Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Med Chem ; 63(5): 2358-2371, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31589041

ABSTRACT

G-protein-coupled receptors like the human Y1 receptor (hY1R) are promising targets in cancer therapy due to their high overexpression on cancer cells and their ability to internalize together with the bound ligand. This mechanism was exploited to shuttle boron atoms into cancer cells for the application of boron neutron capture therapy (BNCT), a noninvasive approach to eliminate cancer cells. A maximized number of carboranes was introduced to the hY1R-preferring ligand [F7,P34]-NPY by solid phase peptide synthesis. Branched conjugates loaded with up to 80 boron atoms per peptide molecule exhibited a maintained receptor activation profile, and the selective uptake into hY1R-expressing cells was demonstrated by internalization studies. In order to ensure appropriate solubility in aqueous solution, we proved the need for eight hydroxyl groups per carborane. Thus, we suggest the utilization of bis-deoxygalactosyl-carborane building blocks in solid phase peptide synthesis to produce selective boron delivery agents for BNCT.


Subject(s)
Boranes/administration & dosage , Boron/administration & dosage , Drug Carriers/metabolism , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/metabolism , Boranes/chemistry , Boranes/pharmacokinetics , Boron/chemistry , Boron/pharmacokinetics , Boron Neutron Capture Therapy , Drug Carriers/chemistry , Drug Delivery Systems , HEK293 Cells , Humans , MCF-7 Cells , Neoplasms/metabolism , Neoplasms/radiotherapy , Neuropeptide Y/chemistry
2.
Redox Biol ; 28: 101314, 2020 01.
Article in English | MEDLINE | ID: mdl-31514051

ABSTRACT

Nuclear factor-erythroid 2 related factor 2 (Nrf2)-mediated signaling plays a central role in maintaining cellular redox homeostasis of hepatic cells. Carbon monoxide releasing molecule-A1 (CORM-A1) has been reported to stimulate up-regulation and nuclear translocation of Nrf2 in hepatocytes. However, the role of CORM-A1 in improving lipid metabolism, antioxidant signaling and mitochondrial functions in nonalcoholic steatohepatitis (NASH) is unknown. In this study, we report that CORM-A1 prevents hepatic steatosis in high fat high fructose (HFHF) diet fed C57BL/6J mice, used as model of NASH. The beneficial effects of CORM-A1 in HFHF fed mice was associated with improved lipid homeostasis, Nrf2 activation, upregulation of antioxidant responsive (ARE) genes and increased ATP production. As, mitochondria are intracellular source of reactive oxygen species (ROS) and important sites of lipid metabolism, we further investigated the mechanisms of action of CORM-A1-mediated improvement in mitochondrial function in palmitic acid (PA) treated HepG2 cells. Cellular oxidative stress and cell viability were found to be improved in PA + CORM-A1 treated cells via Nrf2 translocation and activation of cytoprotective genes. Furthermore, in PA treated cells, CORM-A1 improved mitochondrial oxidative stress, membrane potential and rescued mitochondrial biogenesis thru upregulation of Drp1, TFAM, PGC-1α and NRF-1 genes. CORM-A1 treatment improved cellular status by lowering glycolytic respiration and maximizing OCR. Improvement in mitochondrial respiration and increment in ATP production in PA + CORM-A1 treated cells further corroborate our findings. In summary, our data demonstrate for the first time that CORM-A1 ameliorates tissue damage in steatotic liver via Nrf2 activation and improved mitochondrial function, thus, suggesting the anti-NASH potential of CORM-A1.


Subject(s)
Boranes/administration & dosage , Carbonates/administration & dosage , Diet, High-Fat/adverse effects , High Fructose Corn Syrup/adverse effects , NF-E2-Related Factor 2/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Boranes/pharmacology , Carbonates/pharmacology , Cell Survival , Disease Models, Animal , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress/drug effects , Palmitic Acid/pharmacology , Signal Transduction/drug effects
3.
J Cell Physiol ; 233(5): 4156-4165, 2018 05.
Article in English | MEDLINE | ID: mdl-29034470

ABSTRACT

Concanavalin A (ConA)-induced hepatitis is an experimental model of human autoimmune hepatitis induced in rodents by i.v. injection of Con A. The disease is characterized by increase in serum levels of transaminases and massive immune infiltration of the livers. Type 1, type 2, and type 17 cytokines play a pathogenic role in the development of ConA-induced hepatitis. To understand further the immunoregulatory mechanisms operating in the development and regulation of ConA-induced hepatitis, we have evaluated the role of the anti-inflammatory pathway Nrf2/HO-1/CO (Nuclear Factor E2-related Factor 2/Heme Oxygenase-1/Carbon Monoxide) in this condition and determined whether the in vivo administration of CO via the CO-releasing molecule (CORM) CORM-A1, influences serological and histological development of Con-A-induced hepatitis. We have firstly evaluated in silico the genes belonging to the Nrf2/HO-1/CO pathway that are involved in the pathogenesis of autoimmune hepatitis (AIH). The data obtained from the in silico study demonstrate that a significant number of genes modulated in the liver of ConA-challenged mice belong to the Nrf2 pathway; on the other hand, the administration of CORM-A1 determines an improvement in several sero-immunological and histological parameters, and it is able to modulate genes identified by the in silico analysis. Collectively, our data indicate that the Nrf2/HO-1/CO pathway is fundamental for the regulation of the immune responses, and that therapeutic intervention aimed at its modulation by CORM-A1 may represent a valuable strategy to be considered for the treatment of autoimmune hepatitis in humans.


Subject(s)
Heme Oxygenase-1/genetics , Hepatitis, Autoimmune/genetics , Inflammation/genetics , Membrane Proteins/genetics , NF-E2-Related Factor 2/genetics , Animals , Boranes/administration & dosage , Carbon Monoxide/metabolism , Carbonates/administration & dosage , Concanavalin A/toxicity , Cytokines/metabolism , Disease Models, Animal , Hepatitis, Autoimmune/etiology , Hepatitis, Autoimmune/physiopathology , Humans , Inflammation/metabolism , Inflammation/physiopathology , Liver/metabolism , Liver/physiopathology , Mice , Signal Transduction , Tumor Necrosis Factor-alpha
4.
Biochem Biophys Res Commun ; 478(1): 279-285, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27402268

ABSTRACT

Carboranes are a class of carbon-containing polyhedral boron cluster compounds with globular geometry and hydrophobic surface that interact with hormone receptors such as estrogen receptor (ER) and androgen receptor (AR). We have synthesized BA321, a novel carborane compound, which binds to AR. We found here that it also binds to ERs, ERα and ERß. In orchidectomized (ORX) mice, femoral bone mass was markedly reduced due to androgen deficiency and BA321 restored bone loss in the male, whilst the decreased weight of seminal vesicle in ORX mice was not recovered by administration of BA321. In female mice, BA321 acts as a pure estrogen agonist, and restored both the loss of bone mass and uterine atrophy due to estrogen deficiency in ovariectomized (OVX) mice. In bone tissues, the trabecular bone loss occurred in both ORX and OVX mice, and BA321 completely restored the trabecular bone loss in both sexes. Cortical bone loss occurred in ORX mice but not in OVX mice, and BA321 clearly restored cortical bone loss due to androgen deficiency in ORX mice. Therefore, BA321 is a novel selective androgen receptor modulator (SARM) that may offer a new therapy option for osteoporosis in the male.


Subject(s)
Androgens/metabolism , Boranes/administration & dosage , Osteoporosis/drug therapy , Osteoporosis/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Boranes/pharmacokinetics , Dose-Response Relationship, Drug , Female , Gonads/drug effects , Gonads/metabolism , Humans , Male , Mice , Mice, Inbred Strains , Orchiectomy , Osteoporosis/pathology , Ovariectomy , Treatment Outcome
5.
Immunol Lett ; 165(1): 39-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25839127

ABSTRACT

We have recently shown that carbon monoxide releasing molecule (CORM)-A1 prevents type 1 diabetes induced in C57BL/6 mice with multiple low doses of streptozotocin (MLDS) by shifting the Th1/Th17/M1 balance towards a Th2/M2 response. In the present work we tested the hypothesis that CORM-A1 might influence regulatory arm of the immune response, as well as beta cell regeneration. CORM-A1 (2 mg/kg/day) was administered for 10 days to mice induced with MLDS and/or depleted of low dose cyclophosphamide (CY)-sensitive FoxP3+ T regulatory (Treg) cells. Besides monitoring hyperglycaemia, ex vivo analysis of spleen, pancreatic lymph nodes (PLN) and pancreas was performed at the end of treatment. In CORM-A1-treated MLDS-induced mice the improvement of hyperglycaemia was observed only without depletion of CY-sensitive FoxP3+ Treg cells. This was accompanied by decreased levels of interleukin (IL)-12, IL-2 and early activation marker CD25 in the spleen and PLN and increased transforming growth factor (TGF)-ß, resulting in reduced lymphocyte proliferation in both organs. In parallel, decreased transcript levels of IL-2, but increased mRNA expression of TGF-ß, accompanied with up-regulation of Ki-67 protein expression was observed within pancreas. Together, the data suggested that besides the immunomodulatory potential, CORM-A1 probably induces beta cell regeneration.


Subject(s)
Boranes/pharmacology , Carbonates/pharmacology , Hypoglycemic Agents/pharmacology , Animals , Boranes/administration & dosage , Carbonates/administration & dosage , Cytokines/biosynthesis , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Hypoglycemic Agents/administration & dosage , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacology , Immunomodulation/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Male , Mice , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
6.
Xenobiotica ; 44(1): 36-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23786350

ABSTRACT

1. Dimethylamine borane (DMAB) is used as a reducing agent in the manufacturing of a variety of products and in chemical synthesis. National Toxicology Program is evaluating the toxicity of DMAB in rodents following dermal application. The objective of this study was to evaluate the metabolism and disposition of DMAB in male Harlan Sprague Dawley (HSD) rats. 2. Disposition of radioactivity was similar between gavage and intravenous administration of 1.5 mg/kg [(14)C] DMAB, with nearly 84%-89% of the administered radioactivity recovered in urine 24 h post dosing. At 72 h, only 1% or less was recovered in feces, 0.3% as CO2, and 0.5%-1.4% as volatiles and 0.3%-0.4 % in tissues. 3. The absorption of [(14)C]DMAB following dermal application was moderate; percent dose absorbed increased with the dose, with 23%, 32% and 46% of dose absorbed at 0.15, 1.5 and 15 mg/kg, respectively. Urinary and fecal excretion ranged from 18%-37% and 2%-4% of dose, respectively, and 0.1%-0.2% as CO2, and 1%-3% as volatiles. Tissue retention of the radiolabel was low ∼1%, but was higher than following the gavage or intravenous administration. 4. Following co-adminsitration of DMAB and sodium nitrite by gavage, N-nitrosodimethylamine was not detected in blood or urine above the limit of quantitation of the analytical method of 10 ng/mL. 5. Absorption of DMAB in fresh human skin in vitro was ∼41% of the applied dose: the analysis of the receptor fluid shows that the intact DMAB complex can be absorbed through the skin.


Subject(s)
Boranes/administration & dosage , Boranes/metabolism , Dimethylamines/administration & dosage , Dimethylamines/metabolism , Administration, Cutaneous , Administration, Intravenous , Animals , Boranes/pharmacokinetics , Carbon Radioisotopes/administration & dosage , Carbon Radioisotopes/pharmacokinetics , Carbon Radioisotopes/urine , Dimethylamines/pharmacokinetics , Dimethylnitrosamine/blood , Dimethylnitrosamine/urine , Feces/chemistry , Humans , Male , Rats , Rats, Sprague-Dawley , Sodium Nitrite/administration & dosage
7.
Int J Pharm ; 458(2): 340-6, 2013 Dec 31.
Article in English | MEDLINE | ID: mdl-24140178

ABSTRACT

In this paper we propose a bottom-up approach to obtain new boron carriers built with ortho-carborane functionalized gold nanoparticles (GNPs) for applications in Boron Neutron Capture Therapy. The interaction between carboranes and the gold surface was assured by one or two SH-groups directly linked to the boron atoms of the B10C2 cage. This allowed obtaining stable, nontoxic systems, though optimal biological performance was hampered by low solubility in aqueous media. To improve cell uptake, the hydrophilic character of carborane functionalized GNPs was enhanced by further coverage with an appropriately tailored diblock copolymer (PEO-b-PCL). This polymer also contained pendant carboranes to provide anchoring to the pre-functionalized GNPs. In vitro tests, carried out on osteosarcoma cells, showed that the final vectors possessed excellent biocompatibility joint to the capacity of concentrating boron atoms in the target, which is encouraging evidenced to pursue applications in vivo.


Subject(s)
Boranes/chemistry , Boron Neutron Capture Therapy/methods , Boron/chemistry , Gold/chemistry , Nanoparticles/chemistry , Osteosarcoma/radiotherapy , Animals , Boranes/administration & dosage , Boron/administration & dosage , Cell Line, Tumor , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Gold/administration & dosage , Hydrophobic and Hydrophilic Interactions , Nanoparticles/administration & dosage , Polymers/administration & dosage , Polymers/chemistry , Rats , Solubility , Water/chemistry
8.
Biomaterials ; 34(4): 902-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23174143

ABSTRACT

Multidrug resistance (MDR) of bacteria is still an unsolved serious problem to threaten the health of human beings. Developing new antibacterial agents, therefore, are urgently needed. Herein, we have explored the possibility to design and synthesize some novel antibacterial agents including ferrocene-substituted carborane derivative (Fc(2)SBCp(1)) and have evaluated the relevant antibacterial action against two clinical common MDR pathogens (i.e., Gram-positive Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa) in vitro and in vivo. The results demonstrate that in vitro antimicrobial activity of Fc(2)SBCp(1) could be gradually transformed into a bactericidal effect from a bacteriostatic effect with the increasing concentration of the active carborane derivative, which can also prevent biofilm formation at concentrations below MIC (i.e., minimal inhibitory concentration). Biocompatibility studies indicate that there exists no/or little toxic effect of Fc(2)SBCp(1) on normal cells/tissues and leads to little hemolysis. In vivo studies illustrate that the new carborane derivative Fc(2)SBCp(1) is highly effective in treating bacteremia caused by S. aureus and P. aeruginosa as well as interstitial pneumonia caused by S. aureus. This raises the possibility for the potential utilization of the new ferrocene-substituted carborane derivatives as promising antibacterial therapeutic agents against MDR bacterial infections in future clinical applications.


Subject(s)
Bacteremia/drug therapy , Boranes/administration & dosage , Boranes/chemistry , Drug Resistance, Multiple, Bacterial , Ferrous Compounds/administration & dosage , Ferrous Compounds/chemistry , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemical synthesis , Bacteremia/pathology , Bacterial Physiological Phenomena/drug effects , Carbon/chemistry , Carbon/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Male , Metallocenes , Mice , Treatment Outcome
9.
Arterioscler Thromb Vasc Biol ; 32(9): 2149-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22772756

ABSTRACT

OBJECTIVE: We compared the antithrombotic effects in vivo of 2 chemically different carbon monoxide-releasing molecules (CORM-A1 and CORM-3) on arterial and venous thrombus formation and on hemostatic parameters such as platelet activation, coagulation, and fibrinolysis. The hypotensive response to CORMs and their effects on whole blood gas analysis and blood cell count were also examined. METHODS AND RESULTS: CORM-A1 (10-30 µmol/kg, i.v.), in a dose-dependent fashion, significantly decreased weight of electrically induced thrombus in rats, whereas CORM-3 inhibited thrombosis only at the highest dose used (30 µmol/kg). CORM-A1 showed a direct and stronger inhibition of platelet aggregation than CORM-3 in healthy rats, both in vitro and in vivo. The antiaggregatory effect of CORM-A1, but not CORM-3, correlated positively with weight of the thrombus. Concentration of active plasminogen activator inhibitor-1 in plasma also decreased in response to CORM-A1, but not to CORM-3. Neither CORM-A1 nor CORM-3 had an effect on plasma concentration of active tissue plasminogen activator. CORM-3, but not CORM-A1, decreased the concentration of fibrinogen, fibrin generation, and prolonged prothrombin time. Similarly, laser-induced venous thrombosis observed intravitally via confocal system in green fluorescent protein mice was significantly decreased by CORMs. Although both CORM-A1 and CORM-3 (30 µmol/kg) decreased platelets accumulation in thrombus, only CORM-A1 (3-30 µmol/kg) inhibited platelet activation to phosphatidylserine on their surface. CONCLUSIONS: CORM-3 and CORM-A1 inhibited thrombosis in vivo, however CORM-A1, which slowly releases carbon monoxide, and displayed a relatively weak hypotensive effect had a more pronounced antithrombotic effect associated with a stronger inhibition of platelet aggregation associated with a decrease in active plasminogen activator inhibitor-1 concentration. In contrast, the fast CO releaser CORM-3 that displayed a more pronounced hypotensive effect inhibited thrombosis primarily through a decrease in fibrin generation, but had no direct influence on platelet aggregation and fibrynolysis.


Subject(s)
Arterial Occlusive Diseases/prevention & control , Boranes/pharmacology , Carbon Monoxide/metabolism , Carbonates/pharmacology , Fibrinolytic Agents/pharmacology , Organometallic Compounds/pharmacology , Thrombosis/prevention & control , Venous Thrombosis/prevention & control , Water/chemistry , Animals , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/etiology , Arterial Occlusive Diseases/physiopathology , Blood Coagulation/drug effects , Blood Gas Analysis , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Pressure/drug effects , Boranes/administration & dosage , Boranes/chemistry , Boranes/metabolism , Carbonates/administration & dosage , Carbonates/chemistry , Carbonates/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrin/metabolism , Fibrinogen/metabolism , Fibrinolysis/drug effects , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Organometallic Compounds/administration & dosage , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Plasminogen Activator Inhibitor 1/blood , Platelet Aggregation/drug effects , Prothrombin Time , Rats , Rats, Wistar , Solubility , Thrombosis/blood , Thrombosis/etiology , Thrombosis/physiopathology , Time Factors , Venous Thrombosis/blood , Venous Thrombosis/etiology , Venous Thrombosis/physiopathology
10.
Clin Exp Immunol ; 167(2): 179-87, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22235993

ABSTRACT

Carbon monoxide (CO) is produced during the catabolism of free haem, catalyzed by haem oxygenase (HO) enzymes, and its physiological roles include vasodilation, neurotransmission, inhibition of platelet aggregation and anti-proliferative effects on smooth muscle. In vivo preclinical studies have shown that exogenously administered quantities of CO may represent an effective treatment for conditions characterized by a dysregulated immune response. The carbon monoxide-releasing molecules (CORMs) represent a group of compounds capable of carrying and liberating controlled quantities of CO in the cellular systems. This review covers the physiological and anti-inflammatory properties of the HO/CO pathway in the central nervous system. It also discusses the effects of CORMs in preclinical models of inflammation. The accumulating data discussed herein support the possibility that CORMs may represent a novel class of drugs with disease-modifying properties in multiple sclerosis.


Subject(s)
Boranes/therapeutic use , Carbon Monoxide/therapeutic use , Carbonates/therapeutic use , Multiple Sclerosis/drug therapy , Organometallic Compounds/therapeutic use , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Autoimmunity/drug effects , Boranes/administration & dosage , Carbon Monoxide/administration & dosage , Carbon Monoxide/metabolism , Carbonates/administration & dosage , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/therapeutic use , Cytokines/biosynthesis , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Guanylate Cyclase/metabolism , Heme/metabolism , Heme Oxygenase (Decyclizing)/physiology , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/physiology , Humans , Inflammation/drug therapy , Multiple Sclerosis/immunology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Organometallic Compounds/administration & dosage , Oxidation-Reduction , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Soluble Guanylyl Cyclase , Vasodilator Agents/administration & dosage , Vasodilator Agents/therapeutic use
11.
J Nanobiotechnology ; 9: 6, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21338526

ABSTRACT

BACKGROUND: The new ruthenium(II)-arene complex, which bearing a carborane unit, ruthenium and ferrocenyl functional groups, has a novel versatile synthetic chemistry and unique properties of the respective material at the nanoscale level. The ruthenium(II)-arene complex shows significant cytotoxicity to cancer cells and tumor-inhibiting properties. However, ruthenium(II)-arene complex of mechanism of anticancer activity are scarcely explored. Therefore, it is necessary to explore ruthenium(II)-arene complex mechanism of anticancer activity for application in this area. RESULTS: In this study, the ruthenium(II)-arene complex could significantly induce apoptosis in human lung cancer HCC827 cell line. At the concentration range of 5 µM-100 µM, ruthenium(II)-arene complex had obvious cell cytotoxicity effect on HCC827 cells with IC(50) values ranging 19.6 ± 5.3 µM. Additionally, our observations demonstrate that the ruthenium(II)-arene complex can readily induce apoptosis in HCC827 cells, as evidenced by Annexin-V-FITC, nuclear fragmentation as well as DNA fragmentation. Treatment of HCC827 cells with the ruthenium(II)-arene complex resulted in dose-dependent cell apoptosis as indicated by high cleaved Caspase-8,9 ratio. Besides ruthenium(II)-arene complex caused a rapid induction of cleaved Caspase-3 activity and stimulated proteolytic cleavage of poly-(ADP-ribose) polymerase (PARP) in vitro and in vivo. CONCLUSION: In this study, the ruthenium(II)-arene complex could significantly induce apoptosis in human lung cancer HCC827 cell line. Treatment of HCC827 cells with the ruthenium(II)-arene complex resulted in dose-dependent cell apoptosis as indicated by high cleaved Caspase-8,9 ratio. Besides ruthenium(II)-arene complex caused a rapid induction of cleaved Caspase-3 activity and stimulated proteolytic cleavage of poly-(ADP-ribose) polymerase (PARP) in vitro and in vivo. Our results suggest that ruthenium(II)-arene complex could be a candidate for further evaluation as a chemotherapeutic agent for human cancers, especially lung cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Boranes/administration & dosage , Lung Neoplasms/drug therapy , Nanostructures/administration & dosage , Ruthenium Compounds/administration & dosage , Animals , Boranes/chemistry , Caspases/metabolism , Cell Line, Tumor , Humans , Mice , Mice, Nude , Poly(ADP-ribose) Polymerases/metabolism
12.
Am J Physiol Cell Physiol ; 300(2): C256-65, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21123734

ABSTRACT

We investigated the role of reactive oxygen species (ROS) in promoting cell survival during oxidative stress induced by the inflammatory mediator tumor necrosis factor-α (TNF-α) in cerebral microvascular endothelial cells (CMVEC) from newborn piglets. Nox4 is the major isoform of NADPH oxidase responsible for TNF-α-induced oxidative stress and apoptosis in CMVEC. We present novel data that Nox4 NADPH oxidase-derived ROS also initiate a cell survival mechanism by increasing production of a gaseous antioxidant mediator carbon monoxide (CO) by constitutive heme oxygenase-2 (HO-2). TNF-α rapidly enhanced endogenous CO production in a superoxide- and NADPH oxidase-dependent manner in CMVEC with innate, but not with small interfering RNA (siRNA)-downregulated Nox4 activity. CORM-A1, a CO-releasing compound, inhibited Nox4-mediated ROS production and enhanced cell survival in TNF-α-challenged CMVEC. The ROS-induced CO-mediated survival mechanism requires functional interactions between the protein kinase B/Akt and extracellular signal-related kinase (ERK)/p38 MAPK signaling pathways activated by TNF-α. In Akt siRNA-transfected CMVEC and in cells with pharmacologically inhibited Akt, Erk1/2, and p38 mitogen-activated protein kinase (MAPK) activities, CORM-A1 was no longer capable of blocking Nox4 activation and apoptosis caused by TNF-α. Overall, Nox4 NADPH oxidase-derived ROS initiate both death and survival pathways in TNF-α-challenged CMVEC. The ROS-dependent cell survival pathway is mediated by an endogenous antioxidant CO, which inhibits Nox4 activation via a mechanism that includes Akt, ERK1/2, and p38 MAPK signaling pathways. The ability of CO to inhibit TNF-α-induced ERK1/2 and p38 MAPK activities in an Akt-dependent manner appears to be the key element in ROS-dependent survival of endothelial cells during TNF-α-mediated brain inflammatory disease.


Subject(s)
Apoptosis , Brain/metabolism , Carbon Monoxide/metabolism , Endothelial Cells/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Newborn , Boranes/administration & dosage , Brain/drug effects , Carbonates/administration & dosage , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Humans , Isoenzymes/metabolism , Microvessels/drug effects , Microvessels/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Swine/metabolism , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Radiat Res ; 51(5): 621-6, 2010.
Article in English | MEDLINE | ID: mdl-20921830

ABSTRACT

Boron Neutron Capture Therapy (BNCT) effectiveness depends on the preferential sequestration of boron in cancer cells relative to normal tissue cells. We present a novel strategy for sequestering boron using an adenovirus expressing the sodium iodide symporter (NIS). Human glioma grown subcutaneously in athymic mice and orthotopic rat brain tumors were transfected with NIS using a direct tumor injection of adenovirus. Boron bound as sodium tetrafluoroborate (NaBF(4)) was administered systemically several days after transfection. Tumors were excised hours later and assessed for boron concentration using inductively coupled plasma atomic emission spectroscopy. In the human glioma transfected with NIS, boron concentration was more than 10 fold higher with 100 mg/kg of NaBF(4), compared to tumor not transfected. In the orthotopic tumor model, the presence of NIS conferred almost 4 times the boron concentration in rat tumors transfected with human virus compared with contralateral normal brain not transfected. We conclude that adenovirus expressing NIS has the potential to be used as a novel boron delivery agent and should be explored for future clinical applications.


Subject(s)
Boranes/administration & dosage , Boron Neutron Capture Therapy , Brain Neoplasms/therapy , Glioma/therapy , Symporters/genetics , Symporters/therapeutic use , Adenoviridae , Animals , Boron Neutron Capture Therapy/methods , Genetic Vectors , Humans , Mice , Rats
14.
Bioconjug Chem ; 17(1): 15-20, 2006.
Article in English | MEDLINE | ID: mdl-16417247

ABSTRACT

A new type of boron-rich, DSPC-free, unilamellar liposomes was formed using the novel dual-chain, ionic, nido-carborane lipid, K[nido-7-(C16H33OCH2)2CHOCH2-7,8-C2B9H11] (DAC-16), and cholesterol for encapsulation of an aqueous buffer core. Since DSPC was not necessary for the formation of stable DAC-16 liposomes, the boron concentration of these vesicles was increased dramatically to approximately 8.8 wt % in the dry lipid; these liposomes had a high bilayer boron incorporation efficiency of 98%. DSPC-free liposomes exhibited a size distribution pattern of 40-60 nm, which was in the range normally associated with selective tumor uptake. This size distribution was maintained throughout storage at room temperature for several months. Additionally, optimized liposome formulations incorporating DAC-16, DSPC, and cholesterol were identified having stable size distribution patterns after storage for more than two months at a variety of temperatures. Although animal studies indicate that DAC-16 liposomes are toxic, this new ionic nido-carborane lipid allows the formation of liposomes of high boron content for in vitro applications that require the delivery of large amounts of boron.


Subject(s)
Boranes/chemistry , Liposomes/chemical synthesis , Animals , Boranes/administration & dosage , Boranes/toxicity , Boron Neutron Capture Therapy , Liposomes/administration & dosage , Liposomes/toxicity , Male , Mice , Mice, Inbred BALB C , Toxicity Tests, Acute
15.
Cancer Res ; 62(22): 6552-8, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12438250

ABSTRACT

Convection enhanced delivery (CED) is potentially a powerful method to improvethe targeting of macromolecules to the central nervous system by applying a pressure gradient to establish bulk flow through the brain interstitium during infusion. The purpose of the present study was to evaluate CED as a means to improve the intracerebral and intratumoral (i.t.) uptake of a heavily boronated macromolecule (dendrimer; BD) linked to epidermal growth factor (EGF) for neutron capture therapy in rats bearing a syngeneic epidermal growth factor receptor (EGFR) + glioma. Boronated EGF was radiolabeled with 125I and administered by CED at a rate of 0.33 micro l/min for 15, 30, and 60 min [infusion volumes (V(I)) of 5, 10, and 20 micro l, respectively], using a syringe pump connected to an indwelling cannula implanted into the right caudate nucleus of normal rats or i.t. in rats bearing either F98(EGFR) or F98 wild-type (F98(WT)) gliomas. After infusion, rats were euthanized, and their brains were removed and serially sectioned. The uptake and biodistribution of (125)I-boronated EGF in tumor or brain was studied by quantitative autoradiography and gamma-scintillation counting. The volume of distribution (V(d)) in brain was assessed using a computer interfaced image analysis system. After CED, the V(d) increased from 34.4 to 123.5 micro l with corresponding V(i) ranging from 5 to 20 micro l. The V(d) of BD-EGF in the brain was 64.8 +/- 13.4 micro l with CED (V(i) 10 micro ), and the V(d):V(i) ratio was 6.5 compared with a V(d) of 9.4 +/- 1.6 micro l and a V(d):V(i) ratio of 0.9 after direct intracerebral injection. As determined by quantitative autoradiography and gamma-scintillation counting at 24 h after CED, 47.4% of the injected dose per gram tissue (%ID/g) was localized in F98(EGFR) gliomas compared with 33.2%ID/g after direct i.t. injection and 12.3%ID/g in F98(WT) gliomas. On the basis of these observations, we have concluded that CED is more effective than i.t. injection as a way to deliver boronated EGF to EGFR (+) gliomas for boron neutron capture therapy.


Subject(s)
Boranes/pharmacokinetics , Boron Neutron Capture Therapy/methods , Brain Neoplasms/metabolism , Epidermal Growth Factor/pharmacokinetics , ErbB Receptors/metabolism , Glioma/metabolism , Animals , Autoradiography , Boranes/administration & dosage , Brain/diagnostic imaging , Brain/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/radiotherapy , Dendrimers , Epidermal Growth Factor/administration & dosage , Glioma/diagnostic imaging , Glioma/radiotherapy , Iodine Radioisotopes , Polyamines/administration & dosage , Polyamines/pharmacokinetics , Radionuclide Imaging , Rats , Rats, Inbred F344 , Tissue Distribution
16.
Eur J Med Chem ; 37(8): 649-57, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12161062

ABSTRACT

The synthesis and biological in vitro and in vivo activities of possible new compounds for Boron Neutron Capture Therapy (BNCT) are reported. The azanonaboranes of the type [(RNH(2))B(8)H(11)NHR] are water-soluble when hydrophilic groups are introduced. The reaction of B(9)H(13)SMe(2) with primary amines yields azanonaboranes. Five compounds with different numbers of hydroxypropyl groups have been isolated: [(HO(CH(2))(3)NH(2))B(8)H(11)NHCH(3)] (4), [(HO(CH(2))(3)NH(2))B(8)H(11)NH(CH(2))(3)OH] (2), [((HO(CH(2))(3))(2)NH)B(8)H(11)NHCH(3)] (6), [((HO(CH(2))(3))(2)NH)B(8)H(11)NH(CH(2))(3)OCH(3)] (11) and [((HO(CH(2))(3))(2)NH)B(8)H(11)NH(CH(2))(3)OH] (8). In vitro experiments as judged by cloning survival tests showed that two of the synthesised compounds are not toxic. The in vivo experiments were carried out with C3H/He mice bearing SCCVII tumours and C57 mice bearing B16 tumours. Compounds 2 and 6 have no particular affinity to any tissue, but are excluded from the brain.


Subject(s)
Boranes/chemical synthesis , Boranes/pharmacology , Boron Neutron Capture Therapy , Animals , Boranes/administration & dosage , CHO Cells , Cell Survival/drug effects , Cricetinae , Mice , Mice, Inbred C3H , Molecular Structure , Neoplasms, Experimental/drug therapy , Organ Specificity , Polyamines/chemical synthesis , Propanolamines/chemical synthesis , Radiography , Structure-Activity Relationship , Tissue Distribution
17.
Jpn J Cancer Res ; 92(9): 996-1007, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11572769

ABSTRACT

C57BL mice bearing EL4 tumors and C3H / He mice bearing SCC VII tumors received 5-bromo-2'-deoxyuridine (BrdU) continuously for 5 days via implanted mini-osmotic pumps to label all proliferating (P) cells. Three hours after oral administration of l-p-boronophenylalanine-(10)B (BPA), or 30 min after intraperitoneal injection of sodium borocaptate-(10)B (BSH) or l-p-boronophenylalaninol (BPA-ol), a newly developed (10)B-containing alpha-amino alcohol, the tumors were irradiated with thermal neutron beams. For the combination with mild temperature hyperthermia (MTH) and / or tirapazamine (TPZ), the tumors were heated at 40 degrees C for 30 min immediately before neutron exposure, and TPZ was intraperitoneally injected 30 min before irradiation. The tumors were then excised, minced and trypsinized. The tumor cell suspensions thus obtained were incubated with cytochalasin-B (a cytokinesis blocker), and the micronucleus (MN) frequency in cells without BrdU labeling ( = quiescent (Q) cells) was determined using immunofluorescence staining for BrdU. Meanwhile, 6 h after irradiation, tumor cell suspensions obtained in the same manner were used for determining the apoptosis frequency in Q cells. The MN and apoptosis frequency in total (P + Q) tumor cells were determined from tumors that were not pretreated with BrdU. Without TPZ or MTH, BPA-ol increased both frequencies most markedly, especially for total cells. However, as with BPA, the sensitivity difference between total and Q cells was much larger than with BSH. On combined treatment with both MTH and TPZ, this sensitivity difference was markedly reduced, similarly to when BPA was used. MTH increased the (10)B uptake of all (10)B-compounds into both tumor cells. BPA-ol has good potential as a (10)B-carrier in neutron capture therapy, especially when combined with both MTH and TPZ.


Subject(s)
Boranes/pharmacokinetics , Boron Neutron Capture Therapy , Carcinoma, Squamous Cell/therapy , Lymphoma/therapy , Phenylalanine/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Boranes/administration & dosage , Boranes/chemistry , Boranes/radiation effects , Bromodeoxyuridine/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Combined Modality Therapy , Cytochalasin B/pharmacology , Drug Screening Assays, Antitumor , Fluorescent Antibody Technique, Indirect , Hindlimb , Hyperthermia, Induced , Injections, Intraperitoneal , Interphase , Lymphoma/drug therapy , Lymphoma/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Micronucleus Tests , Molecular Structure , Neutrons , Phenylalanine/administration & dosage , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/radiation effects , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/therapeutic use , Radiometry , Tirapazamine , Triazines/administration & dosage , Triazines/therapeutic use
18.
J Neurooncol ; 33(1-2): 53-8, 1997 May.
Article in English | MEDLINE | ID: mdl-9151223

ABSTRACT

The successful treatment of cancer by boron neutron capture therapy (BNCT) requires the selective concentration of boron-10 within malignant tumors. The potential of liposomes to deliver boron-rich compounds to tumors has been assessed by examination of the biodistribution of boron delivered by liposomes in tumor-bearing mice. Small unilamellar vesicles have been found to stably encapsulate high concentrations of water-soluble ionic boron compounds. Alternatively, lipophilic boron-containing species have been embedded within the phospholipid bilayer of liposomes, and both hydrophilic and lipophilic boron compounds have been incorporated within the same liposome formulation. The biodistribution of boron was determined at several time points over 48 hr after i.v. injection of liposomal suspensions in BALB/c mice bearing EMT6 tumors. The tumor-selective delivery of boron by the liposomes was demonstrated as tumor-boron concentrations increased for several hours post-injection. Even at the low injected doses employed (6-18 mg boron/kg body weight) therapeutic tumor boron concentrations were observed (> 30 micrograms boron/g tissue) and high tumor/blood ratios were achieved (> 5). The most favorable results were obtained with the polyhedral borane Na3[a2-B20H1-NH2CH2CH2NH2]. Liposomes encapsulating this species produced a tumor boron concentration of 45 micrograms/g tissue at 30 hr post-injection, at which time the tumor/blood boron ratio was 9.3.


Subject(s)
Adenocarcinoma/metabolism , Boranes/pharmacokinetics , Mammary Neoplasms, Experimental/metabolism , Adenocarcinoma/radiotherapy , Animals , Boranes/administration & dosage , Boranes/chemistry , Boron Neutron Capture Therapy , Drug Carriers , Female , Liposomes , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred BALB C , Molecular Structure , Tissue Distribution
19.
Radiother Oncol ; 38(1): 41-50, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8850425

ABSTRACT

In boron neutron capture therapy (BNCT), 10B is delivered selectively to the tumour cells and the nuclide then forms high-LET radiation (4He2+ and 7Li3+) upon neutron capture. Today much research is focused on development of a variety of boron compounds aimed for BNCT. The compounds must be thoroughly analysed in preclinical tests regarding basic characteristics such as binding and subcellular distribution to enable accurate estimations of dose-modifying factors. DAC-1,2-[2-(3-amino-propyl)-1,2-dicarba-closo-dodecaboran (12)-1-yl-methoxy]- 1,3-propanediol was synthesized at our laboratories and the human colon carcinoma cells LS-174T were used as an in vitro model. The boron compound showed a remarkable intracellular accumulation, 20-100 times higher than the boron content in the culture medium, in cultured cells and was not removed by extensive washes. Approximately half of the boron taken up also remained within the cells for at least 4 days. The DAC-1 compound alone was not toxic at boron concentrations below 2.5 micrograms B/g. The intracellular distribution of the boron compound was investigated by subcellular fractionation experiments and low pH treatments. It is possible that DAC-1 binds to some intracellular molecules or to membranes connected with organelles in the cytoplasm or even to the inside of the outer cell membrane. Another possibility is that the compound, due to the somewhat lipophilic properties, is embedded in the membranes. Thermal neutron irradiations were carried out at the Brookhaven Medical Research Reactor (BMRR). At a survival level of 0.1, DAC-1 + thermal neutrons were about 10.5 times more effective in cell inactivation than the thermal neutrons alone. Monte Carlo calculations gave a mean value of the 10B-dependent specific energy, the dose, of 0.22 Gy. The total physical dose during irradiation of DAC-1-containing cells with a neutron fluence of 0.18 x 10(12) n/cm2 was 0.39 Gy. The dose-modifying factor, at survival level 0.1, when comparing irradiation with thermal neutrons with and without DAC-1 was 3.4, while the dose-modifying factor when comparing neutron irradiations of cells with DAC-1 and irradiation of the cells with 60Co-gamma was 7.3. The results are encouraging and in vivo tests of tissue distributions and tumour uptake should now be carried out.


Subject(s)
Boranes/pharmacokinetics , Boron Neutron Capture Therapy , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Biological Availability , Boranes/administration & dosage , Boranes/adverse effects , Boranes/therapeutic use , Carcinoma/pathology , Carcinoma/radiotherapy , Cell Membrane/metabolism , Cell Survival , Cobalt Radioisotopes/pharmacokinetics , Cobalt Radioisotopes/therapeutic use , Colonic Neoplasms/pathology , Colonic Neoplasms/radiotherapy , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/metabolism , Monte Carlo Method , Neutrons/therapeutic use , Organelles/metabolism , Radiotherapy Dosage , Subcellular Fractions/metabolism , Tumor Cells, Cultured
20.
Proc Natl Acad Sci U S A ; 91(8): 3029-33, 1994 Apr 12.
Article in English | MEDLINE | ID: mdl-8159700

ABSTRACT

The polyhedral borane ion [n-B20H18]2- reacts with liquid ammonia in the presence of a suitable base to produce an apical-equatorial (ae) isomer of the [B20H17NH3]3- ion, [1-(2'-B10H9)-2-NH3B10H8]3-. The structure of this product has been confirmed by 11B NMR spectroscopy and x-ray crystallography. This species undergoes acid-catalyzed rearrangement to an apical-apical (a2) isomer, [1-(1'-B10H9)-2-NH3B10H8]3-, whose structure has been determined by 11B NMR spectroscopy. The sodium salts of both the ae and the a2 isomers of [B20H17NH3]3- have been encapsulated within small unilamellar liposomes, composed of distearoyl phosphatidylcholine/cholesterol (1:1), and investigated as boron-delivery agents for boron neutron capture therapy (BNCT) of cancer. The biodistribution of boron was determined after the injection of liposomal suspensions into BALB/c mice bearing EMT6 tumors. Both [B20H17NH3]3- isomers exhibited excellent tumor uptake and selectivity at very low injected doses, achieving peak tumor boron concentrations of 30-40 micrograms of B/g of tissue and tumor/blood boron ratios of approximately 5. The enhanced retention of the [B20H17NH3]3- isomers by EMT6 tumors may be attributed to their facile intracellular oxidation to an extremely reactive NH3-substituted [n-B20H18]2- ion, the electrophilic [B20H17NH3]- ion. Both isomers of [B20H17NH3]3- are at least 0.5 V more easily oxidized than other previously investigated species containing 20 boron atoms. In another experiment, [ae-B20H17NH3]3- was encapsulated in liposomes prepared with 5% PEG-2000-distearoyl phosphatidylethanolamine in the liposome membrane. As expected, these liposomes exhibited a longer circulation lifetime in the biodistribution experiment, resulting in the continued accumulation of boron in the tumor over the entire 48-hr experiment and reaching a maximum of 47 micrograms of B/g of tumor.


Subject(s)
Boranes/administration & dosage , Boron Neutron Capture Therapy/methods , Neoplasms, Experimental/metabolism , Animals , Boranes/pharmacokinetics , Female , Liposomes , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...