Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Transl Oncol ; 6(5): 596-606, 2013.
Article in English | MEDLINE | ID: mdl-24151540

ABSTRACT

Intestinal commensal bacteria have recently been shown to trigger macrophages to produce diffusible clastogens (or chromosome-breaking factors) through a bystander effect (BSE) that mediates DNA damage and induces chromosomal instability in neighboring cells. Colon macrophages appear central to colon carcinogenesis and BSE through the expression of tumor necrosis factor-α (TNF-α) and cyclooxygenase-2 (COX-2). The former induces netrin-1, a regulator of intestinal epithelial cell apoptosis, and the latter generates trans-4-hydroxy-2-nonenal (4-HNE), an endogenous mutagen. To test whether colon macrophages are key effectors for BSE, we depleted these cells in interleukin-10 knockout mice colonized with Enterococcus faecalis using encapsulated liposomal clodronate (ELC), a bisphosphonate that causes macrophage apoptosis. We observed that E. faecalis polarizes colon macrophages to an M1 phenotype. In addition, depleting these cells suppressed COX-2 and TNF-α, blocked the formation of 4-HNE protein adducts, and inhibited up-regulation of netrin-1-all markers for BSE. Finally, treatment with ELC prevented colitis, ß-catenin activation, and cancer formation. These results show that selected human commensals can polarize colon macrophages to the M1 phenotype and, when activated, serve as the key effector for bacterial-induced BSE. Our findings suggest that depleting M1-polarized macro-phages is a mechanism for the chemopreventive activity of bisphosphonates and that it represents a new strategy for preventing colon cancer induced by intestinal commensals.

2.
Cancer Prev Res (Phila) ; 6(3): 206-16, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23321929

ABSTRACT

Infection of macrophages by the human intestinal commensal Enterococcus faecalis generates DNA damage and chromosomal instability in mammalian cells through bystander effects. These effects are characterized by clastogenesis and damage to mitotic spindles in target cells and are mediated, in part, by trans-4-hydroxy-2-nonenal (4-HNE). In this study, we investigated the role of COX and lipoxygenase (LOX) in producing this reactive aldehyde using E. faecalis-infected macrophages and interleukin (IL)-10-knockout mice colonized with this commensal. 4-HNE production by E. faecalis-infected macrophages was significantly reduced by COX and LOX inhibitors. The infection of macrophages led to decreased Cox1 and Alox5 expression whereas COX-2 and 4-HNE increased. Silencing Alox5 and Cox1 with gene-specific siRNAs had no effect on 4-HNE production. In contrast, silencing Cox2 significantly decreased 4-HNE production by E. faecalis-infected macrophages. Depleting intracellular glutathione increased 4-HNE production by these cells. Next, to confirm COX-2 as a source for 4-HNE, we assayed the products generated by recombinant human COX-2 and found 4-HNE in a concentration-dependent manner using arachidonic acid as a substrate. Finally, tissue macrophages in colon biopsies from IL-10-knockout mice colonized with E. faecalis were positive for COX-2 by immunohistochemical staining. This was associated with increased staining for 4-HNE protein adducts in surrounding stroma. These data show that E. faecalis, a human intestinal commensal, can trigger macrophages to produce 4-HNE through COX-2. Importantly, it reinforces the concept of COX-2 as a procarcinogenic enzyme capable of damaging DNA in target cells through bystander effects that contribute to colorectal carcinogenesis.


Subject(s)
Aldehydes/metabolism , Cyclooxygenase 2/metabolism , Enterococcus faecalis , Gram-Positive Bacterial Infections/metabolism , Macrophages/metabolism , Macrophages/microbiology , Animals , Blotting, Western , Cyclooxygenase Inhibitors/pharmacology , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Knockout , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
3.
Cancer Res ; 72(20): 5219-29, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22915753

ABSTRACT

Macrophage-induced bystander effects have been implicated as an important mediator of chromosomal instability and colon cancer triggered by Enterococcus faecalis, a human intestinal commensal bacteria. There is little understanding about how inflammatory cytokines mediate bystander effects, but questions in this area are important because of the pivotal contributions made by inflammatory processes to cancer initiation and progression. Here, we report that the central proinflammatory cytokine TNF-α acts as a diffusible mediator of the bystander effects induced by macrophages, an effect caused by a proliferation of macrophages that trigger epithelial cell production of Netrin-1, a neuronal guidance molecule. TNF-α-mediated bystander assays used a murine coculture system of primary colonic epithelial cells and E. faecalis-infected macrophages (in vitro), with an interleukin 10 (IL-10)-deficient mouse model of colon cancer that involves long-term colonization with E. faecalis (in vivo). In cell cocultures, we observed increased expression of the TNF-α receptor Tnfrsf1b and Netrin-1. These effects were blocked by anti-TNF-α antibody or by pretreatment with an inhibitor of NF-κB signaling. RNAi-mediated attenuation of Tnfrsf1b decreased TNF-α-induced netrin-1 production and augmented epithelial cell apoptosis in culture. Extending these observations, colon biopsies from E. faecalis-colonized IL-10(-/-) mice exhibited crypt hyperplasia and increased staining for macrophages, TNF-α, netrin-1, NF-κB, Tnfrsf1b, and the proliferation marker proliferating cell nuclear antigen while also displaying a reduction in epithelial cell apoptosis. Together, our results define a pathway for macrophage-induced bystander effects in which TNF-α triggers TNFRSF1b receptor signaling leading to increased production of Netrin-1, crypt hyperplasia, and decreased epithelial cell apoptosis. In elucidating an important commensal-associated proinflammatory mechanism in the intestinal microenvironment, our work highlights the role of Netrin-1 and a specific TNF-α receptor as candidate targets to prevent or treat colorectal cancer.


Subject(s)
Bystander Effect/physiology , Macrophages/cytology , Nerve Growth Factors/physiology , Tumor Necrosis Factor-alpha/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis/physiology , Cell Line , Coculture Techniques , Enterococcus faecalis/physiology , Gene Silencing , Immunohistochemistry , Mice , NF-kappa B/metabolism , Nerve Growth Factors/biosynthesis , Netrin-1 , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/biosynthesis
4.
Gastroenterology ; 142(3): 543-551.e7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22108198

ABSTRACT

BACKGROUND & AIMS: Enterococcus faecalis is a human intestinal commensal that produces extracellular superoxide and promotes chromosome instability via macrophage-induced bystander effects. We investigated the ability of 4-hydroxy-2-nonenal (4-HNE), a diffusible breakdown product of ω-6 polyunsaturated fatty acids, to mediate these effects. METHODS: 4-HNE was purified from E faecalis-infected macrophages; its genotoxicity was assessed in human colon cancer (HCT116) and primary murine colon epithelial (YAMC) cell lines. RESULTS: 4-HNE induced G(2)-M cell cycle arrest, led to formation γH2AX foci, and disrupted the mitotic spindle in both cell lines. Binucleate tetraploid cells that formed after incubation with 4-HNE were associated with the activation of stathmin and microtubule catastrophe. Silencing glutathione S-transferase α4, a scavenger of 4-HNE, increased the susceptibility of epithelial cells to 4-HNE-induced genotoxicity. Interleukin-10 knockout mice colonized with superoxide-producing E faecalis developed inflammation and colorectal cancer, whereas colonization with a superoxide-deficient strain resulted in inflammation but not cancer. 4-HNE-protein adducts were found in the lamina propria and macrophages in areas of colorectal inflammation. CONCLUSIONS: 4-HNE can act as an autochthonous mitotic spindle poison in normal colonic epithelial and colon cancer cells. This finding links the macrophage-induced bystander effects to colorectal carcinogenesis.


Subject(s)
Aldehydes/metabolism , Autocrine Communication , Bystander Effect , Colon/microbiology , DNA Damage , Enterococcus faecalis/pathogenicity , Epithelial Cells/microbiology , Gram-Positive Bacterial Infections/microbiology , Macrophages/microbiology , Animals , Biopsy , Coculture Techniques , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Disease Models, Animal , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , G2 Phase Cell Cycle Checkpoints , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Gram-Positive Bacterial Infections/genetics , Gram-Positive Bacterial Infections/metabolism , Gram-Positive Bacterial Infections/pathology , HCT116 Cells , Histones/metabolism , Humans , Interleukin-10/deficiency , Interleukin-10/genetics , Macrophages/metabolism , Mice , Mice, Knockout , RNA Interference , Spindle Apparatus/metabolism , Spindle Apparatus/pathology , Stathmin/metabolism , Tetraploidy , Time Factors , Transfection
5.
J Med Microbiol ; 57(Pt 10): 1193-1204, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18809545

ABSTRACT

Enterococcus faecalis is an intestinal commensal that cannot synthesize porphyrins and only expresses a functional respiratory chain when provided with exogenous haematin. In the absence of haematin, E. faecalis reverts to fermentative metabolism and produces extracellular superoxide that can damage epithelial-cell DNA. The acute response of the colonic mucosa to haematin-starved E. faecalis was identified by gene array. E. faecalis was inoculated into murine colons using a surgical ligation model that preserved tissue architecture and homeostasis. The mucosa was exposed to haematin-starved E. faecalis and compared with a control consisting of the same strain grown with haematin. At 1 h post-inoculation, 6 mucosal genes were differentially regulated and this increased to 42 genes at 6 h. At 6 h, a highly significant biological interaction network was identified with functions that included nuclear factor-kappaB (NF-kappaB) signalling, apoptosis and cell-cycle regulation. Colon biopsies showed no histological abnormalities by haematoxylin and eosin staining. Immunohistochemical staining, however, detected NF-kappaB activation in tissue macrophages using antibodies to the nuclear localization sequence for p65 and the F4/80 marker for murine macrophages. Similarly, haematin-starved E. faecalis strongly activated NF-kappaB in murine macrophages in vitro. Furthermore, primary and transformed colonic epithelial cells activated the G2/M checkpoint in vitro following exposure to haematin-starved E. faecalis. Modulation of this cell-cycle checkpoint was due to extracellular superoxide produced as a result of the respiratory block in haematin-starved E. faecalis. These results demonstrate that the uniquely dichotomous metabolism of E. faecalis can significantly modulate gene expression in the colonic mucosa for pathways associated with inflammation, apoptosis and cell-cycle regulation.


Subject(s)
Colon/metabolism , Colon/microbiology , Enterococcus faecalis/metabolism , Gene Expression Regulation/physiology , Hemin , Animals , Cell Cycle/physiology , Cell Line, Tumor , Colon/cytology , Enterococcus faecalis/genetics , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrin-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Exp Biol Med (Maywood) ; 229(11): 1186-95, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564446

ABSTRACT

Enterococcus faecalis is a human intestinal commensal that produces extracellular superoxide, hydrogen peroxide, and hydroxyl radical while colonizing the intestinal tract. To determine whether dietary factors implicated in colorectal cancer affect oxidant production by E. faecalis, radicals were measured in rats colonized with this microorganism while on diets supplemented with iron or phytic acid. Hydroxyl radical activity was measured by assaying for aromatic hydroxylation products of D-phenylalanine using reverse-phase high-performance liquid chromatography and electrochemical detection. In vitro, as expected, iron enhanced, and phytic acid decreased, hydroxyl radical formation by E. faecalis. For rats colonized with E. faecalis given supplemental dietary iron (740 mg elemental iron as ferric phosphate per kg diet) or phytic acid (1.2% w/w), no differences were found in concentrations of urinary ortho- or meta- isomers of D-phenylalanine compared to rats on a basal diet. Aqueous radicals in colonic contents were further assessed ex vivo by electron spin resonance using 5,5-dimethyl-1-pyrroline-N-oxide as a spin trap. Mixtures of thiyl (sulfur-centered) and oxygen-centered radicals were detected across all diets. In vitro, similar spectra were observed when E. faecalis was incubated with hydrogen sulfide, air-oxidized cysteine, or an alkylsulfide, as typical sulfur-containing compounds that might occur in colonic contents. In conclusion, intestinal colonization with E. faecalis in a rat model generates both thiyl and oxygen-centered radicals in colonic contents. Radical formation, however, was not significantly altered by short-term dietary supplementation with iron or phytic acid.


Subject(s)
Enterococcus faecalis/metabolism , Free Radicals/metabolism , Intestines/microbiology , Iron/pharmacology , Phytic Acid/pharmacology , Animals , Chromatography, High Pressure Liquid , Diet , Dietary Supplements/microbiology , Electron Spin Resonance Spectroscopy , Enterococcus faecalis/drug effects , Free Radicals/analysis , Gram-Positive Bacterial Infections/metabolism , Intestines/drug effects , Male , Phenylalanine/urine , Rats
7.
Diabetes ; 52(8): 1994-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12882915

ABSTRACT

Free radical formation evoked by proinflammatory cytokines has been suggested to be involved in the destruction of beta-cells in the course of type 1 diabetes development. However, there is no direct evidence to support this hypothesis. In this study, we used electron paramagnetic resonance spectroscopy in conjunction with spin-trapping methodology to directly determine whether cytokines give rise to free radical formation in the islets. Our results demonstrate that direct, in vivo administration of tumor necrosis factor-alpha (1,000 units), interleukin-1beta (1,000 units), and interferon-gamma (2,000 units) into the rat pancreas through a bile duct cannula leads to the formation of lipid-derived free radicals in this tissue. These free radicals most likely are generated by the beta-cells because previous depletion of these cells by streptozotocin abolished the cytokine-induced free radical formation. Furthermore, macrophage depletion was found to decrease the production of free radicals. Inhibition of the enzyme inducible cyclooxygenase (COX-2) and the transcription factor nuclear factor-kappaB (NF-kappaB) significantly diminished the free radicals' signal intensity, implicating these factors in the formation of free radicals. We have also demonstrated that cytokine treatment leads to the activation of NF-kappaB in the pancreatic islets of the rats.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/enzymology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Antineoplastic Agents/pharmacology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Free Radicals/metabolism , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Isoenzymes/antagonists & inhibitors , Macrophages/cytology , Male , NF-kappa B/metabolism , Nitrobenzenes/pharmacology , Rats , Rats, Wistar , Sulfonamides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
8.
Shock ; 19(3): 252-6, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12630525

ABSTRACT

Preadministration of antioxidants such as pyrrolidine dithiocarbamate (PDTC) and phenyl N-tert-butyl nitrone (PBN) protects animals from lethality in sepsis models. However, the requirement of preadministration greatly diminishes the clinical significance of these studies. Although the synthetic antioxidant PBN has been shown to effectively protect rodents from lethality in endotoxemia (lipopolysaccharide [LPS] model), preliminary screening indicates that pre- or postadministration of PBN does not protect in the rat cecal ligation and puncture (CLP) model. We show in this report that in a rat CLP model, the administration of PBN (150 mg/kg, 30 min after CLP) followed by the antibiotic imipenem (IMP; 10 mg/kg, 1 h after CLP) significantly increased survival compared with other single treatment groups. Previously, we have shown that PBN's protection in a rat LPS model is mediated by the overproduction of the anti-inflammatory cytokine interleukin (IL)-10. We show in this study that the increase in survival found in the PBN + IMP-treated group was abrogated by immunoneutralization with anti-IL-10 antibody, indicating that endogenous IL-10 is an effective protective factor. Plasma LPS levels were shown to be elevated after imipenem treatment, and the increased LPS level could have assisted to overproduce endogenous IL-10, as in the case of the PBN-treated LPS model. Statistical analysis indicated that the increase of IL-10 in PBN + IMP-treated group at early time period has significant association to the improvement of survival.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antioxidants/therapeutic use , Bacterial Infections/prevention & control , Cecum/physiology , Proline/analogs & derivatives , Sepsis/prevention & control , Animals , Cyclic N-Oxides , Disease Models, Animal , Drug Synergism , Interleukin-10/blood , Interleukin-6/blood , Kinetics , Lipopolysaccharides/blood , Lipopolysaccharides/toxicity , Male , Nitrogen Oxides/therapeutic use , Proline/therapeutic use , Punctures , Rats , Rats, Wistar , Thiocarbamates/therapeutic use , Time Factors , Tumor Necrosis Factor-alpha/metabolism
9.
Free Radic Biol Med ; 33(6): 818-26, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12208369

ABSTRACT

Enterococcus faecalis is an intestinal commensal that produces extracellular superoxide (O(2)(*-)) through autoxidation of membrane-associated demethylmenaquinone. To assess free radical production by E. faecalis in vivo, intestinal tracts of rats were colonized using wild-type E. faecalis or a mutant strain with attenuated O(2)(*-) production. Ex vivo electron paramagnetic resonance spin trapping study of colonic contents (mean +/- SD) showed 1.4 +/- 1.5 and 0.094 +/- 0.24 microM 5,5-dimethyl-1-pyrroline-N-oxide-hydroxyl radical adduct/gm stool for rats colonized with wild-type and mutant strains, respectively (p = .002). In vivo hydroxyl radical production was further assayed by aromatic hydroxylation using phenyl N-tert-butylnitrone (PBN) and D-phenylalanine. Hydroxylated PBN and D-phenylalanine products were recovered from stool (microM/gm colonic contents/10(9) colony forming units) and urine (microM/h/ml), respectively, and quantified using electrochemical detection. Hydroxylated (OH) PBNs and isomeric tyrosines (hydroxylated phenylalanine) were significantly increased (mean +/- SD) for rats colonized with wild-type E. faecalis (2-OH PBN, 63 +/- 58; 3-OH PBN, 63 +/- 84; ortho-tyrosine, 31 +/- 27; meta-tyrosine, 17 +/- 14) compared to the mutant strain (2-OH PBN, 2.5 +/- 7.3 (p < .001); 3-OH PBN, 3.9 +/- 12.3 (p = .01); ortho-tyrosine, 1.9 +/- 6.0 (p < .001); meta-tyrosine, 1.5 +/- 3.4 (p = .03)). Similar differences were observed following in vitro incubations of these bacteria with aromatic targets. These results confirm in vivo production of hydroxyl radical by E. faecalis colonizing the intestine, and indicate this bacterium may be a potent source of oxidative stress on the intestinal epithelium.


Subject(s)
Enterococcus faecalis/metabolism , Hydroxyl Radical , Intestines/microbiology , Animals , Chromatography, High Pressure Liquid , Colon/microbiology , Colon/pathology , Electron Spin Resonance Spectroscopy , Free Radical Scavengers/pharmacology , Free Radicals , Hydrogen Peroxide/pharmacology , Nitrobenzenes , Nitrogen Oxides/pharmacology , Oxidative Stress , Phenylalanine/pharmacology , Rats , Reactive Oxygen Species , Superoxide Dismutase/metabolism , Tyrosine/urine
10.
Shock ; 17(3): 210-6, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11900340

ABSTRACT

The free radical trapping compound phenyl N-tert-butylnitrone (PBN) provides potent protection against lethal endotoxemia in rodents, but the mechanism of this protection is not well understood. The objective of this study was to show that PBN administration in lipopolysaccharide- (LPS) induced endotoxemia promotes enhanced production of endogenous interleukin 10 (IL-10), and the expressed IL-10 is a causal factor in the protection from endotoxemia. We show the amplified expression of IL-10 in liver and plasma in PBN- (150 mg/kg) plus LPS- (4 mg/kg) treated rats using ribonuclease protection assay (RPA) and ELISA. In situ hybridization was utilized to visualize the overexpression of the IL-10 gene, and ELISA was used to determine plasma IL-10 and TNFalpha levels. Plasma IL-10 showed a 3-fold increase in PBN/LPS- treated rats compared to those treated with LPS alone, and in contrast, TNFalpha level decreased by more than 90%. However, the administration of PBN alone induced no IL-10 production. Immunoneutralization of IL-10 through anti-IL-10 antibody administration to PBN/LPS-treated rats abrogated PBN's suppression of systemic nitric oxide (NO) formation, a surrogate marker for the severity of endotoxemia, indicating that IL-10 is a causal factor for the protection. In these experiments, systemic NO level was quantified using an in vivo electron paramagnetic resonance (EPR) NO-trapping technique. Gel-shift and immunohistochemical analyses indicated that the transcription factor NF-kappaB was deactivated after PBN treatment, suggesting that NF-kappaB deactivation is closely involved in IL-10 overexpression.


Subject(s)
Endotoxemia/metabolism , Endotoxemia/prevention & control , Interleukin-10/metabolism , Nitrogen Oxides/pharmacology , Animals , Cyclic N-Oxides , Cytokines/drug effects , Cytokines/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , In Situ Hybridization , Interleukin-10/genetics , Interleukin-10/immunology , Lipopolysaccharides , Male , NF-kappa B/metabolism , Nitric Oxide/metabolism , RNA, Messenger , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism
11.
Carcinogenesis ; 23(3): 529-36, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11895869

ABSTRACT

Enterococcus faecalis is a commensal microorganism of the human intestinal tract that produces substantial extracellular superoxide (O(-)(2)), and derivative reactive oxygen species such as H(2)O(2) and hydroxyl radical, through autoxidation of membrane-associated demethylmenaquinone. Because these oxidants may be important as a cause of chromosomal instability (CIN) associated with sporadic adenomatous polyps and colorectal cancer, the ability of E.faecalis to damage eukaryotic cell DNA was examined using the alkaline lysis single cell gel electrophoresis (comet) assay. Both Chinese hamster ovary and HT-29 intestinal epithelial cells showed increased DNA damage after co-incubation with wild-type E. faecalis strain OG1RF, but not a transposon-inactivated mutant with attenuated extracellular O(-)(2) production. E. faecalis-mediated DNA damage was prevented by catalase, but not manganese superoxide dismutase, indicating H(2)O(2) arising from O(-)(2) was the genotoxin. In a rat model of intestinal colonization, OG1RF resulted in significantly higher stool concentrations of H(2)O(2) and 5,5-dimethyl-1-pyrroline N-oxide adducts of hydroxyl and thiyl radicals, as identified by electron spin resonance-spin trapping, compared with rats colonized with a mutant strain having attenuated O(-)(2) production. Using the comet assay, luminal cells from the colon of rats colonized with O(-)(2)-producing E. faecalis showed significantly increased DNA damage compared with control rats colonized with the mutant. These findings suggest a potentially profound role for extracellular free radical production by E. faecalis in promoting CIN associated with sporadic adenomatous polyps and colorectal cancer.


Subject(s)
Colon/microbiology , Colon/pathology , DNA Damage , Enterococcus faecalis/metabolism , Epithelial Cells/pathology , Hydrogen Peroxide/metabolism , Superoxides/metabolism , Animals , CHO Cells , Catalase/metabolism , Cells, Cultured , Colon/metabolism , Comet Assay , Cricetinae , DNA Damage/drug effects , Electron Spin Resonance Spectroscopy , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Ileum/cytology , Ileum/metabolism , Rats , Xanthine/pharmacology , Xanthine Oxidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL