1
|
Abdelmoez WA. Evaluation of histological and ultrastructural changes provoked by prenatal tramadol on postnatal cortical cerebellar neuronal development in rats: possible implication of Ki67, GFAP and MicroRNA-7/P53 signalling trajectories. J Mol Histol 2024; 55:279-301. [PMID: 38639812 PMCID: PMC11102883 DOI: 10.1007/s10735-024-10189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
Tramadol is a novel centrally acting analgesic. Despite, its implementation during pregnancy may impair neuronal survival and synaptic development in neonatal cerebella. The current investigation assessed the histological and ultrastructural alterations in postnatal cortical cerebellar neuronal development induced by prenatal tramadol. 30 offsprings were divided to control group I: fifteen pups born to mothers given saline from D10 till D21 of gestation. Tramadol-treated group II: fifteen pups born to mothers received tramadol HCL (50 mg/kg/day) from D10 till D21 of gestation. Pups were categorized into three subgroups (a, b, and c) and offered for sacrifice on the seventh, fourteenth and twenty-first post-natal days. Light microscopic examination revealed the overcrowding and signs of red degeneration affecting purkinje cell layer. Neurodegenerative signs of both purkinje and granule cell neurons were also confirmed by TEM in form of chromatin condensation, dilated Golgi channels, disrupted endoplasmic reticulum, marked infolding of the nuclear envelope and decrease in granule cell precursors. In addition, the astrocytic processes and terminal nerve axons appeared with different degrees of demyelination and decreased number of oligodendrocytes and degenerated mitochondria. Furthermore, group II exhibited an increase in P53 immune expression. The area percentage of apoptotic cells detected by TUNEL assay was significantly increased. Besides to the significant decrease of Ki67 immunoreactivity in the stem neuronal cell progenitors. Quantitative PCR results showed a significant decline in micro RNA7 gene expression in tramadol treated groups resulting in affection of multiple target genes in P53 signaling pathways, improper cortical size and defect in neuronal development.
Collapse
Affiliation(s)
- Walaa Adel Abdelmoez
- Department of Anatomy and Embryology, Faculty of Medicine, Ain-Shams University, Postal Code: 11591, Abbassia, Cairo, Egypt.
| |
Collapse
|
2
|
Zarewa SA, Binobaid L, Sulaiman AAA, Sobeai HMA, Alotaibi M, Alhoshani A, Isab AA. Synthesis, Characterization, and Anticancer Activity of Phosphanegold(i) Complexes of 3-Thiosemicarbano-butan-2-one Oxime. Biomedicines 2023; 11:2512. [PMID: 37760953 PMCID: PMC10525815 DOI: 10.3390/biomedicines11092512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/23/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Four novel phosphanegold(I) complexes of the type [Au(PR3)(DMT)].PF6 (1-4) were synthesized from 3-Thiosemicarbano-butan-2-one oxime ligand (TBO) and precursors [Au(PR3)Cl], (where R = methyl (1), ethyl (2), tert-butyl (3), and phenyl (4)). The resulting complexes were characterized by elemental analyses and melting point as well as various spectroscopic techniques, including FTIR and (1H, 13C, and 31P) NMR spectroscopy. The spectroscopic data confirmed the coordination of TBO ligands to phosphanegold(I) moiety. The solution chemistry of complexes 1-4 indicated their stability in both dimethyl sulfoxide (DMSO) and a mixture of EtOH:H2O (1:1). In vitro cytotoxicity of the complexes was evaluated relative to cisplatin using an MTT assay against three different cancer cell lines: HCT116 (human colon cancer), MDA-MB-231 (human breast cancer), and B16 (murine skin cancer). Complexes 2, 3, and 4 exhibited significant cytotoxic effects against all tested cancer cell lines and showed significantly higher activity than cisplatin. To elucidate the mechanism underlying the cytotoxic effects of the phosphanegold(I) TBO complexes, various assays were employed, including mitochondrial membrane potential, ROS production, and gene expression analyses. The data obtained suggest that complex 2 exerts potent anticancer activity against breast cancer cells (MDA-MB-231) through the induction of oxidative stress, mitochondrial dysfunction, and apoptosis. Gene expression analyses showed an increase in the activity of the proapoptotic gene caspase-3 and a reduction in the activity of the antiapoptotic gene BCL-xL, which supported the findings that apoptosis had occurred.
Collapse
Affiliation(s)
- Sani A. Zarewa
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia;
| | - Lama Binobaid
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Adam A. A. Sulaiman
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia;
- Core Research Facilities (CRF), King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Homood M. As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Moureq Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Anvarhusein A. Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia;
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| |
Collapse
|
3
|
Mokhtar HEL, Hulail MAE, Mahmoud SM, Yousef DM. Impact of cisplatin administration on cerebellar cortical structure and locomotor activity of infantile and juvenile albino rats: the role of oxidative stress. Anat Sci Int 2021; 97:30-47. [PMID: 34386931 DOI: 10.1007/s12565-021-00624-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/01/2021] [Indexed: 11/30/2022]
Abstract
The central neurotoxicity of cisplatin (CisPt) has always raised questions especially during development, but few studies are available. Hence, this work was designed to assess the CisPt's impacts on the postnatal rat cerebellum via evaluation of locomotor activity, histological and immunohistochemical studies, and to focus on cerebellar oxidative stress-related alterations. Eighty newborn pups were divided into 2 equal experimental groups: the control group was kept without any treatment and CisPt-treated group received a single subcutaneous injection of CisPt (5 μg /g b.w.) in their nape at PD10. Ten rats at PD11, PD17, and PD30 ages were weighed, then deeply anesthetized and sacrificed. For locomotor assessment, 20 pups were divided equally into control and CisPt-treated groups and tested at PD11-13, PD15-17, and PD28-30 ages. CisPt-treated rats suffered from decreased motor activity and showed decreased body and cerebellar weights, reduced levels of enzymatic antioxidants (SOD and CAT), and non-enzymatic antioxidant defense (GSH), and increase of lipid peroxidation marker (MDA). Histopathologically, CisPt sowed deleterious changes within cerebellar cortical layers in the form of vacuolations, decreased thickness, and hemorrhage (in PD17), while Purkinje cells exhibited profound degenerative changes in the form of swelling, disrupted arrangement, distortion, and nuclear shrinkage. In CisPt-treated rats, GFAP demonstrated upregulated, hypertrophied, and branched Bergmann glial fibers and reactive astrogliosis. Immuno-localization of Ki-67-positive cells revealed defective migration associated with decreased proliferation in early ages in addition to glial proliferation in PD30. In conclusion, CisPt causes oxidative stress-related deleterious effects on structure of developing cerebellar cortex and affects locomotor activity.
Collapse
Affiliation(s)
- Hanan E L Mokhtar
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohey A E Hulail
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samar Mortada Mahmoud
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Doaa Mohammed Yousef
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
4
|
Astesana V, Faris P, Ferrari B, Siciliani S, Lim D, Biggiogera M, De Pascali SA, Fanizzi FP, Roda E, Moccia F, Bottone MG. [Pt(O,O'-acac)(γ-acac)(DMS)]: Alternative Strategies to Overcome Cisplatin-Induced Side Effects and Resistance in T98G Glioma Cells. Cell Mol Neurobiol 2021; 41:563-587. [PMID: 32430779 PMCID: PMC11448674 DOI: 10.1007/s10571-020-00873-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (CDDP) is one of the most effective chemotherapeutic agents, used for the treatment of diverse tumors, including neuroblastoma and glioblastoma. CDDP induces cell death through different apoptotic pathways. Despite its clinical benefits, CDDP causes several side effects and drug resistance.[Pt(O,O'-acac)(γ-acac)(DMS)], namely PtAcacDMS, a new platinum(II) complex containing two acetylacetonate (acac) and a dimethylsulphide (DMS) in the coordination sphere of metal, has been recently synthesized and showed 100 times higher cytotoxicity than CDDP. Additionally, PtAcacDMS was associated to a decreased neurotoxicity in developing rat central nervous system, also displaying great antitumor and antiangiogenic activity both in vivo and in vitro. Thus, based on the knowledge that several chemotherapeutics induce cancer cell death through an aberrant increase in [Ca2+]i, in the present in vitro study we compared CDDP and PtAcacDMS effects on apoptosis and intracellular Ca2+ dynamics in human glioblastoma T98G cells, applying a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, electron microscopy, Western blotting, qRT-PCR, and epifluorescent Ca2+ imaging. The results confirmed that (i) platinum compounds may induce cell death through an aberrant increase in [Ca2+]i and (ii) PtAcacDMS exerted stronger cytotoxic effect than CDDP, associated to a larger increase in resting [Ca2+]i. These findings corroborate the use of PtAcacDMS as a promising approach to improve Pt-based chemotherapy against gliomas, either by inducing a chemosensitization or reducing chemoresistance in cell lineages resilient to CDDP treatment.
Collapse
Affiliation(s)
- Valentina Astesana
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, 27100, Pavia, Italy
- Department of Biology, Cihan University-Erbil, Erbil, 44001, Iraq
| | - Beatrice Ferrari
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Stella Siciliani
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, 28100, Novara, Italy
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Sandra Angelica De Pascali
- General and Inorganic Chemistry Laboratory, Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Francesco Paolo Fanizzi
- General and Inorganic Chemistry Laboratory, Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Elisa Roda
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, via Ferrata 9, 27100, Pavia, Italy
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, ICS Maugeri Spa, IRCCS Pavia, Pavia, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, 27100, Pavia, Italy
| | - Maria Grazia Bottone
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology ''L. Spallanzani'', University of Pavia, via Ferrata 9, 27100, Pavia, Italy.
| |
Collapse
|
5
|
Ferrari B, Roda E, Priori EC, De Luca F, Facoetti A, Ravera M, Brandalise F, Locatelli CA, Rossi P, Bottone MG. A New Platinum-Based Prodrug Candidate for Chemotherapy and Its Synergistic Effect With Hadrontherapy: Novel Strategy to Treat Glioblastoma. Front Neurosci 2021; 15:589906. [PMID: 33828444 PMCID: PMC8019820 DOI: 10.3389/fnins.2021.589906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common tumor of the central nervous system. Current therapies, often associated with severe side effects, are inefficacious to contrast the GBM relapsing forms. In trying to overcome these drawbacks, (OC-6-44)-acetatodiamminedichlorido(2-(2-propynyl)octanoato)platinum(IV), also called Pt(IV)Ac-POA, has been recently synthesized. This new prodrug bearing as axial ligand (2-propynyl)octanoic acid (POA), a histone deacetylase inhibitor, has a higher activity due to (i) its high cellular accumulation by virtue of its high lipophilicity and (ii) the inhibition of histone deacetylase, which leads to the increased exposure of nuclear DNA, permitting higher platination and promoting cancer cell death. In the present study, we investigated the effects induced by Pt(IV)Ac-POA and its potential antitumor activity in human U251 glioblastoma cell line using a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, TEM, and Western blotting analyses. In addition, the synergistic effect of Pt(IV)Ac-POA associated with the innovative oncological hadrontherapy with carbon ions was investigated, with the aim to identify the most efficient anticancer treatment combination. Our in vitro data demonstrated that Pt(IV)Ac-POA is able to induce cell death, through different pathways, at concentrations lower than those tested for other platinum analogs. In particular, an enduring Pt(IV)Ac-POA antitumor effect, persisting in long-term treatment, was demonstrated. Interestingly, this effect was further amplified by the combined exposure to carbon ion radiation. In conclusion, Pt(IV)Ac-POA represents a promising prodrug to be incorporated into the treatment regimen for GBM. Moreover, the synergistic efficacy of the combined protocol using chemotherapeutic Pt(IV)Ac-POA followed by carbon ion radiation may represent a promising approach, which may overcome some typical limitations of conventional therapeutic protocols for GBM treatment.
Collapse
Affiliation(s)
- Beatrice Ferrari
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy.,Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Fabrizio De Luca
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Angelica Facoetti
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Mauro Ravera
- Department of Sciences and Technological Innovation (DiSIT), University of Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
6
|
Gattoni G, Bernocchi G. Calcium-Binding Proteins in the Nervous System during Hibernation: Neuroprotective Strategies in Hypometabolic Conditions? Int J Mol Sci 2019; 20:E2364. [PMID: 31086053 PMCID: PMC6540041 DOI: 10.3390/ijms20092364] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/06/2019] [Accepted: 05/11/2019] [Indexed: 02/07/2023] Open
Abstract
Calcium-binding proteins (CBPs) can influence and react to Ca2+ transients and modulate the activity of proteins involved in both maintaining homeostatic conditions and protecting cells in harsh environmental conditions. Hibernation is a strategy that evolved in vertebrate and invertebrate species to survive in cold environments; it relies on molecular, cellular, and behavioral adaptations guided by the neuroendocrine system that together ensure unmatched tolerance to hypothermia, hypometabolism, and hypoxia. Therefore, hibernation is a useful model to study molecular neuroprotective adaptations to extreme conditions, and can reveal useful applications to human pathological conditions. In this review, we describe the known changes in Ca2+-signaling and the detection and activity of CBPs in the nervous system of vertebrate and invertebrate models during hibernation, focusing on cytosolic Ca2+ buffers and calmodulin. Then, we discuss these findings in the context of the neuroprotective and neural plasticity mechanisms in the central nervous system: in particular, those associated with cytoskeletal proteins. Finally, we compare the expression of CBPs in the hibernating nervous system with two different conditions of neurodegeneration, i.e., platinum-induced neurotoxicity and Alzheimer's disease, to highlight the similarities and differences and demonstrate the potential of hibernation to shed light into part of the molecular mechanisms behind neurodegenerative diseases.
Collapse
Affiliation(s)
- Giacomo Gattoni
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Graziella Bernocchi
- Former Full Professor of Zoology, Neurogenesis and Comparative Neuromorphology, (Residence address) Viale Matteotti 73, I-27100 Pavia, Italy.
| |
Collapse
|
7
|
Kursunluoglu G, Taskiran D, Kayali HA. The Investigation of the Antitumor Agent Toxicity and Capsaicin Effect on the Electron Transport Chain Enzymes, Catalase Activities and Lipid Peroxidation Levels in Lung, Heart and Brain Tissues of Rats. Molecules 2018; 23:E3267. [PMID: 30544766 PMCID: PMC6320812 DOI: 10.3390/molecules23123267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/12/2018] [Accepted: 11/19/2018] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is one of the most active cytotoxic agents in cancer treatment. To clarify the interaction with mitochondria, we hypothesize that the activities of mitochondrial electron transport chain (ETC) enzymes succinate dehydrogenase (SDH) and cytochrome c oxidase (COX), nucleotide levels, as well as levels of catalase (CAT) enzyme and membrane lipid peroxidation (LPO) can be affected by cisplatin. There was a significant decrease of both SDH and COX activities in the lung, heart, and brain tissues at the 1st day after cisplatin exposure, and the observed decreased levels of adenosine triphosphate (ATP) and adenosine diphosphate (ADP) in comparison with the control could be because of cisplatin-induced mitochondrial dysfunction. The investigations suggested that cisplatin inhibits SDH, COX, and ATP synthase. The higher LPO level in the studied tissues after 1 and 4 days post-exposure to cisplatin compared to control can be inferred to be a result of elevated electron leakage from the ETC, and reactive oxygen species (ROS) can lead to wide-ranging tissue damage such as membrane lipid damage. Consequently, it was observed that capsaicin may have a possible protective effect on ETC impairment caused by cisplatin. The activities of SDH and COX were higher in heart and brain exposed to cisplatin + capsaicin compared to cisplatin groups, while LPO levels were lower. The investigated results in the cisplatin + capsaicin groups suggested that the antioxidant capacity of capsaicin scavenges ROS and prevents membrane destruction.
Collapse
Affiliation(s)
- Gizem Kursunluoglu
- Izmir Biomedicine and Genome Center (IBG), İzmir 35340, Turkey.
- Department of Chemistry, The Graduate School of Natural and Applied Sciences, Dokuz Eylul University, İzmir 35160, Turkey.
| | - Dilek Taskiran
- Department of Physiology, Ege University School of Medicine, İzmir 35100, Turkey.
| | - Hulya Ayar Kayali
- Izmir Biomedicine and Genome Center (IBG), İzmir 35340, Turkey.
- Department of Chemistry, Division of Biochemistry, Faculty of Science, Dokuz Eylul University, İzmir 35160, Turkey.
| |
Collapse
|
8
|
Abstract
Survival rates of children with cancer are steadily increasing. This urges our attention to neurocognitive and psychiatric outcomes, as these can markedly influence the quality of life of these children. Neurobehavioral morbidity in childhood cancer survivors affects diverse aspects of cognitive function, which can include attention, memory, processing speed, intellect, academic achievement, and emotional health. Reasons for neurobehavioral morbidity are multiple with one major contributor being chemotherapy-induced central nervous system (CNS) toxicity. Clinical studies investigating the effects of chemotherapy on the CNS in children with cancer have reported causative associations with the development of leukoencephalopathies as well as smaller regional grey and white matter volumes, which have been found to correlate with neurocognitive deficits.Preclinical work has provided compelling evidence that chemotherapy drugs are potent neuro- and gliotoxins in vitro and in vivo and can cause brain injury via excitotoxic and apoptotic mechanisms. Furthermore, chemotherapy triggers DNA (deoxyribonucleic acid) damage directly or through increased oxidative stress. It can shorten telomeres and accelerate cell aging, cause cytokine deregulation, inhibit hippocampal neurogenesis, and reduce brain vascularization and blood flow. These mechanisms, when allowed to operate on the developing brain of a child, have high potential to not only cause brain injury, but also alter crucial developmental events, such as myelination, synaptogenesis, neurogenesis, cortical thinning, and formation of neuronal networks.This short review summarizes key publications describing neurotoxicity of chemotherapy in pediatric cancers and potential underlying pathomechanisms.
Collapse
Affiliation(s)
- Chrysanthy Ikonomidou
- Department of Neurology, Section of Child Neurology, University of Wisconsin Madison, Madison, WI, 53705, USA.
| |
Collapse
|
9
|
Martí J, Molina V, Santa-Cruz MC, Hervás JP. Developmental Injury to the Cerebellar Cortex Following Hydroxyurea Treatment in Early Postnatal Life: An Immunohistochemical and Electron Microscopic Study. Neurotox Res 2016; 31:187-203. [PMID: 27601242 DOI: 10.1007/s12640-016-9666-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/17/2016] [Accepted: 08/30/2016] [Indexed: 11/30/2022]
Abstract
Postnatal development of the cerebellar cortex was studied in rats administered with a single dose (2 mg/g) of the cytotoxic agent hydroxyurea (HU) on postnatal day (P) 9 and collected at appropriate times ranging from 6 h to 45 days. Quantification of several parameters such as the density of pyknotic, mitotic, BrdU-positive, and vimentin-stained cells revealed that HU compromises the survival of the external granular layer (EGL) cells. Moreover, vimentin immunocytochemistry revealed overexpression and thicker immunoreactive glial processes in HU-treated rats. On the other hand, we also show that HU leads to the activation of apoptotic cellular events, resulting in a substantial number of dying EGL cells, as revealed by TUNEL staining and at the electron microscope level. Additionally, we quantified several features of the cerebellar cortex of rats exposed to HU in early postnatal life and collected in adulthood. Data analysis indicated that the analyzed parameters were less pronounced in rats administered with this agent. Moreover, we observed several alterations in the cerebellar cortex cytoarchitecture of rats injected with HU. Anomalies included ectopic placement of Purkinje cells and abnormities in the dendritic arbor of these macroneurons. Ectopic granule cells were also found in the molecular layer. These findings provide a clue for investigating the mechanisms of HU-induced toxicity during the development of the central nervous system. Our results also suggest that it is essential to avoid underestimating the adverse effects of this hydroxylated analog of urea when administered during early postnatal life.
Collapse
Affiliation(s)
- Joaquín Martí
- Unidad de Citología e Histología, Facultad de Biociencias, Universidad Autónoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.
| | - Vanesa Molina
- Unidad de Citología e Histología, Facultad de Biociencias, Universidad Autónoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - M C Santa-Cruz
- Unidad de Citología e Histología, Facultad de Biociencias, Universidad Autónoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - José P Hervás
- Unidad de Citología e Histología, Facultad de Biociencias, Universidad Autónoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| |
Collapse
|
10
|
Sleurs C, Deprez S, Emsell L, Lemiere J, Uyttebroeck A. Chemotherapy-induced neurotoxicity in pediatric solid non-CNS tumor patients: An update on current state of research and recommended future directions. Crit Rev Oncol Hematol 2016; 103:37-48. [PMID: 27233118 DOI: 10.1016/j.critrevonc.2016.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 03/21/2016] [Accepted: 05/10/2016] [Indexed: 11/26/2022] Open
Abstract
Neurocognitive sequelae are known to be induced by cranial radiotherapy and central-nervous-system-directed chemotherapy in childhood Acute Lymphoblastic Leukemia (ALL) and brain tumor patients. However, less evidence exists for solid non-CNS-tumor patients. To get a better understanding of the potential neurotoxic mechanisms of non-CNS-directed chemotherapy during childhood, we performed a comprehensive literature review of this topic. Here, we provide an overview of preclinical and clinical studies investigating neurotoxicity associated with chemotherapy in the treatment of pediatric solid non-CNS tumors. Research to date suggests that chemotherapy has deleterious biological and psychological effects, with animal studies demonstrating histological evidence for neurotoxic effects of specific agents and human studies demonstrating acute neurotoxicity. Although the existing literature suggests potential neurotoxicity throughout neurodevelopment, research into the long-term neurocognitive sequelae in survivors of non-CNS cancers remains limited. Therefore, we stress the critical need for neurodevelopmental focused research in children who are treated for solid non-CNS tumors, since they are at risk for potential neurocognitive impairment.
Collapse
Affiliation(s)
- Charlotte Sleurs
- Department of Pediatric Hematology and Oncology, University Hospital Leuven, Leuven, Belgium; Department of Radiology, University Hospital Leuven, Leuven, Belgium.
| | - Sabine Deprez
- Department of Radiology, University Hospital Leuven, Leuven, Belgium
| | - Louise Emsell
- Department of Radiology, University Hospital Leuven, Leuven, Belgium
| | - Jurgen Lemiere
- Department of Pediatric Hematology and Oncology, University Hospital Leuven, Leuven, Belgium; Department of Child & Adolescent Psychiatry, University Hospital Leuven, Leuven, Belgium
| | - Anne Uyttebroeck
- Department of Pediatric Hematology and Oncology, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Grimaldi M, Santin G, Insolia V, Dal Bo V, Piccolini VM, Veneroni P, Barni S, Verri M, De Pascali SA, Fanizzi FP, Bernocchi G, Bottone MG. [Pt(O,O'-acac)(γ-acac)(DMS)] versus cisplatin: apoptotic effects in B50 neuroblastoma cells. Histochem Cell Biol 2016; 145:587-601. [PMID: 26748644 DOI: 10.1007/s00418-015-1396-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2015] [Indexed: 12/19/2022]
Abstract
Cisplatin is one of the most active chemotherapeutic agents used in the treatment of childhood and adult malignancies. Cisplatin induces cell death through different pathways. Despite its effectiveness, the continued clinical use of cisplatin is limited by onset of severe side effects (nephrotoxicity, ototoxicity and neurotoxicity) and drug resistance. Therefore, one of the main experimental oncology purpose is related to the search for new platinum-based drugs to create different types of adducts or more specific and effective subcellular targets. Thus, [Pt(O,O'-acac)(γ-acac)(DMS)], which reacts preferentially with protein thiols or thioether, was synthesized. In our research, different approaches were used to compare cisplatin and [Pt(O,O'-acac)(γ-acac)(DMS)] effects in B50 rat neuroblastoma cells. Our results, using immunocytochemical, cytometric and morphological techniques, showed that these compounds exert a cytostatic action and activate apoptosis with different pathways. Long-term effects demonstrated that [Pt(O,O'-acac)(γ-acac)(DMS)] exerts cytotoxic effects in neuronal B50 cell line not inducing drug resistance. Analysis was performed both to compare the ability of these platinum compounds to induce cell death and to investigate the intracellular mechanisms at the basis of their cytotoxicity.
Collapse
Affiliation(s)
- Maddalena Grimaldi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Giada Santin
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Violetta Insolia
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Veronica Dal Bo
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Valeria Maria Piccolini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Paola Veneroni
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Sergio Barni
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Sandra Angelica De Pascali
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Graziella Bernocchi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy.
| |
Collapse
|
12
|
Bernocchi G, Fanizzi FP, De Pascali SA, Piccolini VM, Gasperini C, Insolia V, Bottone MG. Neurotoxic Effects of Platinum Compounds: Studies in vivo on Intracellular Calcium Homeostasis in the Immature Central Nervous System. TOXICS 2015; 3:224-248. [PMID: 29056659 PMCID: PMC5634691 DOI: 10.3390/toxics3020224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/09/2015] [Accepted: 06/09/2015] [Indexed: 02/08/2023]
Abstract
Platinum compounds cause significant clinical neurotoxicity. Several studies highlight neurological complications especially in paediatric oncology patients with Central Nervous System (CNS) and non-CNS malignancies. To understand the toxicity mechanisms of platinum drugs at cellular and molecular levels in the immature brain, which appears more vulnerable to injury than in the adult one, we compared the effects in vivo of the most used platinum compounds, i.e., cisdichlorodiammineplatinum (cisplatin, cisPt), and the new [Pt(O,O′-acac)(γ-acac)(DMS)] (PtAcacDMS). As models of developing brain areas, we have chosen the cerebellum and hippocampus dentate gyrus. Both areas show the neurogenesis events, from proliferation to differentiation and synaptogenesis, and therefore allow comparing the action of platinum compounds with DNA and non-DNA targets. Here, we focused on the changes in the intracellular calcium homeostasis within CNS architecture, using two immunohistochemical markers, the calcium buffer protein Calbindin and Plasma Membrane Calcium ATPase. From the comparison of the cisPt and PtAcacDMS effects, it emerges how essential the equilibrium and synergy between CB and PMCA1 is or how important the presence of at least one of them is to warrant the morphology and function of nervous tissue and limit neuroarchitecture damages, depending on the peculiar and intrinsic properties of the developing CNS areas.
Collapse
Affiliation(s)
- Graziella Bernocchi
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Francesco P Fanizzi
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Università del Salento, via provinciale Lecce-Monteroni centro Ecotekne, 73100 Lecce, Italy.
| | - Sandra A De Pascali
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Università del Salento, via provinciale Lecce-Monteroni centro Ecotekne, 73100 Lecce, Italy.
| | - Valeria M Piccolini
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Caterina Gasperini
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Violetta Insolia
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Maria Grazia Bottone
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| |
Collapse
|
13
|
Tundo GR, Sbardella D, De Pascali SA, Ciaccio C, Coletta M, Fanizzi FP, Marini S. Novel Platinum(II) compounds modulate insulin-degrading enzyme activity and induce cell death in neuroblastoma cells. J Biol Inorg Chem 2015; 20:101-108. [PMID: 25450414 DOI: 10.1007/s00775-014-1217-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/02/2014] [Indexed: 01/12/2023]
Abstract
The properties of three novel Platinum(II) compounds toward the insulin-degrading enzyme (IDE) enzymatic activity have been investigated under physiological conditions. The rationale of this study resides on previous observations that these compounds, specifically designed and synthesized by some of us, induce apoptosis in various cancer cell lines, whereas IDE has been proposed as a putative oncogene involved in neuroblastoma onset and progression. Two of these compounds, namely [PtCl(O,O'-acac)(DMSO)] and [Pt(O,O'-acac)(γ-acac)(DMS)], display a modulatory behavior, wherefore activation or inhibition of IDE activity occurs over different concentration ranges (suggesting the existence of two binding sites on the enzyme). On the other hand, [Pt(O,O'-acac)(γ-acac)(DMSO)] shows a typical competitive inhibitory pattern, characterized by a meaningful affinity constant (K i = 0.95 ± 0.21 μM). Although all three compounds induce cell death in neuroblastoma SHSY5Y cells at concentrations exceeding 2 μM, the two modulators facilitate cells' proliferation at concentrations ≤ 1.5 μM, whereas the competitive inhibitor [Pt(O,O'-acac)(γ-acac)(DMSO)] only shows a pro-apoptotic activity at all investigated concentrations. These features render the [Pt(O,O'-acac)(γ-acac)(DMSO)] a promising "lead compound" for the synthesis of IDE-specific inhibitors (not characterized yet) with therapeutic potentiality.
Collapse
Affiliation(s)
- Grazia R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,CIRCMSB, Via C. Ulpiani 27, 70125, Bari, Italy
| | - Diego Sbardella
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,CIRCMSB, Via C. Ulpiani 27, 70125, Bari, Italy
| | - Sandra A De Pascali
- CIRCMSB, Via C. Ulpiani 27, 70125, Bari, Italy.,Department of Environmental Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,CIRCMSB, Via C. Ulpiani 27, 70125, Bari, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,CIRCMSB, Via C. Ulpiani 27, 70125, Bari, Italy
| | - Francesco P Fanizzi
- CIRCMSB, Via C. Ulpiani 27, 70125, Bari, Italy.,Department of Environmental Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - Stefano Marini
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy. .,CIRCMSB, Via C. Ulpiani 27, 70125, Bari, Italy.
| |
Collapse
|
14
|
Piccolini VM, Esposito A, Dal Bo V, Insolia V, Bottone MG, De Pascali SA, Fanizzi FP, Bernocchi G. Cerebellum neurotransmission during postnatal development: [Pt(O,O'-acac)(γ-acac)(DMS)] vs cisplatin and neurotoxicity. Int J Dev Neurosci 2014; 40:24-34. [PMID: 25450526 DOI: 10.1016/j.ijdevneu.2014.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/20/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022] Open
Abstract
Several chemotherapeutic drugs are known to cause neurotoxicity. Platinum-based agents in use or in clinical trials display neurotoxic potential accompanied by neurological complications; recent studies have identified a large number of behavioural issues in paediatric oncology patients. To understand the toxicity of platinum drugs at the molecular and cellular levels, this study compares the possible cytotoxic effects of an older platinum compound, cisplatin and a new platinum compound, [Pt(O,O'-acac)(γ-acac)(DMS)], on the CNS of postnatally developing rats, which is much more vulnerable to injury than the CNS of adult rats. Since several drugs interact with neurotransmitters during neuronal maturation, we performed immunostainings with antibodies raised against markers of glutamate and GABA, the major neurotransmitters in the cerebellum. After a single injection of cisplatin at postnatal day 10 (PD10), the labelling of Purkinje cells with the neurotransmitter markers evidenced alterations between PD11 and PD30, i.e. atrophy of the dendrite tree, changes in the distribution of synaptic contacts of parallel and climbing fibres, delay in the elimination of transient synapses on cell soma and severely impaired pinceau formation at the axon hillock. After treatment with [Pt(O,O'-acac)(γ-acac)(DMS)], the sole relevant change concerned the timing of climbing fibres elimination; the transient synapses disappearance on the Purkinje cell soma was delayed in some cells; instead, the growth of Purkinje cell dendrite tree was normal as was the formation of inhibitory synaptic contacts on these neurons. These findings add new evidence not only on the lower neurotoxicity of [Pt(O,O'-acac)(γ-acac)(DMS)] vs cisplatin but also on the involvement of neurotransmitters and relative synaptic connections in the maturation of central nerve tissue.
Collapse
Affiliation(s)
- Valeria Maria Piccolini
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy; Istituto di Genetica Molecolare del CNR, Sezione di Istochimica e Citometria, via Ferrata 9, 27100 Pavia, Italy.
| | - Alessandra Esposito
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Veronica Dal Bo
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Violetta Insolia
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Maria Grazia Bottone
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy; Istituto di Genetica Molecolare del CNR, Sezione di Istochimica e Citometria, via Ferrata 9, 27100 Pavia, Italy.
| | - Sandra Angelica De Pascali
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, 73100 Lecce, Italy.
| | - Francesco Paolo Fanizzi
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, 73100 Lecce, Italy.
| | - Graziella Bernocchi
- Dipartimento di Biologia e Biotecnologie "L. Spallanzani" Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| |
Collapse
|
15
|
Vetrugno C, Muscella A, Fanizzi FP, Cossa LG, Migoni D, De Pascali SA, Marsigliante S. Different apoptotic effects of [Pt(O,O'-acac)(γ-acac)(DMS)] and cisplatin on normal and cancerous human epithelial breast cells in primary culture. Br J Pharmacol 2014; 171:5139-53. [PMID: 24990093 DOI: 10.1111/bph.12831] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 06/16/2014] [Accepted: 06/24/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND AND PURPOSE The aim of this study was to determine whether [platinum (Pt)(O,O'-acetylacetonate (acac))(γ-acac)(dimethylsulphide (DMS))] is differentially cytotoxic in normal and cancer cells, and to measure comparative levels of cytotoxicity compared with cisplatin in the same cells. EXPERIMENTAL APPROACH We performed experiments on cancerous and normal epithelial breast cells in primary culture obtained from the same patients. The apoptotic effects [Pt(O,O'-acac)(γ-acac)(DMS)] and cisplatin in cancerous and normal breast cells were compared. KEY RESULTS Cancer cells were more sensitive to [Pt(O,O'-acac)(γ-acac)(DMS)] (IC50 = 5.22 ± 1.2 μmol·L(-1)) than normal cells (IC50 = 116.9 ± 8.8 μmol·L(-1)). However, the difference was less strong when cisplatin was used (IC50 = 96.0 ± 6.9 and 61.9 ± 6.1 μmol·L(-1) for cancer and normal cells respectively). Both compounds caused reactive oxygen species (ROS) production with different mechanisms: [Pt(O,O'-acac)(γ-acac)(DMS)] quickly activated NAD(P)H oxidase while cisplatin caused a slower formation of mitochondrial ROS. Cisplatin and [Pt(O,O'-acac)(γ-acac)(DMS)] caused activation of caspases, proteolysis of PARP and modulation of Bcl-2, Bax and Bid. [Pt(O,O'-acac)(γ-acac)(DMS)] also caused leakage of cytochrome c from the mitochondria. Overall, these processes proceeded more quickly in cells treated with [Pt(O,O'-acac)(γ-acac)(DMS)] compared with cisplatin. [Pt(O,O'-acac)(γ-acac)(DMS)] effects were faster and quantitatively greater in cancer than in normal cells. [Pt(O,O'-acac)(γ-acac)(DMS)] caused a fast decrease of mitochondrial membrane potential, especially in cancer cells. CONCLUSIONS AND IMPLICATIONS [Pt(O,O'-acac)(γ-acac)(DMS)] was specific to breast cancer cells in primary culture, and this observation makes this compound potentially more interesting than cisplatin.
Collapse
Affiliation(s)
- Carla Vetrugno
- Cell Physiology Lab, Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Universita' del Salento, Lecce, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
De Pascali SA, Muscella A, Vetrugno C, Marsigliante S, Fanizzi FP. Synthesis, characterization and cytotoxicity of novel Pt(II) κ2O,O′-acetylacetonate complexes with nitrogen ligands. Inorganica Chim Acta 2014. [DOI: 10.1016/j.ica.2013.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
17
|
Abstract
Platinum-complexes represent some of the most successful groups of clinically used anticancer drugs. Their mechanism of action relies on the formation of stable DNA adducts occurring at the nitrogen in position 7 of guanine (N7) and involving one or two spatially close residues. The formation of stable DNA adducts is recognized as a DNA damaging event and, ultimately, drives cells to death. Nevertheless, nucleobases are not the only reliable targets of these drugs and other biomolecules can be involved. Among them large interest has been devoted to proteins since they contain several potential reactive sites for platinum (His, Met, and Cys) and, in particular, because the reaction of the metal with sulfur containing groups is a kinetically favored process. As a result, the occurrence of protein adducts and DNA-protein cross-links must be further taken into account in order to fully define cisplatin mechanism of action. Herein, we will summarize the most recent experimental evidence collected so far on protein-cisplatin adduct formation to better dissect its correlation with the drug pharmacological profile. Indeed, in addition to modulation of drug bioavailability and toxicity, the potential role of proteins as reaction intermediates or reservoir systems in platinum drugs can be envisaged. Additionally, the effects of Pt-coordinating groups on the chemical reactivity of the metal complexes will be reviewed. From all these outcomes a general model for Pt-based drugs mechanism of action can be drawn which is more articulate than the one currently supported. It claims proteins as reactive intermediates for DNA platination and it defines them as relevant to fully describe the clinical potential of this class of anticancer drugs.
Collapse
Affiliation(s)
- O Pinato
- Department of Pharmaceutical and Pharmacological Science, v. Marzolo 5, 35131 Padova, Italy.
| | | | | |
Collapse
|
18
|
Synthesis, Crystal Structure, and Biological Study of PtIIComplexes with 4-Acyl-5-pyrazolones. Eur J Inorg Chem 2014. [DOI: 10.1002/ejic.201301479] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
19
|
Antitumor activity of [Pt(O,O'-acac)(γ-acac)(DMS)] in mouse xenograft model of breast cancer. Cell Death Dis 2014; 5:e1014. [PMID: 24457958 PMCID: PMC4040677 DOI: 10.1038/cddis.2013.554] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 12/08/2013] [Accepted: 12/11/2013] [Indexed: 11/17/2022]
Abstract
The higher and selective cytotoxicity of [Pt(O,O′-acac)(γ-acac)(DMS)] toward cancer cell in both immortalized cell lines and in breast cancer cells in primary cultures, stimulated a pre-clinical study so as to evaluate its therapeutic potential in vivo. The efficacy of [Pt(O,O′-acac)(γ-acac)(DMS)] was assessed using a xenograft model of breast cancer developed by injection of MCF-7 cells in the flank of BALB/c nude mice. Treatment of solid tumor-bearing mice with [Pt(O,O′-acac)(γ-acac)(DMS)] induced up to 50% reduction of tumor mass compared with an average 10% inhibition recorded in cisplatin-treated animals. Thus, chemotherapy with [Pt(O,O′-acac)(γ-acac)(DMS)] was much more effective than cisplatin. We also demonstrated enhanced in vivo pharmacokinetics, biodistribution and tolerability of [Pt(O,O′-acac)(γ-acac)(DMS)] when compared with cisplatin administered in Wistar rats. Pharmacokinetics studies with [Pt(O,O′-acac)(γ-acac)(DMS)] revealed prolonged Pt persistence in systemic blood circulation and decreased nefrotoxicity and hepatotoxicity, major target sites of cisplatin toxicity. Overall, [Pt(O,O′-acac)(γ-acac)(DMS)] turned out to be extremely promising in terms of greater in vivo anticancer activity, reduced nephrotoxicity and acute toxicity compared with cisplatin.
Collapse
|
20
|
Fang L, Wuptra K, Chen D, Li H, Huang SK, Jin C, Yokoyama KK. Environmental-stress-induced Chromatin Regulation and its Heritability. ACTA ACUST UNITED AC 2014; 5. [PMID: 25045581 PMCID: PMC4101908 DOI: 10.4172/2157-2518.1000156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chromatin is subject to proofreading and repair mechanisms during the process of DNA replication, as well as repair to maintain genetic and epigenetic information and genome stability. The dynamic structure of chromatin modulates various nuclear processes, including transcription and replication, by altering the accessibility of the DNA to regulatory factors. Structural changes in chromatin are affected by the chemical modification of histone proteins and DNA, remodeling of nucleosomes, incorporation of variant histones, noncoding RNAs, and nonhistone DNA-binding proteins. Phenotypic diversity and fidelity can be balanced by controlling stochastic switching of chromatin structure and dynamics in response to the environmental disruptors and endogenous stresses. The dynamic chromatin remodeling can, therefore, serve as a sensor, through which environmental and/or metabolic agents can alter gene expression, leading to global cellular changes involving multiple interactive networks. Furthermore its recent evidence also suggests that the epigenetic changes are heritable during the development. This review will discuss the environmental sensing system for chromatin regulation and genetic and epigenetic controls from developmental perspectives.
Collapse
Affiliation(s)
- Lei Fang
- Department of Environmental Medicine, NYU School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | - Kenly Wuptra
- Center of Environmental Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd, San Ming District, Kaohsiung 807, Taiwan ; Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 35 Keyan Rd, Zhunan, Miaoli County 350, Taiwan
| | - Danqi Chen
- Department of Environmental Medicine, NYU School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | - Hongjie Li
- Department of Environmental Medicine, NYU School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | - Shau-Ku Huang
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 35 Keyan Rd, Zhunan, Miaoli County 350, Taiwan ; Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Chunyuan Jin
- Department of Environmental Medicine, NYU School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | - Kazunari K Yokoyama
- Center of Environmental Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd, San Ming District, Kaohsiung 807, Taiwan
| |
Collapse
|
21
|
A new platinum(II) compound anticancer drug candidate with selective cytotoxicity for breast cancer cells. Cell Death Dis 2013; 4:e796. [PMID: 24030148 PMCID: PMC3789173 DOI: 10.1038/cddis.2013.315] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 06/27/2013] [Accepted: 07/01/2013] [Indexed: 01/12/2023]
Abstract
[Pt(O,O′-acac)(γ-acac)(DMS)] (PtAcD) is able to induce apoptosis in various human cancer cells, including the cisplatin-resistant human breast carcinoma MCF-7 cells. Here, to confirm that PtAcD has the potentiality for therapeutic intervention, we studied its effects in primary cultured epithelial breast cells obtained from cancers and also from the corresponding histologically proven non-malignant tissue adjacent to the tumor. We demonstrated that PtAcD (1) is more cytotoxic in cancer than in normal breast cells; (2) activated NAD(P)H oxidase, leading to PKC-ζ and PKC-α tanslocations; (3) activated antiapoptotic pathways based on the PKC-α, ERK1/2 and Akt kinases; (4) activated PKC-ζ and, only in cancer cell PKC-δ, responsible for the sustained phosphorylation of p38 and JNK1/2, kinases both of which are involved in the mitochondrial apoptotic process. Moreover, crosstalk between ERK/Akt and JNK/p38 pathways affected cell death and survival in PtAcD-treated breast cell. In conclusion, this study adds and extends data that highlight the pharmacological potential of PtAcD as an anti breast cancer drug.
Collapse
|
22
|
Platinum drugs and neurotoxicity: effects on intracellular calcium homeostasis. Cell Biol Toxicol 2013; 29:339-53. [DOI: 10.1007/s10565-013-9252-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 07/22/2013] [Indexed: 10/26/2022]
|
23
|
Collotta M, Bertazzi PA, Bollati V. Epigenetics and pesticides. Toxicology 2013; 307:35-41. [PMID: 23380243 DOI: 10.1016/j.tox.2013.01.017] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 01/08/2013] [Accepted: 01/16/2013] [Indexed: 01/23/2023]
Abstract
Pesticides, a wide class of environmental contaminants, may cause both acute and delayed health effects in exposed subjects. These effects can range from simple irritation of the skin and eyes to more severe effects such as affecting the nervous system, the reproductive system and cancer. The molecular mechanisms underlying such effects are still under investigation. Epigenetics is the study of heritable changes in gene expression that occur without a change in the DNA sequence. Several epigenetic mechanisms, including DNA methylation, histone modifications and microRNA expression, can be triggered by environmental factors. We review current evidences indicating that epigenetic modifications may mediate pesticide effects on human health. In vitro, animal, and human investigations have identified several classes of pesticides that modify epigenetic marks, including endocrine disruptors, persistent organic pollutants, arsenic, several herbicides and insecticides. Several investigations have examined the effects of environmental exposures and epigenetic markers, and identified toxicants that modify epigenetic states. These modifications are similar to the ones found in pathological tissue samples. In spite of the current limitations, available evidence supports the concept that epigenetics holds substantial potential for furthering our understanding of the molecular mechanisms of pesticides health effects, as well as for predicting health-related risks due to conditions of environmental exposure and individual susceptibility.
Collapse
Affiliation(s)
- M Collotta
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Via San Barnaba 8, Milan 20122, Italy
| | | | | |
Collapse
|
24
|
Gulec M, Oral E, Dursun OB, Yucel A, Hacimuftuoglu A, Akcay F, Suleyman H. Mirtazapine protects against cisplatin-induced oxidative stress and DNA damage in the rat brain. Psychiatry Clin Neurosci 2013; 67:50-8. [PMID: 23279761 DOI: 10.1111/j.1440-1819.2012.02395.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 08/08/2012] [Accepted: 09/11/2012] [Indexed: 01/05/2023]
Abstract
AIM Cisplatin chemotherapy is associated with neurotoxicity, and oxidative stress might play an important role in the pathogenesis. Mirtazapine may be a preventative agent via its less-known antioxidant properties. The aim of this study was to examine the potential chemoprotective effects of mirtazapine against cisplatin-induced oxidative stress and DNA damage. METHODS Twenty-four rats were divided equally into four groups: control; cisplatin (10 mg/kg i.p.); cisplatin plus mirtazapine (10-30 mg/kg, respectively i.p and p.o.); and mirtazapine (30 mg/kg p.o.). The rats were killed at the end of the 14th day of treatment. Brain tissue was examined with regard to antioxidant/oxidant biochemical parameters. RESULTS Although glutathione (tGSH) and nitric oxide (NO) end product mean scores were found to be statistically higher in the control group when compared with the cisplatin group (72.44% and 61.99% percentage change [PC], respectively), malondialdehyde (MDA), myeloperoxidase (MPO), and 8-hydroxyguanine (8-OH-GUA) mean scores were statistically lower in the control group in comparison with the cisplatin group (-55.48%, -67.99%, and -48.81% PC, respectively; P < 0.01). Finally, tGSH and NO end product levels were restored to normal (85.90% and 55.30% PC, respectively), and MDA, MPO, and 8-OH-GUA were significantly reduced by treatment with mirtazapine (-60.50%, -78.59%, and -38.10% PC, respectively; P < 0.01). CONCLUSION Mirtazapine has chemoprotective effects against cisplatin-induced oxidative stress and DNA damage in the rat brain, which may be attributed to its antioxidant capabilities. It would be useful to investigate whether cisplatin at the desired doses can be given concurrently with mirtazapine.
Collapse
Affiliation(s)
- Mustafa Gulec
- Department of Psychiatry, Ataturk University Medical Faculty, Erzurum, Turkey.
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The products obtained by forcing the reaction with nucleosides (guanosine, Guo, and adenosine, Ado) of potential anticancer drugs for nongenomic targets [PtCl(O,O'-acac)(L)] (L = dimethyl sulfoxide, DMSO; dimethyl sulfide, DMS), closely related to their very powerful organometallic analogues [Pt(O,O'-acac)(γ-acac)(L)], have been studied. [PtCl(O,O'-acac)(L)] and [Pt(O,O'-acac)(γ-acac)(L)] complexes were reported unreactive toward nucleobases. Aquo species [Pt(O,O'-acac)H2O(L)]+, obtained from [PtCl(O,O'-acac)(L)] by Ag+ driven coordinated Cl– removal, gave access to [Pt(O,O'-acac)(L)(nucleoside)]+ ([Pt(O,O'-acac)(DMSO)(Guo)]+, [Pt(O,O'-acac)(DMS)(Guo)]+, [Pt(O,O'-acac)(DMSO)(Ado)]+). The effect of the chelate oxygen donor acac (with respect to a chelate diammine), the role of the sulfur ligand (DMSO, DMS), and the influence of the purinic nucleoside itself on the coordinated Guo or Ado dynamic motions in [Pt(O,O'-acac)(L)(nucleoside)]+ complexes have been investigated by NMR spectroscopy. Interestingly, a slow rotation of nucleobase around the Pt–N(7) bond with formation of two rotamers was observed already at room temperature only in the case of [Pt(O,O'-acac)(DMSO)(Guo)]+. On the other hand, no hindered rotation at room temperature was detected in the analogous [Pt(O,O'-acac)(DMS)(Guo)]+ and [Pt(O,O'-acac)(DMSO)(Ado)]+ complexes. Data suggest that rotation of the nucleoside in [Pt(O,O'-acac)(L)(nucleoside)]+ is very different with respect to the analogous [Pt(diammine)(L)(nucleoside)]2+ systems, due to specific interactions between the acac chelate ligand, the DMSO, and the nucleobase.
Collapse
|
26
|
Bottone MG, Veronica DB, Piccolini VM, Bottiroli G, De Pascali SA, Fanizzi FP, Bernocchi G. Developmental expression of cellular prion protein and apoptotic molecules in the rat cerebellum: Effects of platinum compounds. J Chem Neuroanat 2012; 46:19-29. [DOI: 10.1016/j.jchemneu.2012.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 09/07/2012] [Accepted: 09/14/2012] [Indexed: 01/08/2023]
|
27
|
Santin G, Piccolini VM, Barni S, Veneroni P, Giansanti V, Dal Bo V, Bernocchi G, Bottone MG. Mitochondrial fusion: a mechanism of cisplatin-induced resistance in neuroblastoma cells? Neurotoxicology 2012; 34:51-60. [PMID: 23103224 DOI: 10.1016/j.neuro.2012.10.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 12/20/2022]
Abstract
Cisplatin induces apoptosis through different pathways. The intrinsic apoptotic pathway is mediated by mitochondria, which, as a result of cisplatin treatment, undergo morphological alterations. The aim of this study was to investigate cisplatin-induced mitochondrial functional and morphological long-term effects in neuroblastoma B50 rat cells. To this purpose, we followed evaluated different several apoptotic markers by means of flow cytometry, confocal and electron microscopy and western blotting techniques. We applied different treatment protocols based on the incubation of the neuroblastoma B50 rat cells with 40 μM cisplatin: (i) for 48 h and harvesting of the cells at the end of the treatment; (ii) further recovery in drug-free medium for 7 days post-treatment; (iii) conditions as in (ii) followed by re-seeding in normal medium and growth for a further 4 days. We observed apoptosis induction after the first treatment and after the recovery from cell death after long-term culture in drug-free medium. Interestingly, the latter phenomenon was characterized by mitochondrial elongation and mitochondrial protein rearrangement. In recovered and re-seeded cells, mitochondrial equilibrium moved toward fusion, possibly protecting cells from apoptosis.
Collapse
Affiliation(s)
- Giada Santin
- Istituto di Genetica Molecolare del CNR, Università di Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Muscella A, Calabriso N, Vetrugno C, Fanizzi FP, De Pascali SA, Marsigliante S. The signalling axis mediating neuronal apoptosis in response to [Pt(O,O′-acac)(γ-acac)(DMS)]. Biochem Pharmacol 2011; 81:1271-85. [DOI: 10.1016/j.bcp.2011.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2011] [Revised: 03/09/2011] [Accepted: 03/10/2011] [Indexed: 12/28/2022]
|
29
|
De Pascali SA, Lugoli F, De Donno A, Fanizzi FP. Mutagenic Tests Confirm That New Acetylacetonate Pt(II) Complexes Induce Apoptosis in Cancer Cells Interacting with Nongenomic Biological Targets. Met Based Drugs 2011; 2011:763436. [PMID: 21792272 PMCID: PMC3094815 DOI: 10.1155/2011/763436] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 02/04/2023] Open
Abstract
New platinum(II) complexes [PtCl(O,O'-acac)(L)] (1) and [Pt(O,O'-acac)(γ-acac)(L)] (2) (L = DMSO, a; DMS, b) containing a single chelated (O,O'-acac) (1), or one chelated and one σ-bonded (γ-acac) acetylacetonate (2) have been synthesized. The new Pt(II) complexes exhibited high in vitro cytotoxicity on cisplatin sensitive and resistant cell lines and showed negligible reactivity with nucleobases (Guo and 5'-GMP) but selective substitution of DMSO/DMS with soft biological nucleophiles, such as L-methionine. In order to assess the ability of the new complexes with respect to cisplatin to induce apoptosis by interaction with nongenomic targets, the Ames' test, a standard reverse mutation assay, was carried out on two Salmonella typhimurium strains (TA98 and TA100). Interestingly, the new complexes did not show the well-known mutagenic activity exhibited by cisplatin and are, therefore, able to activate apoptotic pathways without interacting with DNA.
Collapse
Affiliation(s)
- Sandra Angelica De Pascali
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, Prov. le Lecce/Monteroni, 73100 Lecce, Italy
| | - Federica Lugoli
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, Prov. le Lecce/Monteroni, 73100 Lecce, Italy
| | - Antonella De Donno
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, Prov. le Lecce/Monteroni, 73100 Lecce, Italy
| | - Francesco Paolo Fanizzi
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, Prov. le Lecce/Monteroni, 73100 Lecce, Italy
| |
Collapse
|
30
|
Developing central nervous system and vulnerability to platinum compounds. CHEMOTHERAPY RESEARCH AND PRACTICE 2011; 2011:315418. [PMID: 22312552 PMCID: PMC3265252 DOI: 10.1155/2011/315418] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 12/21/2010] [Indexed: 12/12/2022]
Abstract
Comparative studies on the effects of the platinum complexes in use or in clinical trials are carried out in order to discover differences in the neurotoxic potential and the reversibility of neurotoxicity. In this paper, we summarized the current literature on neurotoxicity and chemoresistance of cisplatin (cisPt) and discussed our recent efforts on the interference of cisPt and a new platinum compound [Pt(O,O′-acac)(γ-acac)(DMS)] (PtAcacDMS), with high specific reactivity with sulphur ligands instead of nucleobases as cisPt, on some crucial events of rat postnatal cerebellum development. The acute effects of drug treatments on cell proliferation and death in the external granular layer and granule cell migration and the late effects on the dendrite growth of Purkinje cells were evaluated. Together with the demonstrated antineoplastic effectiveness in vitro, compared with cisPt, data suggest a lower neurotoxicity of PtAcacDMS, in spite of its presence in the brain that involves considerations on the blood brain barrier permeability.
Collapse
|