Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Biomed Pharmacother ; 92: 989-997, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28605880

ABSTRACT

Hepatocellular carcinoma (HCC) is the most important cause of cancer-related death, and 85% of HCC is caused by chronic HBV infection, the prognosis of patients and the reduction of HBV DNA levels remain unsatisfactory. JS-K, a nitric oxide-releasing diazeniumdiolates, is effective against various tumors, but little is known on its effects on HBV positive HCC. We found that JS-K reduced the expression of HBsAg and HBeAg in HBV-positive HepG2.2.15 cells. This study aimed to further examine anti-tumor effects of JS-K on HepG2.2.15 cells. The MTT assay and colony forming assay were used to study the cell growth inhibition of JS-K; scratch assay and transwell assay were performed to detect cell migration. The cell cycle was detected by flow cytometry. The immunofluorescence, flow cytometry analysis, and western blot were used to study DNA damage and cell apoptosis. JS-K inhibited HepG2.2.15 cell growth in a dose-dependent manner, suppressed cell colony formation and migration, arrested cells gather in the G2 phase. JS-K (1-20µM) increased the expression of DNA damage-associated protein phosphorylation H2AX (γH2AX), phosphorylation of checkpoint kinase 1 (p-Chk1), phosphorylation of checkpoint kinase 2 (p-Chk2), ataxia-telangiectasia mutated (ATM), phosphorylation of ataxia-telangiectasia mutated rad3-related (p-ATR) and apoptotic-associated proteins cleaved caspase-3, cleaved caspase-7, cleaved poly ADP-ribose polymerase (cleaved PARP). The study demonstrated JS-K is effective against HBV-positive HepG2.2.15 cells, the mechanisms are not only related to inhibition of HBsAg and HBeAg secretion, but also related with induction of DNA damage and apoptosis. JS-K is a promising anti-cancer candidate against HBV-positive HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Apoptosis/drug effects , Azo Compounds/pharmacology , Carcinoma, Hepatocellular/drug therapy , DNA Damage , Hepatitis B virus/drug effects , Liver Neoplasms/drug therapy , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Antineoplastic Agents/metabolism , Antiviral Agents/metabolism , Apoptosis Regulatory Proteins/metabolism , Azo Compounds/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Cycle Proteins/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/metabolism , Hepatitis B virus/immunology , Hepatitis B virus/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/virology , Nitric Oxide Donors/metabolism , Piperazines/metabolism , Signal Transduction/drug effects
2.
Tumour Biol ; 39(6): 1010428317703922, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28653883

ABSTRACT

As a potent radiosensitizer nitric oxide (NO) may be a putative adjuvant in the treatment of malignant gliomas which are known for their radio- and chemoresistance. The NO donor prodrug JS-K (O2-(2.4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen-1-ium-1,2-diolate) allows cell-type specific intracellular NO release via enzymatic activation by glutathione-S-transferases overexpressed in glioblastoma multiforme. The cytotoxic and radiosensitizing efficacy of JS-K was assessed in U87 glioma cells in vitro focusing on cell proliferation, induction of DNA damage, and cell death. In vivo efficacy of JS-K and repetitive irradiation were investigated in an orthotopic U87 xenograft model in mice. For the first time, we could show that JS-K acts as a potent cytotoxic and radiosensitizing agent in U87 cells in vitro. This dose- and time-dependent effect is due to an enhanced induction of DNA double-strand breaks leading to mitotic catastrophe as the dominant form of cell death. However, this potent cytotoxic and radiosensitizing effect could not be confirmed in an intracranial U87 xenograft model, possibly due to insufficient delivery into the brain. Although NO donor treatment was well tolerated, neither a retardation of tumor growth nor an extended survival could be observed after JS-K and/or radiotherapy.


Subject(s)
Azo Compounds/administration & dosage , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Nitric Oxide Donors/administration & dosage , Piperazines/administration & dosage , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/pathology , Humans , Mice , Nitric Oxide/metabolism , Radiation-Sensitizing Agents/administration & dosage , Xenograft Model Antitumor Assays
3.
Cell Death Discov ; 3: 17006, 2017.
Article in English | MEDLINE | ID: mdl-28250971

ABSTRACT

Glioblastoma is associated with poor survival and a high recurrence rate in patients due to inevitable uncontrolled infiltrative tumor growth. The elucidation of the molecular mechanisms may offer opportunities to prevent relapses. In this study we investigated the role of the activating transcription factor 3 (ATF3) in migration of GBM cells in vitro. RNA microarray revealed that gene expression of ATF3 is induced by a variety of chemotherapeutics and experimental agents such as the nitric oxide donor JS-K (O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate). We found NFκB and STAT3 to be downstream targets inhibited by overexpression of ATF3. We demonstrate that ATF3 is directly involved in the regulation of matrix metalloproteinase expression and activation. Overexpression of ATF3 therefore leads to a significantly reduced migration capacity and induction of tissue inhibitors of matrix metalloproteinases. Our study for the first time identifies ATF3 as a potential novel therapeutic target in glioblastoma.

4.
Biomed Pharmacother ; 88: 367-373, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28122301

ABSTRACT

JS-K is a novel anticancer nitric oxide (NO) prodrug effective against a variety of cancer cells, including the inhibition of AM-1 hepatoma cell growth in rats. To further evaluate anticancer effects of JS-K, human hepatoma Hep3B cells were treated with JS-K and the compound control JS-43-126 at various concentrations (0-100µM) for 24h, and cytotoxicity was determined by the MTS assay. The compound control JS-43-126 was not cytotoxic to Hep3B cells at concentrations up to 100µM, while the LC50 for JS-K was about 10µM. To examine the molecular mechanisms of antitumor effects of JS-K, Hep3B cells were treated with 1-10µM of JS-K for 24h, and then subjected to gene expression analysis via real time RT-PCR and protein immunostain via confocal images. JS-K is a GST-α targeting NO prodrug, and decreased immunostaining for GST-α was associated with JS-K treatment. JS-K activated apoptosis pathways in Hep3B cells, including induction of caspase-3, caspase-9, Bax, TNF-α, and IL-1ß, and immunostaining for caspase-3 was intensified. The expressions of thrombospondin-1 (TSP-1) and the tissue inhibitors of metalloproteinase-1 (TIMP-1) were increased by JS-K at both transcript and protein levels. JS-K treatment also increased the expression of differentiation-related genes CD14 and CD11b, and depressed the expression of c-myc in Hep3B cells. Thus, multiple molecular events appear to be associated with anticancer effects of JS-K in human hepatoma Hep3B cells, including activation of genes related to apoptosis and induction of genes involved in antiangiogenesis and tumor cell migration.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/genetics , Nitric Oxide/metabolism , Prodrugs/pharmacology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Antineoplastic Agents/chemistry , Azo Compounds/chemistry , Azo Compounds/pharmacology , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Glutathione Transferase/metabolism , Humans , Isoenzymes/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Matrix Metalloproteinases/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Piperazines/chemistry , Piperazines/pharmacology , Thrombospondins/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism
5.
Cell Death Dis ; 7(9): e2349, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27584787

ABSTRACT

The nitric oxide (NO) donor JS-K is specifically activated by glutathione S-transferases (GSTs) in GST-overexpressing cells. We have shown the induction of cell death in glioblastoma multiforme (GBM) cells at high JS-K doses but the mechanism remains unclear. The aim of this study was to determine whether NO-induced cell death is triggered by induction of apoptotic or necrotic pathways. For the first time, we demonstrate that NO induces cell death via mitotic catastrophe (MC) with non-apoptotic mechanisms in GBM cells. Moreover, the level of morphological changes indicating MC correlates with increased necrosis. Therefore, we conclude that MC is the main mechanism by which GBM cells undergo cell death after treatment with JS-K associated with necrosis rather than apoptosis. In addition, we show that PARP1 is not an exclusive marker for late apoptosis but is also involved in MC. Activating an alternative way of cell death can be useful for the multimodal cancer therapy of GBM known for its strong anti-apoptotic mechanisms and drug resistance.


Subject(s)
Apoptosis/drug effects , Azo Compounds/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Mitosis/drug effects , Nitric Oxide/pharmacology , Piperazines/pharmacology , Adenosine Triphosphate/metabolism , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cyclic GMP/pharmacology , Enzyme Activation/drug effects , Flow Cytometry , Glioblastoma/enzymology , Humans , In Situ Nick-End Labeling , Necrosis , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Time Factors
6.
Proc Natl Acad Sci U S A ; 112(34): E4651-60, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26224837

ABSTRACT

Experimental evidence suggests that nitric oxide (NO) and hydrogen sulfide (H2S) signaling pathways are intimately intertwined, with mutual attenuation or potentiation of biological responses in the cardiovascular system and elsewhere. The chemical basis of this interaction is elusive. Moreover, polysulfides recently emerged as potential mediators of H2S/sulfide signaling, but their biosynthesis and relationship to NO remain enigmatic. We sought to characterize the nature, chemical biology, and bioactivity of key reaction products formed in the NO/sulfide system. At physiological pH, we find that NO and sulfide form a network of cascading chemical reactions that generate radical intermediates as well as anionic and uncharged solutes, with accumulation of three major products: nitrosopersulfide (SSNO(-)), polysulfides, and dinitrososulfite [N-nitrosohydroxylamine-N-sulfonate (SULFI/NO)], each with a distinct chemical biology and in vitro and in vivo bioactivity. SSNO(-) is resistant to thiols and cyanolysis, efficiently donates both sulfane sulfur and NO, and potently lowers blood pressure. Polysulfides are both intermediates and products of SSNO(-) synthesis/decomposition, and they also decrease blood pressure and enhance arterial compliance. SULFI/NO is a weak combined NO/nitroxyl donor that releases mainly N2O on decomposition; although it affects blood pressure only mildly, it markedly increases cardiac contractility, and formation of its precursor sulfite likely contributes to NO scavenging. Our results unveil an unexpectedly rich network of coupled chemical reactions between NO and H2S/sulfide, suggesting that the bioactivity of either transmitter is governed by concomitant formation of polysulfides and anionic S/N-hybrid species. This conceptual framework would seem to offer ample opportunities for the modulation of fundamental biological processes governed by redox switching and sulfur trafficking.


Subject(s)
Hydrogen Sulfide/metabolism , Nitric Oxide/metabolism , Nitrogen Oxides/metabolism , Sulfides/metabolism , Animals , Biological Availability , Male , Nitrogen/metabolism , Rats, Wistar , Sulfur/metabolism
7.
Bioorg Med Chem ; 23(15): 4980-4988, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26043946

ABSTRACT

PABA/NO [O(2)-{2,4-dinitro-5-[4-(N-methylamino)benzoyloxy]phenyl} 1-(N,N-dimethylamino) diazen-1-ium-1,2-diolate] is a nitric oxide (NO)-releasing arylating agent designed to be selectively activated by reaction with glutathione (GSH) on catalysis by glutathione S-transferase P1 (GSTP1), an enzyme frequently overexpressed in cancer cells. PABA/NO has proven active in several cancer models in vitro and in vivo, but its tendency to be metabolized via a variety of pathways, some that generate inactive metabolites and hydrolysis products, limits its potential as a drug. Here we show that a simple replacement of cyano for nitro at the 4 position to give compound 4b ('p-cyano-PABA/NO') has the dual effect of slowing the undesired side reactions while enhancing the proportion of NO release and arylating activity on catalysis by GSTP1. Compound 4b showed increased resistance to hydrolysis and uncatalyzed reaction with GSH, along with a more favorable product distribution in the presence of GSTP1. It also showed significant proapoptotic activity. The data suggest p-cyano-PABA/NO to be a more promising prodrug than PABA/NO, with better selectivity toward cancer cells.


Subject(s)
4-Aminobenzoic Acid/chemistry , Glutathione S-Transferase pi/metabolism , Nitric Oxide/chemistry , 4-Aminobenzoic Acid/metabolism , 4-Aminobenzoic Acid/pharmacology , Biocatalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Glutathione/chemistry , Glutathione/metabolism , Glutathione S-Transferase pi/chemistry , HL-60 Cells , Humans , Isomerism , Kinetics , Molecular Conformation , Nitric Oxide/metabolism , Nitric Oxide/toxicity
8.
Drug Metab Dispos ; 43(7): 1028-36, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870102

ABSTRACT

V-PYRRO/NO [O(2)-vinyl-1-(pyrrolidin-1-yl)diazen-1-ium-1,2-diolate] and V-PROLI/NO (O2-vinyl-[2-(carboxylato)pyrrolidin-1-yl]diazen-1-ium-1,2-diolate), two structurally similar diazeniumdiolate derivatives, were designed as liver-selective prodrugs that are metabolized by cytochrome P450 isoenzymes, with subsequent release of nitric oxide (NO). Yet, their efficacy in the treatment of nonalcoholic fatty liver disease (NAFLD) and their comparative pharmacokinetic and metabolic profiles have not been characterized. The aim of the present work was to compare the effects of V-PYRRO/NO and V-PROLI/NO on liver steatosis, glucose tolerance, and liver fatty acid composition in C57BL/6J mice fed a high-fat diet, as well as to comprehensively characterize the ADME (absorption, distribution, metabolism and excretion) profiles of both NO donors. Despite their similar structure, V-PYRRO/NO and V-PROLI/NO showed differences in pharmacological efficacy in the murine model of NAFLD. V-PYRRO/NO, but not V-PROLI/NO, attenuated liver steatosis, improved glucose tolerance, and favorably modified fatty acid composition in the liver. Both compounds were characterized by rapid absorption following i.p. administration, rapid elimination from the body, and incomplete bioavailability. However, V-PYRRO/NO was eliminated mainly by the liver, whereas V-PROLI/NO was excreted mostly in unchanged form by the kidney. V-PYRRO/NO was metabolized by CYP2E1, CYP2C9, CYP1A2, and CYP3A4, whereas V-PROLI/NO was metabolized mainly by CYP1A2. Importantly, V-PYRRO/NO was a better NO releaser in vivo and in the isolated, perfused liver than V-PROLI/NO, an effect compatible with the superior antisteatotic activity of V-PYRRO/NO. In conclusion, V-PYRRO/NO displayed a pronounced antisteatotic effect associated with liver-targeted NO release, whereas V-PROLI/NO showed low effectiveness, was not taken up by the liver, and was eliminated mostly in unchanged form by the kidney.


Subject(s)
Nitric Oxide Donors/pharmacokinetics , Nitric Oxide Donors/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Pyrrolidines/pharmacology , Pyrrolidines/pharmacokinetics , Pyrrolidines/therapeutic use , Triazenes/pharmacology , Triazenes/therapeutic use , Animals , Biotransformation , Cytochrome P-450 Enzyme System/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Glucose Intolerance , Intestinal Absorption , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Tissue Distribution
9.
Biochem Pharmacol ; 93(3): 389-400, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25534988

ABSTRACT

BACKGROUND AND PURPOSE: There is an unmet medical need for novel NAFLD treatments. Here we have examined the effects of liver-selective NO donor (V-PYRRO/NO) as compared with metformin on hepatic steatosis and glucose tolerance in mice fed high fat diet. MATERIAL AND METHODS: Effects of V-PYRRO/NO (5 mgkg(-1)) or metformin (616 mgkg(-1)) were examined in C57BL/6J mice fed high fat diet (HF, 60 kcal% fat). Quantitative determination of steatosis, liver fatty acid composition and western blot analysis of selected proteins involved in mitochondrial biogenesis, fatty acid de novo synthesis and oxidation, triacylglycerols and cholesterol transport from the liver were performed. Liver NOx and nitrate concentration and blood biochemistry were also analyzed. RESULTS: V-PYRRO/NO and metformin reduced liver steatosis with simultaneous reduction of total liver triacylglycerols, diacylglycerols and ceramides fraction and reversed HF-induced decrease in UFA/SFA ratio. V-PYRRO/NO substantially improved postprandial glucose tolerance, while the effect of metformin was modest and more pronounced on HOMA IR index. The anti-steatotic mechanism of V-PYRRO/NO was dependent on NO release, differed from that of metformin and involved improved glucose tolerance and inhibition of de novo fatty acid synthesis by Akt activation and ACC phosphorylation. In turn, major mechanism of metformin action involved increased expression of proteins implicated in mitochondrial biogenesis and metabolism (PGC-1α, PPARα, COX IV, cytochrome c, HADHSC). CONCLUSIONS: V-PYRRO/NO acts as a liver-specific NO donor prodrug affording pronounced anti-steatotic effects and may represent an efficient, mechanistically novel approach to prevent liver steatosis and insulin resistance.


Subject(s)
Blood Glucose/drug effects , Diet, High-Fat/adverse effects , Fatty Liver/prevention & control , Nitric Oxide Donors/therapeutic use , Postprandial Period/drug effects , Pyrrolidines/therapeutic use , Animals , Blood Glucose/metabolism , Fatty Liver/blood , Fatty Liver/etiology , Glucose Tolerance Test/methods , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Donors/pharmacology , Postprandial Period/physiology , Protective Agents/pharmacology , Protective Agents/therapeutic use , Pyrrolidines/pharmacology
10.
J Org Chem ; 79(19): 9389-93, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25210948

ABSTRACT

We report the apparently unprecedented direct reaction of nitric oxide (NO) with amides to generate ions of structure R(C═O)NH-N(O)═NO(-), with examples including R = Me (1a) or 3-pyridyl (1b). The sodium salts of both released NO in pH 7.4 buffer, with 37 °C half-lives of 1-3 min. As NO-releasing drug candidates, diazeniumdiolated amides would have the advantage of generating only 1 equiv of base on hydrolyzing exhaustively to NO, in contrast to their amine counterparts, which generate 2 equiv of base.


Subject(s)
Amides/chemistry , Azo Compounds/chemistry , Azo Compounds/chemical synthesis , Nitric Oxide/chemistry , Pyridines/chemistry , Molecular Structure
11.
J Org Chem ; 79(10): 4512-6, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24766285

ABSTRACT

Recent theoretical studies have suggested that the parent diazeniumdiolate ion, H2N-N(O)═NO(-) ("diazeniumdiolated ammonia"), might be stable enough to be isolated and that it could potentially serve as a uniquely advantageous prodrug form of bioactive nitroxyl (HNO). Here, we report on an attempt to isolate its O(2)-benzylated derivative by aminolysis of the C═N bond in PhC(NH2)═N-N(O)═NOBn. The reaction proved remarkably sluggish in comparison to aminolysis of unsubstituted benzamidine, and the desired product could not be isolated, apparently because of base sensitivity of the NH2 group. Consistent with this interpretation, O-benzylhydroxylamine and N2O were recovered from the reaction mixture in high yields, along with N,N'-dibutylbenzamidine. Theoretical calculations rationalize the observed slow aminolysis by demonstrating that the diazeniumdiolate group greatly suppresses the electrophilicity of the adjacent C═N carbon center, rendering attack at that position endothermic. The data provide significant insights into the challenges inherent to the pursuit of diazeniumdiolated ammonia.


Subject(s)
Amidines/chemical synthesis , Ammonia/chemistry , Azo Compounds/chemical synthesis , Benzamidines/chemistry , Nitrogen Oxides/chemistry , Amidines/chemistry , Azo Compounds/chemistry
12.
J Med Chem ; 57(6): 2292-302, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24521039

ABSTRACT

We report the antitumor effects of nitric oxide (NO) releasing derivatives of the PARP-1 inhibitor olaparib (1). Compound 5b was prepared by coupling the carboxyl group of 3b and the free amino group of arylated diazeniumdiolated piperazine 4. Analogue 5a has the same structure except that the F is replaced by H. Compound 13 is the same as 5b except that a Me2N-N(O)═NO- group was added para and ortho to the nitro groups of the dinitrophenyl ring. The resulting prodrugs are activated by glutathione in a reaction accelerated by glutathione S-transferase P1 (GSTP1), an enzyme frequently overexpressed in cancers. This metabolism generates NO plus a PARP-1 inhibitor simultaneously, consuming reducing equivalents, leading to DNA damage concomitant with inhibition of DNA repair, and in the case of 13 inducing cross-linking glutathionylation of proteins. Compounds 5b and 13 reduced the growth rates of A549 human lung adenocarcinoma xenografts with no evidence of systemic toxicity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Glutathione S-Transferase pi/metabolism , Neoplasms/enzymology , Nitric Oxide/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Comet Assay , DNA Damage , Drug Design , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/drug effects , Models, Molecular , Neoplasms/drug therapy , Poly (ADP-Ribose) Polymerase-1 , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Pyrazines/pharmacology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
13.
Leuk Res ; 38(3): 377-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24461365

ABSTRACT

The nitric oxide (NO) prodrug JS-K, a promising anti-cancer agent, consists of a diazeniumdiolate group necessary for the release of NO as well as an arylating ring. In this study, we research the mechanism by which JS-K kills a murine erythroleukemia cell line and determine the roles of NO and arylation in the process. Our studies indicate that JS-K inhibits the PI 3-kinase/Akt and MAP kinase pathways. This correlates with the activation of the tumor suppressor FoxO3a and increased expression of various caspases, leading to apoptosis. The arylating capability of JS-K appears to be sufficient for inducing these biological effects. Overall, these data suggest that JS-K kills tumor cells by arylating and inactivating signaling molecules that block the activation of a tumor suppressor.


Subject(s)
Azo Compounds/pharmacology , Cytotoxins/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Prodrugs/pharmacology , Animals , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Forkhead Box Protein O3 , Forkhead Transcription Factors/agonists , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
14.
Nitric Oxide ; 35: 131-6, 2013 Nov 30.
Article in English | MEDLINE | ID: mdl-24126017

ABSTRACT

We introduce a strategy for generating mixtures of nitric oxide (NO) and nitroxyl (HNO) at tunable rates in physiological media. The approach involves converting a spontaneously HNO/NO-generating ion to a caged (prodrug) form that is essentially stable in neutral media, but that can be activated for HNO/NO release by adding an enzyme capable of efficiently opening the cage to regenerate the ion. By judiciously choosing the enzyme, substrate, and reaction conditions, unwanted scavenging of the HNO and NO by the protein can be minimised and the catalytic efficiency of the enzyme can be maintained. We illustrate this approach with a proof-of-concept study wherein the prodrug is Gal-IPA/NO, a diazeniumdiolate of structure iPrHN-N(O)NOR, with R=ß-d-galactosyl. Escherichia coli-derived ß-d-galactosidase at concentrations of 1.9-15nM hydrolysed 56µM substrate with half-lives of 140-19min, respectively, producing the IPA/NO anion (iPrHN-N(O)NO(-), half-life ∼3min), which in turn spontaneously hydrolysed to mixtures of HNO with NO. Using saturating substrate concentrations furnished IPA/NO generation rates that were directly proportional to enzyme concentration. Consistent with these data, the enzyme/substrate combination applied to ventricular myocytes isolated from wild-type mouse hearts resulted not only in a significant positive inotropic effect, but also rescued the cells from the negative inotropy, hypercontractions, and occasional cell death seen with the enzyme alone. This mechanism represents an alternate approach for achieving controlled fluxes of NO/HNO to investigate their biological actions.


Subject(s)
Nitric Oxide/metabolism , Nitrogen Oxides/metabolism , beta-Galactosidase/metabolism , Animals , Anions , Azo Compounds/metabolism , Hydrolysis , Mice , Muscle Cells/metabolism , Myocardium/cytology , Myocardium/metabolism
15.
Redox Biol ; 1: 115-24, 2013.
Article in English | MEDLINE | ID: mdl-24024144

ABSTRACT

JS-K is a nitric oxide (NO)-releasing prodrug of the O (2)-arylated diazeniumdiolate family that has demonstrated pronounced cytotoxicity and antitumor properties in a variety of cancer models both in vitro and in vivo. The current study of the metabolic actions of JS-K was undertaken to investigate mechanisms of its cytotoxicity. Consistent with model chemical reactions, the activating step in the metabolism of JS-K in the cell is the dearylation of the diazeniumdiolate by glutathione (GSH) via a nucleophilic aromatic substitution reaction. The resulting product (CEP/NO anion) spontaneously hydrolyzes, releasing two equivalents of NO. The GSH/GSSG redox couple is considered to be the major redox buffer of the cell, helping maintain a reducing environment under basal conditions. We have quantified the effects of JS-K on cellular GSH content, and show that JS-K markedly depletes GSH, due to JS-K's rapid uptake and cascading release of NO and reactive nitrogen species. The depletion of GSH results in alterations in the redox potential of the cellular environment, initiating MAPK stress signaling pathways, and inducing apoptosis. Microarray analysis confirmed signaling gene changes at the transcriptional level and revealed alteration in the expression of several genes crucial for maintenance of cellular redox homeostasis, as well as cell proliferation and survival, including MYC. Pre-treating cells with the known GSH precursor and nucleophilic reducing agent N-acetylcysteine prevented the signaling events that lead to apoptosis. These data indicate that multiplicative depletion of the reduced glutathione pool and deregulation of intracellular redox balance are important initial steps in the mechanism of JS-K's cytotoxic action.


Subject(s)
Azo Compounds/pharmacology , Leukemia/metabolism , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Prodrugs/chemical synthesis , Acetylcysteine/pharmacology , Azo Compounds/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glutathione/metabolism , Humans , Leukemia/pathology , Membrane Potential, Mitochondrial/drug effects , Nitric Oxide/metabolism , Nitric Oxide Donors/chemical synthesis , Oxidation-Reduction/drug effects , Piperazines/chemical synthesis , Prodrugs/pharmacology , Reactive Nitrogen Species/metabolism
16.
Biomater Sci ; 1(5): 456-459, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23930221

ABSTRACT

O2-protected N-diazeniumdiolate-based silanes were grafted onto mesoporous silica nanoparticles to yield a scaffold with an NO payload of 2.4 µmol NO/mg and NO release half-life of 23 d. Reduced (3-log) Streptococcus mutans viable adhesion was observed for NO-releasing dental restorative materials modified with these particles relative to controls.

17.
Nitric Oxide ; 30: 17-25, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23370169

ABSTRACT

Nitric oxide (NO) released from NO donors can be cytotoxic in tumor cells and can enhance the transport of drugs into brain tumors by altering blood-tumor barrier permeability. The NO donor JS-K [O(2)-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate] releases NO upon enzymatic activation selectively in cells overexpressing glutathione-S-transferases (GSTs) such as gliomas. Thus, JS-K-dependent NO effects - especially on cell viability and vascular permeability - were investigated in U87 glioma cells in vitro and in an orthotopic U87 xenograft model in vivo by magnetic resonance imaging (MRI). In vitro experiments showed dose-dependent antiproliferative and cytotoxic effects in U87 cells. In addition, treatment of U87 cells with JS-K resulted in a dose-dependent activation of soluble guanylate cyclase and intracellular accumulation of cyclic guanosine monophosphate (cGMP) which was irreversibly inhibited by the selective inhibitor of soluble guanylate cyclase ODQ (1H-[1,2,4]oxadiazolo(4,3a)quinoxaline-1-one). Using dynamic contrast enhanced MRI (DCE-MRI) as a minimally invasive technique, we demonstrated for the first time a significant increase in the DCE-MRI read-out initial area under the concentration curve (iAUC60) indicating an acute increase in blood-tumor barrier permeability after i.v. treatment with JS-K. Repeated MR imaging of animals with intracranial U87 gliomas under treatment with JS-K (3.5 µmol/kg JS-K 3×/week) and of untreated controls on day 12 and 19 after tumor inoculation revealed no significant changes in tumor growth, edema formation or tumor perfusion. Immunohistochemical workup of the brains showed a significant antiproliferative effect of JS-K in the gliomas. Taken together, in vitro and in vivo data suggest that JS-K has antiproliferative effects in U87 gliomas and opens the blood-tumor barrier by activation of the NO/cGMP signaling pathway. This might be a novel approach to facilitate entry of therapeutic drugs into brain tumors. DCE-MRI is a non-invasive, repeatable imaging modality to monitor biological effects of NO donors and other experimental therapeutics in intracranial tumor models.


Subject(s)
Azo Compounds/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Magnetic Resonance Imaging/methods , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic GMP/metabolism , Glioma/blood supply , Glioma/metabolism , Humans , Immunohistochemistry , Rats , Rats, Nude , Xenograft Model Antitumor Assays
18.
J Org Chem ; 77(23): 10804-10, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23137026

ABSTRACT

Diazeniumdiolates that have the structure RHN-N(O)═NOR' are of interest as prodrug (caged) forms of the bioeffectors nitric oxide (NO) and nitroxyl (HNO). Previous work has focused on examples possessing α-branched R groups, with isopropylamine (IPA)/NO (R = isopropyl) being the smallest examined to date. To probe the effect of minimizing the alkyl-group size on the chemistry of IPA/NO, we prepared the corresponding methylamine derivative as a sodium salt that was highly unstable but could be trapped in very low overall yield as the stable O(2)-benzyl derivative. To prepare enough for efficient characterization, we devised an alternate synthesis involving a novel N-dealkylation route. CH(3)HN-N(O)═NOBn, synthesized in high yield and crystallized as the Z isomer as determined by X-ray crystallography, was observed to exist as a 11:1 mixture of two isomeric forms in dynamic equilibrium in solution. Similar results were seen for the O(2)-ethyl derivative, whose two equilibrium constituents were partially separated by HPLC to reveal essentially identical UV and mass spectra, indicating them to be Z and E isomers of CH(3)HN-N(O)═NOEt. The results could lead the way to a fuller understanding of the chemistry of the acyclic (E)-diazeniumdiolates.


Subject(s)
Aza Compounds/chemistry , Aza Compounds/chemical synthesis , Methylamines/chemistry , Methylamines/chemical synthesis , Nitric Oxide/chemistry , Alkylation , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Molecular Structure
19.
Chem Res Toxicol ; 25(12): 2670-7, 2012 Dec 17.
Article in English | MEDLINE | ID: mdl-23106594

ABSTRACT

Attachment of glutathione (GSH) to cysteine residues in proteins (S-glutathionylation) is a reversible post-translational modification that can profoundly alter protein structure and function. Often serving in a protective role, for example, by temporarily saving protein thiols from irreversible oxidation and inactivation, glutathionylation can be identified and semiquantitatively assessed using anti-GSH antibodies, thought to be specific for recognition of the S-glutathionylation modification. Here, we describe an alternate mechanism of protein glutathionylation in which the sulfur atoms of the GSH and the protein's thiol group are covalently bound via a cross-linking agent, rather than through a disulfide bond. This form of thiol cross-linking has been shown to occur and has been confirmed by mass spectrometry at the solution chemistry level, as well as in experiments documenting the potent antiproliferative activity of the bis-diazeniumdiolate Double JS-K in H1703 cells in vitro and in vivo. The modification is recognized by the anti-GSH antibody as if it were authentic S-glutathionylation, requiring mass spectrometry to distinguish between them.


Subject(s)
Antineoplastic Agents/pharmacology , Azo Compounds/pharmacology , Glutathione/metabolism , Piperazines/pharmacology , Acetylcysteine/chemistry , Actins/metabolism , Animals , Antineoplastic Agents/chemistry , Azo Compounds/chemistry , Cell Line, Tumor , Dinitrofluorobenzene/analogs & derivatives , Dinitrofluorobenzene/chemistry , Female , Glutathione/chemistry , Humans , Mice , Mice, Nude , Nanoparticles/chemistry , Nitric Oxide/metabolism , Oxygen/metabolism , Piperazines/chemistry
20.
Chem Commun (Camb) ; 48(47): 5931-3, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22572739

ABSTRACT

We report an unexpected ß-elimination pathway by which diazeniumdiolated benzylamines of structure Bn-N(R)-N(O)=N-OR' undergo base-mediated fragmentation to generate N(2)O as the only gaseous product. The reaction is especially rapid for R = 2-hydroxyethyl, in which the hydroxyl group anchimerically assists benzylic proton removal with concomitant expulsion of PhCH=NR and R'OH.


Subject(s)
Azo Compounds/chemistry , Benzylamines/chemistry , Nitrous Oxide/chemistry , Prodrugs
SELECTION OF CITATIONS
SEARCH DETAIL
...