51
|
Mendonça LM, Dos Santos GC, Antonucci GA, Dos Santos AC, Bianchi MDLP, Antunes LMG. Evaluation of the cytotoxicity and genotoxicity of curcumin in PC12 cells. Mutat Res 2009; 675:29-34. [PMID: 19386244 DOI: 10.1016/j.mrgentox.2009.02.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 12/23/2008] [Accepted: 02/04/2009] [Indexed: 05/27/2023]
Abstract
Neurotoxicity induced by reactive oxygen species can appear as an adverse effect of chemotherapy treatment with platinum compounds, such as cisplatin. The use of this drug in clinical practice is limited due to its adverse effects, including nephrotoxicity, ototoxicity, neurotoxicity and genotoxicity. Functional foods or nutraceuticals have demonstrated potential neuroprotective activity in several experiments and models. This study aimed to investigate the possible cytotoxicity and genotoxicity/antigenotoxic effects of curcumin in PC12 cells exposed to cisplatin. Cell viability and genotoxicity/antigenotoxicity were evaluated by the MTT assay and micronucleus test, respectively. PC12 cells were treated with different concentrations of cisplatin and curcumin (0.5 -- 128 microg/mL). Analysis of the results showed that high concentrations of curcumin were cytotoxic and increased micronuclei frequency compared to the control group. In the associated treatments, at all three concentrations evaluated, curcumin significantly reduced the total frequency of micronuclei induced by cisplatin. Determining the cytotoxic and genotoxic/antigenotoxic effects of this frequently used antioxidant in a neuronal model is important to assess possible hazards when combined with other chemical agents, including chemotherapy drugs used in cancer therapy.
Collapse
Affiliation(s)
- Leonardo Meneghin Mendonça
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | |
Collapse
|
52
|
Kagiava A, Tsingotjidou A, Emmanouilides C, Theophilidis G. The effects of oxaliplatin, an anticancer drug, on potassium channels of the peripheral myelinated nerve fibres of the adult rat. Neurotoxicology 2008; 29:1100-6. [PMID: 18845186 DOI: 10.1016/j.neuro.2008.09.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Revised: 09/04/2008] [Accepted: 09/09/2008] [Indexed: 12/27/2022]
Abstract
Oxaliplatin is a novel chemotherapeutic agent which is effective against advanced colorectal cancer, but at the same time causes severe neuropathy in the peripheral nerve fibres, affecting mainly the voltage-gated sodium (Na(+)) channels (VGNaCs), according to literature. In this study the effects of oxaliplatin on the peripheral myelinated nerve fibres (PMNFs) were investigated in vitro using the isolated sciatic nerve of the adult rat. The advantage of this nerve-preparation was that stable in amplitude evoked compound action potentials (CAP) were recorded for over 1000min. Incubation of the sciatic nerve fibres in 25, 100 and 500microM oxaliplatin, for 300-700min caused dramatic distortion of the waveform of the CAP, namely broadening the repolarization phase, repetitive firing and afterhyperpolarization (AHP), related to the malfunction of voltage-gated potassium (K(+)) channels (VGKCs). At a concentration of 5microM, oxaliplatin caused broadening of the repolarization phase of the CAP only, while the no observed effect concentration was estimated to be 1microM. These findings are indicative of severe effects of oxaliplatin on the VGKCs. In contrast, the amplitude and the rise-time of the depolarization of the CAP did not change significantly, a clear indication that the VGNaCs of the particular nerve preparation were not affected by oxaliplatin. The effects of oxaliplatin on the PMNFs were similar to those of 4-aminopyridine (4-AP), a classical antagonist of VGKCs. These similarities in the pattern of action between oxaliplatin and 4-AP combined with the fact that the effects of oxaliplatin were more pronounced and developed at lower concentrations suggest that oxaliplatin acts as a potent VGKCs antagonist.
Collapse
Affiliation(s)
- Alexia Kagiava
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, University Campus, Thessaloniki 54 124, Greece
| | | | | | | |
Collapse
|
53
|
Chiu SJ, Chao JI, Lee YJ, Hsu TS. Regulation of gamma-H2AX and securin contribute to apoptosis by oxaliplatin via a p38 mitogen-activated protein kinase-dependent pathway in human colorectal cancer cells. Toxicol Lett 2008; 179:63-70. [PMID: 18499365 DOI: 10.1016/j.toxlet.2008.04.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 03/15/2008] [Accepted: 04/03/2008] [Indexed: 12/27/2022]
Abstract
Oxaliplatin, a chemotherapeutic drug, induces DNA strand breaks leading to apoptosis in colorectal cancer cells. gamma-H2AX is a phosphorylated histone H2AX that can act as a marker of DNA double-strand breaks (DSBs). It has been shown that securin proteins were over-expressed in a variety of cancer cells. However, the roles of gamma-H2AX and securin on the oxaliplatin-induced apoptosis in human colorectal cancer cells remain unclear. Treatment of oxaliplatin (1-10 microM for 6-24h) persistently induced gamma-H2AX formation and inhibited securin protein expression via a time- and concentration-dependent manner in HCT116 securin-wild type colorectal cancer cells. Compared with HCT116 securin-wild type cells, the induction of apoptosis and persistent gamma-H2AX formation by oxaliplatin was reduced in the HCT116 securin-null colorectal cancer cells. Furthermore, the blockage of caspases by specific caspase inhibitors reduced the levels of gamma-H2AX proteins and cytotoxicity but increased securin protein expression in the oxaliplatin-exposed cells. The gene knockdown of H2AX by transfection with a short interfering RNA of H2AX enhanced the oxaliplatin-induced cell death. Interestingly, the phosphorylation of p38 mitogen-activated protein kinase (MAPK) was markedly increased by oxaliplatin. Pre-treatment of a specific p38 MAPK inhibitor SB202190 reduced gamma-H2AX proteins and increased securin protein expression in the oxaliplatin-treated cells. Our findings suggest that p38 MAPK may oppositely regulate securin protein expression and gamma-H2AX formation in the oxaliplatin-induced apoptosis of human colorectal cancer cells.
Collapse
Affiliation(s)
- Shu-Jun Chiu
- Department of Life Science, Tzu Chi University, Hualien 970, Taiwan.
| | | | | | | |
Collapse
|
54
|
Cavaletti G, Miloso M, Nicolini G, Scuteri A, Tredici G. Emerging role of mitogen-activated protein kinases in peripheral neuropathies. J Peripher Nerv Syst 2007; 12:175-194. [PMID: 17868245 DOI: 10.1111/j.1529-8027.2007.00138.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Among the different families of intracellular molecules that can be modulated during cell damage and repair, mitogen-activated protein kinases (MAPKs) are particularly interesting because they are involved in several intracellular pathways activated by injury and regeneration signals. Despite most of the studies have been performed in non-neurological models, recently a causal role for MAPKs has been postulated in central nervous system disorders. However, also in some peripheral neuropathies, MAPK changes can occur and these modifications might be relevant in the pathogenesis of the damage as well as during regeneration and repair. In this review, the current knowledge on the role of MAPKs in peripheral neuropathies will be discussed.
Collapse
Affiliation(s)
- Guido Cavaletti
- Department of Neurosciences and Biomedical Technologies, University of Milano Bicocca, Monza, Italy.
| | | | | | | | | |
Collapse
|
55
|
Morís G, Ribacoba R, González C. Delayed posterior encephalopathy syndrome following chemotherapy with oxaliplatin and gemcitabine. J Neurol 2007; 254:534-5. [PMID: 17361348 DOI: 10.1007/s00415-006-0273-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 03/05/2006] [Accepted: 03/07/2006] [Indexed: 10/23/2022]
|
56
|
Donzelli E, Salvadè A, Mimo P, Viganò M, Morrone M, Papagna R, Carini F, Zaopo A, Miloso M, Baldoni M, Tredici G. Mesenchymal stem cells cultured on a collagen scaffold: In vitro osteogenic differentiation. Arch Oral Biol 2007; 52:64-73. [PMID: 17049335 DOI: 10.1016/j.archoralbio.2006.07.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Revised: 04/21/2006] [Accepted: 07/18/2006] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Management of periodontal defects has always been a challenge in clinical periodontics. Recently mesenchymal stem cells (MSC) have been proposed for tissue regeneration in periodontal disease and repair of large bone defects. Bone regeneration has to be supported by a scaffold which has to be biocompatible, biodegradable, and able to support cell growth and differentiation. The aim of this study was to evaluate osteogenic differentiation of MSC seeded on a collagen scaffold. DESIGN MSC were obtained from adult rat bone marrow, expanded and cultured in plastic dishes or seeded in a collagen scaffold (Gingistat). MSC were induced towards osteogenic differentiation using osteogenic supplements. Cell differentiation and calcium deposits were evaluated by immunoblotting, immunohistochemistry, histochemical techniques, enzymatic activity assay, and SEM-EDX analysis. Biomaterial in vitro degradation was evaluated by measuring mass reduction after incubation in culture medium. RESULTS Rat MSC osteogenic differentiation was demonstrated by osteopontin and osteocalcin expression and an increase in alkaline phosphatase activity. MSC were distributed homogeneously in the collagen scaffold. Nodular aggregates and alizarin red stained calcium deposits were observed in MSC induced towards osteogenic differentiation cultured in dishes or seeded in the collagen scaffold. SEM-EDX analysis demonstrated that calcium co-localized with phosphorous. The biomaterial in vitro degraded in 4-5 weeks. CONCLUSIONS MSC from bone marrow differentiate towards osteogenic lineage, representing a suitable cell source for bone formation in periodontal regeneration. Gingistat collagen scaffold supports MSC distribution and differentiation, but its short degradation time may be a limitation for a future application in bone tissue regeneration.
Collapse
Affiliation(s)
- E Donzelli
- Dipartimento di Neuroscienze e Tecnologie Biomediche, Università degli Studi di Milano-Bicocca, Via Cadore 48, 20052 Monza, MI, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Rabik CA, Dolan ME. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev 2006; 33:9-23. [PMID: 17084534 PMCID: PMC1855222 DOI: 10.1016/j.ctrv.2006.09.006] [Citation(s) in RCA: 1191] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2006] [Revised: 09/06/2006] [Accepted: 09/11/2006] [Indexed: 12/17/2022]
Abstract
Platinating agents, including cisplatin, carboplatin, and oxaliplatin, have been used clinically for nearly 30years as part of the treatment of many types of cancers, including head and neck, testicular, ovarian, cervical, lung, colorectal and relapsed lymphoma. The cytotoxic lesion of platinating agents is thought to be the platinum intrastrand crosslink that forms on DNA, although treatment activates a number of signal transduction pathways. Treatment with these agents is characterized by resistance, both acquired and intrinsic. This resistance can be caused by a number of cellular adaptations, including reduced uptake, inactivation by glutathione and other anti-oxidants, and increased levels of DNA repair or DNA tolerance. Here we investigate the pathways that treatment with platinating agents activate, the mechanisms of resistance, potential candidate genes involved in the development of resistance, and associated clinical toxicities. Although the purpose of this review is to provide an overview of cisplatin, carboplatin, and oxaliplatin, we have focused primarily on preclinical data that has clinical relevance generated over the past five years.
Collapse
Affiliation(s)
- Cara A Rabik
- Department of Medicine, Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, 5841 S. Maryland Avenue, Box MC2115, Section of Hem-Onc, Chicago, IL 60637, United States
| | | |
Collapse
|
58
|
Florea AM, Büsselberg D. Occurrence, use and potential toxic effects of metals and metal compounds. Biometals 2006; 19:419-27. [PMID: 16841251 DOI: 10.1007/s10534-005-4451-x] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Accepted: 10/26/2005] [Indexed: 11/26/2022]
Abstract
Metals and metal compounds are constituents of our natural environment. Their distribution depends on the existence of natural sources (e.g. volcanoes or erosion) and their use in human's activity. They are transformed naturally (e.g. by bacterial activity) with formation of organic species that influence their mobility and accumulation in abiotic as well as biotic systems. Up to date metal species are released into the environment questioning their influence on human health. Due to their widespread use in human activities such as industry, agriculture and even as medicine (e.g. As, Se, Pt), numerous health risks may be associated with exposure to these substances. Different reports on metal intoxication are documented and studies especially on neurotoxicity, genotoxicity, or carcinogenicity, are previously published in numerous articles. This mini-review gives an overview on the use and the actions of selected metal species of actual scientific concern, with a focus on neuronal cells.
Collapse
Affiliation(s)
- Ana-Maria Florea
- Institut für Physiologie Universitätsklinium Essen, Universität Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | | |
Collapse
|
59
|
Ta LE, Espeset L, Podratz J, Windebank AJ. Neurotoxicity of oxaliplatin and cisplatin for dorsal root ganglion neurons correlates with platinum-DNA binding. Neurotoxicology 2006; 27:992-1002. [PMID: 16797073 DOI: 10.1016/j.neuro.2006.04.010] [Citation(s) in RCA: 253] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 04/24/2006] [Accepted: 04/25/2006] [Indexed: 12/27/2022]
Abstract
Cisplatin has been in use for 40 years, primarily for treatment of ovarian and testicular cancer. Oxaliplatin is the only effective treatment for metastatic colorectal cancer. Neurotoxicity occurs in up to 30% of patients and is dose-limiting for both drugs. The neuropathy is characterized by selective sensory loss in the extremities. Cisplatin treatment is associated with high levels of Pt-DNA binding and apoptosis of dorsal root ganglion (DRG) neurons. In this study, we directly compared the effects of oxaliplatin on DRG in vitro. Compared with cisplatin, oxaliplatin formed fewer Pt-DNA adducts following 6, 12, 24, and 48h (0.007ng Pt/mug DNA, 0.012ng/microg, 0.011ng/microg, 0.011ng/microg versus 0.014ng/microg, 0.022ng/microg, 0.041ng/microg, 0.030ng/microg), respectively. These findings closely correlated with data on cell survival where equimolar concentrations of oxaliplatin induced less cell death than cisplatin. Oxaliplatin-induced DRG death was associated with the morphological characteristics of apoptosis defined by 4'-6-diamidino-2-phenylindole and annexin/propidium iodide staining. Death was completely inhibited by the caspase inhibitor z-VAD-fmk. Our results demonstrate that both compounds cause apoptosis of DRG neurons but compared to cisplatin, oxaliplatin forms fewer Pt-DNA adducts and is less neurotoxic to DRG neurons in vitro.
Collapse
Affiliation(s)
- Lauren E Ta
- Molecular Neuroscience Program and Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
60
|
Coecke S, Eskes C, Gartlon J, Kinsner A, Price A, van Vliet E, Prieto P, Boveri M, Bremer S, Adler S, Pellizzer C, Wendel A, Hartung T. The value of alternative testing for neurotoxicity in the context of regulatory needs. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2006; 21:153-67. [PMID: 21783653 DOI: 10.1016/j.etap.2005.07.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Detection and characterisation of chemical-induced toxic effects in the central and peripheral nervous system represent a major challenge for employing newly developed technologies in the field of neurotoxicology. Precise cellular predictive test batteries for chemical-induced neurotoxicity are increasingly important for regulatory decision making, but also the most efficient way to keep costs and time of testing within a reasonable margin. Current in vivo test methods are based on behavioural and sensory perturbations coupled with routine histopathological investigations. In spite of the empirical usefulness of these tests, they are not always sensitive enough and often, they do not provide information that facilitates a detailed understanding of potential mechanisms of toxicity, thus enabling predictions. In general, such in vivo tests are unsuitable for screening large number of agents. One way to meet the need for more powerful and comprehensive tests via an extended scientific basis is to study neurotoxicity in specific cell types of the brain and to derive generalised mechanisms of action of the toxicants from such series of experiments. Additionally, toxicokinetic models are to be developed in order to give a rough account for the whole absorption, distribution, metabolism, excretion (ADME) process including the blood-brain barrier (BBB). Therefore, an intensive search for the development of alternative methods using animal and human-based in vitro and in silico models for neurotoxic hazard assessment is appropriate. In particular, neurotoxicology represents one of the major challenges to the development of in vitro systems, as it has to account also for heterogeneous cell interactions of the brain which require new biochemical, biotechnological and electrophysiological profiling methods for reliable alternative ways with a high throughput.
Collapse
Affiliation(s)
- Sandra Coecke
- European Centre for the Validation of Alternative Methods (ECVAM), Institute for Health & Consumer Protection, European Commission Joint Research Centre, Ispra (VA), Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Villa D, Miloso M, Nicolini G, Rigolio R, Villa A, Cavaletti G, Tredici G. Low-dose cisplatin protects human neuroblastoma SH-SY5Y cells from paclitaxel-induced apoptosis. Mol Cancer Ther 2005; 4:1439-1447. [PMID: 16170037 DOI: 10.1158/1535-7163.mct-05-0053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Combined anticancer therapy using platinum compounds and antitubulins has increased the risk of neurotoxicity. However, the combination of low-dose cisplatin (CDDP) with toxic doses of paclitaxel significantly reduces cellular death in a human neuroblastoma SH-SY5Y cell line. To analyze the mechanisms of this protection, we evaluated various signaling molecules possibly involved in apoptosis and some relevant cell cycle regulatory proteins. CDDP does not interfere with the tubulin-stabilizing action of paclitaxel. The evaluation of molecular pathways involved in apoptosis indicates that the Bcl-2 but not the caspases may be involved in the CDDP protection of paclitaxel-induced apoptosis. The increase in p53 protein and its nuclear accumulation suggests a possible involvement of p53 in CDDP protection. The use of the chemical inhibitor of p53, pifithrin alpha, excluded this possibility. The study of cyclins and the flow cytometric analysis (fluorescence-activated cell sorting) suggest that CDDP exerts a protective action by blocking cells early in the cell cycle. The determination of the mitotic index indicates that CDDP prevents cells from reaching the mitosis. We concluded that low doses of CDDP are protective against toxic doses of paclitaxel and that the possible mechanism of this protection is that the CDDP prevents human neuroblastoma SH-SY5Y cells from achieving mitosis.
Collapse
Affiliation(s)
- Daniela Villa
- Dipartimento di Neuroscienze e Tecnologie Biomediche, Facoltà di Medicina e Chirurgia, Monza, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|