Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cancer Prev Res (Phila) ; 17(4): 157-167, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38286439

ABSTRACT

Cigarette smoke is a rich source of free radicals that can promote oxidative stress and carcinogenesis, including head and neck squamous cell carcinoma (HNSCC) development; importantly, 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) and 8-iso-prostaglandin F2α (8-isoprostane) are biomarkers of oxidative stress. Several mechanisms, including the antioxidant properties of black raspberry (BRB), account for their chemopreventive effects. In the present clinical trial, we tested the hypothesis that BRB administration reduces biomarkers levels of oxidative stress in buccal cells and urine of smokers. One week after enrolling 21 smokers, baseline buccal cells and urine samples were collected before the administration of BRB lozenges for 8 weeks (5/day, 1 gm BRB/lozenge). Buccal cells and urine samples were collected at the middle and the end of BRB administration. The last samples were collected after the BRB cessation (washout period). We analyzed levels of 8-oxodG and 8-isoprostane (LC/MS-MS), urinary cotinine (ELISA), and creatinine (spectrophotometry). BRB significantly reduced the levels of 8-oxodG by 17.08% (P = 0.00079) in buccal cells and 12.44% (P = 0.034) in urine at the middle of BRB administration as compared with baseline; the corresponding values at the end of BRB administration were 16.46% (P = 0.026) in buccal cells and 25.72% (P = 0.202) in urine. BRB had no significant effect on the levels of urinary 8-isoprostane. BRB's capacity to inhibit 8-oxodG formation of smokers' buccal cells and urine is clearly evident and the reduction in 8-oxodG suggests that antioxidant abilities are central to BRB's HNSCC chemopreventive properties. PREVENTION RELEVANCE: Cigarette smoke contains highly active components namely free radicals that can promote oxidative stress and oral cancer. We found that black raspberry (BRB) inhibited the formation of oxidative stress markers in the oral cavity and urine of smokers suggesting the antioxidant abilities of BRB in preventing oral cancer.


Subject(s)
Head and Neck Neoplasms , Mouth Neoplasms , Rubus , Humans , 8-Hydroxy-2'-Deoxyguanosine/pharmacology , 8-Hydroxy-2'-Deoxyguanosine/therapeutic use , Antioxidants/pharmacology , Biomarkers/urine , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , Deoxyguanosine/urine , Free Radicals/pharmacology , Free Radicals/therapeutic use , Mouth Mucosa/pathology , Mouth Neoplasms/etiology , Mouth Neoplasms/prevention & control , Mouth Neoplasms/drug therapy , Oxidative Stress , Smokers , Squamous Cell Carcinoma of Head and Neck
2.
Pharmacol Res ; 199: 106990, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37984506

ABSTRACT

Resistance to temozolomide (TMZ), the frontline chemotherapeutic agent for glioblastoma (GBM), has emerged as a formidable obstacle, underscoring the imperative to identify alternative therapeutic strategies to improve patient outcomes. In this study, we comprehensively evaluated a novel agent, O6-methyl-2'-deoxyguanosine-5'-triphosphate (O6-methyl-dGTP) for its anti-GBM activity both in vitro and in vivo. Notably, O6-methyl-dGTP exhibited pronounced cytotoxicity against GBM cells, including those resistant to TMZ and overexpressing O6-methylguanine-DNA methyltransferase (MGMT). Mechanistic investigations revealed that O6-methyl-dGTP could be incorporated into genomic DNA, disrupting nucleotide pools balance, and inducing replication stress, resulting in S-phase arrest and DNA damage. The compound exerted its anti-tumor properties through the activation of AIF-mediated apoptosis and the parthanatos pathway. In vivo studies using U251 and Ln229 cell xenografts supported the robust tumor-inhibitory capacity of O6-methyl-dGTP. In an orthotopic transplantation model with U87MG cells, O6-methyl-dGTP showcased marginally superior tumor-suppressive activity compared to TMZ. In summary, our research, for the first time, underscores the potential of O6-methyl-dGTP as an effective candidate against GBM, laying a robust scientific groundwork for its potential clinical adoption in GBM treatment regimens.


Subject(s)
Glioblastoma , Polyphosphates , Humans , Glioblastoma/drug therapy , Glioblastoma/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Nucleosides/pharmacology , Nucleosides/therapeutic use , Caspases , Cell Line, Tumor , Temozolomide/pharmacology , Temozolomide/therapeutic use , Nucleotides , O(6)-Methylguanine-DNA Methyltransferase/metabolism , O(6)-Methylguanine-DNA Methyltransferase/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/therapeutic use , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , DNA , Drug Resistance, Neoplasm
3.
Med Gas Res ; 13(2): 72-77, 2023.
Article in English | MEDLINE | ID: mdl-36204786

ABSTRACT

Diabetic peripheral neuropathy (DPN) is a complex disorder caused by long-standing diabetes. Oxidative stress was considered the critical creed in this DPN pathophysiology. Hydrogen has antioxidative effects on diabetes mellitus and related complications. However, there is still no concern on the beneficial effects of hydrogen in DPN. This paper aimed to evaluate the effects of exogenous hydrogen to reduce the severity of DPN in streptozotocin-induced diabetic rats. Compared with hydrogen-rich saline treatment, hydrogen inhalation significantly reduced blood glucose levels in diabetic rats in the 4th and 8th weeks. With regard to nerve function, hydrogen administration significantly attenuated the decrease in the velocity of motor nerve conduction in diabetic animals. In addition, hydrogen significantly attenuated oxidative stress by reducing the level of malondialdehyde, reactive oxygen species, and 8-hydroxy-2-deoxyguanosine and meaningfully enhanced the antioxidant capability by partially restoring the activities of superoxide dismutase. Further studies showed that hydrogen significantly upregulated the expression of nuclear factor erythroid-2-related factor 2 and downstream proteins such as catalase and hemeoxygenase-1 in the nerves of diabetic animals. Our paper showed that hydrogen exerts significant protective effects in DPN by downregulating oxidative stress via the pathway of nuclear factor erythroid-2-related factor 2, which suggests its potential value in clinical applications.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Neuroprotective Agents , Animals , Rats , Antioxidants/metabolism , Antioxidants/pharmacology , Blood Glucose , Catalase/metabolism , Catalase/pharmacology , Catalase/therapeutic use , Deoxyguanosine/metabolism , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Hydrogen , Malondialdehyde , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Reactive Oxygen Species , Streptozocin , Superoxide Dismutase/metabolism , Superoxide Dismutase/pharmacology , Superoxide Dismutase/therapeutic use
4.
Cancer Cell ; 38(3): 400-411.e6, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32619407

ABSTRACT

Telomerase is an attractive target for anti-tumor therapy as it is almost universally expressed in cancer cells. Here, we show that treatment with a telomere-targeting drug, 6-thio-2'-deoxyguanosine (6-thio-dG), leads to tumor regression through innate and adaptive immune-dependent responses in syngeneic and humanized mouse models of telomerase-expressing cancers. 6-thio-dG treatment causes telomere-associated DNA damages that are sensed by dendritic cells (DCs) and activates the host cytosolic DNA sensing STING/interferon I pathway, resulting in enhanced cross-priming capacity of DCs and tumor-specific CD8+ T cell activation. Moreover, 6-thio-dG overcomes resistance to checkpoint blockade in advanced cancer models. Our results unveil how telomere stress increases innate sensing and adaptive anti-tumor immunity and provide strong rationales for combining telomere-targeting therapy with immunotherapy.


Subject(s)
Deoxyguanosine/analogs & derivatives , Membrane Proteins/immunology , Neoplasms/drug therapy , Telomerase/antagonists & inhibitors , Telomere/genetics , Thionucleosides/pharmacology , Adaptive Immunity/drug effects , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , HCT116 Cells , Humans , Immunity, Innate/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Telomerase/metabolism , Telomere/enzymology , Thionucleosides/therapeutic use , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Burden/immunology
5.
Cancer Res ; 80(5): 929-936, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31948943

ABSTRACT

Cell membrane transporters facilitate the passage of nucleobases and nucleosides for nucleotide synthesis and metabolism, and are important for the delivery of nucleoside analogues used in anticancer drug therapy. Here, we investigated if cell membrane transporters are involved in the cellular uptake of the nucleoside analogue DNA damage mediator 6-thio-2'-deoxyguanosine (6-thio-dG). A large panel of non-small cell lung cancer (NSCLC) cell lines (73 of 77) were sensitive to 6-thio-dG; only four NSCLC lines were resistant to 6-thio-dG. When analyzed by microarray and RNA sequencing, the resistant NSCLC cell lines clustered together, providing a molecular signature for patients that may not respond to 6-thio-dG. Significant downregulation of solute carrier family 43 A3 (SLC43A3), an equilibrative nucleobase transporter, was identified as a candidate in this molecular resistance signature. High levels of SLC43A3 mRNA predicted sensitivity to 6-thio-dG and therefore SLC43A3 could serve as a promising biomarker for 6-thio-dG sensitivity in patients with NSCLC. SIGNIFICANCE: These findings identify a biomarker of resistance to the telomeric DNA damage mediator 6-thio-2'-deoxyguanosine.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Amino Acid Transport Systems/metabolism , Biomarkers, Tumor/metabolism , Deoxyguanosine/analogs & derivatives , Lung Neoplasms/drug therapy , Thionucleosides/pharmacology , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Amino Acid Transport Systems/genetics , Animals , Cell Line, Tumor , DNA Damage/drug effects , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , Down-Regulation , Drug Resistance, Neoplasm , Female , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Lung/pathology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , RNA, Small Interfering/metabolism , Telomere/drug effects , Thionucleosides/therapeutic use , Xenograft Model Antitumor Assays
6.
Cornea ; 37(10): 1311-1317, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29923862

ABSTRACT

PURPOSE: To evaluate the therapeutic effects of topical 8-oxo-2'-deoxyguanosine (8-oxo-dG) on experimental ocular chemical injury models. METHODS: We created ocular chemical injury models with 8-week-old BALB/c mice (n = 70) by applying 100% ethanol; the mice were then treated with 8-oxo-dG eye drops 10 and 5 mg/mL and phosphate-buffered saline (PBS) twice daily. After 7 days, clinical findings such as corneal integrity, clarity, and neovascularization were assessed. Histology, immunohistochemistry findings, and inflammatory cytokine levels using real-time polymerase chain reactions in the corneas of the mice were also analyzed. RESULTS: Topical application of 8-oxo-dG eye drops resulted in a significant improvement of epithelial defects and clarity, dose dependently (each P < 0.001). Inflammatory cell infiltration and corneal stromal edema were also decreased in the 8-oxo-dG-treated mice compared with PBS-treated controls, based on hematoxylin and eosin staining. The expressions of F4/80 and neutrophil elastase-positive inflammatory cells and IL-1 and TNF-α cytokine levels were significantly reduced in the 8-oxo-dG group compared with the PBS group (each P < 0.01). CONCLUSIONS: Topical 8-oxo-dG application showed an excellent therapeutic effect in ocular chemical injury models by suppressing inflammation.


Subject(s)
Burns, Chemical/drug therapy , Corneal Injuries/drug therapy , Deoxyguanosine/analogs & derivatives , Eye Burns/drug therapy , Ophthalmic Solutions/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine , Analysis of Variance , Animals , Burns, Chemical/metabolism , Burns, Chemical/pathology , Cornea/pathology , Corneal Injuries/chemically induced , Corneal Injuries/pathology , Corneal Neovascularization/pathology , Corneal Stroma/pathology , Cytokines/metabolism , Deoxyguanosine/therapeutic use , Disease Models, Animal , Epithelium, Corneal/pathology , Ethanol , Eye Burns/metabolism , Eye Burns/pathology , Male , Mice , Mice, Inbred BALB C
7.
Biochemistry (Mosc) ; 82(13): 1686-1701, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29523066

ABSTRACT

8-Oxo-7,8-dihydroguanine (8-oxo-G) is a key biomarker of oxidative damage to DNA in cells, and its genotoxicity is well-studied. In recent years, it has been confirmed experimentally that free 8-oxo-G and molecules containing it are not merely inert products of DNA repair or degradation, but they are actively involved in intracellular signaling. In this review, data are systematized indicating that free 8-oxo-G and oxidized (containing 8-oxo-G) extracellular DNA function in the body as mediators of stress signaling and initiate inflammatory and immune responses to maintain homeostasis under the action of external pathogens, whereas exogenous 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dGuo) exhibits pronounced antiinflammatory and antioxidant properties. This review describes known action mechanisms of oxidized guanine and 8-oxo-G-containing molecules. Prospects for their use as a therapeutic target are considered, as well as a pharmaceutical agent for treatment of a wide range of diseases whose pathogenesis is significantly contributed to by inflammation and oxidative stress.


Subject(s)
Deoxyguanosine/analogs & derivatives , 8-Hydroxy-2'-Deoxyguanosine , Animals , Biomarkers , DNA Damage/drug effects , DNA Repair/drug effects , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , Guanine/analogs & derivatives , Humans , Inflammation/drug therapy , Oxidative Stress/drug effects
8.
Curr Atheroscler Rep ; 16(11): 452, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25252787

ABSTRACT

Oxidative stress due to an excess of reactive oxygen species (ROS) may play a role in the development and progression of cardiovascular disease (CVD). 8-hydroxy-2'-deoxyguanosine (8-OHdG) is a marker of oxidative DNA damage caused by ROS. This review aimed to assess the association between 8-OHdG and CVD by reviewing the literature. Studies in human subjects using either plasma or urine to determine 8-OHdG concentrations were surveyed. Eighteen relevant studies were found, of which 13 were case-control studies and five had a prospective design. Without exception, the case-control studies showed significant positive associations between 8-OHdG and CVD. In agreement, two prospective studies showed a significant association of 8-OHdG and heart failure. Furthermore, two prospective studies found a significant association between 8-OHdG and stroke, and finally, one prospective study showed a borderline significant (p = 0.08) association between coronary artery disease (CAD) patients developing a cardiac event and 8-OHdG concentrations. In conclusion, high levels of 8-OHdG in blood and urine are associated with atherosclerosis and heart failure, but further large prospective studies are needed to investigate 8-OHdG as a predictor for cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/drug therapy , Deoxyguanosine/analogs & derivatives , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Clinical Trials as Topic , DNA Damage , Deoxyguanosine/chemistry , Deoxyguanosine/therapeutic use , Humans
9.
Biochem Biophys Res Commun ; 443(2): 610-6, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24333420

ABSTRACT

Metabolic syndrome describes a group of clinical features that together increase the incidence of coronary artery disease, stroke and type 2 diabetes. Insulin resistance is a major risk factor for developing metabolic syndrome. A chronic state of inflammation accompanies the accumulation of surplus lipids in adipose and liver tissue, frequently involved in insulin resistance. 8-Oxo-2'-deoxyguanosine (8-Oxo-dG) is a potent anti-inflammatory agent that inactivates both Rac1 and Rac2 which are critical to initiating the inflammatory responses in various cell types, including macrophages. In this study, we explored whether 8-Oxo-dG suppressed a series of systemic inflammatory cascades, resulting in the amelioration of typical features of metabolic syndrome in obese mice. The results demonstrate that 8-Oxo-dG effectively improved hyperglycemia, dyslipidemia and fatty liver changes in obese mice. The level of biochemical markers indicative of systemic inflammation were reduced in 8-Oxo-dG treated mice, whereas serum levels of adiponectin, a crucial factor associated with improved metabolic syndrome, were enhanced. Our results demonstrate that 8-Oxo-dG effectively disrupts the pathogenesis of insulin resistance and obesity-associated metabolic syndrome.


Subject(s)
Cytokines/immunology , Deoxyguanosine/analogs & derivatives , Metabolic Syndrome/immunology , Metabolic Syndrome/prevention & control , Obesity/immunology , Obesity/prevention & control , 8-Hydroxy-2'-Deoxyguanosine , Animals , Deoxyguanosine/therapeutic use , Male , Mice , Mice, Inbred C57BL , Treatment Outcome
10.
World J Gastroenterol ; 18(4): 302-8, 2012 Jan 28.
Article in English | MEDLINE | ID: mdl-22294836

ABSTRACT

Reactive oxygen species (ROS) attack guanine bases in DNA easily and form 8-hydroxydeoxyguanosine (8-OHdG), which can bind to thymidine rather than cytosine, based on which, the level of 8-OHdG is generally regarded as a biomarker of mutagenesis consequent to oxidative stress. For example, higher levels of 8-OHdG are noted in Helicobacter pylori-associated chronic atrophic gastritis as well as gastric cancer. However, we have found that exogenous 8-OHdG can paradoxically reduce ROS production, attenuate the nuclear factor-κB signaling pathway, and ameliorate the expression of proinflammatory mediators such as interleukin (IL)-1, IL-6, cyclo-oxygenase-2, and inducible nitric oxide synthase in addition to expression of nicotinamide adenine dinucleotide phosphate oxidase (NOX)-1, NOX organizer-1 and NOX activator-1 in various conditions of inflammation-based gastrointestinal (GI) diseases including gastritis, inflammatory bowel disease, pancreatitis, and even colitis-associated carcinogenesis. Our recent finding that exogenous 8-OHdG was very effective in either inflammation-based or oxidative-stress-associated diseases of stress-related mucosal damage has inspired the hope that synthetic 8-OHdG can be a potential candidate for the treatment of inflammation-based GI diseases, as well as the prevention of inflammation-associated GI cancer. In this editorial review, the novel fact that exogenous 8-OHdG can be a functional molecule regulating oxidative-stress-induced gastritis through either antagonizing Rac-guanosine triphosphate binding or blocking the signals responsible for gastric inflammatory cascade is introduced.


Subject(s)
Anti-Inflammatory Agents/metabolism , Antineoplastic Agents/metabolism , Biomarkers/metabolism , Deoxyguanosine/analogs & derivatives , Gastrointestinal Diseases/metabolism , Oxidative Stress , 8-Hydroxy-2'-Deoxyguanosine , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Colitis/drug therapy , Deoxyguanosine/metabolism , Deoxyguanosine/therapeutic use , Gastrointestinal Diseases/physiopathology , Humans , Inflammation/drug therapy , Reactive Oxygen Species/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/physiopathology
11.
Biochem Pharmacol ; 81(1): 111-22, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20816670

ABSTRACT

Reactive oxygen species (ROS) attack guanine bases in DNA and form 8-hydroxydeoxyguanosine (8-OHdG), which has been regarded simply as an oxidative mutagenic by-product. On the other hand, our previous report showed paradoxically ROS attenuating action of generated 8-OHdG. In the current study, both in vitro and in vivo experiments were executed in order to document anti-oxidative and anti-inflammatory actions of 8-OHdG in cell model and to elucidate the therapeutic efficacy against water immersion restraint stress (WIRS)-induced gastritis animal model. Electron spin resonance measurements showed that 8-OHdG at >5µg/ml completely scavenged OH(-) radicals, which was further confirmed by checking 2'-7'-dichlorodihydrofluorescein diacetate (DCFDA) spectroscopy. On molecular assay, 8-OHdG antagonized the action of GTP on Rac, a small GTP binding protein, without affecting Rac-guanosine exchange factor (GEF) or phosphoinositide 3-kinases (PI3K) activity. In Raw264.7 cells, 8-OHdG was found to be associated with marked attenuations of NOX1, NOXO1, and NOXA1 accompanied with the decreased expressions of LPS-induced inflammatory mediators including COX-2, iNOS, IL-1ß, and IL-6. Similarly, 8-OHdG attenuated hypoxia-induced angiogenesis and platelet endothelial cell adhesion molecule-1 (PECAM-1), COX-2, iNOS, IL-8, and VEGF expressions in HUVEC cells. At transcriptional level, 8-OHdG inhibited the nuclear translocation of NF-κB, inhibitory κB kinase (IKK) ß kinase activation, and decreased phospho-IκBα levels. 8-OHdG efficiently ameliorated WIRS-induced gastric mucosal injury as evidenced with improvement of gross lesion index and attenuation of engaging mediators. Taken together, exogenous 8-OHdG can be a functional molecule regulating oxidative stress-induced gastritis through either antagonizing Rac-GTP binding or blocking the signals responsible for gastric inflammatory cascade.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Deoxyguanosine/analogs & derivatives , Gastritis/drug therapy , Stress, Physiological/physiology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Cell Line , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Specific Pathogen-Free Organisms , Transcription, Genetic
12.
Gene Ther ; 14(1): 86-92, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16885999

ABSTRACT

The multisubstrate deoxyribonucleoside kinase of Drosophila melanogaster (Dm-dNK) is investigated for possible use as a suicide gene in combined gene/chemotherapy of cancer. The enzyme has broader substrate specificity and higher catalytic rate compared to herpes simplex type 1 thymidine kinase and other known dNKs. Although the enzyme has broad substrate specificity, it has a preference for pyrimidine nucleosides and nucleoside analogs. We have evaluated the substrate specificity and kinetic properties of Dm-dNK proteins containing M88R, V84A+M88R or V84A+M88R+A110D mutations in the amino-acid sequence. These engineered enzymes showed a relative increase in phosphorylation of purine nucleoside analogs such as ganciclovir, 9-beta-D-arabinofuranosylguanine and 2',2'-difluorodeoxyguanosine compared to the wild-type enzyme. The mutant enzymes were expressed in an osteosarcoma thymidine kinase-deficient cell line and the sensitivity of the cell line to nucleoside analogs was determined. The cells expressing the M88R mutant enzyme showed the highest increased sensitivity to purine nucleoside analogs with 8- to 80-fold decreased inhibition constant IC(50) compared to untransduced control cells or cells expressing the wild-type nucleoside kinase. In summary, our data show that enzyme engineering can be used to shift the substrate specificity of the Dm-dNK to selectively increase the sensitivity of cells expressing the enzyme to purine nucleoside analogs.


Subject(s)
Antineoplastic Agents/therapeutic use , Drosophila melanogaster/enzymology , Genes, Transgenic, Suicide , Genetic Therapy/methods , Neoplasms/therapy , Phosphotransferases/genetics , Amino Acid Sequence , Animals , Antineoplastic Agents/metabolism , Arabinonucleosides/therapeutic use , Cell Line, Tumor , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/therapeutic use , Drosophila melanogaster/genetics , Ganciclovir/therapeutic use , Gene Expression , Genetic Engineering , Molecular Sequence Data , Mutagenesis, Site-Directed , Neoplasms/enzymology , Plasmids/administration & dosage , Sequence Alignment , Substrate Specificity , Transfection/methods
13.
Exp Mol Med ; 38(4): 417-27, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16953121

ABSTRACT

To elucidate the roles of 8-hydroxydeoxyguanosine (oh(8)dG), the nucleoside of 8-hydroxyguanine (oh(8)Gua), we examined the effects of oh(8)dG upon LPS-induced intercellular adhesion molecule-1 (ICAM-1) expression and the underlying mechanisms in brain microglial cells. We found that oh(8)dG reduces LPS-induced reactive oxygen species (ROS) production, STAT3 activation, and ICAM-1 expression. oh(8)dG also suppresses pro-inflammatory cytokines, such as TNF-alpha, IL-6 and IFN-gamma. Overexpression of dominant negative STAT3 completely diminshed STAT3-mediated ICAM-1 transcriptional activity. Chromatin immunoprecipitation studies revealed that oh(8)dG inhibited recruitment of STAT3 to the ICAM-1 promoter, followed by a decrease in ICAM-1 expression. Using mice lacking a functional Toll-like receptor 4 (TLR4), we demonstrated that, while TLR4+/+ microglia were activated by LPS, TLR4-/- microglia exhibited inactivated STAT3 in response to LPS. Evidently, LPS modulates STAT3-dependent ICAM-1 induction through TLR4-mdiated cellular responses. Oh(8)dG apparently plays a role in anti-inflammatory actions via suppression of ICAM-1 gene expression by blockade of the TLR4-STAT3 signal cascade in inflammation-enhanced brain microglia. Therefore, oh(8)dG in the cytosol probably functions as an anti-inflammatory molecule and should be considered as a candidate for development of anti-inflammatory agents.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Deoxyguanosine/analogs & derivatives , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/pharmacology , Microglia/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Brain/cytology , Brain/drug effects , Cell Survival/drug effects , Cytokines/biosynthesis , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , Encephalitis/drug therapy , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/physiology , Toll-Like Receptor 4/genetics
15.
Int J Oncol ; 24(4): 901-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15010828

ABSTRACT

Bacterial and synthetic DNAs containing CpG dinucleotides in specific sequence contexts (CpG DNA) activate the vertebrate immune system and produce potent Th1 immune responses. Recently, we reported immunomodulatory oligonucleotides (IMOs) containing 3'-3'-attached novel structures (immunomers) and synthetic immunostimulatory CpR (R=2'-deoxy-7-deazguanosine) dinucleotides show potent stimulatory activity with distinct cytokine secretion profiles. In the present study, we evaluated in vivo immunopharmacological and antitumor properties of second-generation immunomodulatory oligonucleotides (IMOs) either alone or in combination with chemotherapeutic agents. Repeated peritumoral administration of IMOs at 1 mg/kg to mice bearing established subcutaneous CT26 colon tumor or B16.F0 melanoma resulted in complete regression or strong inhibition of tumor growth. Direct peritoneal injection of IMOs at 2.5 mg/kg to mice bearing peritoneally implanted ascites CT26 or B16.F0 tumors completely eradicated or inhibited tumor growth. Treatment of mice bearing beta-gal expressing CT26.CL25 tumor with IMOs resulted in a significant tumor-specific CTL responses compared with treatment with a control non-CpG DNA or PBS. These responses correlated with IFN-gamma, but not IL-4 secreted in IMO, treated mice. A 5-fold increase in beta-gal specific IgG2a antibodies was found in mice, significantly increasing the IgG2a/IgG1 ratio. IMOs showed similar antitumor activity in both wt and IL-6 knockout (ko) C57BL/6 mice but failed to elicit activity in IL-12 ko C57BL/6 mice. Tumor-free mice from the IMO treatment group rejected the same tumor cell rechallenge, suggesting an adaptive immune response against these cells. Moreover, naïve mice quickly developed specific antitumor response without IMO treatment following adoptive transfer of splenocytes obtained from tumor free mice from the IMO treated group. Additionally, the co-administration of IMOs with chemotherapeutic agents, docetaxel and doxorubicin, resulted in synergistic antitumor effects in both B16.F0 melanoma and 4T1 breast carcinoma models. These results demonstrate potent antitumor activity of second-generation IMO compounds containing a synthetic CpR stimulatory motif in a broad spectrum of tumor models through induction of strong Th1 immune responses. IMO treatment resulted in the development of tumor-specific memory immune responses. No treatment-related toxicity was observed in mice at the doses and treatment schedules studied.


Subject(s)
Deoxyguanosine/analogs & derivatives , Deoxyguanosine/therapeutic use , Neoplasms/drug therapy , Oligonucleotides/therapeutic use , Th1 Cells/immunology , Adoptive Transfer , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , CpG Islands/immunology , Cytotoxicity, Immunologic , Drug Synergism , Female , Humans , Immunoglobulin G/blood , Interleukin-12/blood , Interleukin-12/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Survival Rate , Th1 Cells/metabolism , Tumor Cells, Cultured
16.
Hum Gene Ther ; 13(4): 543-51, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11874632

ABSTRACT

Ganciclovir (GCV) is widely used as a prodrug for selective activation in tumor cells expressing herpes simplex virus thymidine kinase (HSV-TK) because of its ability to induce multi-log cytotoxicity to HSV-TK-expressing as well as nonexpressing bystander cells. We now report that another substrate for HSV-TK, D-carbocyclic 2'-deoxyguanosine (CdG), induces multi-log cytotoxicity in HSV-TK-expressing and bystander cells at concentrations

Subject(s)
Antiviral Agents/pharmacology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/pharmacology , Ganciclovir/pharmacology , Prodrugs/pharmacology , Thymidine Kinase/biosynthesis , Viral Proteins/biosynthesis , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Cell Death/drug effects , Cell Death/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/virology , Deoxyguanosine/metabolism , Deoxyguanosine/therapeutic use , Enzyme Activation/drug effects , Ganciclovir/metabolism , Ganciclovir/therapeutic use , Gene Transfer Techniques , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/virology , Prodrugs/metabolism , Prodrugs/therapeutic use , Simplexvirus/enzymology , Thymidine Kinase/genetics , Tumor Cells, Cultured
17.
Br J Pharmacol ; 134(7): 1498-504, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11724756

ABSTRACT

1. We investigated the ability of a newly synthesized sugar derivative, OJ-R9188, [N-(2-tetradecylhexadecanoyl)-O-(L-alpha-fucofuranosyl)-D-seryl]-L-glutamic acid 1-methylamide 5-L-arginine salt, to block binding of selectins to their ligands in vitro and inhibit the infiltration of leukocytes in vivo. 2. OJ-R9188 prevented the binding of human E-, P- and L-selectin-IgG fusion proteins to immobilized sialyl Lewis(x) (sLe(x))-pentasaccharide glycolipid, with IC(50) values of 4.3, 1.3, and 1.2 microM, respectively. 3. In a mouse model of thioglycollate-induced peritonitis, OJ-R9188 at 10 mg kg(-1), i.v. inhibited neutrophil accumulation in the peritoneal cavity. In the IgE-mediated skin reaction, OJ-R9188 at 3 and 10 mg kg(-1), i.v. significantly inhibited extravasation of neutrophils and eosinophils into the inflammatory sites and at 10 mg kg(-1), i.v. also inhibited infiltration caused by picryl chloride-induced delayed-type hypersensitivity in mice. These results suggest that OJ-R9188 may be a useful selectin blocker, with activity against human and mouse E-, P- and L-selectins in vitro and in vivo, and that blocking selectin-sLe(x) binding is a promising strategy for the treatment of allergic skin diseases.


Subject(s)
Deoxyguanosine/pharmacology , Dermatitis/prevention & control , Free Radical Scavengers/pharmacology , Hypersensitivity, Delayed/prevention & control , Selectins/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Binding, Competitive/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/therapeutic use , Dermatitis/immunology , Dermatitis/metabolism , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , E-Selectin/metabolism , Free Radical Scavengers/therapeutic use , Humans , Hypersensitivity, Delayed/chemically induced , Hypersensitivity, Delayed/metabolism , Immunoglobulin G/metabolism , L-Selectin/metabolism , Male , Mice , Mice, Inbred BALB C , Oligosaccharides/metabolism , P-Selectin/metabolism , Peritonitis/chemically induced , Peritonitis/metabolism , Peritonitis/prevention & control , Picryl Chloride/administration & dosage , Polysaccharides/pharmacology , Recombinant Fusion Proteins/metabolism , Sialyl Lewis X Antigen , Thioglycolates/administration & dosage
18.
J Neurooncol ; 51(1): 19-24, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11349876

ABSTRACT

Treatment of malignant brain tumors with chloroethylnitrosoureas (CENUs) in addition to surgical resection and radiotherapy remains the foundation of glioma therapy. However, the clinical response to CENUs is at best modest. A novel analogue of nitrosoureas, 2-chloroethyl-3-sarcosinamide-1-nitrosourea (SarCNU), as compared to the standard CENU, 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), has been demonstrated to have increased anticancer effects both in vitro and in vivo. Unfortunately, many human tumors have been known to be resistant to CENUs since they express DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). In order to assess whether SarCNU has an effect on MGMT positive tumors, we evaluated its antitumor efficacy using an MGMT positive human glioma (SF-767) nude mouse xenograft model. Since SF-767 has high MGMT levels, BCNU treatment (20 mg/kg, Q4D x 3 i.p.) alone did not result in a satisfactory anticancer effect (p > 0.05). As expected, O6-benzylguanine (O6-BG) (100 mg/kg), which was given prior to BCNU treatment, by depleting MGMT activity, significantly enhanced BCNU antitumor efficacy (p < 0.001). Moreover, SarCNU treatment (167 mg/kg, Q4D x 3 i.p.) alone had a better antitumor effect than O6-BG plus BCNU treatment (F = 51.7, p = 0.0004). However, in this xenograft model, O6-BG did not significantly enhance the anticancer efficacy of SarCNU (F = 0.8, p = 0.411). The SF-767 human glioma xenograft is positive for extraneuronal monoamine transporter EMT (EMT) as determined by reverse-transcription polymerase chain reaction (RT-PCR). Our present results suggest that SarCNU is also effective for MGMT positive tumor if they exhibit EMT.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Carmustine/therapeutic use , Glioma/drug therapy , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Organic Cation Transport Proteins , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Carmustine/analogs & derivatives , Carrier Proteins/metabolism , Cell Line , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/therapeutic use , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous
19.
Clin Cancer Res ; 7(2): 421-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11234899

ABSTRACT

Bis-2-chloroethylnitrosourea (BCNU) or temozolomide (TMZ) were tested alone or in combination with the AGT inhibitors O6-benzyl-2'-deoxyguanosine (dBG) or O6-benzylguanine (BG) against human glial tumor xenografts growing s.c. in athymic mice. Four glioblastoma (SWB77, SWB40, SWB39, and D-54) and one anaplastic oligodendroglioma (SWB61) xenografts having O6-alkylguanine-DNA alkyltransferase (AGT) activities of 75, 45, 10, < 10, and 16 fmol/mg protein, respectively, were used. BCNU at 35 mg/m2 was ineffective against these tumors, although 70 mg/m2 (LD10, 75 mg/m2) produced a marked tumor growth delay (T-C) in D54 but had no effect against SWB40 or SWB77. Coadministration of BG or dBG and BCNU necessitated reduction of the BCNU dose to a maximum of 30 and 35 mg/m2, respectively, because of increased toxicity. Optimized treatment with dBG (250 mg/m2) and BCNU (35 mg/m2) resulted in T-Cs of 30, 29, 11, 16, and 14 days for SWB77, SWB40, SWB39, D-54 and SWB61, respectively. These delays were more pronounced than those induced with optimized, isotoxic treatments with BG (180 mg/m2) and BCNU (30 mg/m2). In comparison to BCNU, TMZ was less toxic, with an LD10 of 400 mg/m2. TMZ (300 mg/m2) was more effective than BCNU against SWB77, SWB40, and SWB61, inducing T-Cs of 23, 53, and 56 days, respectively. BG and dBG enhanced the toxicity of TMZ in athymic mice by decreasing the LD10 from 400 to 200 mg/m2. TMZ (180 mg/m2) with either BG (180 mg/m2) or dBG (250 mg/m2) resulted in T-Cs of 31 and 49 days in SWB77, respectively, as compared with 16 days for TMZ (180 mg/m2) alone. In SWB40, the combination of TMZ with dBG, but not with BG, was significantly more effective than the maximum tolerated dose of TMZ (300 mg/m2) alone. The combination of TMZ with AGT inactivators had no benefit, as compared with TMZ alone, against xenografts with marginal AGT activity. In conclusion, at equimolar doses dBG was less toxic than BG in athymic mice when combined with either BCNU or TMZ. In this regard, BCNU or TMZ can be used at higher doses in combination with dBG than with BG. This study further demonstrates that there is a significant benefit of depleting AGT with nonspecific AGT inhibitors prior to treatment with either BCNU or TMZ in tumors having AGT activity >45 fmol/mg protein.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Carmustine/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Deoxyguanosine/analogs & derivatives , Glioma/drug therapy , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis , Deoxyguanosine/therapeutic use , Drug Resistance, Neoplasm , Enzyme Inhibitors/therapeutic use , Glioma/enzymology , Glioma/pathology , Guanine/analogs & derivatives , Guanine/therapeutic use , Humans , Mice , Mice, Nude , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Temozolomide , Transplantation, Heterologous , Tumor Cells, Cultured
20.
Clin Cancer Res ; 5(11): 3676-81, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589786

ABSTRACT

O6-Benzyl-2'-deoxyguanosine (dBG), a water-soluble inhibitor of O6-methylguanine-DNA methyltransferase (MGMT), potentiates the efficacy of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) against MGMT-positive, BCNU-resistant Daoy human medulloblastoma tumor xenografts in athymic mice (S. C. Schold et al., Cancer Res., 56: 2076-2081, 1996). Such potentiation was comparable to that observed for O6-benzylguanine, the prototype MGMT inhibitor that is currently undergoing clinical trials. In this study, we optimized the therapeutic effect of the dBG and BCNU combination against brain tumor xenografts without inducing substantial toxicity in the host by adjusting the doses of both compounds. dBG was escalated from 133 mg/m2 to 200 and 300 mg/m2, whereas corresponding doses of BCNU were reduced from 25 mg/m2 to 17 and 11 mg/m2, respectively. The growth delays of 30.2, 38.4, and 22.3 days, respectively, observed for the above regimens suggest that the optimal drug combination is not achieved with maximum doses of dBG. In fact, the highest doses of dBG (300 mg/m2) contributed to more frequent BCNU-related toxicities, despite the reduced BCNU dosage, and a reduction of the therapeutic effect. Toxicity was related to the depletion of MGMT activity in the gut of host mice and was manifested by edema, inflammation, and hemorrhage in the bowel wall by subsequent BCNU administration. With additional dosage adjustments, we found that tumor suppression of >90 days without toxicity was observed at 200 mg/m2 dBG and 23 mg/m2 BCNU. At these doses, tumors were eradicated (regressed to an undetectable size for >90 days) in 8 of 12 animals. Thus, dBG is the first of the MGMT inhibitors to show a curative effect in combination with BCNU against a human central nervous system tumor xenograft in athymic mice.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cerebellar Neoplasms/drug therapy , Deoxyguanosine/analogs & derivatives , Medulloblastoma/drug therapy , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Animals , Apoptosis , Carmustine/administration & dosage , Carmustine/toxicity , Cerebellar Neoplasms/pathology , Deoxyguanosine/administration & dosage , Deoxyguanosine/therapeutic use , Deoxyguanosine/toxicity , Humans , Inflammation , Intestinal Mucosa/enzymology , Medulloblastoma/pathology , Mice , Mice, Nude , Mitosis , Transplantation, Heterologous , Tumor Cells, Cultured , Weight Loss/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL