Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Int J Cancer ; 131(9): 2008-15, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22336951

ABSTRACT

Acrylamide is a high-volume industrial chemical, a component of cigarette smoke, and a product formed in certain foods prepared at high temperatures. Previously, we compared the extent of DNA adduct formation and mutations in B6C3F(1) /Tk mice treated neonatally with acrylamide or glycidamide to obtain information concerning the mechanism of acrylamide genotoxicity. We have now examined the tumorigenicity of acrylamide and glycidamide in mice treated neonatally. Male B6C3F(1) mice were injected intraperitoneally on postnatal days 1, 8 and 15 with 0.0, 0.14 or 0.70 mmol acrylamide or glycidamide per kg body weight per day and the tumorigenicity was assessed after 1 year. Survival in each of the groups was >87%, there were no differences in body weights among the groups, and the only treatment-related neoplasms involved the liver. The incidence of combined hepatocellular adenoma or carcinoma was 3.8% in the control group, 8.3% in the 0.14 mmol acrylamide and glycidamide per kg body weight groups, 4.2% in the 0.70 mmol acrylamide per kg body weight group and 71.4% in the 0.70 mmol glycidamide per kg body weight group. Analysis of the hepatocellular tumors indicated that the increased incidence observed in mice administered 0.70 mmol glycidamide per kg body weight was associated with A → G and A → T mutations at codon 61 of H-ras. These results, combined with our previous data on DNA adduct formation and mutation induction, suggest that the carcinogenicity of acrylamide is dependent on its metabolism to glycidamide, a pathway that is deficient in neonatal mice.


Subject(s)
Acrylamide/toxicity , Carcinogens/toxicity , Epoxy Compounds/toxicity , Genes, ras/drug effects , Acrylamide/metabolism , Adenoma, Liver Cell/chemically induced , Animals , Animals, Newborn , Carcinogenicity Tests , DNA Adducts/biosynthesis , DNA Damage/drug effects , Female , Liver/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation , Random Allocation
2.
Bioorg Chem ; 44: 19-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22784829

ABSTRACT

The enzymatic (tyrosinase) and chemical (NaIO(4), Ag(2)O or Frémys's salt) oxidation of biologically relevant catecholamines, such as dopamine (DA), N-acetyldopamine (NADA) and the Ecstasy metabolites (α-MeDA and N-Me-α-MeDA) generates the corresponding o-quinone which can be trapped with nitrogen bionucleophiles such as N-acetyl-histidine and imidazole in a regioselective reaction that takes place predominantly at the 6-position of the catecholamine.


Subject(s)
Agaricales/enzymology , Catecholamines/chemistry , Catecholamines/metabolism , Monophenol Monooxygenase/metabolism , Nitrogen/metabolism , Nitrogen/chemistry , Oxidation-Reduction , Quinones/chemistry , Quinones/metabolism
3.
Eur J Med Chem ; 172: 16-25, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30939350

ABSTRACT

Herein we report novel hybrid compounds based on valproic acid and DNA-alkylating triazene moieties, 1, with therapeutic potential for glioblastoma multiforme chemotherapy. We identified hybrid compounds 1d and 1e to be remarkably more potent against glioma and more efficient in decreasing invasive cell properties than temozolomide and endowed with chemical and plasma stability. In contrast to temozolomide, which undergoes hydrolysis to release an alkylating metabolite, the valproate hybrids showed a low potential to alkylate DNA. Key physicochemical properties align for optimal CNS penetration, highlighting the potential of these effective triazene based-hybrids for enhanced anticancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Triazenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Brain Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioma/pathology , Humans , Molecular Structure , Structure-Activity Relationship , Triazenes/chemical synthesis , Triazenes/chemistry , Tumor Cells, Cultured
4.
Eur J Pharm Sci ; 105: 47-54, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28487145

ABSTRACT

Efavirenz is an anti-HIV drug that presents relevant short- and long-term central nervous system adverse reactions. Its main metabolite (8-hydroxy-efavirenz) was demonstrated to be a more potent neurotoxin than efavirenz itself. This work was aimed to understand how efavirenz biotransformation to 8-hydroxy-efavirenz is related to its short- and long-term neuro-adverse reactions. To access those mechanisms, the expression and activity of Cyp2b enzymes as well as the thiolomic signature (low molecular weight thiols plus S-thiolated proteins) were longitudinally evaluated in the hepatic and brain tissues of rats exposed to efavirenz during 10 and 36days. Efavirenz and 8-hydroxy-efavirenz plasma concentrations were monitored at the same time points. Cyp2b induction had a delayed onset in liver (p<0.001), translating into increases in Cyp2b activity in liver and 8-hydroxy-efavirenz plasma concentration (p<0.001). Moreover, an increase in S-cysteinyl-glycinylated proteins (p<0.001) and in free low molecular weight thiols was also observed in liver. A distinct scenario was observed in hippocampus, which showed an underexpression of Cyp2b as well as a decrease in S-cysteinylated and S-glutathionylated proteins. Additionally, the observed changes in tissues were associated with a marked increase of S-glutathionylation in plasma. Our data suggest that the time course of efavirenz biotransformation results from different mechanisms for its short- and long-term neurotoxicity. The difference in the redox profile between liver and hippocampus might explain why, despite being mostly metabolized by the liver, this drug is neurotoxic. If translated to clinical practice, this evidence will have important implications in efavirenz short- and long-term neurotoxicity prevention and management.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Benzoxazines/pharmacokinetics , Neurotoxicity Syndromes/metabolism , Alkynes , Animals , Anti-HIV Agents/adverse effects , Anti-HIV Agents/blood , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Benzoxazines/adverse effects , Benzoxazines/blood , Benzoxazines/metabolism , Biotransformation , Cyclopropanes , Cytochrome P-450 CYP2B1/genetics , Cytochrome P-450 CYP2B1/metabolism , Hippocampus/metabolism , Liver/metabolism , Male , Neurotoxicity Syndromes/blood , Prefrontal Cortex/metabolism , Rats, Wistar , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Sulfhydryl Compounds/metabolism
5.
Mutat Res ; 506-507: 41-8, 2002 Sep 30.
Article in English | MEDLINE | ID: mdl-12351143

ABSTRACT

2,6-Dimethylaniline (2,6-DMA) is an intermediate in the manufacture of several products, including pesticides, dyestuffs, and synthetic resins. It is also present in nanogram amounts in tobacco smoke, and is a major metabolite of the potent anesthetic and antiarrhythmic drug lidocaine, as well as a nasal carcinogen in rats. As with other aromatic amines, 2,6-DMA can undergo metabolic activation through cytochrome p450-mediated N-hydroxylation, followed by O-esterification to a reactive derivative capable of forming DNA adducts. We have recently characterized four DNA adducts resulting from this metabolic pathway. Three of the adducts arose from reaction of the exocyclic heteroatoms of deoxyadenosine and deoxyguanosine with the carbon para to the arylamine nitrogen. The fourth adduct resulted from reaction of the 2,6-DMA nitrogen with the C8 atom of deoxyguanosine. In order to investigate the relative contribution of the exocyclic heteroatom adducts as compared to the C8-deoxyguanosine adduct to the toxicities elicited by 2,6-DMA, we synthesized and compared the mutagenicity of N-hydroxy-2,6-DMA, N-hydroxy-4-deutero-2,6-DMA, 2,6-dimethylnitrosobenzene, 4-deutero-2,6-dimethylnitrosobenzene, and N-hydroxy-4-fluoro-2,6-DMA. In Salmonella typhimurium TA100, the two deuterated compounds and their non-deuterated analogues gave similar mutagenic responses ( approximately 25 revertants/nmol). Likewise in S. typhimurium TA98, a similar mutant frequency ( approximately 0.7 revertants/nmol) was obtained with the four compounds. With N-hydroxy-4-fluoro-2,6-DMA, the mutant frequency was reduced by approximately 90% in S. typhimurium TA100 and approximately 50% in S. typhimurium TA98. The results suggest that multiple adducts contribute to base substitution mutations detected by S. typhimurium TA100 while the C8-deoxyguanosine adduct is primarily responsible for the frameshift mutations detected by S. typhimurium TA98.


Subject(s)
Aniline Compounds/toxicity , Mutagens/toxicity , Animals , DNA Adducts/drug effects , DNA Damage , DNA, Bacterial/drug effects , Deoxyguanosine/metabolism , Deuterium , Fluorine , Mutagenicity Tests , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Structure-Activity Relationship
6.
Toxicol Lett ; 219(1): 59-64, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23467017

ABSTRACT

The anti-HIV drug abacavir is associated with idiosyncratic hypersensitivity reactions and cardiotoxicity. Although the mechanism underlying abacavir-toxicity is not fully understood, drug bioactivation to reactive metabolites may be involved. This work was aimed at identifying abacavir-protein adducts in the hemoglobin of HIV patients as biomarkers of abacavir bioactivation and protein modification. The protocol received prior approval from the Hospital Ethics Committee, patients gave their written informed consent and adherence was controlled through a questionnaire. Abacavir-derived Edman adducts with the N-terminal valine of hemoglobin were analyzed by an established liquid chromatography-electrospray ionization-tandem mass spectrometry method. Abacavir-valine adducts were detected in three out of ten patients. This work represents the first evidence of abacavir-protein adduct formation in humans. The data confirm the ability of abacavir to modify self-proteins and suggest that the molecular mechanism(s) of some abacavir-induced adverse reactions may require bioactivation.


Subject(s)
Aldehydes/metabolism , Anti-HIV Agents/adverse effects , Anti-HIV Agents/metabolism , Dideoxynucleosides/adverse effects , Dideoxynucleosides/metabolism , Adult , Aged , Aged, 80 and over , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Biotransformation , Dideoxynucleosides/pharmacokinetics , Dideoxynucleosides/therapeutic use , Drug Monitoring , Female , HIV Infections/blood , HIV Infections/drug therapy , Hemoglobins/metabolism , Humans , Male , Middle Aged , Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL