Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Inorg Biochem ; 181: 56-64, 2018 04.
Article in English | MEDLINE | ID: mdl-29407908

ABSTRACT

Dysfunctional accumulation of amyloid-ß (Aß) protein stimulated by Cu2+ is considered as a key process in the pathogenesis of Alzheimer's disease (AD). Thus, bifunctional substances capable of chelating Cu2+ and inhibiting Aß aggregation are promising therapeutic agents against AD. Herein, a novel bifunctional decapeptide RTHLVFFARK-NH2 (RK10) was developed by integrating a metal chelating tripeptide (RTH) and an Aß aggregation inhibitor Ac-LVFFARK-NH2 (LK7). The high selectivity of RK10 for Cu2+ over other biologically relevant metal ions was demonstrated by isothermal titration calorimetry. RK10 bound Cu2+ with a dissociation constant of 0.02 µM. This enabled RK10 to sequester Cu2+ from Aß40-Cu2+ species and to arrest the production of reactive oxygen species (ROS) catalyzed by Cu2+ or Aß40-Cu2+ species. Extensive physical, biophysical and biological studies indicate that RK10 targeted free and Cu2+-bound Aß40 species, suppressed Aß40 aggregation, and diminished the cytotoxicity induced by Aß40 and Cu2+-mediated Aß40 in cultured SH-SY5Y cells. Taken together, the results proved the excellent selective roles of RK10 in inhibiting Cu2+-mediated Aß40 aggregation and eliminating ROS generation catalyzed by Cu2+/Aß40-Cu2+ species. Thus, this work provided new insight into the design and development of potent bifunctional inhibitors against Aß aggregation and cytotoxicity.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid/drug effects , Chelating Agents/pharmacology , Copper/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Peptide Fragments/antagonists & inhibitors , Algorithms , Amyloid/chemistry , Amyloid/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Calorimetry , Cell Line, Tumor , Cell Survival/drug effects , Chelating Agents/chemistry , Chelating Agents/metabolism , Coordination Complexes/antagonists & inhibitors , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Coordination Complexes/toxicity , Copper/chemistry , Copper/metabolism , Drug Design , Humans , Kinetics , Microscopy, Atomic Force , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , Oxidative Stress/drug effects , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Aggregation, Pathological/prevention & control , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Thermodynamics
2.
Biochem Pharmacol ; 118: 109-120, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27565891

ABSTRACT

Mercury compounds are well-known toxic environmental pollutants and potently induce severe neurotoxicological effects in human and experimental animals. Previous studies showed that one of the mechanisms of mercury compounds neurotoxicity arose from the over-activation of the N-methyl d-aspartate (NMDA)-type glutamate receptor induced by increased glutamate release. In this work, we aimed to investigate the molecular mechanisms of Hg compounds neurotoxicities by identifying their biological targets in cells. Firstly, the inhibitory effects of four Hg compounds, including three organic (methyl-, ethyl- and phenyl-mercury) and one inorganic (Hg2+) Hg compounds, on the activity of arginine decarboxylase (ADC), a key enzyme in the central agmatinergic system, were evaluated. They were found to inhibit the ADC activity significantly with methylmercury (MeHg) being the strongest (IC50=7.96nM). Furthermore, they showed remarkable inhibitory effects on ADC activity in PC12 cells (MeHg>EtHg>PhHg>HgCl2), and led to a marked loss in the level of agmatine, an endogenous neuromodulatory and neuroprotective agent that selectively blocks the activation of NMDA receptors. MeHg was detected in the immunoprecipitated ADC from the cells, providing unequivocal evidence for the direct binding of MeHg with ADC in the cell. Molecular dynamics simulation revealed that Hg compounds could form the coordination bond not only with cofactor PLP of ADC, but also with substrate arginine. Our finding indicated that MeHg could attenuate the neuroprotective effects of agmatine by the inhibition of ADC, a new cellular target of MeHg, which might be implicated in molecular mechanism of MeHg neurotoxicity.


Subject(s)
Carboxy-Lyases/antagonists & inhibitors , Environmental Pollutants/toxicity , Enzyme Inhibitors/toxicity , Methylmercury Compounds/toxicity , Models, Molecular , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/drug effects , Absorption, Physiological , Agmatine/antagonists & inhibitors , Agmatine/metabolism , Animals , Arginine/metabolism , Binding Sites , Biocatalysis/drug effects , Carboxy-Lyases/chemistry , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/antagonists & inhibitors , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Decarboxylation/drug effects , Environmental Pollutants/antagonists & inhibitors , Environmental Pollutants/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Ethylmercuric Chloride/antagonists & inhibitors , Ethylmercuric Chloride/metabolism , Ethylmercuric Chloride/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Mercuric Chloride/antagonists & inhibitors , Mercuric Chloride/metabolism , Mercuric Chloride/toxicity , Methylmercury Compounds/antagonists & inhibitors , Methylmercury Compounds/metabolism , Molecular Dynamics Simulation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Neurons/metabolism , Phenylmercury Compounds/antagonists & inhibitors , Phenylmercury Compounds/metabolism , Phenylmercury Compounds/toxicity , Rats
3.
Free Radic Res ; 50(4): 426-46, 2016.
Article in English | MEDLINE | ID: mdl-26733073

ABSTRACT

Multidrug resistance (MDR) in cancer represents a variety of strategies employed by tumor cells to evade the beneficial cytotoxic effects of structurally different anticancer drugs and thus confers impediments to the successful treatment of cancers. Efflux of drugs by MDR protein-1, functional P-glycoprotein and elevated level of reduced glutathione confer resistance to cell death or apoptosis and thus provide a possible therapeutic target for overcoming MDR in cancer. Previously, we reported that a Schiff base ligand, potassium-N-(2-hydroxy 3-methoxy-benzaldehyde)-alaninate (PHMBA) overcomes MDR in both in vivo and in vitro by targeting intrinsic apoptotic/necrotic pathway through induction of reactive oxygen species (ROS). The present study describes the synthesis and spectroscopic characterization of a copper chelate of Schiff base, viz., copper (II)-N-(2-hydroxy-3-methoxy-benzaldehyde)-alaninate (CuPHMBA) and the underlying mechanism of cell death induced by CuPHMBA in vitro. CuPHMBA kills both the drug-resistant and sensitive cell types irrespective of their drug resistance phenotype. The cell death induced by CuPHMBA follows apoptotic pathway and moreover, the cell death is associated with intrinsic mitochondrial and extrinsic receptor-mediated pathways. Oxidative stress plays a pivotal role in the process as proved by the fact that antioxidant enzyme; polyethylene glycol conjugated-catalase completely blocked CuPHMBA-induced ROS generation and abrogated cell death. To summarize, the present work provides a compelling rationale for the future clinical use of CuPHMBA, a redox active copper chelate in the treatment of cancer patients, irrespective of their drug-resistance status.


Subject(s)
Antineoplastic Agents/pharmacology , Chelating Agents/pharmacology , Coordination Complexes/pharmacology , Copper/chemistry , Reactive Oxygen Species/agonists , Schiff Bases/pharmacology , Alanine/analogs & derivatives , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Benzaldehydes/chemistry , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Catalase/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chelating Agents/chemical synthesis , Coordination Complexes/antagonists & inhibitors , Coordination Complexes/chemical synthesis , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Oxidation-Reduction , Polyethylene Glycols/chemistry , Primary Cell Culture , Reactive Oxygen Species/metabolism , Schiff Bases/antagonists & inhibitors , Schiff Bases/chemical synthesis
4.
Curr Cancer Drug Targets ; 14(4): 371-9, 2014.
Article in English | MEDLINE | ID: mdl-24720338

ABSTRACT

The α(v)ß3 integrin is highly expressed in prostate cancer (PCa), in which it is a key player in tumour invasion, angiogenesis and metastasis formation. Therefore, α(v)ß3 integrin is considered a very promising target for molecular imaging of PCa. This study tested the potential of the novel α(v)ß3 integrin affine agent [68Ga]NOTA-RGD in comparison with the established [¹8F]fluoroethylcholine (FEC) and [¹8F]fluorodeoxyglucose (FDG) for assessing PCa using positron emission tomography (PET). [68Ga]NOTA-RGD showed a lower uptake in PC-3 and DU-145 cells compared with FEC and FDG. µPET imaging studies showed a good delineation of the PCa xenografts in mice. The means tumor-to-muscle and tumor-to-bone-ratio amounted 5.1 ± 1.4 and 5.2 ± 1.2 for [68Ga]NOTA-RGD compared with 2.6 ± 0.9 and 2.9 ± 1.6 for FDG, and 2.4 ± 0.7 and 0.8 ± 0.2 for FEC, respectively. The uptake of [68Ga]NOTA-RGD into tumor was fully inhibited by c(RGDfV), known to bind specifically to α(v)ß3 integrin, confirming the specificity of the tumor uptake in vivo. These results suggest that [68Ga]NOTA-RGD is a promising candidate for PET imaging of α(v)ß3 integrin expression in PCa and warrant further in vivo validations to ascertain its potential as an imaging agent for clinical use. The simple and fast preparation of [68Ga]NOTA-RGD may greatly facilitate its translation to a clinical setting.


Subject(s)
Affinity Labels , Coordination Complexes , Gallium Radioisotopes , Integrin alphaV/metabolism , Integrin beta3/metabolism , Oligopeptides , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Affinity Labels/chemistry , Affinity Labels/metabolism , Animals , Biological Transport/drug effects , Cell Line, Tumor , Choline/analogs & derivatives , Choline/metabolism , Coordination Complexes/antagonists & inhibitors , Coordination Complexes/metabolism , Fluorodeoxyglucose F18/metabolism , Gallium Radioisotopes/chemistry , Gallium Radioisotopes/metabolism , Humans , Integrin alphaV/chemistry , Integrin beta3/chemistry , Integrins/antagonists & inhibitors , Integrins/metabolism , Isotope Labeling , Male , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Oligopeptides/antagonists & inhibitors , Oligopeptides/metabolism , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Positron-Emission Tomography , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism
5.
J Inorg Biochem ; 128: 174-82, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23932552

ABSTRACT

The chelating properties toward iron(III) and aluminium(III) of variously substituted salicyl-aldehydes and salicylic acids have been evaluated, together with the effect of methoxy and nitro substituents in ortho and para position with respect to the phenolic group. The protonation and iron and aluminium complex formation equilibria have been studied by potentiometry, UV-visible spectrophotometry and (1)H NMR spectroscopy. The overall results highlight that salicyl-aldehydes present good chelating properties toward iron(III), with pFe ranging from 14.2 with nitro to 15.7 with methoxy substituent, being ineffective toward aluminium; the pFe values for salicylic acids are generally lower than those for salicyl-aldehydes, and about 4 units higher than the corresponding pAl values. The effect of the two substituents on the chelating properties of the ligands can be rationalized in terms of the Swain-Lupton treatment which accounts for the field and resonance effects. The structural characterization of the 1:2 iron complex with p-nitro salicylic acid shows that iron(III) ion exhibits an octahedral surrounding where two salicylate chelating ligands supply two O-phenolate and two O-carboxylate donor atoms in a roughly equatorial plane. The trans-apical sites are occupied by two aqua ligands.


Subject(s)
Aldehydes/chemistry , Aluminum/chemistry , Coordination Complexes/chemistry , Iron/chemistry , Salicylates/chemistry , Salicylic Acid/chemistry , Chelating Agents/chemistry , Chelating Agents/pharmacology , Coordination Complexes/antagonists & inhibitors , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Potentiometry , Spectrophotometry
6.
Neurotoxicology ; 38: 1-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23727015

ABSTRACT

Various forms of mercury possess different rates of absorption, metabolism and excretion, and consequently, toxicity. Methylmercury (MeHg) is a highly neurotoxic organic mercurial. Human exposure is mostly due to ingestion of contaminated fish. Ethylmercury (EtHg), another organic mercury compound, has received significant toxicological attention due to its presence in thimerosal-containing vaccines. This study was designed to compare the toxicities induced by MeHg and EtHg, as well as by their complexes with cysteine (MeHg-S-Cys and EtHg-S-Cys) in the C6 rat glioma cell line. MeHg and EtHg caused significant (p<0.0001) decreases in cellular viability when cells were treated during 30min with each mercurial following by a washing period of 24h (EC50 values of 4.83 and 5.05µM, respectively). Significant cytotoxicity (p<0.0001) was also observed when cells were treated under the same conditions with MeHg-S-Cys and EtHg-S-Cys, but the respective EC50 values were significantly increased (11.2 and 9.37µM). l-Methionine, a substrate for the l-type neutral amino acid carrier transport (LAT) system, significantly protected against the toxicities induced by both complexes (MeHg-S-Cys and EtHg-S-Cys). However, no protective effects of l-methionine were observed against MeHg and EtHg toxicities. Corroborating these findings, l-methionine significantly decreased mercurial uptake when cells were exposed to MeHg-S-Cys (p=0.028) and EtHg-S-Cys (p=0.023), but not to MeHg and EtHg. These results indicate that the uptake of MeHg-S-Cys and EtHg-S-Cys into C6 cells is mediated, at least in part, through the LAT system, but MeHg and EtHg enter C6 cells by mechanisms other than LAT system.


Subject(s)
Amino Acid Transport System L/metabolism , Cysteine/toxicity , Ethylmercuric Chloride/metabolism , Ethylmercuric Chloride/toxicity , Glioma/pathology , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Animals , Biological Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/antagonists & inhibitors , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Coordination Complexes/toxicity , Cysteine/chemistry , Ethylmercuric Chloride/antagonists & inhibitors , Ethylmercuric Chloride/chemistry , Glioma/metabolism , Glutathione/drug effects , Glutathione/metabolism , Hippocampus/metabolism , Methionine/pharmacology , Methylmercury Compounds/antagonists & inhibitors , Methylmercury Compounds/chemistry , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...