Resveratrol-3-O-glucuronide and resveratrol-4'-O-glucuronide reduce DNA strand breakage but not apoptosis in Jurkat T cells treated with camptothecin

  • Authors:
    • Susan J. Zunino
    • David H. Storms
  • View Affiliations

  • Published online on: June 16, 2017     https://doi.org/10.3892/ol.2017.6392
  • Pages: 2517-2522
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Resveratrol has been reported to inhibit or induce DNA damage, depending upon the type of cell and the experimental conditions. Dietary resveratrol is present in the body predominantly as metabolites and limited data is available concerning the activities of these metabolic products. In the present study, physiologically obtainable levels of the resveratrol metabolites resveratrol-3-O-glucuronide, resveratrol‑4'‑O‑glucuronide and resveratrol-3-O-sulfate were evaluated for their ability to protect Jurkat T cells against DNA damage induced by the topoisomerase I inhibitors camptothecin and topotecan. The cells were pretreated for 24 h with 10 µM resveratrol aglycone or each resveratrol metabolite prior to the induction of DNA damage with camptothecin or topotecan. In separate experiments, the cells were co‑treated with resveratrol or its metabolites, and a topoisomerase I inhibitor. The detection of histone 2AX phosphorylation and terminal deoxynucleotidyl transferase dUTP nick end‑labeling (TUNEL) were used to determine DNA damage, and apoptosis was measured using an antibody against cleaved poly ADP‑ribose polymerase. It was identified that pretreatment of the cells with resveratrol‑3‑O‑glucuronide and resveratrol‑4'‑O‑glucuronide reduced the mean fluorescence intensity of staining for DNA strand breaks following treatment with camptothecin, while the percentage of cells undergoing apoptosis was unchanged. However, pretreatment of the cells with resveratrol aglycone increased the DNA damage and apoptosis induced by the drugs. These results suggest that the glucuronide metabolites of resveratrol partially protected the cells from DNA damage, but did not influence the induction of cell death by camptothecin and topotecan. These data suggest that resveratrol aglycone treatment may be beneficial for treating types of cancer that have direct contact with resveratrol prior to its metabolism, including gastrointestinal cancers, which are routinely treated with topoisomerase I inhibitors.

Introduction

Resveratrol, an anti-fungal phytochemical that occurs in grapes, blueberries, mulberries, cranberries and red wine, is rapidly metabolized by the intestine and liver following oral consumption (16). The major phase II metabolites of resveratrol include glucuronidated, sulfated and methylated products (26). Intestinal bacteria have been reported to break down resveratrol to metabolic products including benzoic, phenylacetic and propionic acids (7,8). Dihydroresveratrol, another metabolic product of gut bacteria, has been identified in the form of glucuronidated and sulfated products in plasma and urine following the consumption of resveratrol (9,10). Gut microbiota produce other conversion products of resveratrol subsequent to ingestion, including 3,4′-dihydroxy-trans-stilbene and 3,4′-dihydroxybibenzyl (1113). A previous human study with oral doses of ≤5 g of resveratrol per day demonstrated that the peak plasma levels of resveratrol, resveratrol-3-O-glucuronide, resveratrol-4′-O-glucuronide and resveratrol-3-O-sulfate were 4.2, 17.1, 10.2 and 18.3 µM, respectively (2). These data indicate that high concentrations of resveratrol glucuronide and sulfate metabolites are achieved in the plasma following the dietary intake of resveratrol. Pharmacokinetic analysis of resveratrol and its metabolites in that study revealed that the phytochemical products remain in the plasma for 5–8 h (2).

Camptothecin is a topoisomerase I inhibitor that induces cytotoxicity by the generation of DNA strand breaks (14). Topotecan is a water-soluble derivative of camptothecin. Camptothecin compounds act primarily by binding to and stabilizing the topoisomerase I-DNA complex, which then collides with the replication fork during S phase of the cell cycle. This collision results in topoisomerase I-linked DNA breaks, the formation of double-strand DNA breaks and the irreversible arrest of DNA replication. Camptothecin compounds inhibit transcription by a similar mechanism: By binding to the topoisomerase I-DNA complex and colliding with the RNA polymerase complex, leading to the arrest of RNA synthesis and the generation of single-strand DNA breaks (14).

DNA strand breaks result in the activation of the DNA repair machinery and, if repair is not possible, apoptosis. DNA strand breaks can be quantified by several methods, including by the antibody-mediated detection of phosphorylated histone subunit 2AX (H2AX) and labeling the ends of broken strands with the terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) assay (15,16). H2AX is a histone 2A isoform that is present at levels of 2–25% in the histone core of the DNA complex. It is phosphorylated, in response to DNA strand breaks, by the phosphatidylinositol-3 kinase-like family of kinases, including ataxia telangiectasia mutated (ATM) and ATM-and Rad3-related (ATR), and by DNA-dependent protein kinase (DNA-PK) (17,18).

Resveratrol aglycone has been studied extensively in vitro to determine its mechanisms of action; however, the majority of these studies have utilized high concentrations of resveratrol aglycone (50–100 µM) that are not yet achievable in vivo, particularly in the plasma. Considering that resveratrol aglycone reached a peak concentration of 4.2 µM in the blood following the ingestion of 5 g per day (2), the data from the majority of in vitro studies are not likely to be reflective of the actual activity once resveratrol is absorbed and metabolized. There is also limited information on the activity of resveratrol metabolites.

In the present study, using Jurkat T cells as a model representing a cell type that occurs in the blood, camptothecin and topotecan were applied in order to determine whether or not glucuronidated and sulfated metabolites of resveratrol were able to increase or inhibit the DNA damage induced by these drugs. DNA damage was measured by determining the extent of H2AX phosphorylation and TUNEL staining. Apoptosis in the cells was determined by measuring the extent of the cleavage of poly ADP-ribose polymerase (PARP). The cells were pretreated with physiological levels of resveratrol-3-O-glucuronide, resveratrol-4′-O-glucuronide or resveratrol-3-O-sulfate prior to the induction of DNA damage by camptothecin and topotecan, or co-treated with metabolites and drugs. Flow cytometry was used to measure the extent of DNA damage and apoptosis, and the activities of the resveratrol metabolites were compared with equivalent amounts of the resveratrol aglycone.

Materials and methods

Cell culture and chemicals

Jurkat acute lymphoblastic T leukemia cells (American Type Culture Collection, Manassas, VA, USA) were cultured at 37°C with 5% CO2 in RPMI-1640 medium (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), 50 IU/ml penicillin, 50 µg/ml streptomycin, 0.25 µg/ml amphotericin B, 1 mM sodium pyruvate and 2 mM L-glutamine (Invitrogen; Thermo Fisher Scientific, Inc.). Trans-resveratrol (≥99% pure), dimethyl sulfoxide (DMSO; vehicle) and topotecan hydrochloride hydrate (≥98%) were purchased from Sigma-Aldrich (Merck KGaA). Trans-resveratrol 3-O-D-glucuronide (≥95%), trans-resveratrol 4′-O-D-glucuronide (≥95%) and trans-resveratrol-3-O-sulfate (≥98%) were purchased from Cayman Chemical Company (Ann Arbor, MI, USA). Camptothecin (≥98%) was purchased from MP Biomedicals, LLC (Santa Ana, CA, USA).

H2AX phosphorylation and apoptosis measurements

Jurkat T cells were cultured at a concentration of 0.5×106 cells/ml in 24-well plates (Sarstedt, Inc., Newton, NC, USA) with 0.1% DMSO (control) or 10 µM each of resveratrol aglycone, resveratrol-3-O-glucuronide, resveratrol-4′-O-glucuronide or resveratrol-3-O-sulfate for 24 h in the previously described conditions. Cells were then washed with RPMI-1640 and incubated with 5 µM camptothecin or 10 µM topotecan for a further 4 h in the same conditions. The cells were fixed and permeabilized for intracellular staining using the Fixation/Permeabilization Solution kit (BD Biosciences, San Jose, CA, USA) according to the manufacturer's protocol. For each treatment group, 106 cells were stained with phycoerythrin-conjugated anti-cleaved PARP (0.06 µg/20 µl test; cat no. 552933) and Alexa Fluor-conjugated anti-phosphorylated H2AX (0.125 µg/5 µl test; cat no. 56-447) (both from BD Biosciences) antibodies. Following 30 min incubation on ice, the cells were fixed in 1% paraformaldehyde prepared in phosphate-buffered saline (Sigma-Aldrich; Merck KGaA). The cells were assessed with an LSRFortessa flow cytometer and the results analyzed using FACSDiva software v8.0.1 (both from BD Biosciences). A total of 30,000 events were collected per measurement, subsequent to gating to exclude debris.

TUNEL assay

Cells were pretreated with resveratrol and its metabolites for 24 h as previously described. Cells were then washed with RPMI medium and treated with 5 µM camptothecin or 10 µM topotecan for a further 4 h. In separate experiments, the cells were co-treated with resveratrol or its metabolites, plus one of the topoisomerase-inhibitor drugs at doses as previously described for a total of 4 h. DNA strand breaks were labeled with a TUNEL assay (Apo-Direct kit; BD Biosciences) with fluorescein isothiocyanate (FITC)-labelled dUTP according to the manufacturer's protocol, with the following modifications: The fixation and membrane permeabilization step using 70% v/v ethanol was performed for >18 h at −20°C and the end-labeling reaction with the terminal transferase was 4 h at 37°C. Labeled cells were treated at room temperature for 30 min with RNase and propidium iodide (PI) as provided in the Apo-Direct kit at the concentrations recommended by the manufacturer. A total of 30,000 events were collected on the LSRFortessa flow cytometer for each treatment. The end-labeling of DNA strand breaks was evaluated, subsequent to gating singlet populations, based on the fluorescence of the PI area vs. width.

Statistical analysis

Statistical analyses were performed using GraphPad Prism v6.05 (GraphPad Software, Inc., La Jolla, CA, USA) and the data were presented as the mean ± standard error of the mean. P-values were obtained using one-way analysis of variance with Tukey's multiple comparisons test to evaluate the significance of differences between the means of the treatment groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Phosphorylation of H2AX

The histone protein H2AX is phosphorylated in response to DNA damage. The extent of phosphorylated H2AX was assessed by flow cytometry to determine the effects of resveratrol and its metabolites on DNA damage induced by camptothecin and topotecan. The cells were pretreated with 10 µM of resveratrol, or its metabolites, for 24 h prior to a 4-h treatment with the DNA-damaging agents. Resveratrol aglycone was used at a concentration of 10 µM as a comparative control for DNA damage and apoptosis. The proportion of cells with phosphorylated H2AX and the mean fluorescence intensity (MFI) of this population were measured to determine alterations in DNA damage attributable to the phytochemicals. Treatment of cells with resveratrol aglycone alone for 24 h increased the percentage of cells with phosphorylated H2AX compared with untreated control cells (Fig. 1A; P<0.05). Additionally, resveratrol aglycone pretreatment with the subsequent addition of camptothecin or topotecan was associated with an increased percentage of cells with phosphorylated H2AX compared with cells treated only with the topoisomerase inhibitors (Fig. 1B and C; P<0.05), suggesting an additive effect. A corresponding increase in the MFI of H2AX staining was observed following the pretreatment of Jurkat cells with resveratrol aglycone followed by the DNA damaging agents (data not shown). In camptothecin-treated cells, resveratrol aglycone increased the MFI from 5,332±156 in the control group to 7,000±198; and in topotecan-treated cells, resveratrol aglycone increased the MFI from 5,105±115 (control) to 6,747±209 (P<0.05), indicating an increase in DNA damage. The resveratrol glucuronide and sulfate metabolites did not significantly alter the percentage of H2AX+ cells or the MFI of H2AX staining following drug treatments.

DNA strand breaks following pretreatment of cells with phytochemicals

The TUNEL assay provides a direct measurement of DNA damage by labeling the DNA break-point ends. In cells treated with camptothecin or topotecan, only pretreatment with resveratrol aglycone resulted in a significant increase of the percentage of cells with detectable DNA breaks compared with the control group (Fig. 2; P<0.05), which confirms the result observed for H2AX phosphorylation. However, in camptothecin-treated cells, pretreatment with resveratrol-3-glucuronide or resveratrol-4′-glucuronide significantly reduced the MFI compared with the control group, suggesting that the extent of damage (or number of DNA strand breaks) per cell was attenuated by pretreatment with these metabolites (Fig. 3, P<0.05). In the topotecan-treated cells, there were no differences in MFI between any of the groups (data not shown).

Induction of apoptosis following pretreatment with resveratrol and metabolites

The topoisomerase I inhibitors camptothecin and topotecan function to produce DNA damage, cell cycle arrest and apoptosis. The extent of apoptosis was measured in the Jurkat cells using an antibody against the cleaved form of PARP, as cleavage of PARP occurs at a late stage in the apoptotic pathway. The cells were pretreated with resveratrol or its metabolites for 24 h, and DNA damage was induced by the topoisomerase I inhibitors. Cells treated only with resveratrol or its metabolites for 24 h did not induce PARP cleavage compared with untreated cells (Fig. 4A). In the topoisomerase I inhibitor-treated cells, only pretreatment with 10 µM resveratrol aglycone increased the percentage of cells with cleaved PARP compared with the cells treated only with the drugs (Fig. 4B and C; P<0.05), whereas the resveratrol metabolites had no effect on the level of apoptosis induced by treatment with camptothecin and topotecan.

Co-treatment with resveratrol metabolites and topoisomerase I inhibitors

Jurkat cells were co-treated with resveratrol aglycone or its metabolites plus a topoisomerase I inhibitor for 4 h, and DNA damage was analyzed using the TUNEL assay. Treatment with only resveratrol or one of its metabolites for 4 h did not increase the extent of DNA strand breakage detected by the assay (Fig. 5A). Co-treatment of cells with resveratrol aglycone and camptothecin for 4 h decreased the percentage of cells with DNA damage compared with treatment with camptothecin alone (Fig. 5B; P<0.05). A similar reduction in the percentage of cells with DNA damage was observed for cells co-treated with topotecan and resveratrol aglycone, although this reduction did not reach statistical significance. A reduction in DNA damage was observed between topotecan treatment alone (control) and co-treatment with topotecan plus resveratrol 4′-O-D-glucuronide (Fig. 5C; P<0.05). Co-treatment with resveratrol 3-O-D-glucuronide or resveratrol-3-O-sulfate did not alter DNA damage induced by camptothecin or topotecan, and no differences in MFI were observed for any of the treatment groups (data not shown).

Discussion

The topoisomerase I inhibitors camptothecin and topotecan were utilized in the present study as a means to induce DNA damage and test the ability of glucuronidated and sulfated metabolites of resveratrol to prevent DNA strand breaks. The pretreatment of Jurkat T cells with resveratrol-3-O-glucuronide and resveratrol-4′-O-glucuronide significantly decreased the mean fluorescence signal from the FITC-UTP used for end-labeling DNA strand breaks induced by camptothecin, suggesting that these resveratrol metabolites were able to reduce the number of strand breaks per cell caused by this drug. No differences in camptothecin- or topotecan-induced DNA damage, as measured by H2AX phosphorylation, were observed between the control and any of the pretreatment groups, with the exception of resveratrol aglycone. DNA strand breaks induce a rapid response by the DNA repair/apoptotic machinery. H2AX is phosphorylated following single-strand DNA breaks and replication stress (by ATR), double-strand DNA breaks (by ATM), and fragmentation of DNA during apoptosis (by DNA-PK) (19,20). It has been reported that the region of phosphorylated H2AX extends up to 1 megabase on either side of a double-strand DNA break in mammalian cells; this post-translational modification is the first step for the recruitment of DNA repair complexes to the damaged sites (1922). The labeling of DNA ends by the TUNEL assay is a direct method of determining DNA strand breakage and is a more sensitive method, compared with phosphorylated H2AX, for determining the extent of DNA damage on a per-cell basis.

Camptothecin is a lipophilic compound and topotecan is its water-soluble analogue. It has been reported that lipophilic camptothecin has a greater topoisomerase inhibitory effect and cytotoxicity than its water-soluble counterparts (23,24). However, the two compounds demonstrated a similar ability to induce the phosphorylation of H2AX and DNA strand breaks in the present study. Therefore, it remains unclear why pretreatment with the glucuronide metabolites of resveratrol produced a reduction in DNA strand breaks (as determined by the TUNEL assay) following camptothecin, but not topotecan, treatment.

Resveratrol aglycone has been reported to have anticancer and chemosensitizing activities against cancer cells; mechanisms of action for this have previously been proposed (1,2528). Anticancer mechanisms of action for resveratrol, which have been described predominantly from in vitro studies, include the inhibition of transcription factors, kinases and other molecules involved in cell proliferation and survival. In animal models, resveratrol has demonstrated efficacy against breast, esophageal, lung and colon cancer, and was reported to decrease the extent of metastasis of melanoma and lung or colon carcinoma (2937). However, resveratrol aglycone has been reported to protect certain types of cells from DNA damage. For example, a previous study demonstrated that resveratrol could inhibit DNA damage in the kidneys of rats treated with the carcinogen KBrO3 (38). In addition, resveratrol attenuated DNA damage induced by H2O2 in glioma cells and peripheral blood lymphocytes, protected DNA from damage by chromium, and reduced the number of DNA adducts induced by the carcinogen dibenzo [a,l]pyrene (3942). In the present study, the pretreatment of Jurkat cells with 10 µM resveratrol aglycone consistently increased the level of DNA damage induced by camptothecin and topotecan and was used as a positive control in the DNA damage and apoptosis assays. However, a decrease in DNA strand breaks in cells co-treated for 4 h with resveratrol and camptothecin was observed, as in cells co-treated with resveratrol 4′-O-D-glucuronide and topotecan. As has been observed for a number of chemotherapeutic drugs, camptothecins induce oxidative stress in various tissues (4345); the short co-treatment period with resveratrol in the present study may have provided protective effects against drug-induced oxidative stress due to resveratrol's antioxidant activities (1). Furthermore, camptothecin and resveratrol are lipophilic agents, which may have increased direct interactions between the molecules during co-treatment.

There is limited data available concerning the activity of resveratrol metabolites. Resveratrol-3-O-glucuronide and resveratrol-4′O-glucuronide are two major metabolic products of resveratrol subsequent to ingestion, and the concentrations used in the present study were intended to be physiologically relevant based on a previous study (2). It will be important to understand the potential of these metabolites to protect cells from DNA damage induced by chemotherapeutic drugs. We hypothesize that the effects of these metabolites may be particularly important for the protection of normal cells during chemotherapy. Future research on the protective activities of the metabolic products of resveratrol are necessary to elucidate their interaction with DNA-damaging drugs used in the treatment of cancer. Of note, camptothecins are used in the combinatorial treatment of colorectal cancers (4648). The increased DNA damage and apoptosis of cells treated with camptothecin and topotecan following pretreatment with resveratrol aglycone in the present study suggests that dietary resveratrol may be a useful addition for treating gastrointestinal cancers that have direct contact with unmetabolized resveratrol.

Acknowledgements

This study was supported by the USDA CRIS Project (grant no. 2032-53000-001-00D). USDA is an equal opportunity provider and employer.

References

1 

Aggarwal BB, Bhardwaj A, Aggarwal RS, Seeram NP, Shishodia S and Takada Y: Role of resveratrol in prevention and therapy of cancer: Preclinical and clinical studies. Anticancer Res. 24:2783–2840. 2004.PubMed/NCBI

2 

Brown VA, Patel KR, Viskaduraki M, Crowell JA, Perloff M, Booth TD, Vasilinin G, Sen A, Schinas AM, Piccirilli G, et al: Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 70:9003–9011. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Patel KR, Scott E, Brown VA, Gescher AJ, Steward WP and Brown K: Clinical trials of resveratrol. Ann NY Acad Sci. 1215:161–169. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Walle T, Hsieh F, DeLegge MH, Oatis JE Jr and Walle UK: High absorption but very low bioavailability of oral resveratrol in humans. Drug Metabol Dispos. 32:1377–1382. 2004. View Article : Google Scholar

5 

Vitrac X, Desmoulière A, Brouillaud B, Krisa S, Deffieux G, Barthe N, Rosenbaum J and Mérillon JM: Distribution of [14C]-trans-resveratrol, a cancer chemopreventive polyphenol, in mouse tissues after oral administration. Life Sci. 72:2219–2233. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Wenzel E, Soldo T, Erbersdobler H and Somoza V: Bioactivity and metabolism of trans-resveratrol orally administered to Wistar rats. Mol Nutr Food Res. 49:482–494. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Crozier A, Jaganath IB and Clifford MN: Dietary phenolics: Chemistry, bioavailability and effects on health. Nat Prod Rep. 26:1001–1043. 2009. View Article : Google Scholar : PubMed/NCBI

8 

van Duynhoven J, Vaughan EE, Jacobs DM, Kemperman RA, van Velzen EJ, Gross G, Roger LC, Possemiers S, Smilde AK, Doré J, et al: Metabolic fate of polyphenols in the human superorganism. Proc Natl Acad Sci USA. 108 Suppl 1:pp. 4531–4538. 2011; View Article : Google Scholar : PubMed/NCBI

9 

Rotches-Ribalta M, Andres-Lacueva C, Estruch R, Escribano E and Urpi-Sarda M: Pharmacokinetics of resveratrol metabolic profile in healthy humans after moderate consumption of red wine and grape extract tablets. Pharmacol Res. 66:375–382. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Radko Y, Christensen KB and Christensen LP: Semi-preparative isolation of dihydroresveratrol-3-O-β-d-glucuronide and four resveratrol conjugates from human urine after oral intake of a resveratrol-containing dietary supplement. J Chromatogr B Analyt Technol Biomed Life Sci. 930:54–61. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Bode LM, Bunzel D, Huch M, Cho GS, Ruhland D, Bunzel M, Bub A, Franz CM and Kulling SE: In vivo and in vitro metabolism of trans-resveratrol by human gut microbiota. Am J Clin Nutr. 97:295–309. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Jung CM, Heinze TM, Schnackenberg LK, Mullis LB, Elkins SA, Elkins CA, Steele RS and Sutherland JB: Interaction of dietary resveratrol with animal-associated bacteria. FEMS Microbiol Lett. 297:266–273. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Azorín-Ortuño M, Yáñez-Gascón MJ, Vallejo F, Pallarés FJ, Larrosa M, Lucas R, Morales JC, Tomás-Barberán FA, García-Conesa MT and Espín JC: Metabolites and tissue distribution of resveratrol in the pig. Mol Nutr Food Res. 55:1154–1168. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Liu LF, Desai SD, Li TK, Mao Y, Sun M and Sim SP: Mechanism of action of campothecin. Ann N Y Acad Sci. 922:1–10. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Kuo LJ and Yang LX: Gamma-H2AX-a novel biomarker for DNA double-strand breaks. In Vivo. 22:305–309. 2008.PubMed/NCBI

16 

Gavrieli Y, Sherman Y and Ben-Sasson SA: Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol. 119:493–501. 1992. View Article : Google Scholar : PubMed/NCBI

17 

Rogakou EP, Pilch DR, Orr AH, Ivanova VS and Bonner WM: DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 273:5858–5868. 1998. View Article : Google Scholar : PubMed/NCBI

18 

Shiloh Y: ATM and related protein kinases: Safeguarding genome integrity. Nat Rev Cancer. 3:155–168. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Fernandez-Capetillo O, Lee A, Nussenzweig M and Nussenzweig A: H2AX: The histone guardian of the genome. DNA Repair (Amst). 3:959–967. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Srivastava N, Gochhait S, de Boer P and Bamezai RN: Role of H2AX in DNA damage response and human cancers. Mutat Res. 682:180–188. 2009. View Article : Google Scholar

21 

Redon C, Pilch D, Rogakou E, Sedelnikova O, Newrock K and Bonner W: Histone H2A variants H2AX and H2AZ. Curr Opin Genet Dev. 12:162–169. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Shroff R, Arbel-Eden A, Pilch D, Ira G, Bonner WM, Petrini JH, Haber JE and Lichten M: Distribution and dynamics of chromatin modification induced by a defined DNA double-strand break. Curr Biol. 14:1703–1711. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Bom D, Curran DP, Kruszewski S, Zimmer SG, Strode J Thompson, Kohlhagen G, Du W, Chavan AJ, Fraley KA, Bingcang AL, et al: The novel silatecan 7-tert-butyldimethylsilyl-10-hydroxycamptothecin displays high lipophilicity, improved human blood stability, and potent anticancer activity. J Med Chem. 43:3970–3980. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Van Hattum AH, Pinedo HM, Schlüper HM, Hausheer FH and Boven E: New highly lipophilic camptothecin BNP1350 is an effective drug in experimental human cancer. Int J Cancer. 88:260–266. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Colin D, Limagne E, Jeanningros S, Jacquel A, Lizard G, Athias A, Gambert P, Hichami A, Latruffe N, Solary E and Delmas D: Endocytosis of resveratrol via lipid rafts and activation of downstream signaling pathways in cancer cells. Cancer Prev Res (Phila). 4:1095–1106. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Li G, He S, Chang L, Lu H, Zhang H, Zhang H and Chiu J: GADD45α and Annexin A1 are involved in the apoptosis of HL-60 induced by resveratrol. Phytomedicine. 18:704–709. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Kartal M, Saydam G, Sahin F and Baran Y: Resveratrol triggers apoptosis through ceramide metabolizing genes in human K562 chronic myeloid leukemia cells. Nutr Cancer. 63:637–644. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Gupta SC, Kannappan R, Reuter S, Kim JH and Aggarwal BB: Chemosensitization of tumors by resveratrol. Ann N Y Acad Sci. 1215:150–160. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Banerjee S, Bueso-Ramos C and Aggarwal BB: Suppression of 7,12-dimethylbenz(a)anthracene-induced mammary carcinogenesis in rats by resveratrol: Role of nuclear factor-kappaB, cyclooxygenase 2, and matrix metalloproteinase 9. Cancer Res. 62:4945–4954. 2002.PubMed/NCBI

30 

Li ZG, Hong T, Shimada Y, Komoto I, Kawabe A, Ding Y, Kaganoi J, Hashimoto Y and Imamura M: Suppression of N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumorigenesis in F344 rats by resveratrol. Carcinogenesis. 23:1531–1536. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Lee EO, Lee HJ, Hwang HS, Ahn KS, Chae C, Kang KS, Lu J and Kim SH: Potent inhibition of Lewis lung cancer growth by heyneanol A from the roots of Vitis amurensis through apoptotic and anti-angiogenic activities. Carcinogenesis. 27:2059–2069. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Liu HS, Pan CE, Yang W and Liu XM: Antitumor and immunomodulatory activity of resveratrol on experimentally implanted tumor of H22 in Balb/c mice. World J Gastroenterol. 9:1474–1476. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Tessitore L, Davit A, Sarotto I and Caderni G: Resveratrol depresses the growth of colorectal aberrant crypt foci by affecting bax and p21(CIP) expression. Carcinogenesis. 21:1619–1622. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Baur JA and Sinclair DA: Therapeutic potential of resveratrol: The in vivo evidence. Nat Rev Drug Discov. 5:493–506. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Kimura Y and Okuda H: Resveratrol isolated from Polygonum cuspidatum root prevents tumor growth and metastasis to lung and tumor-induced neovascularization in lewis lung carcinoma-bearing mice. J Nutr. 131:1844–1849. 2001.PubMed/NCBI

36 

Busquets S, Ametller E, Fuster G, Olivan M, Raab V, Argilés JM and López-Soriano FJ: Resveratrol, a natural diphenol, reduces metastatic growth in an experimental cancer model. Cancer Lett. 245:144–148. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Weng YL, Liao HF, Li AF, Chang JC and Chiou RY: Oral administration of resveratrol in suppression of pulmonary metastasis of BALB/c mice challenged with CT26 colorectal adenocarcinoma cells. Mol Nutr Food Res. 54:259–267. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Cadenas S and Barja G: Resveratrol, melatonin, vitamin E, and PBN protect against renal oxidative DNA damage induced by the kidney carcinogen KBrO3. Free Radic Biol Med. 26:1531–1537. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Quincozes-Santos A, Andreazza AC, Nardin P, Funchal C, Gonçalves CA and Gottfried C: Resveratrol attenuates oxidative-induced DNA damage in C6 glioma cells. Neurotoxicology. 28:886–891. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Burkhardt S, Reiter RJ, Tan DX, Hardeland R, Cabrera J and Karbownik M: DNA oxidatively damaged by chromium(III) and H(2)O(2) is protected by the antioxidants melatonin, N(1)-acetyl-N(2)-formyl-5-methoxykynuramine, resveratrol and uric acid. Int J Biochem Cell Biol. 33:775–783. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Liu GA and Zheng RL: Protection against damaged DNA in the single cell by polyphenols. Pharmazie. 57:852–854. 2002.PubMed/NCBI

42 

Russell GK, Gupta RC and Vadhanam MV: Effect of phytochemical intervention on dibenzo[a,l]pyrene-induced DNA adduct formation. Mutat Res. 774:25–32. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Singh K, Bhori M and Marar T: α-Tocopherol mediated amelioration of camptothecin-induced free radical damage to avert cardiotoxicities. Hum Exp Toxicol. 34:380–389. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Singh KC, Kaur R and Marar T: Ameliorative effect of vitamin E on chemotherapy induced side effects in rat liver. J Pharmacol Toxicol. 6:481–492. 2011. View Article : Google Scholar

45 

Singh KC and Marar T: Acute toxicity of camptothecin and influence of α-tocopherol on hematological and biochemical parameters. J Cell Tiss Res. 11:2833–2837. 2011.

46 

Guo Y, Shi M, Shen X, Yang C, Yang L and Zhang J: Capecitabine plus irinotecan versus 5-FU/leucovorin plus irinotecan in the treatment of colorectal cancer: A meta-analysis. Clin Colorectal Cancer. 13:110–118. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Vanhoefer U, Harstrick A, Achterrath W, Cao S, Seeber S and Rustum YM: Irinotecan in the treatment of colorectal cancer: Clinical overview. J Clin Oncol. 19:1501–1518. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Fujita K, Kubota Y, Ishida H and Sasaki Y: Irinotecan, a key chemotherapeutic drug for metastatic colorectal cancer. World J Gastroenterol. 21:12234–12248. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 14 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zunino SJ and Zunino SJ: Resveratrol-3-O-glucuronide and resveratrol-4'-O-glucuronide reduce DNA strand breakage but not apoptosis in Jurkat T cells treated with camptothecin. Oncol Lett 14: 2517-2522, 2017
APA
Zunino, S.J., & Zunino, S.J. (2017). Resveratrol-3-O-glucuronide and resveratrol-4'-O-glucuronide reduce DNA strand breakage but not apoptosis in Jurkat T cells treated with camptothecin. Oncology Letters, 14, 2517-2522. https://doi.org/10.3892/ol.2017.6392
MLA
Zunino, S. J., Storms, D. H."Resveratrol-3-O-glucuronide and resveratrol-4'-O-glucuronide reduce DNA strand breakage but not apoptosis in Jurkat T cells treated with camptothecin". Oncology Letters 14.2 (2017): 2517-2522.
Chicago
Zunino, S. J., Storms, D. H."Resveratrol-3-O-glucuronide and resveratrol-4'-O-glucuronide reduce DNA strand breakage but not apoptosis in Jurkat T cells treated with camptothecin". Oncology Letters 14, no. 2 (2017): 2517-2522. https://doi.org/10.3892/ol.2017.6392