Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 72
1.
Exp Neurol ; 341: 113705, 2021 07.
Article En | MEDLINE | ID: mdl-33753139

Anticancer therapeutics can provoke severe side effects that impair the patient's quality of life. A frequent dose-limiting side effect of platinum-based anticancer therapy is neurotoxicity. Its pathophysiology is poorly understood, and effective preventive or therapeutic measures are missing. Therefore, elucidation of the molecular mechanism of platinating drug-induced neurotoxicity and the development of preventive strategies is urgently needed. To this end, we aim to use C. elegans as a 3R-compliant in vivo model. The 3R principles were conceived for animal welfare in science concerning animal experiments, which should be replaced, reduced or refined. We can analytically demonstrate dose-dependent uptake of cisplatin (CisPt) in C. elegans, as well as genotoxic and cytotoxic effects based on DNA adduct formation (i.e., 1,2-GpG intrastrand crosslinks), induction of apoptosis, and developmental toxicity. Measuring the impairment of pharyngeal pumping as a marker of neurotoxicity, we found that especially CisPt reduces the pumping frequency at concentrations where basal and touch-provoked movement were not yet affected. CisPt causes glutathione (GSH) depletion and RNAi-mediated knockdown of the glutamate-cysteine ligase GCS-1 aggravates the CisPt-induced inhibition of pharyngeal pumping. Moreover, N-acetylcysteine (NAC) mitigated CisPt-triggered toxicity, indicating that GSH depletion contributes to the CisPt-induced pharyngeal damage. In addition to NAC, amifostine (WR1065) also protected the pharynx of C. elegans from the toxic effects of CisPt. Measuring pharyngeal activity by the electrophysiological recording of neurotransmission in the pharynx, we confirmed that CisPt is neurotoxic in C. elegans and that NAC is neuroprotective in the nematode. The data support the hypothesis that monitoring the pharyngeal activity of C. elegans is a useful surrogate marker of CisPt-induced neurotoxicity. In addition, a low GSH pool reduces the resistance of neurons to CisPt treatment, and both NAC and WR1065 are capable of attenuating platinum-induced neurotoxicity during post-incubation in C. elegans. Overall, we propose C. elegans as a 3R-compliant in vivo model to study the molecular mechanisms of platinum-induced neurotoxicity and to explore novel neuroprotective therapeutic strategies to alleviate respective side effects of platinum-based cancer therapy.


Antineoplastic Agents/toxicity , Cisplatin/toxicity , Disease Models, Animal , Neurotoxicity Syndromes/prevention & control , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Chemoprevention/methods , Dose-Response Relationship, Drug , Mercaptoethylamines/pharmacology , Mercaptoethylamines/therapeutic use , Neurotoxicity Syndromes/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Platinum Compounds/toxicity
2.
Exp Neurol ; 325: 113141, 2020 03.
Article En | MEDLINE | ID: mdl-31865195

One of the most relevant dose-limiting adverse effects of platinum drugs is the development of a sensory peripheral neuropathy that highly impairs the patients' quality of life. Nowadays there are no available efficacy strategies for the treatment of platinum-induced peripheral neurotoxicity (PIPN), and the only way to prevent its development and progression is by reducing the dose of the cytostatic drug or even withdrawing the chemotherapy regimen. This clinical issue has been the main focus of hundreds of preclinical research works during recent decades. As a consequence, dozens of in vitro and in vivo models of PIPN have been developed to elucidate the molecular mechanisms involved in its development and to find neuroprotective targets. The apoptosis of peripheral neurons has been identified as the main mechanism involved in PIPN pathogenesis. This mechanism of DRG sensory neurons cell death is triggered by the nuclear and mitochondrial DNA platination together with the increase of the oxidative cellular status induced by the depletion of cytoplasmic antioxidant mechanisms. However, since there has been no successful transfer of preclinical results to clinical practise in terms of therapeutic approaches, some mechanisms of PIPN pathogenesis still remain to be elucidated. This review is focused on the pathogenic mechanisms underlying PIPN described up to now, provided by the critical analysis of in vitro and in vivo models.


Antineoplastic Agents/toxicity , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Peripheral Nervous System Diseases/chemically induced , Platinum Compounds/toxicity , Animals , Disease Models, Animal , Peripheral Nervous System Diseases/pathology
3.
BMC Nephrol ; 19(1): 219, 2018 09 04.
Article En | MEDLINE | ID: mdl-30180818

BACKGROUND: Nephrotoxicity is a major hazard complicating the use of platinum based drugs (PBD), which can hinder using higher doses protocols to maximize the therapeutic gain. Shortage of serum creatinine level as an accurate biomarker for acute kidney injuries (AKI) necessitates searching for novel biomarkers with better sensitivity and specificity in patients on PBD. METHODS: In a prospective cohort design, 132 patients receiving PBD were selected for the study. AKI was diagnosed by continuous follow up of serum creatinine level according to Kidney Disease: Improving Global Outcomes (KDIGO) guidelines 2012. Serum creatinine and urinary biomarkers (KIM-1, NGAL and cystatin C) was measured in the day of treatment and for 3 days after PBD cycle. RESULTS: AKI occurred in 35 patients (26.52% of patients). KIM-1, Cystatin C, and NGAL showed significant increase in samples collected in the day of AKI in comparison to their corresponding basal levels (P <  0.0001). In addition, significant increase in urinary levels of the biomarkers in samples collected 1 day before AKI in comparison to their basal levels (P <  0.0001, P <  0.0001, and P = 0.013 for KIM-1, NGAL and Cystatin C respectively). Furthermore KIM-1 data showed a significant increase 2 days before serum creatinine rise in comparison to the corresponding KIM-1 levels in patients who developed AKI (P = 0.001). CONCLUSIONS: Urinary KIM-1, Cystatin C and NGAL can predict PBD induced AKI in earlier stages than serum createnine. KIM-1 is the most sensitive biomarker for early detection of AKI in patients receiving PBD.


Acute Kidney Injury/chemically induced , Acute Kidney Injury/urine , Hepatitis A Virus Cellular Receptor 1/metabolism , Platinum Compounds/toxicity , Acute Kidney Injury/diagnosis , Adult , Biomarkers/urine , Cohort Studies , Cystatin C/urine , Female , Humans , Lipocalin-2/urine , Male , Middle Aged , Prospective Studies
4.
Inhal Toxicol ; 30(11-12): 472-481, 2018.
Article En | MEDLINE | ID: mdl-30654668

Halogenated platinum salts can trigger the development of occupational asthma. Until recently, laboratory research into the development and manifestation of platinum hypersensitivity responses were hindered by the lack of an animal model suitable for assessing the functional consequences of allergic sensitization. We employed a newly developed mouse model to assess the potential allergenicity of ammonium tetrachloroplatinate (ATCP), compare the relative potency of ATCP and another platinum salt, ammonium hexachloroplatinate (AHCP) and assess potential cross-reactivity. Mice were topically sensitized with ATCP before being challenged by intratracheal aspiration (IA) with ATCP. Ventilatory responses were assessed using whole-body plethysmography (WBP). An immediate response (IR) was observed in ATCP-sensitized and challenged mice. Two days later, responsiveness to the nonspecific stimuli methacholine (Mch) was detected in ATCP-sensitized mice using WBP. Bronchoalveolar lavage fluid collected from sensitized mice contained an average of 3.3% eosinophils compared to less than 0.5% in non-sensitized mice (p<.05). Serum harvested from sensitized mice also contained increased total serum immunoglobulin E (p<.05). These data are the first to demonstrate that topical exposure to ATCP is sufficient to develop immediate type hypersensitivity and that a single intra-airway challenge is capable of triggering pulmonary responses. To investigate potential cross-reactivity, mice were sensitized to AHCP and, challenged by a single IA with a second platinum compound, ATCP. Compared to non-sensitized mice challenged with ATCP, these mice exhibited an IR, responsiveness to Mch, and eosinophilic infiltration in the lungs similar to that achieved with AHCP challenge, thus demonstrating cross-reactivity.


Bronchial Hyperreactivity/etiology , Chlorides/toxicity , Drug Hypersensitivity/etiology , Platinum Compounds/toxicity , Respiratory Hypersensitivity/etiology , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cross Reactions , Disease Models, Animal , Drug Hypersensitivity/immunology , Drug Hypersensitivity/physiopathology , Female , Immunoglobulin E/blood , Mice, Inbred BALB C , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/physiopathology
5.
Sci Rep ; 7(1): 11708, 2017 09 15.
Article En | MEDLINE | ID: mdl-28916831

Using primary melanocytes and HEK293 cells, we found that cAMP signaling accelerates repair of bi- and mono-functional platinum-induced DNA damage. Elevating cAMP signaling either by the agonistic MC1R ligand melanocyte stimulating hormone (MSH) or by pharmacologic cAMP induction by forskolin enhanced clearance of intrastrand cisplatin-adducts in melanocytes or MC1R-transfected HEK293 cells. MC1R antagonists human beta-defensin 3 and agouti signaling protein blocked MSH- but not forskolin-mediated enhancement of platinum-induced DNA damage. cAMP-enhanced repair of cisplatin-induced DNA damage was dependent on PKA-mediated phosphorylation of ATR on S435 which promoted ATR's interaction with the key NER factor xeroderma pigmentosum A (XPA) and facilitated recruitment of an XPA-ATR-pS435 complex to sites of cisplatin DNA damage. Moreover, we developed an oligonucleotide retrieval immunoprecipitation (ORiP) assay using a novel platinated-DNA substrate to establish kinetics of ATR-pS435 and XPA's associations with cisplatin-damaged DNA. Expression of a non-phosphorylatable ATR-S435A construct or deletion of A kinase-anchoring protein 12 (AKAP12) impeded platinum adduct clearance and prevented cAMP-mediated enhancement of ATR and XPA's associations with cisplatin-damaged DNA, indicating that ATR phosphorylation at S435 is necessary for cAMP-enhanced repair of platinum-induced damage and protection against cisplatin-induced mutagenesis. These data implicate cAMP signaling as a critical regulator of genomic stability against platinum-induced mutagenesis.


Cyclic AMP/metabolism , DNA Damage/drug effects , DNA Repair/drug effects , Melanocortins/physiology , Mutagenesis/drug effects , Cell Line , Cells, Cultured , Genomic Instability/drug effects , HEK293 Cells , Humans , Platinum Compounds/toxicity , Signal Transduction/physiology
6.
Drug Chem Toxicol ; 40(2): 228-234, 2017 Apr.
Article En | MEDLINE | ID: mdl-27387363

Circulating platinum (Pt) is detectable in the blood of Pt-treated cancer patients for over a decade after the treatment. Prolonged exposure to Pt, in combination with adverse compounds from nutrition and lifestyle, such as cadmium (Cd), could increase the risk from second cancers. The aim of this study was to investigate the effects of simultaneous exposure to Cd- and Pt-compounds on oxidative and DNA damage and the possible protective effects of zinc (Zn) and selenium (Se). The aqueous solutions of PtCl4, CdCl2 × H2O, ZnCl2 and Na2SeO3 were added, alone or in combination, to whole blood and isolated erythrocytes to produce the final concentrations of 2000 µg/L of Pt, 8 µg/L of Cd, 100 µg/L of Se, and 1000 µg/L of Zn. The activity of copper, zinc-superoxide dismutase, glutathione peroxidase and glutathione in whole blood was determined after 1 h exposure in in vitro conditions. The induction of DNA strand-breaks in human peripheral blood leukocytes was determined with the alkaline comet assay after 24 h exposure. Exposure to Pt and/or Cd decreased the activities of antioxidant enzymes and elevated DNA damage compared to control. A statistically significant change in the activity of both enzymes and in the induction of DNA strand-breaks was observed in the cells treated with Pt + Cd combination, while the addition of Se and/or Zn resulted in partial recovery of these effects. The results indicate that combined exposure to Pt and Cd could disrupt antioxidant protection of the organism and increase DNA damage, whereas Se and Zn could partially ameliorate these harmful effects.


Antioxidants/pharmacology , Cadmium Chloride/toxicity , Chlorides/pharmacology , DNA Damage/drug effects , Enzymes/blood , Mutagens/toxicity , Oxidative Stress/drug effects , Platinum Compounds/toxicity , Sodium Selenite/pharmacology , Zinc Compounds/pharmacology , Adult , Biomarkers/blood , Comet Assay , Cytoprotection , Erythrocytes/drug effects , Erythrocytes/enzymology , Glutathione/blood , Glutathione Peroxidase/blood , Humans , Leukocytes/drug effects , Leukocytes/enzymology , Leukocytes/pathology , Male , Superoxide Dismutase/blood , Young Adult
7.
Brain Imaging Behav ; 11(6): 1652-1663, 2017 Dec.
Article En | MEDLINE | ID: mdl-27766586

Women with ovarian cancer often undergo chemotherapy involving multiple agents. However, little is known about treatment-related central neurotoxicity in this population. The goal of this cross-sectional study was to assess brain structure and function and neurocognitive abilities in patients with ovarian cancer following first-line chemotherapy. Eighteen patients with ovarian, peritoneal and fallopian tube cancer and eighteen healthy controls matched for gender, age and education participated in the study. The patients were evaluated 1-4 months following completion of first-line taxane/platinum chemotherapy. All participants underwent structural and functional magnetic resonance imaging (MRI), and completed neuropsychological tests of attention, memory and executive functions. Neuroimaging assessments included voxel-based morphometry (VBM) for measuring gray matter (GM) volume, and functional MRI (fMRI) during the N-back working memory task. The results of VBM showed that patients had significantly reduced GM volume compared to healthy controls in the right middle/superior frontal gyrus, and in the left supramarginal gyrus and left inferior parietal lobule. fMRI results indicated significantly decreased activation in patients relative to healthy controls in the left middle frontal gyrus and left inferior parietal lobule during the N-back task (1/2/3-back >0-back). There were no statistically significant differences between the two groups on the neuropsychological tests. This is the first study showing structural and functional alterations involving frontal and parietal regions in patients with ovarian cancer treated with first-line chemotherapy. These findings are congruent with studies involving women with breast cancer, and provide additional supporting evidence for central neurotoxicity associated with taxane/platinum chemotherapy.


Antineoplastic Agents/toxicity , Brain/diagnostic imaging , Brain/physiopathology , Ovarian Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Brain/drug effects , Brain/pathology , Brain Mapping , Bridged-Ring Compounds/therapeutic use , Bridged-Ring Compounds/toxicity , Cohort Studies , Cross-Sectional Studies , Female , Gray Matter/diagnostic imaging , Gray Matter/drug effects , Gray Matter/pathology , Gray Matter/physiopathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Memory, Short-Term/physiology , Middle Aged , Neuropsychological Tests , Organ Size , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/physiopathology , Ovarian Neoplasms/psychology , Pilot Projects , Platinum Compounds/therapeutic use , Platinum Compounds/toxicity , Preliminary Data , Taxoids/therapeutic use , Taxoids/toxicity
8.
J Neurosci Methods ; 273: 1-9, 2016 11 01.
Article En | MEDLINE | ID: mdl-27485087

BACKGROUND: It is currently unclear how the platinum (Pt) species released from platinum-containing stimulating electrodes may affect the health of the surrounding tissue. This study develops an effective system to assess the cytotoxicity of any electrode-liberated Pt over a short duration, to screen systems before future in vivo testing. NEW METHOD: A platinum electrode was stimulated for two hours under physiologically relevant conditions to induce the liberation of Pt species. The total concentration of liberated Pt species was quantified and the concentration found was used to develop a range of Pt species for our model system comprised of microglia and neuron-like cells. RESULTS: Under our stimulation conditions (k=2.3, charge density of 57.7µC/cm2), Pt was liberated to a concentration of 1ppm. Interestingly, after 24h of Pt exposure, the dose-dependent cytotoxicity plots revealed that cell death became statistically significant at 10ppm for microglia and 20ppm for neuronal cells. However, in neuron-like cell cultures, concentrations above 1ppm resulted in significant neurite loss after 24h. COMPARISON WITH EXISTING METHODS: To our knowledge, there does not exist a simple, in vitro assay system for assessing the cytotoxicity of Pt liberated from stimulating neural electrodes. CONCLUSIONS: This work describes a simple model assay that is designed to be applicable to almost any electrode and stimulation system where the electrode is directly juxtaposed to the neural target. Based on the application, the duration of stimulation and Pt exposure may be varied.


Cell Death/drug effects , Cytotoxins/toxicity , Electrodes/adverse effects , Microglia/drug effects , Motor Neurons/drug effects , Platinum Compounds/toxicity , Animals , Cell Line, Transformed , Culture Media, Conditioned/toxicity , Dose-Response Relationship, Drug , Electric Stimulation , Mice , Microglia/chemistry
9.
Drug Resist Updat ; 27: 72-88, 2016 07.
Article En | MEDLINE | ID: mdl-27449599

It is now widely accepted that organic anion-transporting polypeptides (OATPs), especially members of the OATP1A/1B family, can have a major impact on the disposition and elimination of a variety of endogenous molecules and drugs. Owing to their prominent expression in the sinusoidal plasma membrane of hepatocytes, OATP1B1 and OATP1B3 play key roles in the hepatic uptake and plasma clearance of a multitude of structurally diverse anti-cancer and other drugs. Here, we present a thorough assessment of the currently available OATP1A and OATP1B knockout and transgenic mouse models as key tools to study OATP functions in vivo. We discuss recent studies using these models demonstrating the importance of OATPs, primarily in the plasma and hepatic clearance of anticancer drugs such as taxanes, irinotecan/SN-38, methotrexate, doxorubicin, and platinum compounds. We further discuss recent work on OATP-mediated drug-drug interactions in these mouse models, as well as on the role of OATP1A/1B proteins in the phenomenon of hepatocyte hopping, an efficient and flexible way of liver detoxification for both endogenous and exogenous substrates. Interestingly, glucuronide conjugates of both the heme breakdown product bilirubin and the protein tyrosine kinase-targeted anticancer drug sorafenib are strongly affected by this process. The clinical relevance of variation in OATP1A/1B activity in patients has been previously revealed by the effects of polymorphic variants and drug-drug interactions on drug toxicity. The development of in vivo tools to study OATP1A/1B functions has greatly advanced our mechanistic understanding of their functional role in drug pharmacokinetics, and their implications for therapeutic efficacy and toxic side effects of anticancer and other drug treatments.


Antineoplastic Agents/toxicity , Inactivation, Metabolic/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Neoplasms/metabolism , Organic Cation Transport Proteins/genetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Camptothecin/toxicity , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/toxicity , Drug Interactions , Gene Expression , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Irinotecan , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver-Specific Organic Anion Transporter 1/metabolism , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Methotrexate/toxicity , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Niacinamide/analogs & derivatives , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Niacinamide/toxicity , Organic Cation Transport Proteins/deficiency , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Phenylurea Compounds/toxicity , Platinum Compounds/pharmacokinetics , Platinum Compounds/pharmacology , Platinum Compounds/toxicity , Sorafenib , Taxoids/pharmacokinetics , Taxoids/pharmacology , Taxoids/toxicity
10.
Inhal Toxicol ; 27(10): 468-80, 2015.
Article En | MEDLINE | ID: mdl-26309092

Occupational exposure to halogenated platinum salts can trigger the development of asthma. The risk to the general population that may result from the use of platinum in catalytic converters and its emerging use as a diesel fuel additive is unclear. To investigate pulmonary responses to platinum, we developed a mouse model of platinum hypersensitivity. Mice were sensitized through application of ammonium hexachloroplatinate (AHCP) to the shaved back on days 0, 5 and 19, and to each ear on days 10, 11 and 12. On days 24 and 29, mice were challenged by oropharyngeal aspiration with AHCP in saline. Before and immediately after challenge, pulmonary responses were assessed using whole body plethysmography (WBP). A dose-dependent increase in immediate responses was observed in AHCP-sensitized and challenged mice. On days 26 and 31, changes in ventilatory responses to methacholine (Mch) aerosol were assessed by WBP; dose-dependent increases in Mch responsiveness occurred in sensitized mice. Lymph node cell counts indicate a proliferative response in lymph nodes draining the sites of application. Bronchoalveolar lavage fluid harvested from sensitized mice contained an average of 5% eosinophils compared to less than 0.5% in non-sensitized mice (p < 0.05); significant increases in total serum immunoglobulin E were observed for all sensitized mice. Although a second airway challenge on day 29 affected some results, only one airway challenge was needed to observe changes in lung function.


Allergens/toxicity , Chlorides/toxicity , Hypersensitivity/etiology , Lung/drug effects , Platinum Compounds/toxicity , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Disease Models, Animal , Female , Hypersensitivity/blood , Hypersensitivity/immunology , Hypersensitivity/physiopathology , Immunoglobulin E/blood , L-Lactate Dehydrogenase/metabolism , Lung/immunology , Lung/physiopathology , Mice, Inbred BALB C
11.
Int J Clin Exp Pathol ; 8(1): 81-95, 2015.
Article En | MEDLINE | ID: mdl-25755695

BACKGROUND: Cisplatin, carboplatin and oxaliplatin are structurally-related compounds, which are commonly used in cancer therapy. Cisplatin (Platinol(®)) has Boxed Warning stating: "Cumulative renal toxicity associated with PLATINOL is severe", while carboplatin and oxaliplatin are less nephrotoxic. These drugs form platinum adducts with cellular DNA. Their bindings to cellular thiols (e.g., glutathione and metallothionein) are known to contribute to drug resistance while thiol depletion augments platinum toxicity. METHODS: Using phosphorescence oxygen analyzer, this study investigated the effects of platinum drugs on renal cellular respiration (mitochondrial O2 consumption) in the presence and absence of the thiol blocking agent N-ethylmaleimide (used here as a model for thiol depletion). Renal cellular ATP was also determined. Kidney fragments from C57BL/6 mice were incubated at 37 °C in Krebs-Henseleit buffer (gassed with 95% O2:5% CO2) with and without 100 µM platinum drug in the presence and absence of 100 µM N-ethylmaleimide for ≤ 6 h. RESULTS: Platinum drugs alone had no effects on cellular respiration (P ≥ 0.143) or ATP (P ≥ 0.161). N-ethylmaleimide lowered cellular respiration (P ≤ 0.114) and ATP (P = 0.008). The combination of platinum drug and N-ethylmaleimide significantly lowered both cellular respiration (P ≤ 0.006) and ATP (P ≤ 0.003). Incubations with N-ethylmaleimide alone were associated with moderate-to-severe tubular necrosis. Incubations with cisplatin+N-ethylmaleimide vs. cisplatin alone produced similar severities of tubular necrosis. Tubular derangements were more prominent in carboplatin+N-ethylmaleimide vs. carboplatin alone and in oxaliplatin+N-ethylmaleimide vs. oxaliplatin alone. CONCLUSIONS: These results demonstrate the adverse events of thiol depletion on platinum-induced nephrotoxicities. The results suggest cellular bioenergetics is a useful surrogate biomarker for assessing drug-induced nephrotoxicities.


Antineoplastic Agents/toxicity , Cell Respiration/drug effects , Kidney/drug effects , Platinum Compounds/toxicity , Animals , Ethylmaleimide/pharmacology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Sulfhydryl Compounds/metabolism , Sulfhydryl Reagents/pharmacology
12.
Anticancer Res ; 35(2): 739-51, 2015 Feb.
Article En | MEDLINE | ID: mdl-25667453

BACKGROUND/AIM: A new platinum compound, (Pt(O,O'-acac)(γ-acac)(DMS)) (PtAcacDMS), has been shown to possess higher cytotoxic activity than cisplatin on several cancer and chemoresistant cell lines. The aim of the present study was to compare the nephrotoxic effects - particularly renal fibrogenesis- of PtAcacDMS and cisplatin in rats after the subcutaneous administration of a single dose (5 mg/Kg b.w., s.c.) of either compound to ten-day-old rats. MATERIALS AND METHODS: Control and treated rats were killed 1 day (PD11), 7 days (PD17), 21 days (PD31) and 40 days (PD50) after the injection. Kidneys were processed for light and electron microscopy, and platinum determination. Antibodies against E-cadherin (E-cad), vimentin (VIM) and α-smooth muscle actin (αSMA) were used to identify epithelial and mesenchymal cells. RESULTS AND CONCLUSION: Cisplatin produced progressive cortical fibrotic lesions displaying a variable number of VIM-positive tubules and interstitial αSMA-positive cells around. By contrast, PtAcacDMS induced a minimal number of histopathological changes, which declined in the adult samples, while the renal platinum content was generally higher after PtAcacDMS than after cisplatin. The present experimental model was proven suitable to investigate the occurrence of epithelial-mesenchymal transition (EMT) in renal fibrogenesis induced by the platinum-based compounds.


Cisplatin/toxicity , Disease Models, Animal , Fibrosis/chemically induced , Kidney/drug effects , Micelles , Platinum Compounds/toxicity , Animals , Body Weight/drug effects , Female , Kidney/pathology , Rats , Rats, Wistar
13.
Cardiovasc Toxicol ; 15(3): 261-8, 2015 Jul.
Article En | MEDLINE | ID: mdl-25404470

We have compared the cardiotoxicity of five platinum complexes in a model of isolated rat heart using the Langendorff technique. These effects were assessed via coronary flow (CF) and cardiac functional parameters. cis-Diamminedichloroplatinum(II) (cisplatin, CDDP), dichloro-(1,2-diaminocyclohexane)platinum(II) (Pt((II))DACHCl2), dichloro-(ethylenediamine)platinum(II) (Pt((II))ENCl2), tetrachloro-(1,2-diaminocyclohexane)platinum(IV) (Pt((IV))DACHCl4) and tetrachloro-(ethylenediamine)platinum(IV) (Pt((II))ENCl4) were perfused at increasing concentrations of 10(-8), 10(-7), 10(-6), 10(-5) and 10(-4) M during 30 min. In this paper, we report that cisplatin-induced dose-dependent effects on cardiac contractility and coronary flow both manifested as decrease in cardiac contractile force (dP/dt)max, heart rate and significant reduction in CF. Pt((II))ENCl2, Pt((IV))ENCl2 and Pt((IV))DACHCl4 did induce dose-dependent response only in case of CF. Our results could be also important for better understanding dose-dependent side effects of potential metal-based anticancer drugs.


Antineoplastic Agents/toxicity , Cardiotoxicity/physiopathology , Cisplatin/toxicity , Heart/drug effects , Heart/physiopathology , Platinum Compounds/toxicity , Animals , Cardiotoxicity/pathology , Dose-Response Relationship, Drug , Male , Organ Culture Techniques , Platinum Compounds/chemistry , Rats , Rats, Wistar
14.
Neuro Endocrinol Lett ; 35 Suppl 2: 35-42, 2014.
Article En | MEDLINE | ID: mdl-25638364

OBJECTIVES: Road traffic pollutants and the residues of cytostatics that are widely used in anti-cancer therapy are a significant sources of platinum group elements (PGE; Pt, Pd and Rh) in environment. These metals can migrate into sewage and thus pollute surface waters. The purpose of our study was to evaluate the effect of PtCl4 on the antioxidant and enzymatic activity of duckweed (Lemna minor), a bioindicator of the aquatic environment. METHODS: The study was performed using a 7-day conventional test based on the OECD 221 (CSN EN ISO 20079)--Lemna sp. Growth Inhibition Test. We also conducted a microbiotest to analyse the effects of PtC4, PdCl2 and RhCl3 on the morphology and vegetative growth of colonies of this plant and compared their inhibitory effects during the microbiotest. RESULTS: We observed inhibition of colony growth and clear morphological changes. Antioxidant and enzymatic activities increased with platinum doses increased. The 168hEC50 of PtCl4 was 12.16 µM (95% confidence interval = 9.88-14.44) and the 168hEC50 of PdCl2 was 50.39 (95% confidence interval = 23.83-76.96). The greatest inhibition of growth by RhCl3 was observed at 25 µM. CONCLUSIONS: The obtained results suggest that L. minor phytotoxicity tests should be widely used in the biomonitoring.


Palladium/toxicity , Plants , Platinum Compounds/toxicity , Rhodium/toxicity , Water Pollution, Chemical/adverse effects , Environmental Monitoring , Plants/anatomy & histology , Plants/enzymology , Plants/metabolism
15.
Nanoscale ; 5(24): 12464-73, 2013 Dec 21.
Article En | MEDLINE | ID: mdl-24165858

Magnetic resonance (MR) imaging using magnetic nanoparticles as the contrast agent has been extensively explored in biomedical imaging and disease diagnosis. Herein, we develop biocompatible polymer coated ultra-small Pt3Co magnetic nanoparticles as a new T2-weighted MR imaging contrast agent. A unique class of alloy Pt3Co nanoparticles is synthesized through a thermal decomposition method. After being modified with polyethylene glycol (PEG), the obtained Pt3Co-PEG nanoparticles exhibit an extremely high T2-weighted relaxivity rate (r2) up to 451.2 mM s(-1), which is much higher than that of Resovist®, a commercial T2-MR contrast agent used in the clinic. In vitro experiments indicate no obvious cytotoxicity of Pt3Co-PEG nanoparticles to various cell lines. After intravenous injection of Pt3Co-PEG nanoparticles, in vivo T2-weighted MR imaging of tumor-bearing mice reveals strong tumor contrast, which is much higher than that offered by injecting Resovist®. We further study the long-term biodistribution and toxicology of this new type of MR contrast nanoparticles after intravenous injection into healthy mice. Despite the significant retention of Pt3Co-PEG nanoparticles in the mouse liver and spleen, no appreciable toxicity of these nanoparticles to the treated animals has been noted in our detailed histological and hematological analysis over a course of 60 days. Our work demonstrates that functionalized Pt3Co nanoparticles may be a promising new type of T2-weighted MR contrast agent potentially useful in biomedical imaging and diagnosis.


Cobalt/chemistry , Contrast Media/chemical synthesis , Magnetic Resonance Imaging/methods , Metal Nanoparticles/chemistry , Platinum Compounds/chemistry , Polyethylene Glycols/chemistry , Animals , Cells, Cultured , Cobalt/toxicity , Contrast Media/chemistry , Contrast Media/toxicity , HEK293 Cells , Humans , Magnets/chemistry , Magnets/toxicity , Materials Testing , Metal Nanoparticles/toxicity , Mice , Mice, Inbred BALB C , Platinum Compounds/toxicity , Polyethylene Glycols/toxicity , Time Factors
16.
Adv Exp Med Biol ; 755: 47-53, 2013.
Article En | MEDLINE | ID: mdl-22826048

Allergic immediate-type reactions by halogenated compounds of platinum (Pt) (platinum salts) have been described in workers in precious metal refineries and catalyst productions. In both industries there are exposures to many different Pt compounds. It is believed that the most important allergens are those compounds with the highest number of halide ligands. It is unknown whether sensitizations to compounds with a lower number of halide ligands represent co-sensitizations or are due to cross-reactivity. We report a worker engaged in the production of cytotoxic drugs with occupational asthma and exposure to only one Pt salt with four halide ligands. The 22-year-old worker developed work-related sneezing, runny nose, and variable dyspnea about a year after he had started to work in the cytotoxic drugs production with exposure to potassium tetrachloroplatinate(II) (K(2)PtCl(4)). He was immediately removed from his workplace and admitted for a medical opinion about 6 months afterwards. Spirometry was normal, but asthma was corroborated by a positive response to methacholine. The results of skin prick testing could not be interpreted due to urticaria factitia. Challenge with K(2)PtCl(4) by a dosimeter method yielded a clear immediate-type reaction with an increase of exhaled nitric oxide from 32 to 156 ppb after 24 h indicating an increased airway inflammation. Pt salts with four halide ligands like K(2)PtCl(4) may cause an allergic immediate-type reaction and occupational asthma. Workers in the production of Pt-containing cytotoxic drugs with exposure to these substances should be included in medical surveillance programs for the prevention of occupational asthma caused by Pt salts.


Antineoplastic Agents/chemical synthesis , Asthma, Occupational/chemically induced , Chlorides/toxicity , Cisplatin/chemical synthesis , Hypersensitivity, Immediate/chemically induced , Platinum Compounds/toxicity , Adult , Humans , Male
17.
Yakugaku Zasshi ; 132(11): 1281-5, 2012.
Article Ja | MEDLINE | ID: mdl-23123720

Platinum agents are widely used in cancer chemotherapy. Cisplatin, carboplatin, oxaliplatin and nedaplatin have a common chemical structure consisting of platinum, carrier groups and leaving groups, and undergo the similar mechanism of cytotoxicity. Only cisplatin induces nephrotoxicity, although the molecular mechanism involved is unclear. Organic cation transporter (OCT)/SLC22A, and multidrug and toxin extrusion (MATE)/SLC47A play a role in renal handling of cationic drug in the kidney. We focused on a role of transporters in nephrotoxicity of platinum agents. OCT2 mediates the transport of cisplatin and is the determinant of cisplatin-induced nephrotoxicity. In addition, MATE1 protects cisplatin-induced nephrotoxicity. Oxaliplatin, which was a superior substrate of the luminal efflux transporter, MATE2-K as well as OCT2, did not show nephrotoxicity. Moreover, carboplatin and nedaplatin were not transported by these transporters. Substrate specificity could regulate the features of platinum agents. Recent findings indicate that organic cation transporters are key to the nephrotoxicity of platinum agents.


Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney/drug effects , Kidney/metabolism , Organic Cation Transport Proteins/physiology , Organic Cation Transporter 1/physiology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Biological Transport, Active , Cisplatin/chemistry , Cisplatin/metabolism , Humans , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/metabolism , Platinum Compounds/metabolism , Platinum Compounds/toxicity , Substrate Specificity
18.
Neuro Endocrinol Lett ; 33 Suppl 3: 107-12, 2012.
Article En | MEDLINE | ID: mdl-23353852

OBJECTIVES: An enhanced worldwide application of platinum group elements (PGE), in particular platinum, has been observed during recent decades. An increased concentration of PGE was determined in collected samples of great amount of aqueous ecosystems.The aim was to compare phytotoxic effect of platinum (PtCl4) by performing two different bioassays on green algae Pseudokirchneriella subcapitata and macrophyte duckweed, Lemna minor. MEDTHODS: The algal experiment (Pseudokichneriella subcapitata) followed OECD 201, the concentration row for PtCl4 was: 0.05; 0.01; 0.25; 0.5; 1 µM. The duckweed (Lemna minor) experiment was conducted according to OECD 221, employed PtCl4 concentrations were: 5; 10; 25; 50; 100 µM. Plants were cultivated as a microbiotest, using micro-volumes. RESULTS: The results of the algal test showed significant growth inhibition of the final biomass. The values of 72hEC5(µ), 72hEC10(µ), 72hEC20(µ) counted on a basis of average specific growth rate (µ) were 0.31 µM, 0.58 µM and 1.12 µM of PtCl4, respectively. The values, obtained on a basis of the area under the growth curves (A), were 0.04 µM (72hEC5(A)), 0.24 µM (72hEC10(A)) and 0.64 µM (72hEC20(A)). The experiment with duckweed showed 50% of growth inhibition and the values of 168hEC50(µ) were 19.55 µM and 168hEC50(A) 13.63 µM of PtCl4. CONCLUSION: The fronds of duckweed showed strong adverse effect of platinum influence (chlorosis, necrosis). The algal test and the estimation of 72hEC5(A) appears to be the most sensitive.


Chlorophyta/drug effects , Chlorophyta/growth & development , Platinum Compounds/toxicity , Platinum/toxicity , Water Pollutants, Chemical/toxicity , Biological Assay , Biomass , Dose-Response Relationship, Drug , Ecosystem , Toxicity Tests
19.
Neuro Endocrinol Lett ; 33 Suppl 3: 173-8, 2012.
Article En | MEDLINE | ID: mdl-23353863

OBJECTIVES: Regarding the environmental pollution, platinum group elements (PGE) are in the centre of interest of current research. These rare elements are used as effective substances in automotive catalysts to reduce pollution by emissions originating from fuel combustion. Due to their harmful potential, it is necessary to monitor their content and behaviour in different samples. Comprehensive studies on PGE behaviour and effects are still lacking. Their distribution in the food chain and data on bioaccumulation has not been described so far. METHODS: We focused on reproductive effects of platinum (PtCl4), in particular. Our study is based on a collembolan laboratory breed, test optimalization and validation according to the OECD 232 standards [CSN ISO 11267 - Soil quality - Inhibition of reproduction of Collembola (Folsomia candida) by soil pollutants]. The concentrations of PtCl4 tested were as follows: 5, 10, 25, 50 and 100 µM. The EC50 was determined after 28 days of testing. RESULTS: The results were evaluated using the inhibition of reproduction compared with controls. The EC50 was determined after the 28-day test. The value of 28dEC50 of the boric acid test was estimated at 120 mg/kg and the measured 28dEC50 of PtCl4 was 200.4 µM. CONCLUSION: The presented data can be considered as a step forward in the assessment of the potential risk of platinum in the terrestrial environment. However, more toxicity data for various species are needed to evaluate the environmental risk of platinum in soils.


Arthropods/drug effects , Platinum Compounds/toxicity , Platinum/toxicity , Reproduction/drug effects , Soil Pollutants/toxicity , Age Factors , Animals , Arthropods/growth & development , Behavior, Animal/drug effects , Female , Male , Toxicity Tests
...