Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cells ; 11(9)2022 05 09.
Article in English | MEDLINE | ID: mdl-35563891

ABSTRACT

The immunophilin FKBP51, the angiomotin AmotL2, and the scaffoldin IQGAP1 are overexpressed in many types of cancer, with the highest increase in leucocytes from patients undergoing oxaliplatin chemotherapy. Inflammation is involved in the pathogenesis of nephrotoxicity induced by platinum analogs. Cilastatin prevents renal damage caused by cisplatin. This functional and confocal microscopy study shows the renal focal-segmental expression of TNFα after cisplatin administration in rats, predominantly of tubular localization and mostly prevented by co-administration of cilastatin. FKBP51, AmotL2 and IQGAP1 protein expression increases slightly with cilastatin administration and to a much higher extent with cisplatin, in a cellular- and subcellular-specific manner. Kidney tubule cells expressing FKBP51 show either very low or no expression of TNFα, while cells expressing TNFα have low levels of FKBP51. AmotL2 and TNFα seem to colocalize and their expression is increased in tubular cells. IQGAP1 fluorescence increases with cilastatin, cisplatin and joint cilastatin-cisplatin treatment, and does not correlate with TNFα expression or localization. These data suggest a role for FKBP51, AmotL2 and IQGAP1 in cisplatin toxicity in kidney tubules and in the protective effect of cilastatin through inhibition of dehydropeptidase-I.


Subject(s)
Cilastatin , Cisplatin , Angiomotins , Animals , Carrier Proteins/metabolism , Cilastatin/metabolism , Cilastatin/pharmacology , Cilastatin/therapeutic use , Cisplatin/metabolism , Cisplatin/toxicity , Humans , Rats , Tumor Necrosis Factor-alpha/metabolism , ras GTPase-Activating Proteins/metabolism
2.
J Lipid Res ; 59(9): 1561-1574, 2018 09.
Article in English | MEDLINE | ID: mdl-30049708

ABSTRACT

Nephrotoxicity is a major limitation to cisplatin antitumor therapies. Cilastatin, an inhibitor of renal dehydropeptidase-I, was recently proposed as a promising nephroprotector against cisplatin toxicity, preventing apoptotic cell death. In this work, cilastatin nephroprotection was further investigated in a rat model, with a focus on its effect on 76 renal lipids altered by cisplatin, including 13 new cisplatin-altered mitochondrial cardiolipin species. Lipid imaging was performed with MALDI mass spectrometry imaging (MALDI-MSI) in kidney sections from treated rats. Cilastatin was proved to significantly diminish the lipid distribution alterations caused by cisplatin, lipid levels being almost completely recovered to those of control samples. The extent of recovery of cisplatin-altered lipids by cilastatin turned out to be relevant for discriminating direct or secondary lipid alterations driven by cisplatin. Lipid peroxidation induced by cisplatin was also shown to be reduced when cilastatin was administered. Importantly, significant groups separation was achieved during multivariate analysis of cortex and outer-medullary lipids, indicating that damaged kidney can be discerned from the nephroprotected and healthy groups and classified according to lipid distribution. Therefore, we propose MALDI-MSI as a powerful potential tool offering multimolecule detection possibilities to visualize and evaluate nephrotoxicity and nephroprotection based on lipid analysis.


Subject(s)
Cilastatin/metabolism , Cisplatin/adverse effects , Kidney/drug effects , Kidney/metabolism , Lipid Metabolism/drug effects , Molecular Imaging , Animals , Cytoprotection/drug effects , Female , Kidney/diagnostic imaging , Lipid Peroxidation/drug effects , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
3.
World J Microbiol Biotechnol ; 32(7): 117, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27263012

ABSTRACT

Carbapenemase-producing Enterobacteriaceae (CPE) isolates have now emerged worldwide. We therefore modified the phenotypic Carba NP test by use of a filter paper strip for easily and rapidly identifying CPE in routine laboratory. A collection of 56 CPE and carbapenemase-producing Pseudomonas spp. isolates (including 28 NDM-1, 11 IMP-14a, 1 IMP-1, 1 IMP-4, 1 IMP-9, 1 IMP-15, 4 VIM-2, 1 VIM-1, 1 IMP-14a with VIM-2, 3 OXA-48, 3 OXA-181 and 1 KPC-2 producers) and 41 non-CPE isolates (including 19 ESBL, 7 pAmpC, 3 AmpC, 9 ESBL with pAmpC and 3 non-ESBL & non-AmpC producers) as confirmed by the PCR methods were tested by the paper strip method using pharmaceutical imipenem/cilastatin as a substrate. Bacterial colonies of each isolate were applied directly on filter paper strips dropped with either imipenem-phenol red (test strip) or phenol red solution alone (control strip). The reaction was read within 5 min. This test failed to detect 3 OXA-181, 2 OXA-48 and 3 IMP-14a producers (85.7 % sensitivity), whereas no false positives were seen (100 % specificity). Further evaluation of the paper strip test in 267 CPE screening-positive isolates from three hospitals by their medical technologists showed 92.0 % sensitivity (100 % for NDM producers) and 100 % specificity compared with the PCR methods. Because of its ease, rapidness and cost effective, the paper strip test has a potential for routine CPE testing in low-resource laboratories particularly in areas with high prevalence of NDM enzymes, leading to appropriate antimicrobial therapy and infection control strategy.


Subject(s)
Bacterial Proteins/analysis , Enterobacteriaceae/enzymology , Enterobacteriaceae/isolation & purification , Enzyme Assays/methods , Reagent Strips , beta-Lactamases/analysis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriological Techniques/methods , Carbapenems/pharmacology , Cilastatin/metabolism , Cilastatin, Imipenem Drug Combination , Drug Combinations , Enterobacteriaceae/genetics , Enterobacteriaceae Infections/microbiology , Enzyme Assays/standards , Humans , Imipenem/metabolism , Phenotype , Polymerase Chain Reaction , Pseudomonas/enzymology , Pseudomonas/genetics , Pseudomonas/isolation & purification , beta-Lactamases/genetics , beta-Lactamases/metabolism
4.
Appl Environ Microbiol ; 80(23): 7348-55, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25239898

ABSTRACT

A novel nonheme chloroperoxidase (RhEst1), with promiscuous esterase activity for enantioselective hydrolysis of ethyl (S)-2,2-dimethylcyclopropanecarboxylate, was identified from a shotgun library of Rhodococcus sp. strain ECU1013. RhEst1 was overexpressed in Escherichia coli BL21(DE3), purified to homogeneity, and functionally characterized. Fingerprinting analysis revealed that RhEst1 prefers para-nitrophenyl (pNP) esters of short-chain acyl groups. pNP esters with a cyclic acyl moiety, especially that with a cyclobutanyl group, were also substrates for RhEst1. The Km values for methyl 2,2-dimethylcyclopropanecarboxylate (DmCpCm) and ethyl 2,2-dimethylcyclopropane carboxylate (DmCpCe) were 0.25 and 0.43 mM, respectively. RhEst1 could serve as an efficient hydrolase for the bioproduction of optically pure (S)-2,2-dimethyl cyclopropane carboxylic acid (DmCpCa), which is an important chiral building block for cilastatin. As much as 0.5 M DmCpCe was enantioselectively hydrolyzed into (S)-DmCpCa, with a molar yield of 47.8% and an enantiomeric excess (ee) of 97.5%, indicating an extremely high enantioselectivity (E = 240) of this novel and unique biocatalyst for green manufacturing of highly valuable chiral chemicals.


Subject(s)
Chloride Peroxidase/isolation & purification , Chloride Peroxidase/metabolism , Cilastatin/metabolism , Protease Inhibitors/metabolism , Rhodococcus/enzymology , Amino Acid Sequence , Chloride Peroxidase/genetics , Cloning, Molecular , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Molecular Sequence Data , Phylogeny , Rhodococcus/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Substrate Specificity
5.
Appl Microbiol Biotechnol ; 97(17): 7659-67, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23807665

ABSTRACT

(S)-(+)-2,2-Dimethylcyclopropane carboxylic acid [(S)-(+)-DMCPA] is a key chiral intermediate for production of Cilastatin, an excellent renal dehydropeptidase-I inhibitor. In this study, a new method for preparation of (S)-(+)-DMCPA with microbial esterases was investigated. A microbial screening program obtained six esterase-producing isolates that could display relatively high activities and enantioselectivities using racemic ethyl 2,2-dimethylcyclopropane carboxylate (DMCPE) as screening substrate, aiming at forming optically pure (S)-(+)-DMCPA. Further selection was carried out with substrates having different alcohol moieties, including methyl, ethyl, and 2-chloroethyl esters. Finally, one of these strains, numbered ECU1013, with high enantioselectivity toward the hydrolytic resolution of methyl 2,2-dimethylcyclopropane carboxylate (DMCPM), afforded the (S)-product in 92 % ee, and was later identified as Rhodococcus sp. According to our research, there were several active esterases to DMCPM in cells of Rhodococcus sp. ECU1013; however, (S)-preferential esterase was selectively enriched based on the time-dependent profile of esterases biosynthesis, thereby the enantiomeric excess of biotransformation product (ee p) was constantly increased, finally maintained at 95 % (S). To improve the yield, various organic solvents were employed for better dispersion of the hydrophobic substrate. As a result, (±)-DMCPM of up to 400 mM in the organic phase of isooctane was enantioselectively hydrolyzed into (S)-(+)-DMCPA, with an isolation yield of 38 % and a further increase of ee p to 99 %.


Subject(s)
Cilastatin/metabolism , Cyclopropanes/metabolism , Rhodococcus/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cyclopropanes/chemistry , Esterases/genetics , Esterases/metabolism , Hydrolysis , Molecular Sequence Data , Phylogeny , Rhodococcus/classification , Rhodococcus/enzymology , Rhodococcus/isolation & purification , Soil Microbiology , Stereoisomerism
6.
J Pharmacol Exp Ther ; 334(2): 419-29, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20435919

ABSTRACT

A major area in cancer therapy is the search for protective strategies against cisplatin-induced nephrotoxicity. We investigated the protective effect of cilastatin on cisplatin-induced injury to renal proximal tubular cells. Cilastatin is a specific inhibitor of renal dehydrodipeptidase I (DHP-I), which prevents hydrolysis of imipenem and its accumulation in the proximal tubule. Primary cultures of proximal cells were treated with cisplatin (1-30 microM) in the presence or absence of cilastatin (200 microg/ml). Apoptosis and mitochondrial injury were assessed by different techniques. Cisplatin uptake and DNA binding were measured by inductively coupled plasma spectrometry. HeLa cells were used to control the effect of cilastatin on the tumoricidal activity of cisplatin. Cisplatin increased cell death, apoptotic-like morphology, caspase activation, and mitochondrial injury in proximal tubular cells in a dose- and time-dependent way. Concomitant treatment with cilastatin reduced cisplatin-induced changes. Cilastatin also reduced the DNA-bound platinum but did not modify cisplatin-dependent up-regulation of death receptors (Fas) or ligands (tumor necrosis factor alpha, Fas ligand). In contrast, cilastatin did not show any effects on cisplatin-treated HeLa cells. Renal DHP-I was virtually absent in HeLa cells. Cilastatin attenuates cisplatin-induced cell death in proximal tubular cells without reducing the cytotoxic activity of cisplatin in tumor cells. Our findings suggest that the affinity of cilastatin for renal dipeptidase makes this effect specific for proximal tubular cells and may be related to a reduction in intracellular drug accumulation. Therefore, cilastatin administration might represent a novel strategy in the prevention of cisplatin-induced acute renal injury.


Subject(s)
Antineoplastic Agents/toxicity , Cilastatin/pharmacology , Cisplatin/toxicity , Dipeptidases/antagonists & inhibitors , Kidney Tubules, Proximal/drug effects , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Cilastatin/metabolism , DNA/metabolism , Fas Ligand Protein/biosynthesis , Fas Ligand Protein/genetics , HeLa Cells , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/enzymology , Membrane Potential, Mitochondrial/drug effects , RNA, Messenger/biosynthesis , Swine , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , fas Receptor/biosynthesis , fas Receptor/genetics
7.
J Inorg Biochem ; 104(1): 37-46, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19879002

ABSTRACT

The reaction mechanism of the dinuclear zinc enzyme human renal dipeptidase is investigated using hybrid density functional theory. This enzyme catalyzes the hydrolysis of dipeptides and beta-lactam antibiotics. Two different protonation states in which the important active site residue Asp288 is either neutral or ionized were considered. In both cases, the bridging hydroxide is shown to be capable of performing the nucleophilic attack on the substrate carbonyl carbon from its bridging position, resulting in the formation of a tetrahedral intermediate. This step is followed by protonation of the dipeptide nitrogen, coupled with C-N bond cleavage. The calculations establish that both cases have quite feasible energy barriers. When the Asp288 is neutral, the hydrolytic reaction occurs with a large exothermicity. However, the reaction becomes very close to thermoneutral with an ionized Asp288. The two zinc ions are shown to play different roles in the reaction. Zn1 binds the amino group of the substrate, and Zn2 interacts with the carboxylate group of the substrate, helping in orienting it for the nucleophilic attack. In addition, Zn2 stabilizes the oxyanion of the tetrahedral intermediate, thereby facilitating the nucleophilic attack.


Subject(s)
Aspartic Acid/chemistry , Cilastatin/chemistry , Dipeptidases/chemistry , Dipeptides/chemistry , Zinc/chemistry , Catalytic Domain , Cilastatin/metabolism , Crystallography, X-Ray , Dipeptidases/metabolism , Dipeptides/metabolism , Humans , Hydrolysis , Models, Molecular , Molecular Structure
8.
Bioorg Med Chem ; 11(6): 991-8, 2003 Mar 20.
Article in English | MEDLINE | ID: mdl-12614884

ABSTRACT

An analysis of the X-ray structure of cilastatin bound to membrane dipeptidase, together with docking studies, is presented here to reveal how a simple amide may act as a high-affinity, reversible, amidase inhibitor. Cilastatin binds as a normal substrate and is orientated in a perfect near-attack conformer for formation of a tetrahedral intermediate with the zinc-bound water/hydroxide. This intermediate is fated, however, only to revert to its starting components as scission of the amide bond is prevented by the precise fit of cilastatin within the active site. The cilastatin alkyl end groups that are tightly buttressed against amino acid residues on opposite sides of the active site, are aligned along the C-N reaction coordinate axis thereby preventing collapse of the intermediate via rupture of the C-N bond. Such a feature could have more general applicability in the explicit design of substrate variants as selective, tight-binding, and reversible inhibitors.


Subject(s)
Cilastatin/chemistry , Cilastatin/metabolism , Dipeptidases/metabolism , Amidohydrolases/antagonists & inhibitors , Chemical Phenomena , Chemistry, Physical , Cilastatin/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Leukotriene D4/antagonists & inhibitors , Models, Molecular , Protein Binding , Protein Conformation
9.
Clin Infect Dis ; 24 Suppl 2: S249-55, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9126700

ABSTRACT

Meropenem is a carbapenem antibiotic that appears to be widely distributed in tissues and is eliminated by both excretion and metabolism. Approximately 70% of meropenem is excreted via the kidneys, thus dosage adjustments are required for patients with renal impairment. The pharmacokinetic parameters for meropenem are similar to those for imipenem/cilastatin, with the exception of meropenem's smaller volume of distribution. The urinary recovery of meropenem is as high as that of imipenem in combination with cilastatin, an inhibitor of renal dehydropeptidase. Therefore, unlike imipenem, meropenem can be used without dehydropeptidase inhibitors to obtain a consistently high concentration in the urine without nephrotoxic effects.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Bacterial Infections/drug therapy , Thienamycins/pharmacokinetics , Adult , Aged , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacterial Infections/urine , Child , Child, Preschool , Cilastatin/metabolism , Cilastatin/pharmacokinetics , Drug Therapy, Combination , Humans , Imipenem/metabolism , Imipenem/pharmacokinetics , Infant , Kidney/drug effects , Kidney/metabolism , Meropenem , Molecular Structure , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacokinetics , Rabbits , Rats , Thienamycins/chemistry , Thienamycins/metabolism , Urine/chemistry
10.
Toxicol Lett ; 76(3): 239-43, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7762010

ABSTRACT

The present study evaluated the proconvulsant liability of biapenem, a novel carbapenem antibiotic, in in vitro and in vivo experiments, in comparison with the carbapenems, imipenem/cilastatin and meropenem. Imipenem/cilastatin is a carbapenem antibiotic with known proconvulsive liability in man and in animal experiments. In in vivo studies imipenem/cilastatin, at doses of 400/400 mg/kg i.v., significantly lowered the convulsive threshold of pentylenetetrazol (PTZ) in mice and shifted the dose-response curve of PTZ. The effects of biapenem (400 mg/kg i.v.) and another reference carbapenem, meropenem (400 mg/kg i.v.), in the mouse PTZ model were not significantly different from control. In in vitro experiments the carbapenems were tested for their ability to inhibit [3H]muscimol (1.3 mM) binding to rat brain homogenates at concentrations of 1-10 mM. Similar to in vivo results, when compared to imipenem/cilastatin, biapenem and meropenem did not inhibit [3H]muscimol binding to the GABAA receptor complex in brain homogenates while imipenem/cilastatin exhibited significant inhibition (IC50 = 4.6 mM). These results further confirm the correlation between in vitro GABAA binding and in vivo PTZ convulsive testing with carbapenem antibiotics, and suggest that biapenem possesses a low proconvulsive liability.


Subject(s)
Cilastatin/toxicity , Imipenem/toxicity , Seizures/chemically induced , Thienamycins/toxicity , Analysis of Variance , Animals , Binding, Competitive , Cilastatin/administration & dosage , Cilastatin/metabolism , Dose-Response Relationship, Drug , Imipenem/administration & dosage , Imipenem/metabolism , Injections, Intravenous , Male , Meropenem , Mice , Muscimol/metabolism , Pentylenetetrazole/toxicity , Rats , Receptors, GABA-A/metabolism , Thienamycins/administration & dosage , Thienamycins/metabolism , gamma-Aminobutyric Acid/metabolism
11.
Drug Metab Dispos ; 17(4): 426-32, 1989.
Article in English | MEDLINE | ID: mdl-2571484

ABSTRACT

Cilastatin, a potent inhibitor of renal dehydropeptidase I, was specifically designed to inhibit renal metabolism of the antibiotic imipenem in order to achieve therapeutically relevant imipenem concentrations in the urinary tract. In this study the elimination kinetics of cilastatin in rats at doses of 5, 10, 20, 50, 100, and 200 mg/kg iv were demonstrated to be dose dependent, with total plasma clearance and non-renal clearance falling from 20.2 +/- 3.1 ml/min/kg and 17.7 +/- 3.3 ml/min/kg (mean +/- S.D.) at the 5 mg/kg dose to 11.4 +/- 1.2 ml/min/kg and 5.30 +/- 1.2 ml/min/kg, respectively, at the 200 mg/kg dose, whereas the volume of distribution of the drug remained unchanged. Since cilastatin is mainly eliminated by renal excretion as well as by N-acetylation, the non-renal clearance may reasonably reflect the N-acetylation process. Thus, the dose-dependent kinetics of cilastatin might be explained, at least partly, by the saturation of the N-acetylation of the drug. The dose-related decrease in the fraction (fm) of cilastatin converted to its N-acetylated metabolite provided further evidence for the saturable N-acetylation. The fm values decreased from 0.915 at the 10 mg/kg dose to 0.626 at the 100 mg/kg dose. Although both the total plasma clearance and non-renal clearance decreased with increasing dose, the dose had an opposite effect on the renal clearance of cilastatin. The renal clearance of cilastatin increased from 2.50 +/- 0.40 ml/min/kg at the lowest dose to 6.10 +/- 0.50 ml/min/kg at the highest dose as the dose increased.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cilastatin/pharmacokinetics , Animals , Biotransformation , Blood Proteins/metabolism , Cilastatin/blood , Cilastatin/metabolism , Dose-Response Relationship, Drug , Half-Life , Injections, Intravenous , Male , Probenecid/pharmacology , Protein Binding , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...