Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 9(9)2020 Sep 03.
Article in English | MEDLINE | ID: mdl-32899204

ABSTRACT

Gentamicin is a used antibiotic that causes nephrotoxicity in 10-20% of treatment periods, which limits its use considerably. Our results have shown that cilastatin may be a promising therapeutic alternative in toxin-induced acute kidney injury (AKI). Here, we investigated its potential use as a nephroprotector against gentamicin-induced AKI in vitro and in vivo. Porcine renal cells and rats were treated with gentamicin and/or cilastatin. In vivo nephrotoxicity was analyzed by measuring biochemical markers and renal morphology. Different apoptotic, oxidative and inflammatory parameters were studied at cellular and systemic levels. Megalin, mainly responsible for the entry of gentamicin into the cells, was also analyzed. Results show that cilastatin protects cells from gentamicin-induced AKI. Cilastatin decreased creatinine, BUN, kidney injury molecule-1 (KIM-1) and severe morphological changes previously increased by gentamicin in rats. The interference of cilastatin with lipid rafts cycling leads to decreased expression of megalin, and therefore gentamicin uptake and myeloid bodies, resulting in a decrease of apoptotic, oxidative and inflammatory events. Moreover, cilastatin did not prevent bacterial death by gentamicin. Cilastatin reduced gentamicin-induced AKI by preventing key steps in the amplification of the damage, which is associated to the disruption of megalin-gentamicin endocytosis. Therefore, cilastatin might represent a novel therapeutic tool in the prevention and treatment of gentamicin-induced AKI in the clinical setting.

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.
Nephrol Dial Transplant ; 32(10): 1645-1655, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28340076

ABSTRACT

BACKGROUND: Cisplatin is a potent chemotherapeutic drug whose nephrotoxic effect is a major complication and a dose-limiting factor for antitumoral therapy. There is much evidence that inflammation contributes to the pathogenesis of cisplatin-induced nephrotoxicity. We found that cilastatin, a renal dehydropeptidase-I inhibitor, has protective effects in vitro and in vivo against cisplatin-induced renal damage by inhibiting apoptosis and oxidation. Here, we investigated the potential use of cilastatin to protect against cisplatin-induced kidney injury and inflammation in rats. METHODS: Male Wistar rats were divided into four groups: control, cilastatin-control, cisplatin and cilastatin-cisplatin. Nephrotoxicity was assessed 5 days after administration of cisplatin based on blood urea nitrogen, creatinine, glomerular filtration rate (GFR), kidney injury molecule (KIM)-1 and renal morphology. Inflammation was measured using the electrophoretic mobility shift assay, immunohistochemical studies and evaluation of inflammatory mediators. RESULTS: Compared with the control rats, cisplatin-administered rats were affected by significant proximal tubule damage, decreased GFR, increased production of inflammatory mediators and elevations in urea, creatinine and tissue KIM-1 levels. Cilastatin prevented these changes in renal function and ameliorated histological damage in cisplatin-administered animals. Cilastatin also reduced pro-inflammatory cytokine levels, activation of nuclear factor-κB and CD68-positive cell concentrations. CONCLUSIONS: Cilastatin reduces cisplatin-induced nephrotoxicity, which is associated with decreased inflammation in vivo. Although the exact role of decreased inflammation in nephroprotection has not been fully elucidated, treatment with cilastatin could be a novel strategy for the prevention of cisplatin-induced acute kidney injury.


Subject(s)
Antineoplastic Agents/toxicity , Cilastatin/pharmacology , Cisplatin/toxicity , Nephritis/prevention & control , Protease Inhibitors/pharmacology , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Cilastatin/therapeutic use , Creatinine/blood , Cytokines/blood , Cytokines/urine , Drug Evaluation, Preclinical , Glomerular Filtration Rate/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Male , NF-kappa B/metabolism , Nephritis/chemically induced , Nephritis/metabolism , Protease Inhibitors/therapeutic use , Rats , Rats, Wistar
4.
Talanta ; 164: 16-26, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28107912

ABSTRACT

Imaging techniques for mapping molecular distributions in tissue sections can reveal valuable information on biomolecules involved in relevant biochemical processes. A method has been developed for comprehensive, reproducible and sensitive lipid imaging by matrix-assisted laser/desorption ionization-LTQ-Orbitrap mass spectrometry in kidney sections, showing the benefits of exact mass determination. Matrix deposition parameters for positive and negative lipid ion imaging using different matrices such as 2,5-dihydroxybenzoic acid (DHB), 9-aminoacridine (9-AA) or α-cyano-4-hydroxycinnamic acid (CHCA) have been optimized for the broadest detection and identification of renal lipids. The combination of 9-AA and DHB was found as the most suitable for negative and positive ion mode lipid imaging, respectively. Lipid mapping and related identification strategies and limitations have also been discussed. Production of 100-µm resolution images was proved to be enough for discerning lipid distribution in kidney substructures. Imaging reproducibility was assessed on parallel kidney slices with time. This method has been applied to the lipidomics analysis on kidney sections from rats treated with the antitumor drug cisplatin and compared to healthy control rats. Up to 66 different renal lipids out of 450 extracted ion images (mainly phospholipid species, in addition to sulfatides and cholesterol sulfate) have been found and identified showing a modified distribution pattern due to cisplatin-induced nephrotoxicity. These lipid species reflect either topographic, signaling or structural processes in damaged kidney and could potentially be used for nephrotoxicity assessment or as therapeutic targets. This is, to our knowledge, the first imaging lipidomics study for nephrotoxicity assessment of cisplatin chemotherapy.


Subject(s)
Cisplatin/adverse effects , Kidney/drug effects , Kidney/metabolism , Lipid Metabolism/drug effects , Molecular Imaging , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Animals , Female , Rats , Rats, Wistar
5.
Biomed Res Int ; 2016: 2518626, 2016.
Article in English | MEDLINE | ID: mdl-27556029

ABSTRACT

Warm ischemia (WI) produces a significant deleterious effect in potential kidney grafts. Hypothermic machine perfusion (HMP) seems to improve immediate graft function after transplant. Our aim was to analyze the effect of short pretransplant periods of pulsatile HMP on histology and renal injury in warm-ischemic kidneys. Twelve minipigs were used. WI was achieved in the right kidney by applying a vascular clamp for 45 min. After nephrectomy, autotransplant was performed following one of two strategies: cold storage of the kidneys or cold storage combined with perfusion in pulsatile HMP. The graft was removed early to study renal morphology, inflammation (fibrosis), and apoptosis. Proinflammatory activity and fibrosis were less pronounced after cold storage of the kidneys with HMP than after cold storage only. The use of HMP also decreased apoptosis compared with cold storage only. The detrimental effects on cells of an initial and prolonged period of WI seem to improve with a preservation protocol that includes a short period of pulsatile HMP after cold storage and immediately before the transplant, in comparison with cold storage only.


Subject(s)
Kidney Transplantation , Kidney , Perfusion/methods , Tissue Survival , Transplants , Animals , Cold Temperature , Kidney/pathology , Kidney/physiology , Kidney/surgery , Kidney Transplantation/methods , Kidney Transplantation/statistics & numerical data , Swine , Swine, Miniature , Time Factors , Transplants/physiology , Transplants/statistics & numerical data
6.
Anal Bioanal Chem ; 408(9): 2309-18, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26825342

ABSTRACT

The study of the distribution of the cytostatic drugs cisplatin, carboplatin, and oxaliplatin along the kidney may help to understand their different nephrotoxic behavior. Laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) allows the acquisition of trace element images in biological tissues. However, results obtained are affected by several variations concerning the sample matrix and instrumental drifts. In this work, an internal standardization method based on printing an Ir-spiked ink onto the surface of the sample has been developed to evaluate the different distributions and accumulation levels of the aforementioned drugs along the kidney of a rat model. A conventional ink-jet printer was used to print fresh sagittal kidney tissue slices of 4 µm. A reproducible and homogenous deposition of the ink along the tissue was observed. The ink was partially absorbed on top of the tissue. Thus, this approach provides a pseudo-internal standardization, due to the fact that the ablation sample and internal standard take place subsequently and not simultaneously. A satisfactory normalization of LA-ICP-MS bioimages and therefore a reliable comparison of the kidney treated with different Pt-based drugs were achieved even for tissues analyzed on different days. Due to the complete ablation of the sample, the transport of the ablated internal standard and tissue to the inductively coupled plasma-mass spectrometry (ICP-MS) is practically taking place at the same time. Pt accumulation in the kidney was observed in accordance to the dosages administered for each drug. Although the accumulation rate of cisplatin and oxaliplatin is high in both cases, their Pt distributions differ. The strong nephrotoxicity observed for cisplatin and the absence of such side effect in the case of oxaliplatin could explain these distribution differences. The homogeneous distribution of oxaliplatin in the cortical and medullar areas could be related with its higher affinity for cellular transporters such as MATE2-k.


Subject(s)
Carboplatin/toxicity , Cisplatin/toxicity , Ink , Kidney/drug effects , Organoplatinum Compounds/toxicity , Printing , Animals , Carboplatin/metabolism , Cisplatin/metabolism , Kidney/metabolism , Organoplatinum Compounds/metabolism , Oxaliplatin , Rats , Reference Standards
7.
Biomed Res Int ; 2015: 704382, 2015.
Article in English | MEDLINE | ID: mdl-26504822

ABSTRACT

Vancomycin is a very effective antibiotic for treatment of severe infections. However, its use in clinical practice is limited by nephrotoxicity. Cilastatin is a dehydropeptidase I inhibitor that acts on the brush border membrane of the proximal tubule to prevent accumulation of imipenem and toxicity. The aim of this study was to investigate the potential protective effect of cilastatin on vancomycin-induced apoptosis and toxicity in cultured renal proximal tubular epithelial cells (RPTECs). Porcine RPTECs were cultured in the presence of vancomycin with and without cilastatin. Vancomycin induced dose-dependent apoptosis in cultured RPTECs, with DNA fragmentation, cell detachment, and a significant decrease in mitochondrial activity. Cilastatin prevented apoptotic events and diminished the antiproliferative effect and severe morphological changes induced by vancomycin. Cilastatin also improved the long-term recovery and survival of RPTECs exposed to vancomycin and partially attenuated vancomycin uptake by RPTECs. On the other hand, cilastatin had no effects on vancomycin-induced necrosis or the bactericidal effect of the antibiotic. This study indicates that cilastatin protects against vancomycin-induced proximal tubule apoptosis and increases cell viability, without compromising the antimicrobial effect of vancomycin. The beneficial effect could be attributed, at least in part, to decreased accumulation of vancomycin in RPTECs.


Subject(s)
Anti-Bacterial Agents/toxicity , Cell Survival/drug effects , Cilastatin/pharmacology , Kidney Tubules, Proximal/cytology , Protective Agents/pharmacology , Vancomycin/toxicity , Animals , Apoptosis/drug effects , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Swine
9.
J Appl Physiol (1985) ; 114(9): 1282-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23288549

ABSTRACT

Mitochondrial muscle alterations are common in patients with chronic obstructive pulmonary disease (COPD) and manifest mainly as decreased oxidative capacity and excessive production of reactive oxygen species (ROS). The significant loss of oxidative capacity observed in the quadriceps of COPD patients is mainly due to reduced mitochondrial content in the fibers, a finding consistent with the characteristic loss of type I fibers observed in that muscle. Decreased oxidative capacity does not directly limit maximum performance; however, it is associated with increased lactate production at lower exercise intensity and reduced endurance. Since type I fiber atrophy does not occur in respiratory muscles, the loss of such fibers in the quadriceps could be to the result of disuse. In contrast, excessive production of ROS and oxidative stress are observed in both the respiratory muscles and the quadriceps of COPD patients. The causes of increased ROS production are not clear, and a number of different mechanisms can play a role. Several mitochondrial alterations in the quadriceps of COPD patients are similar to those observed in diabetic patients, thus suggesting a role for muscle alterations in this comorbidity. Amino acid metabolism is also altered. Expression of peroxisome proliferator-activated receptor-γ coactivator-1α mRNA is low in the quadriceps of COPD patients, which could also be a consequence of type I fiber loss; nevertheless, its response to exercise is not altered. Patterns of muscle cytochrome oxidase gene activation after training differ between COPD patients and healthy subjects, and the profile is consistent with hypoxic stress, even in nonhypoxic patients.


Subject(s)
Muscle, Skeletal/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Humans , Mitochondria, Muscle/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Models, Biological , Muscle, Skeletal/physiopathology , Oxygen Consumption , Pulmonary Disease, Chronic Obstructive/physiopathology , Reactive Oxygen Species/metabolism
10.
Kidney Int ; 82(6): 652-63, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22718191

ABSTRACT

Cisplatin is an anticancer agent marred by nephrotoxicity; however, limiting this adverse effect may allow the use of higher doses to improve its efficacy. Cilastatin, a small molecule inhibitor of renal dehydropeptidase I, prevents proximal tubular cells from undergoing cisplatin-induced apoptosis in vitro. Here, we explored the in vivo relevance of these findings and the specificity of protection for kidney cells in cisplatin-treated rats. Cisplatin increased serum blood urea nitrogen and creatinine levels, and the fractional excretion of sodium. Cisplatin decreased the glomerular filtration rate, promoted histological renal injury and the expression of many pro-apoptotic proteins in the renal cortex, increased the Bax/Bcl2 ratio, and oxidative stress in kidney tissue and urine. All these features were decreased by cilastatin, which preserved renal function but did not modify the pharmacokinetics of cisplatin area under the curve. The cisplatin-induced death of cervical, colon, breast, and bladder-derived cancer cell lines was not prevented by cilastatin. Thus, cilastatin has the potential to prevent cisplatin nephrotoxicity without compromising its anticancer efficacy.


Subject(s)
Antineoplastic Agents , Apoptosis/drug effects , Cilastatin/pharmacology , Cisplatin , Kidney Diseases/prevention & control , Kidney/drug effects , Protease Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis Regulatory Proteins/metabolism , Area Under Curve , Biomarkers/blood , Blood Urea Nitrogen , Body Weight/drug effects , Cell Line, Tumor , Cisplatin/pharmacokinetics , Cisplatin/toxicity , Creatinine/blood , Cytoprotection , Dipeptidases/antagonists & inhibitors , Dipeptidases/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Glomerular Filtration Rate/drug effects , Kidney/enzymology , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/blood , Kidney Diseases/chemically induced , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Male , Natriuresis/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Swine
11.
Anal Chem ; 83(20): 7933-40, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21913725

ABSTRACT

A laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS)-based methodology is presented for Pt, Cu, and Zn bioimaging on whole kidney 3 µm sagittal sections from rats treated with pharmacological doses of cisplatin, which were sacrificed once renal damage had taken place. Pt turned out to accumulate in the kidney cortex and corticomedullary junction, corresponding to areas where the proximal tubule S3 segments (the most sensitive cells to cisplatin nephrotoxicity) are located. This demonstrates the connection between platinum accumulation and renal damage proved by histological examination of HE-stained sections and evaluation of serum and urine biochemical parameters. Cu and Zn distribution maps revealed a significant displacement in cells by Pt, as compared to control tissues. A dramatic decrease in the Pt accumulation in the cortex was observed when cilastatin was coadministered with cisplatin, which can be related to its nephroprotective effect. Excellent imaging reproducibility, sensitivity (LOD 50 fg), and resolution (down to 8 µm) were achieved, demonstrating that LA-ICP-MS can be applied as a microscopic metal detector at cellular level in certain tissues. A simple and quick approach for the estimation of Pt tissue levels was proposed, based on tissue spiking.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney/pathology , Mass Spectrometry/methods , Animals , Cilastatin/pharmacology , Copper/chemistry , Female , Kidney Cortex/pathology , Kidney Tubules, Proximal/pathology , Mass Spectrometry/instrumentation , Platinum/analysis , Rats , Rats, Wistar , Zinc/chemistry
12.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...