Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nitric Oxide ; 73: 66-73, 2018 02 28.
Article in English | MEDLINE | ID: mdl-28587887

ABSTRACT

Diet is associated with the development of cancer in the gastrointestinal (GI) tract, because dietary nitrate and nitrite are the main nitrosating agents that are responsible for the formation of carcinogenic N-nitroso compounds (NOCs) when nitrosatable substrates, such as amine and amide, are present in the GI tract. However, whether the nitroso compounds become beneficial S-nitroso compounds or carcinogenic NOCs might depend on dietary and environmental factors including food stuffs, gastric acidity, microbial flora, and the mean transit time of digesta. This review focused on GI NOC formation and environmental risk factors affecting its formation to provide appropriate nutritional strategies to prevent the development of GI cancer.


Subject(s)
Colorectal Neoplasms/etiology , Diet , Inflammatory Bowel Diseases/etiology , Nitrates/pharmacokinetics , Nitroso Compounds/metabolism , Colon/microbiology , Dysbiosis/etiology , Esophagus/metabolism , Humans , Nitric Oxide/metabolism , Nitrites/pharmacokinetics , Nitroso Compounds/adverse effects , Nitroso Compounds/pharmacokinetics , Stomach/drug effects
2.
J Pharmacol Exp Ther ; 344(2): 339-47, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23211362

ABSTRACT

Nitroxyl (HNO) donors have potential benefit in the treatment of heart failure and other cardiovascular diseases. 1-Nitrosocyclohexyl acetate (NCA), a new HNO donor, in contrast to the classic HNO donors Angeli's salt and isopropylamine NONOate, predominantly releases HNO and has a longer half-life. This study investigated the vasodilatative properties of NCA in isolated aortic rings and human platelets and its mechanism of action. NCA was applied on aortic rings isolated from wild-type mice and apolipoprotein E-deficient mice and in endothelial-denuded aortae. The mechanism of action of HNO was examined by applying NCA in the absence and presence of the HNO scavenger glutathione (GSH) and inhibitors of soluble guanylyl cyclase (sGC), adenylyl cyclase (AC), calcitonin gene-related peptide receptor (CGRP), and K(+) channels. NCA induced a concentration-dependent relaxation (EC(50), 4.4 ĀµM). This response did not differ between all groups, indicating an endothelium-independent relaxation effect. The concentration-response was markedly decreased in the presence of excess GSH; the nitric oxide scavenger 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide had no effect. Inhibitors of sGC, CGRP, and voltage-dependent K(+) channels each significantly impaired the vasodilator response to NCA. In contrast, inhibitors of AC, ATP-sensitive K(+) channels, or high-conductance Ca(2+)-activated K(+) channels did not change the effects of NCA. NCA significantly reduced contractile response and platelet aggregation mediated by the thromboxane A(2) mimetic 9,11-dideoxy-11α,9α-epoxymethanoprostaglandin F(2)(α) in a cGMP-dependent manner. In summary, NCA shows vasoprotective effects and may have a promising profile as a therapeutic agent in vascular dysfunction, warranting further evaluation.


Subject(s)
Acetates/pharmacology , Nitric Oxide Donors/pharmacology , Nitrogen Oxides/metabolism , Nitroso Compounds/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Acetates/pharmacokinetics , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Blood Platelets/cytology , Blood Platelets/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Half-Life , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Nitric Oxide Donors/pharmacokinetics , Nitroso Compounds/pharmacokinetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacokinetics , Vasodilator Agents/pharmacokinetics
3.
Nitric Oxide ; 33: 1-5, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23680525

ABSTRACT

Nitrite anion is bioactive nitric oxide (NO) species circulating in blood, and represents the NO bioavailability and endothelial function. In this study, we aimed to investigate the nitrite levels and the correlation with hemolysis and severity in Ɵ-thalassemia/hemoglobin E (Ɵ-thal/HbE). 38 Children (12.0Ā±1.9 years of age) with a diagnosis of mild, moderate and severe Ɵ-thalassemia were enrolled in the study. The blood nitrite levels and potential plasma NO consumption were measured by the chemiluminescence method. The nitrite levels in whole blood and erythrocytes of the severe thalassemia subjects were lower than those of the control subjects. At day 7 after transfusion of packed erythrocytes, the nitrite levels in erythrocytes increased. The plasma hemoglobin and NO consumption increased in the severe thalassemia subjects. The nitrite levels in erythrocytes inversely correlated with plasma hemoglobin, lactate dehydrogenase activity, potential NO consumption, and lipid peroxidation. Our studies demonstrate the decreased NO bioavailability in thalassemia, which could result from endothelial dysfunction, the increased potential NO consumption in plasma by cell-free hemoglobin and oxidative stress.


Subject(s)
Hemoglobin E/metabolism , Nitrites/blood , beta-Thalassemia/blood , Adolescent , Analysis of Variance , Case-Control Studies , Child , Erythrocytes , Female , Hemoglobins/metabolism , Humans , Male , Nitric Oxide/blood , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/pharmacokinetics , Nitroso Compounds/administration & dosage , Nitroso Compounds/pharmacokinetics , beta-Thalassemia/drug therapy , beta-Thalassemia/metabolism
4.
Nitric Oxide ; 24(1): 1-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20933602

ABSTRACT

Several studies have implicated the role of Nitric Oxide (NO) in the regulation of tumor cell behavior and have shown that NO either promotes or inhibits tumorigenesis. These conflicting findings have been resolved, in part, by the levels of NO used such that low levels promote tumor growth and high levels inhibit tumor growth. Our studies have focused on the use of high levels of NO provided primarily by the NO donor, DETANONOate. We have shown that treatment of resistant tumor cells with DETANONOate sensitizes them to apoptosis by both chemotherapeutic drugs and cytotoxic immunotherapeutic ligands. The underlying mechanisms by which NO sensitizes tumor cells to apoptosis were shown to be regulated, in part, by NO-mediated inhibition of the NF-κB survival/anti-apoptotic pathways and downstream of NF-κB by inhibition of the transcription factor Yin Yang 1 (YY1). In addition to NO-induced sensitization to apoptosis, we have also shown that NO induced the expression of the metastasis-suppressor/immunosurveillance cancer gene product, Raf-1 kinase inhibitor protein (RKIP). Overexpression of RKIP mimics NO in tumor cells-induced sensitization to apoptosis. The induction of RKIP by NO was the result of the inhibition of the RKIP repressor, Snail, downstream of NF-κB. These findings established the presence of a dysregulated NF-κB/Snail/YY1/ RKIP circuitry in resistance and that treatment with NO modifies this loop in tumor cells in favor of the inhibition of tumor cell survival and the response to cytotoxic drugs. Noteworthy, the NF-κB/Snail/YY1/RKIP loop consists of gene products that regulate the epithelial to mesenchymal transition (EMT) and, thus, tumor metastasis. Hence, we have found that treatment of metastatic cancer cell lines with DETANONOate inhibited the EMT phenotype, through both the inhibition of the metastasis-inducers, NF-κB and Snail and the induction of the metastasis-suppressor, RKIP. Altogether, the above findings establish, for the first time, the dual role of high levels of NO in the sensitization of tumor cells to apoptotic stimuli as well as inhibition of EMT. Hence, NO donors may be considered as novel potential therapeutic agents with dual roles in the treatment of patients with refractory cancer and in the prevention of the initiation of the metastatic cascade via EMT.


Subject(s)
Neoplasms/drug therapy , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Animals , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm , Drug Synergism , Humans , NF-kappa B/metabolism , Neoplasms/metabolism , Nitric Oxide Donors/pharmacokinetics , Nitroso Compounds/pharmacokinetics , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors , Transcription Factors/metabolism , YY1 Transcription Factor/metabolism
5.
Invest New Drugs ; 28(5): 694-702, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19557306

ABSTRACT

PURPOSE: Given the limited options available to treat canine cancers, the use of companion animals for evaluating new drugs may identify better therapies for veterinary and human oncology. The anti-tumor effects of nitrosylcobalamin (NO-Cbl), an apoptosis-inducing, vitamin B12-based carrier of nitric oxide (NO), was evaluated in four dogs with spontaneous cancer. EXPERIMENTAL DESIGN: (1) A 13 year-old female spayed Giant Schnauzer with inoperable thyroid carcinoma and hypercalcemia. (2) A 6 year-old male neutered Golden Retriever with a malignant peripheral nerve sheath tumor (MPNST). (3) A ten yr-old neutered male Bichon Frise with apocrine gland anal sac adenocarcinoma (AGACA). (4) A 7 year-old female spayed Labrador mix with spinal meningioma following partial surgical resection. Tumor regression was measured by physical exam and verified using ultrasound (case 1) and MRI (case 2-4). Serum chemistries and hematologic parameters were monitored throughout the studies. RESULTS: (1) The Giant Schnauzer demonstrated a 77% reduction in tumor volume after ten weeks of daily NO-Cbl treatment. (2) The Golden Retriever demonstrated a 53% reduction in tumor volume after 15 months of daily NO-Cbl therapy. (3) The Bichon Frise demonstrated a 43% regression of the primary tumor and a 90% regression of an iliac lymph node measured by MRI after 15 months of treatment. After 61 months, the dog currently has stable disease, normal liver enzymes, CBC analysis, and no evidence of toxicity. (4) The Labrador demonstrated complete regression of the residual tumor after 6 months of treatment. CONCLUSION: We have shown previously that NO-Cbl is endocytosed by malignant cells, resulting in intra-tumoral NO release. In this study, we have shown that daily long-term use of NO-Cbl induced responses in all dogs without any signs of toxicity. The use of NO-Cbl capitalizes on the tumor-specific properties of the vitamin B12 receptor and represents a promising anti-cancer therapy.


Subject(s)
Dog Diseases/drug therapy , Neoplasms/veterinary , Nitroso Compounds/therapeutic use , Vitamin B 12/analogs & derivatives , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Dog Diseases/diagnostic imaging , Dog Diseases/pathology , Dogs , Dose-Response Relationship, Drug , Female , Magnetic Resonance Imaging , Male , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/pathology , Nitroso Compounds/metabolism , Nitroso Compounds/pharmacokinetics , Tumor Burden , Ultrasonography , Vitamin B 12/metabolism , Vitamin B 12/pharmacokinetics , Vitamin B 12/therapeutic use
6.
Eur J Med Chem ; 208: 112768, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32932211

ABSTRACT

4-Nitroso-3-trifluoromethyl-5-alkyl[(het)aryl]pyrazoles were synthesized via one-pot nitrosation of 1,3-diketones or their lithium salts followed by treatment of hydrazines. Reduction of nitroso-derivatives made it possible to obtain 4-amino-3-trifluoromethylpyrazoles chlorides. According to computer-aided calculations, all synthesized compounds are expected to have acceptable ADME profile for drug design. Tuberculostatic, antibacterial, antimycotic, antioxidant and cytotoxic activities of the compounds were evaluated inĀ vitro, while their analgesic and anti-inflammatory action was tested inĀ vivo along with acute toxicity studies. N-Unsubstituted 4-nitrosopyrazoles were the most effective tuberculostatics (MIC to 0.36Ā Āµg/ml) and antibacterial agents against Streptococcus pyogenes (MIC to 7.8Ā Āµg/ml), Staphylococcus aureus,S. aureus MRSA and Neisseria gonorrhoeae (MIC to 15.6Ā Āµg/ml). 4-Nitroso-1-methyl-5-phenylpyrazole had the pronounced antimycotic action against a wide range of fungi (Trichophytonrubrum, T. tonsurans, T. violaceum, T. interdigitale, Epidermophytonfloccosum, Microsporumcanis with MIC 0.38-12.5Ā Āµg/ml). N-Unsubstituted 4-aminopyrazoles shown high radical-scavenging activity in ABTS test, ORAC/AAPH and oxidative erythrocyte hemolysis assays. 1-Methyl-5-phenyl-3-trifluoromethylpyrazol-4-aminium chloride revealed potential anticancer activity against HeLa cells (SIĀ >Ā 1351). The pronounced analgesic activity was found for 4-nitroso- and 4-aminopyrazoles having phenyl fragment at the position 5 in "hot plate" test. The most of the obtained pyrazoles had a moderate acute toxicity.


Subject(s)
Nitroso Compounds/pharmacology , Pyrazoles/pharmacology , Animals , Bacteria/drug effects , Drug Screening Assays, Antitumor , Epidermophyton/drug effects , HeLa Cells , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure , Nitroso Compounds/chemical synthesis , Nitroso Compounds/pharmacokinetics , Nitroso Compounds/toxicity , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/toxicity , Rats, Sprague-Dawley , Structure-Activity Relationship , Trichophyton/drug effects
7.
Cancer Res ; 65(19): 8604-7, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16204024

ABSTRACT

Beta-catenin plays an important role in colonic tumorigenesis whereas inducible nitric oxide synthase and nitric oxide are elevated in colonic inflammation. Resistance of colonic epithelial cells to the induction of apoptosis may contribute to tumor development. Nitric oxide can stimulate apoptosis and, paradoxically, is implicated in the development of colon cancer. Our hypothesis was that beta-catenin could increase the resistance of colonic cancer cells to nitric oxide-induced apoptotic cell death. Here we show, using a beta-catenin overexpression system, that increased cytosolic beta-catenin renders colonic epithelial cells more resistant to nitric oxide-induced apoptotic cell death, independently of nitric oxide-induced accumulation of p53. Furthermore, we show that this occurs through inhibition of nitric oxide-induced release of cytochrome c from mitochondria and by blocking both the nitric oxide-induced suppression of the antiapoptotic protein, Bcl-xL, and the phosphorylation of Akt. We contend that increased nitric oxide production, such as that which occurs in chronic colonic inflammation, may select the cells with oncogenic mutant beta-catenin regulatory genes and contribute to human colonic carcinogenesis and tumor progression.


Subject(s)
Apoptosis/physiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Nitric Oxide/antagonists & inhibitors , beta Catenin/biosynthesis , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/genetics , Humans , Nitric Oxide/pharmacokinetics , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacokinetics , Nitric Oxide Donors/pharmacology , Nitroso Compounds/antagonists & inhibitors , Nitroso Compounds/pharmacokinetics , Nitroso Compounds/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Transfection , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/metabolism , beta Catenin/genetics
8.
Biomaterials ; 140: 162-169, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28651144

ABSTRACT

In the latest trend of anticancer chemotherapy research, there were many macromolecular anticancer drugs developed based on enhanced permeability and retention (EPR) effect, such as albumin bound paclitaxel nanoparticle (nab- PTX, also called AbraxaneĀ®). However, cancers with low vascular permeability posed a challenge for these EPR based therapeutic systems. Augmenting the intrinsic EPR effect with an intrinsic vascular modulator such as nitric oxide (NO) could be a promising strategy. S-nitrosated human serum albumin dimer (SNO-HSA Dimer) shown promising activity previously was evaluated for the synergistic effect when used as a pretreatment agent in nab-PTX therapy against various tumor models. In the high vascular permeability C26 murine colon cancer subcutaneous inoculation model, SNO-HSA Dimer enhanced tumor selectivity of nab-PTX, and attenuated myelosuppression. SNO-HSA Dimer also augmented the tumor growth inhibition of nab-PTX in low vascular permeability B16 murine melanoma subcutaneous inoculation model. Furthermore, nab-PTX therapy combined with SNO-HSA Dimer showed higher antitumor activity and improved survival rate of SUIT2 human pancreatic cancer orthotopic model. In conclusion, SNO-HSA Dimer could enhance the therapeutic effect of nab-PTX even in low vascular permeability or intractable pancreatic cancers. The possible underlying mechanisms of action of SNO-HSA Dimer were discussed.


Subject(s)
Albumin-Bound Paclitaxel/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Nitroso Compounds/therapeutic use , Serum Albumin, Human/therapeutic use , Albumin-Bound Paclitaxel/pharmacokinetics , Albumin-Bound Paclitaxel/pharmacology , Albumins/pharmacokinetics , Albumins/pharmacology , Albumins/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Capillary Permeability/drug effects , Cell Line, Tumor , Drug Synergism , Female , Humans , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/metabolism , Neoplasms/pathology , Nitric Oxide/metabolism , Nitroso Compounds/pharmacokinetics , Nitroso Compounds/pharmacology , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Multimerization , Serum Albumin, Human/pharmacokinetics , Serum Albumin, Human/pharmacology
9.
Biochem J ; 387(Pt 2): 473-8, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15579132

ABSTRACT

The N-nitroso-derivative of melatonin, NOM (1-nitrosomelatonin), which has been demonstrated to be a NO* [oxidonitrogen*] donor in buffered solutions, is a new potential drug particularly in neurological diseases. The advantage of NOM, a very lipophilic drug, is its ability to release both melatonin and NO*, an easily diffusible free radical. In order to evaluate the distribution and the pharmacokinetics of NOM, [O-methyl-3H]NOM was administered to and followed in mice. A complementary method for monitoring NOM, EPR, was performed in vitro and ex vivo with (MGD)2-Fe2+ (iron-N-methyl-D-glucamine dithiocarbamate) complex as a spin trap. The behaviour of NOM was compared with that of GSNO (S-nitrosoglutathione), a hydrophilic NO* donor. In the first minutes following [O-methyl-3H]NOM intraperitoneal injection, the radioactivity was found in organs (6% in the liver, 1% in the kidney and 0.6% in the brain), but not in the blood. In both liver and brain, the radioactivity content decreased over time with similar kinetics reflecting the diffusion and metabolism of NOM and of its metabolites. Based on the characterization and the quantification of the EPR signal in vitro with NOM or GSNO using (MGD)2-Fe2+ complex in phosphate-buffered solutions, the detection of these nitroso compounds was realized ex vivo in mouse tissue extracts. (MGD)2-Fe2+-NO was observed in the brain of NOM-treated mice in the first 10 min following injection, revealing that NOM was able to cross the blood-brain barrier, while GSNO was not.


Subject(s)
Melatonin/analogs & derivatives , Nitroso Compounds/pharmacokinetics , S-Nitrosoglutathione/pharmacokinetics , Animals , Blood-Brain Barrier , Brain/metabolism , Electron Spin Resonance Spectroscopy , Female , Liver/metabolism , Melatonin/pharmacokinetics , Mice , Mice, Inbred C57BL , Sorbitol/analogs & derivatives , Spin Labels , Thiocarbamates , Tissue Distribution , Tritium
10.
Cancer Res ; 48(14): 4049-52, 1988 Jul 15.
Article in English | MEDLINE | ID: mdl-3383197

ABSTRACT

When five volunteers were given food containing cod and vegetables (a traditional Japanese food, called tara-chiri), their urinary excretion of N-nitrosothioproline increased from 7.9 +/- 4.2 (SD) micrograms/day to 110 +/- 64.5 micrograms/day. This increase was accounted for by in vivo nitrosation of of thioproline by nitrite formed from nitrate in the vegetables. This finding was confirmed by results on a volunteer who ate boiled cod and Japanese radish (daikon) (a simple version of the food containing cod and vegetables). Boiled cod was found to contain 300-500 micrograms/100 g of thioproline, and the level nearly doubled when the cod was boiled with Japanese radish. This increase occurred during the cooking of cod with Japanese radish by the reaction of formaldehyde in the cod with cysteine in the Japanese radish. The nitrosation of thioproline was estimated to be 1000-fold that of proline in the human body. Thus thioproline is a very sensitive probe of in vivo nitrosation. Thioproline formation either in vivo or in vitro may have the following two roles in reducing tumorigenesis in humans: (a) detoxication of formaldehyde, which is genotoxic; and (b) blocking the formation of carcinogenic N-nitroso compounds by trapping nitrite and then being excreted in the urine.


Subject(s)
Fishes , Nitroso Compounds/urine , Thiazoles/urine , Vegetables , Animals , Formaldehyde/analysis , Half-Life , Humans , Nitrates/urine , Nitroso Compounds/pharmacokinetics , Thiazoles/analysis , Thiazoles/pharmacokinetics , Thiazolidines
11.
Cancer Res ; 61(12): 4701-6, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406540

ABSTRACT

High amounts of nitric oxide (NO) produced by activated macrophages or NO donors are required to induce cytotoxicity and apoptosis in pathogens and tumor cells. High concentrations of NO may lead to nonspecific toxicity thereby limiting the use of NO donors in the treatment of cancer. In this study, we tested the possibility of potentiating the apoptotic action of NO in a human breast cancer cell line, MDA-MB-468, by combining it with a farnesyltransferase inhibitor (FTI), which has been shown to induce apoptosis in some other cancer cell lines with minimal toxicity to normal cells. DETA-NONOate, a long acting NO donor which has a half-life of 20 h at 37 degrees C, was used in this study. DETA-NONOate (1 mM), which releases NO in the range produced by activated macrophages, induced apoptosis after 36 h in MDA-MB-468 cells via cytochrome c release and caspase-9 and -3 activation. FTI (25 microM) potentiated the action of lower concentrations of DETA-NONOate (25-100 microM) by inducing apoptosis in these cells within 24 h by increasing cytochrome c release and caspase-9 and -3 activation. This effect was observed preferentially in the cancer cell lines studied with no apoptosis induction in normal breast epithelial cells. This novel combination of FTI and NO may emerge as a promising approach for the treatment of breast cancer.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Apoptosis/drug effects , Breast Neoplasms/pathology , Nitric Oxide/pharmacology , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Drug Synergism , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Humans , Nitric Oxide/pharmacokinetics , Nitric Oxide Donors/pharmacokinetics , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacokinetics , Nitroso Compounds/pharmacology , Tumor Cells, Cultured
12.
Toxicology ; 206(2): 221-31, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15588915

ABSTRACT

Sulfamethoxazole hypersensitivity may be caused by production of the protein-reactive metabolite nitroso sulfamethoxazole (SMX-NO) and interaction of SMX-NO with T-cells. We have characterised the nature of the immune response induced by administration of sulfamethoxazole, sulfamethoxazole metabolites and nitrosobenzene to BALB/c mice. Drugs were administered over a 13-day period to induce polarised cytokine secretion profiles. Proliferation was measured by [(3)H] thymidine incorporation. Cytokine secretion was monitored by ELISA. Results were compared with those provoked by exposure to type 1 and type 2 chemical allergens, 2,4-dinitrochlorobenzene (DNCB) and trimellitic anhydride (TMA). CD4(+) or CD8(+) T-cells were depleted ex vivo to identify the primary source of cytokines. Lymph node activation was observed following treatment with DNCB, TMA, nitrosobenzene and SMX-NO, but not with sulfamethoxazole or sulfamethoxazole hydroxylamine (SMX-NHOH). DNCB and TMA induced type 1 and type 2 cytokine profiles, respectively. SMX-NO treatment stimulated the production of high levels of IL-5, variable amounts of IFN-gamma, and relatively low levels of IL-10 and IL-4. Nitrosobenzene-activated lymph node cells secreted only low levels of IFN-gamma and IL-5. Depletion of CD4(+) or CD8(+) T-cells from SMX-NO stimulated lymph node cells revealed that CD4(+) T-cells were the major source of IL-5. In conclusion, the data presented indicates that subcutaneous administration to mice of SMX-NO, but not the parent drug, stimulated the secretion of high levels of IL-5 from activated CD4(+) T-cells, which is consistent with the clinical profile of the drug.


Subject(s)
Anti-Infective Agents/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Drug Hypersensitivity/etiology , Interleukin-5/biosynthesis , Nitroso Compounds/toxicity , Sulfamethoxazole/analogs & derivatives , Sulfamethoxazole/toxicity , Animals , Anti-Infective Agents/pharmacokinetics , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Cytokines/immunology , Dinitrochlorobenzene/immunology , Drug Hypersensitivity/immunology , Enzyme-Linked Immunosorbent Assay , Female , Interleukin-5/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Nitroso Compounds/immunology , Nitroso Compounds/pharmacokinetics , Phthalic Anhydrides/immunology , Sulfamethoxazole/immunology , Sulfamethoxazole/metabolism , Sulfamethoxazole/pharmacokinetics , Th1 Cells/immunology , Th2 Cells/immunology
13.
Article in English | MEDLINE | ID: mdl-8268774

ABSTRACT

The hypothesis that intragastric synthesis of N-nitroso compounds (NOC) in early life could play a role in gastric carcinogenesis was tested by applying the N-nitrosoproline (NPRO) test to about 50 children living in high- and low-risk areas for stomach cancer in Costa Rica. The median values of excretion of NPRO and the sum of three nitrosamino acids (micrograms/12 h urine) were 10-20% of those in adults from other geographical high-risk areas for stomach cancer. The urinary NPRO level after proline intake was higher in children from the high-risk area (P < 0.04) and markedly reduced after ingestion of ascorbic acid together with proline (P < 0.05). NPRO levels on the day of proline intake were highly correlated with levels of nitrate excretion (P < 0.001). Mean levels of total NOC in an aqueous (pH 2) extract of cooked beans from the high- and low-risk areas were similar. Acid-catalyzed nitrosation of the extract increased the total NOC concentration up to 1000-fold, but there was no difference between samples from the two areas. About 10% of bean extracts from both areas showed weak direct-acting genotoxicity in Escherichia coli; after acid-catalyzed nitrosation, all samples were genotoxic at similar levels. The diet of children in the low-risk area satisfied recommended levels of intake of energy and most nutrients except riboflavin and retinol equivalents. Diets from the high-risk area were deficient in energy intake and all nutrients except protein and vitamin C.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Gastric Mucosa/metabolism , Nitrosamines/urine , Stomach Neoplasms/etiology , Adolescent , Adult , Aged , Ascorbic Acid/administration & dosage , Biomarkers/urine , Child , Costa Rica/epidemiology , Diet , Energy Metabolism , Fabaceae/chemistry , Female , Humans , Incidence , Male , Middle Aged , Nitrates/urine , Nitroso Compounds/analysis , Nitroso Compounds/pharmacokinetics , Plants, Medicinal , Proline/administration & dosage , Risk Factors , Socioeconomic Factors , Stomach Neoplasms/epidemiology , Stomach Neoplasms/urine
14.
J Med Chem ; 43(21): 4005-16, 2000 Oct 19.
Article in English | MEDLINE | ID: mdl-11052806

ABSTRACT

Despite its widespread use, diclofenac has gastrointestinal liabilities common to nonsteroidal antiinflammatory drugs (NSAIDs) that might be reduced by concomitant administration of a gastrointestinal cytoprotectant such as nitric oxide (NO). A series of novel diclofenac esters containing a nitrosothiol (-S-NO) moiety as a NO donor functionality has been synthesized and evaluated in vivo for bioavailability, pharmacological activity, and gastric irritation. All S-NO-diclofenac derivatives acted as orally bioavailable prodrugs, producing significant levels of diclofenac in plasma within 15 min after oral administration to mice. At equimolar oral doses, S-NO-diclofenac derivatives (20a-21b) displayed rat antiinflammatory and analgesic activities comparable to those of diclofenac in the carrageenan-induced paw edema test and the mouse phenylbenzoquinone-induced writhing test, respectively. All tested S-NO-diclofenac derivatives (20a-21b) were gastric-sparing in that they elicited markedly fewer stomach lesions as compared to the stomach lesions caused by a high equimolar dose of diclofenac in the rat. Nitrosothiol esters of diclofenac comprise a novel class of NO-donating compounds having therapeutic potential as nonsteroidal antiinflammatory agents with an enhanced gastric safety profile.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Diclofenac/chemical synthesis , Nitroso Compounds/chemical synthesis , Prodrugs/chemical synthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biological Availability , Diclofenac/chemistry , Diclofenac/pharmacokinetics , Diclofenac/pharmacology , Male , Mice , Nitroso Compounds/chemistry , Nitroso Compounds/pharmacokinetics , Nitroso Compounds/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Stomach/pathology , Structure-Activity Relationship
15.
Invest Ophthalmol Vis Sci ; 41(3): 749-55, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10711690

ABSTRACT

PURPOSE: To determine whether a novel peroxynitrite-based photosensitizer S-nitrosoglutathione (GSNO) can produce specific in vitro light-induced cell death of both standard animal lung and human Tenon's capsule (TC) fibroblasts and to compare this effect with that produced by the established photodynamic porphyrin precursor 5-aminolevulinic acid (ALA). METHODS: V79-4 Chinese hamster lung and human TC fibroblasts were established in tissue culture. GSNO, together with its radioactive tritiated and fluorescent dansylated derivatives, were synthesized. The labeled molecules were prepared to determine the time course of uptake into the fibroblasts. Uptake was monitored by scintillation counting for the tritiated GSNO and confocal fluorescence microscopy for the dansylated GSNO. The uptake of ALA and biosynthesis of its photosensitive product were determined by fluorescence emission spectroscopy of a separate set of fibroblasts. Once uptake was established, both cell lines were incubated with varying concentrations of GSNO or ALA as a function of time (0, 4, or 24 hours) before light exposure (200 msec pulsed visible light, 0.068 W per pulse, for 10 minutes at a distance of 10 cm). After 10 minutes of irradiation, the cells were washed and exposed to fresh tissue culture medium. The effect of the treatment was determined 24 hours later by measuring cell viability. RESULTS: A 2-minute drug treatment time (0 hours incubation) with GSNO, followed by 10 minutes of irradiation, resulted in approximately 78% of fibroblast cell death at the lowest concentration of GSNO used compared with the control, which was exposed to light, but no GSNO. The higher concentrations of GSNO, or longer drug treatment times before irradiation, did not statistically increase cell death. Maximal cell death was thus obtained using the lowest GSNO concentration (50 mM) and drug treatment time (2 minutes). In contrast, the well-established photosensitizer ALA killed only approximately 4% of cells at the lowest concentration and drug treatment time tested. At drug treatment times of 4 hours and less, increased concentrations of ALA did not produce cell death of more statistical significance. It was not until 24 hours of drug treatment that comparable amounts of cell death were produced by ALA and GSNO. In all experiments similar results were obtained with the animal lung and human TC fibroblasts, suggesting that the source of the fibroblast had no effect on the outcome. The differences in treatment effects between GSNO and ALA were statistically significant under all conditions tested. CONCLUSIONS: GSNO is able to cause light-specific cell death of human TC fibroblasts at drug treatment times (2 minutes) and irradiation times (10 minutes) that would be compatible with its use in glaucoma filtering surgery. This in vitro performance was superior to that of the well-established photosensitizer ALA, which required treatment times longer than 4 hours to approach the light-specific cell death produced by only 2 minutes of GSNO treatment.


Subject(s)
Aminolevulinic Acid/pharmacology , Fascia/pathology , Filtering Surgery , Glutathione/analogs & derivatives , Lung/pathology , Nitroso Compounds/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Aminolevulinic Acid/pharmacokinetics , Animals , Cell Death , Cell Division/drug effects , Cell Line , Cricetinae , Cricetulus , Fascia/drug effects , Fascia/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/drug therapy , Glutathione/pharmacokinetics , Glutathione/pharmacology , Humans , Lung/drug effects , Lung/metabolism , Microscopy, Confocal , Nitroso Compounds/pharmacokinetics , Photolysis , Photosensitizing Agents/pharmacokinetics , S-Nitrosoglutathione
16.
Biochem Pharmacol ; 45(10): 2129-34, 1993 May 25.
Article in English | MEDLINE | ID: mdl-8512594

ABSTRACT

We previously postulated that the catalase-mediated oxidation of cyanamide leads to the formation of the unstable intermediate, N-hydroxycyanamide, which spontaneously decomposes to nitroxyl, the putative inhibitor of aldehyde dehydrogenase (EC 1.2.1.3; AlDH). Since it was not possible to provide direct evidence for the inhibition of AlDH by nitroxyl, we examined the activity of three representative substituted nitroxyls (C-nitroso compounds), viz. nitrosobenzene (NB), 1-nitrosoadamantane (NA), and 2-methyl-2-nitrosopropane (MNP), as direct inhibitors of yeast AlDH in vitro. While NB and NA were highly effective inhibitors in this system exhibiting IC50 values of 2.5 and 8.6 microM, respectively, MNP was considerably less effective with an IC50 of 0.15 mM. When tested in vivo, NA did not show any inhibitory activity on the hepatic AlDH, possibly due to the lack of site-specific delivery of the active monomeric form of this compound. However, NB at a low dose did inhibit hepatic AlDH as reflected by an increase in blood acetaldehyde levels. These results attest to the abilities of NB and NA to act as direct inhibitors of AlDH analogous to nitroxyl itself.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Nitrogen Oxides/pharmacology , Nitroso Compounds/pharmacology , Adamantane/analogs & derivatives , Adamantane/pharmacokinetics , Adamantane/pharmacology , Animals , Biotransformation , Free Radicals/pharmacology , Male , Nitrobenzenes/pharmacokinetics , Nitrobenzenes/pharmacology , Nitroso Compounds/pharmacokinetics , Rats , Rats, Sprague-Dawley , Saccharomyces cerevisiae/enzymology
17.
Biochem Pharmacol ; 55(5): 657-65, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9515576

ABSTRACT

Spontaneous release of nitric oxide (NO) from S-nitrosothiols cannot explain their bioactivity, suggesting a role for cellular metabolism or receptors. Using immortalised cells and human platelets, we have identified a cell-mediated mechanism for the biotransformation of the physiological S-nitrosothiol compound S-nitrosoglutathione (GSNO) into nitrite. We suggest the name "GSNO lyase" for this activity. GSNO lyase activity varied between cell types, being highest in a fibroblast cell line and lowest in platelets. In NRK 49F fibroblasts, GSNO lyase mediated a saturable, GSNO concentration-dependent accumulation of nitrite in conditioned medium, which was inhibited both by transition metal chelators, and by subjecting cells to oxidative stress using a combination of the thiol oxidant diamide and Zn2+, a glutathione reductase inhibitor. Activity was resistant, however, to both acivicin, an inhibitor of gamma-glutamyl transpeptidase (EC 2.3.2.2), and to ethacrynic acid, an inhibitor of Pi class glutathione-S-transferases (EC 2.5.1.18), thus neither of these enzymes could account for NO release. Although GSNO lyase does not explain the platelet-selective pharmacological properties of GSNO, cellular biotransformation suggests therapeutic avenues for targeted delivery of NO to other tissues.


Subject(s)
Glutathione/analogs & derivatives , Nitroso Compounds/pharmacokinetics , Animals , Biotransformation , Cell Line , Chelating Agents/pharmacology , Culture Media, Conditioned , Glutathione/metabolism , Glutathione/pharmacokinetics , Homeostasis , Humans , Nitrates/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Nitroso Compounds/metabolism , Rats , S-Nitrosoglutathione , Swine
18.
J Appl Physiol (1985) ; 90(3): 777-88, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181583

ABSTRACT

Nitric oxide (NO) appears in the exhaled breath and is elevated in inflammatory diseases. We developed a steady-state mathematical model of the bronchial mucosa for normal small and large airways to understand NO and S-nitrosoglutathione (GSNO) kinetics and transport using data from the existing literature. Our model predicts that mean steady-state NO and GSNO concentrations for large airways (generation 1) are 2.68 nM and 113 pM, respectively, in the epithelial cells and 0.11 nM (approximately 66 ppb) and 507 nM in the mucus. For small airways (generation 15), the mean concentrations of NO and GSNO, respectively, are 0.26 nM and 21 pM in the epithelial cells and 0.02 nM (approximately 12 ppb) and 132 nM in the mucus. The concentrations in the mucus compare favorably to experimentally measured values. We conclude that 1) the majority of free NO in the mucus, and thus exhaled NO, is due to diffusion of free NO from the epithelial cell and 2) the heterogeneous airway contribution to exhaled NO is due to heterogeneous airway geometries, such as epithelium and mucus thickness.


Subject(s)
Bronchi/physiology , Glutathione/analogs & derivatives , Glutathione/pharmacokinetics , Models, Biological , Nitric Oxide/metabolism , Nitroso Compounds/pharmacokinetics , Respiratory Mucosa/physiology , Humans , Kinetics , Mathematics , Platelet Aggregation Inhibitors/pharmacokinetics , Reproducibility of Results , S-Nitrosoglutathione , Trachea/physiology
19.
Neurosci Lett ; 171(1-2): 89-93, 1994 Apr 25.
Article in English | MEDLINE | ID: mdl-7521951

ABSTRACT

In vivo voltammetry was performed in rat brain cortex and in rat brain endothelial constitutive NO-synthase preparations. The use of a recent microcaptor detecting N-hydroxy- and N-nitroso-L-arginine permitted to find only the latest in biological preparations. The construction of a new membrane selective electrode for nitric oxide (NO) allowed to assert its absence in these preparations at micromolar level. Half-live of N-nitroso-L-arginine was 4 s in rat brain cortex and the washout curve of NO after over brain insufflation gave an half-life of 10.5 min; their diffusion coefficients in brain were 3.810(-5) for NO and 3.910(-6) cm2.s-1 for N-nitroso-L-arginine. These facts indicate that N-nitroso-L-arginine is degraded directly into nitrites and citrulline after its synthesis by endothelial NO-synthase.


Subject(s)
Arginine/analogs & derivatives , Cerebral Cortex/metabolism , Nitric Oxide/pharmacokinetics , Nitroso Compounds/pharmacokinetics , Amino Acid Oxidoreductases/metabolism , Animals , Arginine/pharmacokinetics , Cerebral Cortex/enzymology , Diffusion , Half-Life , Male , Microelectrodes , Nitric Oxide Synthase , Rats , Rats, Sprague-Dawley
20.
Oncol Rep ; 5(4): 965-9, 1998.
Article in English | MEDLINE | ID: mdl-9625856

ABSTRACT

One tenth of the LD50 as a single dose of various N-nitroso compounds (N-nitrosodimethylamine; NDMA, N-nitrosodiethylamine; NDEA, N-nitrosoethylpropylamine; NEPA, N-nitrosodipropylamine; NDPA, N-nitrosomethylethylamine; NMEA, N-nitroso-methylbutylamine; NMBA and N-nitrosoethylbutylamine; NEBA) was administrated into male mice. This dose markedly increased the hepatic contents of cytochrome P450 and cytochrome b5 and activities of NADPH-cytochrome c reductase and aryl hydrocarbon hydroxylase (AHH). The highest increase in the activity of cytochrome P450 (+142% relative to the control value) was shown in animals treated with either N-nitrosoethylpropylamine or N-nitrosodiethylamine. On the other hand, the lowest increase in the activity (+16%) was revealed in animals treated with N-nitrosodimethylamine (not significant compared to the control value). Cytochrome b5 content was increased by 190% of the control value in mice treated with N-nitrosomethylbutylamine, while N-nitrosodibutylamine induced the lowest increase (+20%). The maximum increase (+182%) in the activity of aryl hydrocarbon hydroxylase was shown in animals which received N-nitrosomethylbutylamine, while the lowest increase (+23%) in animals which received N-nitrosodiethylamine. The activity of hepatic AHH was also increased above the control value in animals treated with NDMA, NEBA NDPA, NMEA and NDBA by 138, 98, 90, 89 and 69%, respectively. Identically, NADPH-cytochrome c reductase activity was increased in animals which received NEPA, NMBA, NDMA, NMEA, NDPA, NEBA and NDEA by 202, 150, 110, 95, 94, 77 and 37%, respectively.


Subject(s)
Carcinogens/pharmacokinetics , Liver/metabolism , Nitroso Compounds/pharmacokinetics , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Carcinogens/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytochromes b5/metabolism , Inactivation, Metabolic , Lethal Dose 50 , Male , Mice , Microsomes/drug effects , NADPH-Ferrihemoprotein Reductase/metabolism , Nitroso Compounds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL