Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nanomaterials (Basel) ; 11(8)2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34443862

ABSTRACT

Technological innovation, environmental sustainability, health, and wellness are the trajectories explored by current research to identify new strategies for a general improvement of human quality of life [...].

2.
Adv Exp Med Biol ; 1295: 3-27, 2021.
Article in English | MEDLINE | ID: mdl-33543453

ABSTRACT

Clinical responses and tolerability of conventional nanocarriers (NCs) are sometimes different from those expected in anticancer therapy. Thus, new smart drug delivery systems (DDSs) with stimuli-responsive properties and novel materials have been developed. Several clinical trials demonstrated that these DDSs have better clinical therapeutic efficacy in the treatment of many cancers than free drugs. Composition of DDSs and their surface properties increase the specific targeting of therapeutics versus cancer cells, without affecting healthy tissues, and thus limiting their toxicity versus unspecific tissues. Herein, an extensive revision of literature on NCs used as DDSs for cancer applications has been performed using the available bibliographic databases.


Subject(s)
Nanoparticles , Neoplasms , Drug Carriers/therapeutic use , Drug Delivery Systems , Humans , Neoplasms/drug therapy
3.
Electromagn Biol Med ; 40(1): 11-25, 2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33073612

ABSTRACT

Among haematological parameters of patients seriously ill with the coronavirus infectious disease 2019 (COVID-19), leucocytosis, lymphocytopenia, and the abnormal release of circulating cytokines, termed cytokine storm syndrome (CSS, also known as cytokine release syndrome or CRS), were found associated with disease severity. In particular, according to the serum cytokine profiling, pro-inflammatory interleukin 6 (IL-6) and anti-inflammatory interleukin 10 (IL-10) were observed to be considerably higher in patients experiencing respiratory distress, septic shock and/or multi-organ failure, namely "critical cases" requiring intensive care unit (ICU) admission, very often resulting in death. Interestingly, the production of these cytokines from human lymphocytes was found to be modulated by exposure of 24 h to a 554.2-553.8 mT inhomogeneous static magnetic field (SMF), which elicits IL-10 and suppresses IL-6. Thus, herein, with the aim of restoring lymphocyte count and physiological serum levels of IL-6 and IL-10, the infusion of human leukocyte antigen (HLA)-matched and SMF-exposed allogenic lymphocytes is proposed for the first time as an easy and affordable treatment option for COVID-19 patients. Even if the count of lymphocytes in COVID-19 patients is very low, SMF exposure may be a valuable tool for reprogramming autologous lymphocytes towards physiological conditions. Furthermore, the same procedure could be extended to include the whole autologous or allogenic white blood cells (WBCs). Time-varying/pulsed magnetic fields exerting comparable cell effects could also be employed.


Subject(s)
COVID-19/complications , Cytokine Release Syndrome/therapy , HLA Antigens/immunology , Lymphocytes/cytology , Lymphopenia/therapy , Magnetic Fields , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Humans , Immunotherapy , Interleukin-6/chemistry , Interleukin-6/metabolism , Lymphocytes/immunology , Lymphopenia/complications , Lymphopenia/immunology , Lymphopenia/pathology , Models, Molecular , Protein Conformation , Signal Transduction/immunology
4.
Hematol Oncol Stem Cell Ther ; 14(1): 1-15, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32971031

ABSTRACT

Glioblastoma (GB) is one of the most malignant types of central nervous system tumours, classified as grade IV by the World Health Organization. Despite the therapeutic advances, the prognosis is ominous, with a median survival of about 12-15 months post diagnosis. Although therapeutic options available can increase the survival, they are ineffective in treating patients with GB. Impairing factors such as the blood-brain barrier, cancer stem cells, and infiltration into brain parenchyma lead to failure of current therapies. Therefore, clinicians need novel/alternative effective strategies to treat GB. Due to their ability to preserve healthy tissues and to provide an effective and long-lasting response, stem cells (SCs) with tropism for tumour cells have attracted considerable attention in the scientific community. As is the case here, SCs can be used to target brain tumour cancer cells, especially high-grade malignant gliomas like GB, by overcoming the resistance and exerting benefits for patients affected with such lethal disease. Herein, we will discuss the research knowledge regarding SC-based therapy for the treatment of GB, focalising our attention on SCs and SC-released extracellular vesicles modified to express/load different antitumour payloads, as well as on SCs exploited as a diagnostic tool. Advantages and unresolved issues of anticancer SC-based therapy will also be considered.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Cell- and Tissue-Based Therapy , Glioblastoma , Neoplastic Stem Cells , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioblastoma/diagnosis , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis
5.
Nanomaterials (Basel) ; 10(6)2020 Jun 24.
Article in English | MEDLINE | ID: mdl-32599957

ABSTRACT

The scientific community is becoming increasingly interested in identifying, characterizing, and delivering nutraceuticals, which constitutes a multi-billion-dollar business. These bioactive agents are claimed to exhibit several health benefits, including the prevention and treatment of diseases such as arthritis, cancer, osteoporosis, cataracts, Alzheimer's, and Huntington's diseases, heart, brain and metabolic disorders, etc. Nutraceuticals are typically consumed as part of a regular human diet and are usually present within foods, comprising vegetable oil, although at low levels and variable composition. Thus, it is difficult to control the type, amount and frequency of their ingestion by individuals. Nanoformulations about vegetable oil-based bioactive compounds with nutraceutical properties are useful for overcoming these issues, while improving the uptake, absorption, and bioavailability in the body. The purpose of this current study is to review papers on such nanoformulations, particularly those relevant for health benefits and the prevention and management of diseases, as well as bioactives extracted from vegetable oils enhancing the drug effectiveness, retrieved through bibliographic databases by setting a timespan from January 2000 to April 2020 (about 1758 records).

6.
Molecules ; 25(6)2020 Mar 19.
Article in English | MEDLINE | ID: mdl-32204392

ABSTRACT

One of the most relevant drawbacks in medicine is the ability of drugs and/or imaging agents to reach cells. Nanotechnology opened new horizons in drug delivery, and silver nanoparticles (AgNPs) represent a promising delivery vehicle for their adjustable size and shape, high-density surface ligand attachment, etc. AgNPs cellular uptake involves different endocytosis mechanisms, including lipid raft-mediated endocytosis. Since static magnetic fields (SMFs) exposure induces plasma membrane perturbation, including the rearrangement of lipid rafts, we investigated whether SMF could increase the amount of AgNPs able to pass the peripheral blood lymphocytes (PBLs) plasma membrane. To this purpose, the effect of 6-mT SMF exposure on the redistribution of two main lipid raft components (i.e., disialoganglioside GD3, cholesterol) and on AgNPs uptake efficiency was investigated. Results showed that 6 mT SMF: (i) induces a time-dependent GD3 and cholesterol redistribution in plasma membrane lipid rafts and modulates gene expression of ATP-binding cassette transporter A1 (ABCA1), (ii) increases reactive oxygen species (ROS) production and lipid peroxidation, (iii) does not induce cell death and (iv) induces lipid rafts rearrangement, that, in turn, favors the uptake of AgNPs. Thus, it derives that SMF exposure could be exploited to enhance the internalization of NPs-loaded therapeutic or diagnostic molecules.


Subject(s)
Lymphocytes/cytology , Membrane Microdomains/metabolism , Silver/pharmacokinetics , ATP Binding Cassette Transporter 1 , Adult , Biological Transport , Endocytosis , Female , Humans , Lipid Peroxidation , Lymphocytes/chemistry , Magnetic Fields , Male , Metal Nanoparticles , Reactive Oxygen Species/metabolism , Silver/chemistry
7.
Int J Pharm ; 573: 118826, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31715352

ABSTRACT

Stanozolol (STZ) is a drug used to treat serious disorders like aplastic anemia and hereditary angioedema. It is also indicated as an adjunct therapy for the treatment of vascular disorders and growth failures. Encouraging results obtained using animal models demonstrated that STZ increases bone formation and mineralization, thus improving both density and biomechanical properties. Like natural androgens, such as TST and 5α-dihydrotestosterone (5α-DHT), STZ binds androgen receptor (AR) to activate AR-mediated signaling. Despite its therapeutic effects, this synthetic anabolic-androgenic steroid (AAS), or 5α-DHT derivative, due to its high lipophilicity, is poor soluble in water. Thus, to increase the water solubility and stability of STZ, as well as its bioavailability and efficacy, an innovative PEGylated STZ (STZ conjugated with (MeO-PEG-NH2)10kDa, (MeO-PEG-NH)10kDa-STZ) was synthesized. As confirmed by chromatography (RP-HPLC) and spectrometry (ATR-FTIR, 1H NMR, elemental CHNS(O) analysis, MALDI-TOF/TOF) analyses, a very pure, stable and soluble compound was obtained. Acetylcholinesterase (AChE) competitive ELISA demonstrated that the resulting PEGylated STZ competes against biological TST, especially at lower concentrations. Cytotoxicity of increasing concentrations (1, 10, 25 or 50 µM) of STZ and/or (MeO-PEG-NH)10kDa-STZ was also evaluated for up 80 h by performing the MTT assay on human osteosarcoma Saos-2 cells, which express AR and are responsive to STZ. PEGylation mitigated cytotoxicity of STZ, by increasing the cell viability values, especially at higher drug concentrations. Furthermore, these results suggest that (MeO-PEG-NH)10kDa-STZ is a promising and reliable drug to be used in clinical conditions in which TST is required.


Subject(s)
Anabolic Agents/pharmacokinetics , Androgens/pharmacokinetics , Drug Compounding/methods , Drug Design , Stanozolol/pharmacokinetics , Anabolic Agents/chemistry , Anabolic Agents/therapeutic use , Anabolic Agents/toxicity , Androgens/chemistry , Androgens/therapeutic use , Androgens/toxicity , Biological Availability , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Drug Stability , Hormone Replacement Therapy/methods , Humans , Hydrophobic and Hydrophilic Interactions , Polyethylene Glycols/chemistry , Receptors, Androgen/metabolism , Solubility , Stanozolol/chemistry , Stanozolol/therapeutic use , Stanozolol/toxicity , Testosterone/deficiency , Toxicity Tests , Water/chemistry
8.
Biomed Pharmacother ; 92: 254-264, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28551545

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant brain cancer that causes high mortality in humans. It responds poorly to the most common cancer treatments, such as surgery, chemo- and radiation therapy. Temozolomide (TMZ) is an alkylating agent that has been widely used to treat GBM; resistance to this drug is often found. One unexplored possibility for overcoming this resistance is a treatment based on concomitant exposure to electromagnetic fields (EMF) and TMZ. Indeed, many evidences show that EMF affects cancer cells and drug performance. In this study, we evaluated the potential synergistic effect of 100µM TMZ and EMF (100Hz, 100G) on two human glioma cells line, i.e., U87 and T98G above single treatments, TMZ or EMF. Co-treatment synergistically enhanced apoptosis in U87 and T98G cells, by increasing the expression of P53, Bax, and Caspase-3 and decreasing that of Bcl-2 and Cyclin-D1. We also observed an increase in reactive oxygen species (ROS) production and the overexpression of the heme oxygenase-1 (HO-1) gene in comparison to controls. In conclusion, since EMF enhanced the apoptotic effect of TMZ, possibly through a redox regulation mechanism, the TMZ/EMF combination may be effective for glioma cancer treating. Further studies are needed to reveal the action mechanism of this possible novel therapeutic approach.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Cytotoxins/toxicity , Dacarbazine/analogs & derivatives , Electromagnetic Fields/adverse effects , Glioblastoma/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dacarbazine/toxicity , Glioblastoma/pathology , Humans , Magnetic Field Therapy/methods , Temozolomide
9.
Toxicol In Vitro ; 41: 64-74, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28223142

ABSTRACT

This study aims to determine the interaction (uptake and biological effects on cell viability and cell cycle progression) of glucose capped silver nanoparticles (AgNPs-G) on human epithelioid cervix carcinoma (HeLa) cells, in relation to amount, 2×103 or 2×104 NPs/cell, and exposure time, up to 48h. The spherical and well dispersed AgNPs (30±5nm) were obtained by using glucose as reducing agent in a green synthesis method that ensures to stabilize AgNPs avoiding cytotoxic soluble silver ions Ag+ release. HeLa cells take up abundantly and rapidly AgNPs-G resulting toxic to cells in amount and incubation time dependent manner. HeLa cells were arrested at S and G2/M phases of the cell cycle and subG1 population increased when incubated with 2×104 AgNPs-G/cell. Mitotic index decreased accordingly. The dissolution experiments demonstrated that the observed effects were due only to AgNPs-G since glucose capping prevents Ag+ release. The AgNPs-G influence on HeLa cells viability and cell cycle progression suggest that AgNPs-G, alone or in combination with chemotherapeutics, may be exploited for the development of novel antiproliferative treatment in cancer therapy. However, the possible influence of the cell cycle on cellular uptake of AgNPs-G and the mechanism of AgNPs entry in cells need further investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Glucose/pharmacology , Metal Nanoparticles , Silver/pharmacology , Cell Survival/drug effects , HeLa Cells , Humans , L-Lactate Dehydrogenase/metabolism , Metal Nanoparticles/ultrastructure , Microscopy, Electron, Transmission
10.
Electromagn Biol Med ; 36(3): 238-247, 2017.
Article in English | MEDLINE | ID: mdl-27874284

ABSTRACT

The impact of extremely low-frequency pulsed electromagnetic fields (ELF-PEMFs) at various frequencies and amplitudes was investigated on cell cycle, apoptosis and viability of the Glioblastoma Multiforme (GBM) cell line (U87), in vitro. The GBM is a malignant brain tumor with high mortality in humans and poorly responsive to the most common type of cancer treatments, such as surgery, chemotherapy and radiation therapy. U87 cells with five experimental groups (I-V) were exposed to various ELF-PEMFs for 2, 4 and 24 h, as follows: (I) no exposure, control; (II) 50 Hz 100 ± 15 G; (III) 100 Hz 100 ± 15 G; (IV) 10 Hz 50 ± 10 G; (V) 50 Hz 50 ± 10 G. The morphology properties, cell viability and gene expression of proteins involved in cell cycle regulation (Cyclin-D1 and P53) and apoptosis (Caspase-3) were investigated. After 24 h, the cell viability and Cyclin-D1 expression increased in Group II (30%, 45%), whereas they decreased in Groups III (29%, 31%) and IV (21%, 34%); P53 and Caspase-3 elevated only in Group III; and no significant difference was observed in Group V, respectively, compared with the control (p < 0.05). The data suggest that the proliferation and apoptosis of human GBM are influenced by exposure to ELF-PEMFs in different time-dependent frequencies and amplitudes. The fact that some of the ELF-PEMFs frequencies and amplitudes favor U87 cells proliferation indicates precaution for the use of medical devices related to the MFs on cancer patients. On the other hand, some other ELF-PEMFs frequencies and intensities arresting U87 cells growth could open the way to develop novel therapeutic approaches.


Subject(s)
Electromagnetic Fields , Glioblastoma , Apoptosis , Caspase 3/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Survival , Cyclin D1/metabolism , Humans
11.
Curr Drug Targets ; 16(13): 1512-30, 2015.
Article in English | MEDLINE | ID: mdl-26028043

ABSTRACT

One of the frontier of nanoscience is undoubtedly represented by the use of nanotechnologies in the pharmaceutical research. During the last decades a big family of nanostructures that have a surface-acting action, such as NanoParticles (NPs), lipid nanocarriers and many more, have been developed to be used as Drug Delivery Systems (DDSs). However, these nanocarriers opened also new frontiers in nanometrology, requiring an accurate morphological characterization, near atomic resolution, before they are really available to clinicians to ascertain their elemental composition, to exclude the presence of contaminants introduced during the synthesis procedure and to ensure biocompatibility. Classical Transmission (TEM) and Scanning Electron Microscopy (SEM) techniques frequently have to be adapted for an accurate analysis of formulation morphology, especially in case of hydrated colloidal systems. Specific techniques such as environmental scanning microscopy and/or cryo preparation are required for their investigation. Analytical Electron Microscopy (AEM) techniques such as Electron Energy-Loss Spectroscopy (EELS) or Energy-Dispersive X-ray Spectroscopy (EDXS) are additional assets to determine the elemental composition of the systems. Here we will discuss the importance of Electron Microscopy (EM) as a reliable tool in the pharmaceutical research of the 21(st) century, focalizing our attention on advantages and limitations of different kind of NPs (in particular silver and carbon NPs, cubosomes) and vesicles (liposomes and niosomes).


Subject(s)
Drug Delivery Systems , Microscopy, Electron/methods , Nanoparticles , Animals , Drug Carriers/chemistry , Humans , Lipids/chemistry , Liposomes , Nanotechnology/methods
12.
PLoS One ; 9(11): e113530, 2014.
Article in English | MEDLINE | ID: mdl-25423171

ABSTRACT

Beneficial or adverse effects of Static Magnetic Fields (SMFs) are a large concern for the scientific community. In particular, the effect of SMF exposure during anticancer therapies still needs to be fully elucidated. Here, we evaluate the effects of SMF at induction levels that cisPt-treated cancer patients experience during the imaging process conducted in Low field (200-500 mT), Open field (300-700 mT) and/or inhomogeneous High field (1.5-3 T) Magnetic Resonance Imaging (MRI) machines. Human adrenergic neuroblastoma SH-SY5Y cells treated with 0.1 µM cisPt (i.e. the lowest concentration capable of inducing apoptosis) were exposed to SMF and their response was studied in vitro. Exposure of 0.1 µM cisPt-treated cells to SMF for 2 h decreased cell viability (30%) and caused overexpression of the apoptosis-related cleaved caspase-3 protein (46%). Furthermore, increase in ROS (Reactive Oxygen Species) production (23%) and reduction in the number of mitochondria vs controls were seen. The sole exposure of SMF for up to 24 h had no effect on cell viability but increased ROS production and modified cellular shape. On the other hand, the toxicity of cisPt was significantly prevented during 24 h exposure to SMF as shown by the levels of cell viability, cleaved caspase-3 and ROS production. In conclusion, due to the cytoprotective effect of 31.7-232.0 mT SMF on low-cisPt-concentration-treated SH-SY5Y cells, our data suggest that exposure to various sources of SMF in cancer patients under a cisPt regimen should be strictly controlled.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Magnetics , Neuroblastoma/pathology , Caspase 3/metabolism , Cell Line, Tumor , Humans , Neuroblastoma/enzymology , Neuroblastoma/metabolism , Reactive Oxygen Species/metabolism
13.
Article in English | MEDLINE | ID: mdl-24795765

ABSTRACT

Carica papaya is widely used in folk medicine as herbal remedy to prevent, protect against, and cure several diseases. These curative properties are based on the presence in different parts of the plant of phytochemical nutrients with antioxidant effect. Seeds are the less exploited part; thus this study is aimed at assessing the antioxidant activities of the C. papaya seeds water extract against hydrogen peroxide (H2O2) oxidative stress in human skin Detroit 550 fibroblasts. C. papaya seeds water extract is not toxic and acts as a potent free radical scavenger, providing protection to Detroit 550 fibroblasts that underwent H2O2 oxidative stress. Data show that (i) the maximum protective effect is achieved by the simultaneous administration of the extract with 1 mM H2O2; (ii) the extract in presence of an oxidative stress does not increase catalase activity and prevents the release of cytochrome C and the inner mitochondrial transmembrane potential (Δψ m ) loss; (iii) the extract is more efficient than vitamin C to hamper the oxidative damage; (iv) the purified subfractions of the seeds water extract exert the same antioxidant effect of whole extract. In conclusion, C. papaya seeds water extract is potentially useful for protection against oxidative stress.

14.
PLoS One ; 8(8): e72341, 2013.
Article in English | MEDLINE | ID: mdl-23977284

ABSTRACT

The aim of the present work has been the design and the realization of a Magnetostatic Field System for Exposure of Cell cultures (MaFiSEC) for the uniform and the reproducible exposure of cell cultures to static magnetic fields (SMFs) of moderate magnetic induction. Experimental and computer-simulated physical measurements show that MaFiSEC: i) generates a SMF with magnetic induction that can be chosen in the range of 3 to 20 mT; ii) allows the uniform SMF exposure of cells growing in adhesion and in suspension; iii) is cheap and easy to use. The efficacy and reproducibility of MaFiSEC has been tested by comparing the biological effects exerted on isolated human lymphocytes by 72 h of exposure to a magnet (i.e. Neodymium Magnetic Disk, NMD) placed under the culture Petri dish. Lymphocytes morphology, viability, cell death, oxidative stress and lysosomes activity were the parameters chosen to evaluate the SMF biological effects. The continuous exposure of cells to a uniform SMF, achieved with MaFiSEC, allows highly reproducible biochemical and morphological data.


Subject(s)
Lymphocytes/cytology , Magnetic Fields , Cell Adhesion , Cell Culture Techniques , Cell Death , Cell Survival , Cells, Cultured , Humans , Lymphocytes/metabolism , Lysosomes/metabolism , Magnets , Oxidative Stress , Reproducibility of Results
15.
PLoS One ; 8(8): e72374, 2013.
Article in English | MEDLINE | ID: mdl-23991101

ABSTRACT

The effect of inhomogeneous static magnetic field (SMF)-exposure on the production of different cytokines from human peripheral blood mononuclear cells (PMBC), i.e., lymphocytes and macrophages, was tested in vitro. Some cultures were activated with lipopolysaccharide (LPS) at time point -3 h and were either left alone (positive control) or exposed to SMF continuously from 0 until 6, 18, or 24 h. The secretion of interleukin IL-6, IL-8, tumor necrosis factor TNF-α, and IL-10 was tested by ELISA. SMF-exposure caused visible morphological changes on macrophages as well as on lymphocytes, and also seemed to be toxic to lymphocytes ([36.58; 41.52]%, 0.308≤p≤0.444), but not to macrophages (<1.43%, p≥0.987). Analysis of concentrations showed a significantly reduced production of pro-inflammatory cytokines IL-6, IL-8, and TNF-α from macrophages compared to negative control ([56.78; 87.52]%, p = 0.031) and IL-6 compared to positive control ([45.15; 56.03]%, p = 0.035). The production of anti-inflammatory cytokine IL-10 from macrophages and from lymphocytes was enhanced compared to negative control, significantly from lymphocytes ([-183.62; -28.75]%, p = 0.042). The secretion of IL-6 from lymphocytes was significantly decreased compared to positive control ([-115.15; -26.84]%, p = 0.039). This massive in vitro evidence supports the hypotheses that SMF-exposure (i) is harmful to lymphocytes in itself, (ii) suppresses the release of pro-inflammatory cytokines IL-6, IL-8, and TNF-α, and (iii) assists the production of anti-inflammatory cytokine IL-10; thus providing a background mechanism of the earlier in vivo demonstrated anti-inflammatory effects of SMF-exposure.


Subject(s)
Inflammation Mediators/metabolism , Lymphocytes/cytology , Macrophages/cytology , Magnetics , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Lymphocytes/metabolism , Macrophages/metabolism
16.
Bioelectromagnetics ; 30(5): 352-64, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19189300

ABSTRACT

This study deals with the morphofunctional influence of 72 h exposure to a 6 mT static magnetic field (SMF) during differentiation induced by 50 ng/ml 12-O-tetradecanoyl-13-phorbol acetate (TPA) in human leukaemia U937 cells. The cell morphology of U937 cells was investigated by optic and electron microscopy. Specific antibodies and/or molecules were used to label CD11c, CD14, phosphatidylserine, F-actin and to investigate the distribution and activity of lysosomes, mitochondria and SER. [Ca(2+)](i) was evaluated with a spectrophotometer. The degree of differentiation in SMF-exposed cells was lower than that of non-exposed cells, the difference being exposure time-dependent. SMF-exposed cells showed cell shape and F-actin modification, inhibition of cell attachment, appearance of membrane roughness and large blebs and impaired expression of specific macrophagic markers on the cell surface. The intracellular localization of SER and lysosomes was only partially affected by exposure. A significant localization of mitochondria with an intact membrane potential at the cell periphery in non-exposed, TPA-stimulated cells was observed; conversely, in the presence of SMF, mitochondria were mainly localised near the nucleus. In no case did SMF exposure affect cell viability. The sharp intracellular increase of [Ca(2+)](i) could be one of the causes of the above-described changes.


Subject(s)
Cell Differentiation/drug effects , Magnetics , Tetradecanoylphorbol Acetate/pharmacology , Actin Cytoskeleton/metabolism , Animals , Calcium/metabolism , Cell Shape , Endoplasmic Reticulum, Smooth/metabolism , Humans , Lysosomes/metabolism , Macrophages/cytology , Macrophages/drug effects , Mitochondria/metabolism , Monocytes/cytology , Monocytes/drug effects , Time Factors , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...