yuspa  SH.  Effects  of  tumor  promoters  and  steroidal  anti-­inflammatory  agents  on  skin  of  newborn  mice  in  vivo  and  in  vitro.  J  Natl  Cancer  Inst  1978;  60:  425–31.  

• 12  

Nathan  C.  Specificity  of  a  third  kind:  reactive  oxygen  and  nitrogen  intermediates  in  cell  signalling.  J  Clin  Invest  2003;  111:  769–78.  

• 13  

Kröncke  KD.  Nitrosative  stress  and  transcription.  Biol  Chem  2003;  384:  1365–77.  

• 14  

Valko  M,Leibfritz  D,Moncol  J,Cronin  MTD,Mazur  M,Telser  J.  Free  radicals  and  antioxidant  in  normal  physiological  function  and  human  disease.  Int  J  Biochem  Cell  Biol  2007;  39:  44–84.  

• 15  

Sies  H.  Oxidative  stress:  Introductory  remarks.  In:  SiesH,  ed.  Oxidative  stress.  San  Diego:  Academic  Press,  1985:  1–8.  

• 16  

Sies  H.  Biochemistry  of  oxidative  stress.  Angew  Chem  Int  Ed  Engl  1986;  25:  1058–71.  

Direct  Link:  

• 17  

Sies  H,Cadenas  E.  Oxidative  stress:  damage  to  intact  cells  and  organs.  Philos  Trans  R  Soc  Lond  B  Biol  Sci  1985;  311:  617–31.  

• 18  

Hinson  RM,Williams  JA,Shacter  E.  Elevated  interleukin  6  is  induced  by  prostaglandin  E2  in  a  murine  model  of  inflammation:  possible  role  of  cyclooxygenase-­2.  Proc  Natl  Acad  Sci  USA  1996;  93:  4885–90.  

• 19  

Shacter  E,Arzadon  GK,Williams  J.  Elevation  of  IL-­6  in  response  to  a  chronic  inflammatory  stimulus  in  mice:  inhibition  by  indomethacin.  Blood  1992;  80:  194–202.  

• 20  

Moore  RJ,Owens  DM,Stamp  G,Arnott  C,Burke  F,East  N,Holdsworth  H,Turner  L,Rollins  B,Pasparakis  M,Kollias  G,Balkwill  F.  Mice  deficient  in  tumor  necrosis  factor-­α  are  resistant  to  skin  carcinogenesis.  Nat  Med  1999;  5:  828–31.  

• 21  

Zouki  C,Jozsef  L,Ouellet  S,Paquette  Y,Filep  JG.  Peroxynitrite  mediates  cytokine-­induced  IL-­8  gene  expression  and  production  by  human  leukocytes.  J  Leukoc  Biol  2001;  69:  815–24.  

• 22  

Jackson  JR,Seed  MP,Kircher  CH,Willoughby  DA,Winkler  JD.  The  codependence  of  angiogenesis  and  chronic  inflammation.  FASEB  J  1997;  11:  457–65.  

• 23  

Baron  JA,Sandler  RS.  Nonsteroidal  anti-­inflammatory  drugs  and  cancerprevention.  Annu  Rev  Med  2000;  51:  511–23.  

• 24  

Prescott  SM,Fitzpatrick  FA.  Cyclooxygenase-­2  and  carcinogenesis.  Biochim  Biophys  Acta  2000;  1470:  M69–M78.  

• 25  

Howe  LR,Dannenberg  AJ.  A  role  for  cyclooxygenase-­2  inhibitors  in  the  prevention  and  treatment  of  cancer.  Semin  Oncol  2002;  29:  111–19.  

• 26  

Dannenberg  AJ,Altorki  NK,Boyle  JO,Dang  C,Howe  LR,Weksler  BB,Subbaramaiah  K.  Cyclooxygenase  2:  a  pharmacological  target  for  the  prevention  of  cancer.  Lancet  Oncol  2001;  2:  544–51.  

• 27  

Lin  DT,Subbaramaiah  K,Shah  JP,Dannenberg  AJ,Boyle  JO.  Cyclooxygenase-­2:  a  novel  molecular  target  for  the  prevention  and  treatment  of  head  and  neck  cancer.  Head  Neck  2002;  24:  792–9.  

• 28  

Gupta  RA,DuBois  RN.  Colorectal  cancer  prevention  and  treatment  by  inhibition  of  cyclooxygenase-­2.  Nat  Rev  Cancer  2001;  1:  11.  

• 29  

Williams  CS,Mann  M,DuBois  RN.  The  role  of  cyclooxygenases  in  inflammation,  cancer,  and  development.  Oncogene  1999;  18:  7908–16.  

• 30  

Gately  S.  The  contributions  of  cyclooxygenase-­2  to  tumor  angiogenesis.  Cancer  Metastasis  Rev  2000;  19:  19–27.  

• 31  

Howe  LR,Subbaramaiah  K,Brown  AM,Dannenberg  AJ.  Cyclooxygenase-­2:  a  target  for  the  prevention  and  treatment  of  breast  cancer.  Endocr  Relat  Cancer  2001;  8:  97–114.  

• 32  

Goodwin  DC,Landino  LM,Marnett  LJ.  Effects  of  nitric  oxide  and  nitricoxide-­derived  species  on  prostaglandin  endoperoxide  synthase  and  prostaglandin  biosynthesis.  FASEB  J  1999;  13:  1121–36.  

• 33  

Marnett  LJ,Riggins  JN,West  JD.  Endogenous  generation  of  reactive  oxidants  and  electrophiles  and  their  reactions  with  DNA  and  protein.  J  Clin  Invest  2003;  111:  583–93.  

• 34  

Herrera  B,Alvarez  AM,Sanchez  A,Fernandez  M,Roncero  C,Benito  M,Fabregat  I.  Reactive  oxygen  species  (ROS)  mediates  the  mitochondrial-­dependent  apoptosis  induced  by  transforming  growth  factor  (beta)  in  fetal  hepatocytes.  FASEB  J  2001;  15:  741–51.  

• 35  

Herrera  B,Fernandez  M,Alvarez  AM,Roncero  C,Benito  M,Gil  J,Fabregat  I.  Activation  of  caspases  occurs  downstream  from  radical  oxygen  species  production,  Bcl-­xL  down-­regulation,  and  early  cytochrome  C  release  in  apoptosis  induced  by  transforming  growth-­factor  β  in  rat  fetal  hepatocytes.  Hepatology  2001;  34:  548–56.  

• 36  

Sata  N,Klonowski-­Stumpe  H,Han  B,Haussinger  D,Niederau  C.  Cytotoxicity  of  peroxynitrite  in  rat  pancreatic  acinar  AR4-­2J  cells.  Pancreas  1997;  15:  278–84.  

• 37  

Zhuang  S,Simon  G.  Peroxynitrite-­induced  apoptosis  involves  activation  of  multiple  caspases  in  HL-­60  cells.  Am  J  Physiol  Cell  Physiol  2000;  279:  C341–C351.  

• 38  

Johnstone  RW,Ruefli  AA,Lowe  SW.  Apoptosis:  a  link  between  cancer  genetics  and  chemotherapy.  Cell  2002;  108:  153–64.  

• 39  

Jabs  T.  Reactive  oxygen  intermediates  as  mediators  of  programmed  cell  death  in  plants  and  animals.  Biochem  Pharmacol  1999;  57:  231–45.  

• 40  

Valko  M,Morris  H,Cronin  MT.  Metals,  toxicity  and  oxidative  stress.  Curr  Med  Chem  2005;  12:  1161–208.  

• 41  

Marnett  LJ.  Oxyradicals  and  DNA  damage.  Carcinogenesis  2000;  21:  361–70.  

• 42  

Valko  M,Rhodes  CJ,Moncol  J,Izakovic  M,Mazur  M.  Free  radicals,  metals  and  antioxidants  in  oxidative  stress-­induced  cancer.  Chem  Biol  Interact  2006;  160:  1–40.  

• 43  

Inoue  S,Kawanishi  S.  Oxidative  DNA  damage  induced  by  simultaneous  generation  of  nitric  oxide  and  superoxide.  FEBS  Lett  1995;  371:  86–88.  

• 44  

Yermilov  V,Rubio  J,Becchi  M,Friesen  MD,Pignatelli  B,Ohshima  H.  Formation  of  8-­nitroguanine  by  the  reaction  of  guanine  with  peroxynitrite  in  vitro.  Carcinogenesis  1995;  16:  2045–50.  

• 45  

Akaike  T,Okamoto  S,Sawa  T,Yoshitake  J,Tamura  F,Ichimori  K,Miyazaki  K,Sasamoto  K,Maeda  H.  8-­Nitroguanosine  formation  in  viral  pneumonia  and  its  implication  for  pathogenesis.  Proc  Natl  Acad  Sci  USA  2003;  100:  685–90.  

• 46  

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  2007;  39:  44–84.  

• 47  

Sawa  T,Akaike  T,Ichimori  K,Akuta  T,Kaneko  K,Nakayama  H,Stuehr  DJ,Maeda  H.  Superoxide  generation  mediated  by  8-­nitroguanosine,  a  highly  redox-­active  nucleic  acid  derivative.  Biochem  Biophys  Res  Commun  2003;  311:  300–6.  

• 48  

Zaki  MH,Akuta  T,Akaike  T.  Nitric  oxide-­induced  nitrative  stress  involved  in  microbial  pathogenesis.  J  Pharmacol  Sci  2005;  98:  117–29.  

• 49  

Suzuki  N,Yasui  M,Geacintov  NE,Shafirovich  V,Shibutani  S.  Miscoding  events  during  DNA  synthesis  past  the  nitration-­damaged  base  8-­nitroguanine.  Biochemistry  2005;  44:  9238–45.  

• 50  

Bos  JL.  The  ras  gene  family  and  human  carcinogenesis.  Mutat  Res  1988;  195:  255–71.  

• 51  

Takahashi  T,Nau  MM,Chiba  I,Birrer  MJ,Rosenberg  RK,Vinocour  M,Levitt  M,Pass  H,Gazdar  AF,Minna  JD.  p53:  a  frequent  target  for  genetic  abnormalities  in  lung  cancer.  Science  1989;  246:  491–4.  

• 52  

Hsu  IC,Metcalf  RA,Sun  T,Welsh  JA,Wang  NJ,Harris  CC.  Mutational  hotspot  in  the  p53  gene  in  human  hepatocellular  carcinomas.  Nature  1991;  350:  427–8.  

• 53  

Albring  M,Griffith  J,Attardi  G.  Association  of  a  protein  structure  of  probable  membrane  derivation  with  HeLa  cell  mitochondrial  DNA  near  its  origin  of  replication.  Proc  Natl  Acad  Sci  USA  1977;  74:  1348–52.  

• 54  

Clayton  DA.  Transcription  of  the  mammalian  mitochondrial  genome.  Annu  Rev  Biochem  1984;  53:  573–94.  

• 55  

Yakes  FM,Van  Houten  B.  Mitochondrial  DNA  damage  is  more  extensive  and  persists  longer  than  nuclear  DNA  damage  in  human  cells  following  oxidative  stress.  Proc  Natl  Acad  Sci  USA  1997;  94:  514–19.  

• 56  

Beckman  KB,Ames  BN.  Oxidative  decay  of  DNA.  J  Biol  Chem  1997;  272:  19633–6.  

• 57  

Berlett  BS,Stadtman  ER.  Protein  oxidation  in  aging,  disease,  and  oxidative  stress.  J  Biol  Chem  1997;  272:  20313–16.  

• 58  

Esterbauer  H,Schaur  RJ,Zollner  H.  Chemistry  and  biochemistry  of  4-­hydroxynonenal,  malonaldehyde  and  related  aldehydes.  Free  Radic  Biol  Med  1991;  11:  81–128.  

• 59  

Kowaltowski  AJ,Vercesi  AE.  Mitochondrial  damage  induced  by  conditions  of  oxidative  stress.  Free  Radic  Biol  Med  1999;  26:  463–71.  

• 60  

Fliss  MS,Usadel  H,Caballero  OL,Wu  L,Buta  MR,Eleff  SM,Jen  J,Sidransky  D.  Facile  detection  of  mitochondrial  DNA  mutations  in  tumors  and  bodily  fluids.  Science  2000;  287:  2017–19.  

• 61  

Richard  SM,Bailliet  G,Paez  GL,Bianchi  MS,Peltomaki  P,Bianchi  NO.  Nuclear  and  mitochondrial  genome  instability  in  human  breast  cancer.  Cancer  Res  2000;  60:  4231–7.  

• 62  

Polyak  K,Li  Y,Zhu  H,Lengauer  C,Willson  JK,Markowitz  SD,Trush  MA,Kinzler  KW,Vogelstein  B.  Somatic  mutations  of  the  mitochondrial  genome  in  human  colorectal  tumours.  Nat  Genet  1998;  20:  291–3.  

• 63  

Habano  W,Nakamura  S,Sugai  T.  Microsatellite  instability  in  the  mitochondrial  DNA  of  colorectal  carcinomas:  evidence  for  mismatch  repair  systems  in  mitochondrial  genome.  Oncogene  1998;  17:  1931–7.  

• 64  

Tamura  G,Nishizuka  S,Maesawa  C,Suzuki  Y,Iwaya  T,Sakata  K,Endoh  Y,Motoyama  T.  Mutations  in  mitochondrial  control  region  DNA  in  gastric  tumors  of  Japanese  patients.  Eur  J  Cancer  1999;  35:  316–19.  

• 65  

Horton  TM,Petros  JA,Heddi  A,Shoffner  J,Kaufman  AE,Graham  SD,Gramlich  T,Wallace  DC.  Novel  mitochondrial  DNA  deletion  found  in  renal  cell  carcinoma.  Genes  Chromosomes  Cancer  1996;  15:  95–101.  

• 66  

Uchida  K.  4-­Hydroxy-­2-­nonenal:  a  product  and  mediator  of  oxidative  stress.  Prog  Lipid  Res  2003;  42:  318–43.  

• 67  

Blair  IA.  Lipid  hydroperoxide-­mediated  DNA  damage.  Exp  Gerontol  2001;  36:  1473–81.  

• 68  

Hu  W,Feng  Z,Eveleigh  J,Iver  G,Pan  J,Amin  S,Chung  FL,Tang  MS.  The  major  lipid  peroxidation  product,  trans-­4-­hydroxy-­2-­nonenal,  preferentially  forms  DNA  adducts  at  codon  249  of  human  p53  gene,  a  unique  mutational  hotspot  in  hepatocellular  carcinoma.  Carcinogenesis  2002;  23:  1781–9.  

• 69  

Gago-­Dominguez  M,Castelao  JE,Yuan  JM,Ross  RK,Yu  MC.  Lipid  peroxidation:  a  novel  and  unifying  concept  of  the  etiology  of  renal  cell  carcinoma  (United  States).  Cancer  Causes  Control  2002;  13:  287–93.  

• 70  

Sander  CS,Hamm  F,Elsner  P,Thiele  JJ.  Oxidative  stress  in  malignant  melanoma  and  non-­melanoma  skin  cancer.  Br  J  Dermatol  2003;  148:  913–22.  

• 71  

Kuhn  H,Borchert  A.  Regulation  of  enzymatic  lipid  peroxidation:  the  interplay  of  peroxidizing  and  peroxide  reducing  enzymes.  Free  Radic  Biol  Med  2002;  33:  154–72.  

• 72  

Nair  J,Barbin  A,Velic  I,Bartsch  H.  Etheno  DNA-­base  adducts  from  endogenous  reactive  species.  Mutat  Res  1999;  424:  59–69.  

• 73  

Ruef  J,Rao  GN,Li  F,Bode  C,Patterson  C,Bhatnagar  A,Runge  MS.  Induction  of  rat  aortic  smooth  muscle  cell  growth  by  the  lipid  peroxidation  product  4-­hydroxy-­2-­nonenal.  Circulation  1998;  97:  1071–8.  

• 74  

Cheng  JZ,Singhal  SS,Saini  M,Singhal  J,Piper  JT,VanKuijk  FJ,Zimniak  P,Awasthi  YC,Awasthi  S.  Effects  of  mGST  A4  transfection  on  4-­hydroxynonenal-­mediated  apoptosis  and  differentiation  of  K562  human  erythroleukemia  cells.  Arch  Biochem  Biophys  1999;  372:  29–36.  

• 75  

Dianzani  MU,Barrera  G,Parola  M.  4-­Hydroxy-­2,3-­nonenal  as  a  signal  for  cell  function  and  differentiation.  Acta  Biochim  Pol  1999;  46:  61–75.  

• 76  

Fazio  VM,Rinaldi  M,Ciafre  S,Barrera  G,Farace  MG.  Control  of  neoplastic  cell  proliferation  and  differentiation  by  restoration  of  4-­hydroxynonenal  physiological  concentrations.  Mol  Aspects  Med  1993;  14:  217–28.  

• 77  

Kumagai  T,Kawamoto  Y,Nakamura  Y,Hatayama  I,Satoh  K,Osawa  T,Uchida  K.  4-­hydroxy-­2-­nonenal,  the  end  product  of  lipid  peroxidation,  is  a  specific  inducer  of  cyclooxygenase-­2  gene  expression.  Biochem  Biophys  Res  Commun  2000;  273:  437–41.  

• 78  

Pryor  WA,Stanley  JP.  Letter:  a  suggested  mechanism  for  the  production  of  malonaldehyde  during  the  autoxidation  of  polyunsaturated  fatty  acids.  Nonenzymatic  production  of  prostaglandin  endoperoxides  during  autoxidation.  J  Org  Chem  1975;  40:  3615–17.  

• 79  

Marnett  LJ.  Lipid  peroxidation-­DNA  damage  by  malondialdehyde.  Mutat  Res  1999;  424:  83–95.  

• 80  

Wang  MY,Dhingra  K,Hittelman  WN,Liehr  JG,deAndrade  M,Li  DH.  Lipid  peroxidation-­induced  putative  malondialdehyde–DNA  adducts  in  human  breast  tissues.  Cancer  Epidemiol  Biomark  Prev  1996;  5:  705–10.  

• 81  

Fink  SP,Reddy  GR,Marnett  LJ.  Mutagenicity  in  Escherichia  coli  of  the  major  DNA  adduct  derived  from  the  endogenous  mutagen  malondialdehyde.  Proc  Natl  Acad  Sci  USA  1997;  94:  8652–57.  

• 82  

Mao  H,Schnetz-­Boutaud  NC,Weisenseel  JP,Marnett  LJ,Stone  MP.  Duplex  DNA  catalyzes  the  chemical  rearrangement  of  a  malondialdehyde  deoxyguanosine  adduct.  Proc  Natl  Acad  Sci  USA  1999;  96:  6615–20.  

• 83  

Fedtke  N,Boucheron  JA,Walker  VE,Swenberg  JA.  Vinyl  chloride-­induced  DNA  adducts,  Part  2:  Formation  and  persistence  of  7-­2�-­oxoethylguanine  and  N-­  2,3-­ethenoguanine  in  rat-­tissue  DNA.  Carcinogenesis  1990;  11:  1287–92.  

• 84  

Zachara  BA.  Mammalian  selenoproteins.  J  Trace  Elem  Electrolytes  Health  Dis  1992;  6:  137–51.  

• 85  

Steele  VE,Holmes  CA,Hawk  ET,Kopelovich  L,Lubet  RA,Crowell  JA,Sigman  CC,Kelloff  GJ.  Lipoxygenase  inhibitors  as  potential  cancer  chemopreventives.  Cancer  Epidemiol  Biomarkers  Prev  1999;  8:  467–83.  

• 86  

Tang  DG,Honn  KV.  Apoptosis  of  W256  carcinosarcoma  cells  of  the  monocytoid  origin  induced  by  NDGA  involves  lipid  peroxidation  and  depletion  of  GSH:  role  of  12-­lipoxygenase  in  regulating  tumor  cell  survival.  J  Cell  Physiol  1997;  172:  155–70.  

• 87  

Nie  D,Che  M,Grignon  D,Tang  K,Honn  KV.  Role  of  eicosanoids  in  prostate  cancer  progression.  Cancer  Metastasis  Rev  2001;  20:  195–206.  

• 88  

Eling  TE,Glasgow  WC.  Cellular  proliferation  and  lipid  metabolism:  importance  of  lipoxygenases  in  modulating  epidermal  growth  factor-­dependent  mitogenesis.  Cancer  Metastasis  Rev  1994;  13:  397–410.  

• 89  

Eling  TE,Everhart  AL,Angerman-­Stewart  J,Hui  R,Glasgow  WC.  Modulation  of  epidermal  growth  factor  signal  transduction  by  linoleic  acid  metabolites.  Adv  Exp  Med  Biol  1997;  407:  319–22.  

• 90  

Schnurr  K,Borchert  A,Kuhn  H.  Inverse  regulation  of  lipid-­peroxidizing  and  hydroperoxyl  lipid-­reducing  enzymes  by  interleukins  4  and  13.  FASEB  J  1999;  13:  143–54.  

• 91  

Schnurr  K,Brinckmann  R,Kuhn  H.  Cytokine  induced  regulation  of  15-­lipoxygenase  and  phospholipid  hydroperoxide  glutathione  peroxidase  in  mammalian  cells.  Adv  Exp  Med  Biol  1999;  469:  75–81.  

• 92  

Chen  CJ,Huang  HS,Chang  WC.  Inhibition  of  arachidonate  metabolism  in  human  epidermoid  carcinoma  a431  cells  overexpressing  phospholipid  hydroperoxide  glutathione  peroxidase.  J  Biomed  Sci  2002;  9:  453–9.  

• 93  

Huang  HS,Chen  CJ,Suzuki  H,Yamamoto  S,Chang  WC.  Inhibitory  effect  of  phospholipid  hydroperoxide  glutathione  peroxidase  on  the  activity  of  lipoxygenases  and  cyclooxygenases.  Prostaglandins  Other  Lipid  Mediat  1999;  58:  65–75.  

• 94  

Shitashige  M,Morita  I,Murota  S.  Different  substrate  utilization  between  prostaglandin  endoperoxide  H  synthase-­1  and  -­2  in  NIH3T3  fibroblasts.  Biochim  Biophys  Acta  1998;  1389:  57–66.  

• 95  

Samuelsson  B,Dahlen  SE,Lindgren  JA,Rouzer  CA,Serhan  CN.  Leukotrienes  and  lipoxins:  structures,  biosynthesis,  and  biological  effects.  Science  1987;  237:  1171–6.  

• 96  

Chen  CJ,Huang  HS,Lin  SB,Chang  WC.  Regulation  of  cyclooxygenase  and  12-­lipoxygenase  catalysis  by  phospholipids  hydroperoxide  glutathione  peroxidase  in  A431  cells.  Prostaglandins  Leukot  Essent  Fatty  Acids  2000;  62:  261–8.  

• 97  

Wang  HP,Qian  SY,Schafer  FQ,Domann  FE,Oberley  LW,Buettner  GR.  Phospholipid  hydroperoxide  glutathione  peroxidase  protects  against  singlet  oxygen-­induced  cell  damage  of  photodynamic  therapy.  Free  Radic  Biol  Med  2001;  30:  825–35.  

• 98  

Chen  GQ,Zhu  J,Shi  XG,Ni  JH,Zhong  HJ,Si  GY,Jin  XL,Tang  W,Li  XS,Xong  SM,Shen  ZX,Sun  GL,  et  al.  In  vitro  studies  on  cellular  and  molecular  mechanisms  of  

arsenic  trioxide  (As2O3)  in  the  treatment  of  acute  promyelocytic  leukemia:  As2O3  induces  NB4  cell  apoptosis  with  downregulation  of  Bcl-­2  expression  and  modulation  of  PML-­RAR  α/PML  proteins.  Blood  1996;  88:  1052–61.  

• 99  

Huang  HS,Chang  WC,Chen  CJ.  Involvement  of  reactive  oxygen  species  in  arsenite-­induced  downregulation  of  phospholipids  hydroperoxide  glutathione  peroxidase  in  human  epidermoid  carcinoma  A431  cells.  Free  Radic  Biol  Med  2002;  33:  864–73.  

• 100  

Aoki  Y,Yarchoan  R,Braun  J,Iwamoto  A,Tosato  G.  Viral  and  cellular  cytokines  in  AIDS-­related  malignant  lymphomatous  effusions.  Blood  2000;  96:  1599–601.  

• 101  

Trentin  L,Cerutti  A,Zambello  R,Sancretta  R,Tassinari  C,Facco  M,Adami  F,Rodeghiero  F,Agostini  C,Semenzato  G.  Interleukin-­15  promotes  the  growth  of  leukemic  cells  of  patients  with  B-­cell  chronic  lymphoproliferative  disorders.  Blood  1996;  87:  3327–35.  

• 102  

Fosslien  E.  Molecular  pathology  of  cyclooxygenase-­2  in  neoplasia.  Ann  Clin  Lab  Sci  2000;  30:  3–21.  

• 103  

Subbaramaiah  K,Dannenberg  AJ.  Cyclooxygenase  2:  a  molecular  target  for  cancer  prevention  and  treatment.  Trends  Pharmacol  Sci  2003;  24:  96–102.  

 


Recommended