Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncol Lett ; 24(3): 295, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35949615

ABSTRACT

Previous studies have shown that intermittent exposure to a 50 Hz, 100 µT sinusoidal magnetic field (MF) promotes proliferation of human neuroblastoma cells, NB69. This effect is mediated by activation of the epidermal growth factor receptor through a free radical-dependent activation of the p38 pathway. The present study investigated the possibility that the oxidative stress-sensitive protein p53 is a potential target of the MF, and that field exposure can affect the protein expression. To that end, NB69 cells were exposed to short intervals of 30 to 120 min to the aforementioned MF parameters. Two specific anti-p53 antibodies that allow discrimination between the wild and unfolded forms of p53 were used to study the expression and cellular distribution of both isoforms of the protein. The expression of the antiapoptotic protein Bcl-2, whose regulation is mediated by p53, was also analyzed. The obtained results revealed that MF exposure induced increases in p53 gene expression and in protein expression of the wild-type form of p53. Field exposure also caused overexpression of the unfolded form of p53, together with changes in the nuclear/cytoplasmic distribution of both forms of the protein. The expression of protein Bcl-2 was also significantly increased in response to the MF. As a whole, these results indicated that the MF is capable of interacting with the function, distribution and conformation of protein p53. Such interactions could be involved in previously reported MF effects on NB69 proliferation promotion.

2.
Oncol Lett ; 22(5): 807, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34630714

ABSTRACT

Due to their alleged analgesic, anti-inflammatory and tissue regenerative effects, capacitive-resistive electrothermal therapy (CRET), which is based on non-invasive exposure to radiofrequency (RF) currents, is often applied to chemotherapeutically treated patients with cancer. Our previous studies have demonstrated that subthermal CRET currents can elicit a number of cell responses, including anti-proliferative effects, in the human liver cancer cell line HepG2. Such effects involve significant changes in the regulation of proteins involved in MAPK signaling pathways, which are also implicated in the cancer cell response to standard anticancer drugs such as sorafenib. This overlap in response pathways may lead to competitive, neutralizing or blocking interactions between the electrical and chemical treatments, thus raising questions on the advisability of CRET treatment for their analgesic, anti-inflammatory or other purposes in patients undergoing chemotherapy. The present study analyzed the effects of simultaneous treatment with sorafenib and 448-kHz, subthermal CRET current on the proliferation and viability of HepG2 cell cultures. Cell viability was assessed through Trypan blue or XTT assays, while flow cytometry was applied for cell cycle and apoptosis analysis. The expression of proteins involved in cell proliferation were assessed by immunoblotting and immunofluorescence. The results revealed no evidence to suggest that the electrical treatment counteracted or neutralized the cellular response to sorafenib at the different conditions evaluated. Furthermore, at the standard pharmacological sorafenib concentration, 5 µM, the combined treatment elicited an anti-proliferative response significantly stronger than that induced by each of the treatments when applied separately in HepG2 cells. These data do not support the hypothesis that CRET exposure may inhibit or diminish the effects of a chemotherapeutic drug used in cancer treatment, and highlights the requirement for further investigation into the cell response to the combined action of electrical and chemical treatments.

3.
Electromagn Biol Med ; 40(1): 103-116, 2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33345643

ABSTRACT

Our previous studies have shown that intermittent exposure to a 50-Hz, 100-µT sine wave magnetic field (MF) promotes human NB69 cell proliferation, mediated by activation of the epidermal growth factor receptor (EGFR) and pathways MAPK-ERK1/2 and p38; being the effects on proliferation and p38 activation blocked by the chelator N-acetylcysteine. The present work investigates the MF effects on free radical (FR) production, and the potential involvement of NADPH oxidase, the main source of reactive oxygen species (ROS), in the MF-induced activation of MAPK pathways. To this end, the field effects on MAPK-ERK1/2, -p38 and -JNK activation in the presence or absence of the NADPH oxidase inhibitor, diphenyleneiodonium chloride (DPI), as well as the expression of the p67phox subunit, were analyzed. The results revealed that field exposure increases FR production and induces early, transient expression of the cytosolic component of the NADPH oxidase, p67phox. Also, the MF-induced activation of the MAPK-JNK pathway, but not that of -ERK1/2 or -p38 pathways, was prevented in the presence of the DPI, which has been shown to significantly reduce p67phox expression. These data, together with those from previous studies, identify various, FR-dependent or -independent mechanisms, involved in the MF-induced proliferative response mediated by MAPK signaling activation.


Subject(s)
MAP Kinase Signaling System , Magnetic Fields , NADPH Oxidases/metabolism , Neuroblastoma/pathology , Cell Line, Tumor , Humans
4.
Electromagn Biol Med ; 40(1): 201-209, 2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33073635

ABSTRACT

Capacitive-Resistive Electric Transfer (CRET) thermotherapies aim at tissue repair and regeneration through non-invasive application of RF currents. We have reported that the cellular response to subthermal CRET currents is non-linearly dependent on the signal frequency, and that in vitro exposure to a 448-kHz CRET signal promotes ADSC proliferation, as well as collagen and glycosaminoglycan synthesis in prechondrocytic cells. The present work investigates the response of neonatal fibroblasts to subthermal exposure (100 µA/mm2) to two CRET signals: a 448-kHz, non-modulated sinusoidal wave vs. a 20-kHz amplitude-modulation of the 448-kHz carrier. To that end, cell proliferation and expression of the biomarkers Hsp47, Hsp27 and decorin were assessed by cell count, PCNA and Western blotting. The results revealed that while both signals significantly and equivalently increased early (4 h) expression of Hsp47, the modulated signal was more efficient in inducing Hsp27 and decorin overexpression and promoting cell proliferation. These data indicate that the cellular response is dependent on the RF signal modulation and suggest that the therapeutic effects of CRET could be mediated by promotion of fibroblastic proliferation and overexpression of biomarkers that are essential in skin regeneration.


Subject(s)
Fibroblasts/cytology , Fibroblasts/radiation effects , Radio Waves , Dose-Response Relationship, Radiation , Humans , Infant, Newborn
5.
Cell Physiol Biochem ; 52(4): 893-907, 2019.
Article in English | MEDLINE | ID: mdl-30964607

ABSTRACT

BACKGROUND/AIMS: Previous studies have shown that a 63-hour, intermittent exposure to a 50 Hz, 100 µT magnetic field (MF) induces in the NB69 line of human neuroblastoma a proliferative response that is mediated by activation of the MAPK pathways ERK1/2 and p38. The present study aims to investigate the potential involvement of the epidermal growth factor receptor (EGFR) in the field-induced cell proliferation and activation of MAPK pathways. METHODS: NB69 cultures were MF- or sham-exposed for 5 to 30 minute intervals and 63 hours. Cell proliferation and activation of MAPK-ERK1/2, -p38 and -JNK was analyzed in the presence or absence of erlotinib, an effective inhibitor of EGFR tyrosine kinase. The expression of p-EGFR and MMP-9 in the presence or absence of MF was also studied. Between 3 and 7 replicates of each experiment were performed, using between 3 and 4 samples per experimental condition and replicate. At the end of each replicate, the samples were analyzed at short times (5-30 min) through immunofluorescence and Western blotting, and the growth response was assessed (63 hours interval) through dye exclusion with Trypan blue. RESULTS: The results confirmed that field exposure induces cell proliferation and activation of ERK1/2, p38 and JNK, and revealed that these effects were blocked with erlotinib. The data also showed that, compared to shamexposed controls, the MF exposure induces early and transient increases in the expression of p-EGFR and MMP-9 at 15 and 5 min from the exposure onset, respectively. CONCLUSION: The obtained results reveal that the activation of the MAPK-ERK1/2 and -p38 pathways by the MF is mediated by the EGF receptor. Taken together with our previously published results, this dataset suggests that the proliferative response induced in NB69 by a 63-hour exposure to a weak, power frequency MF, is mediated by early transient activation of EGFR in which MMP-9 would be involved.


Subject(s)
Cell Proliferation , MAP Kinase Signaling System , Magnetic Fields , Neoplasm Proteins/metabolism , Neuroblastoma/metabolism , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Neoplasm Proteins/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology
6.
Mol Med Rep ; 13(5): 3895-903, 2016 May.
Article in English | MEDLINE | ID: mdl-27035334

ABSTRACT

The 448 kHz capacitive­resistive electric transfer (CRET) is an electrothermal therapy currently applied in anticellulite and antiobesity treatments. The aim of the present study was to determine whether exposure to the CRET electric signal at subthermal doses affected early adipogenic processes in adipose­derived stem cells (ADSC) from human donors. ADSC were incubated for 2 or 9 days in the presence of adipogenic medium, and exposed or sham­exposed to 5 min pulses of 448 kHz electric signal at 50 µA/mm2 during the last 48 h of the incubation. Colorimetric, immunofluorescence, western blotting and reverse transcription­quantitative polymerase chain reaction assays were performed to assess adipogenic differentiation of the ADSC. Electric stimulation significantly decreased cytoplasmic lipid content, after both 2 and 9 days of differentiation. The antiadipogenic response in the 9 day samples was accompanied by activation of mitogen­activated protein kinase kinase 1/2, decreased expression and partial inactivation of peroxisome proliferator­activated receptor (PPAR) Î³, which was translocated from the nucleus to the cytoplasm, together with a significant decrease in the expression levels of the PPARG1 gene, perilipin, angiopoietin­like protein 4 and fatty acid synthase. These results demonstrated that subthermal stimulation with CRET interferes with the early adipogenic differentiation in ADSC, indicating that the electric stimulus itself can modulate processes controlling the synthesis and mobilization of fat, even in the absence of the concomitant thermal and mechanical components of the thermoelectric therapy CRET.


Subject(s)
Adipose Tissue/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Adipose Tissue/cytology , Cells, Cultured , Electric Stimulation , Humans , Mesenchymal Stem Cells/cytology , Mitogen-Activated Protein Kinase Kinases/metabolism , PPAR gamma/metabolism , Perilipin-1/metabolism
7.
Int J Mol Sci ; 17(4): 510, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27058530

ABSTRACT

The proliferative response of the neuroblastoma line NB69 to a 100 µT, 50 Hz magnetic field (MF) has been shown mediated by activation of the MAPK-ERK1/2 pathway. This work investigates the MF effect on the cell cycle of NB69, the participation of p38 and c-Jun N-terminal (JNK) kinases in the field-induced proliferative response and the potential involvement of reactive oxygen species (ROS) in the activation of the MAPK-ERK1/2 and -p38 signaling pathways. NB69 cultures were exposed to the 100 µT MF, either intermittently for 24, 42 or 63 h, or continuously for periods of 15 to 120 min, in the presence or absence of p38 or JNK inhibitors: SB203580 and SP600125, respectively. Antioxidant N-acetylcysteine (NAC) was used as ROS scavenger. Field exposure induced transient activation of p38, JNK and ERK1/2. The MF proliferative effect, which was mediated by changes in the cell cycle, was blocked by the p38 inhibitor, but not by the JNK inhibitor. NAC blocked the field effects on cell proliferation and p38 activation, but not those on ERK1/2 activation. The MF-induced proliferative effects are exerted through sequential upregulation of MAPK-p38 and -ERK1/2 activation, and they are likely mediated by a ROS-dependent activation of p38.


Subject(s)
Cell Proliferation , MAP Kinase Signaling System , Magnetic Fields/adverse effects , Neuroblastoma/etiology , Neuroblastoma/metabolism , Reactive Oxygen Species/metabolism , Cell Cycle , Cell Line, Tumor , Humans , Neuroblastoma/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Cell Physiol Biochem ; 37(6): 2297-310, 2015.
Article in English | MEDLINE | ID: mdl-26625130

ABSTRACT

BACKGROUND/AIMS: Semicircular lipoatrophy (SL) is an idiopathic condition characterized by atrophy of subcutaneous fatty tissue. Although several studies have suggested a possible association between SL and occupational exposure to power frequency magnetic fields (MF), no mechanism has been proposed so far that explains an influence of these fields on adipogenesis. METHODS: The study investigates the effects of a 50 Hz, 100 µT MF on the adipogenesis of stem cells isolated from human adipose tissue (ADSC). Cells were plated in Petri dishes and either exposed intermittently to the field for 42 hours or sham-exposed. RESULTS: Field exposure significantly reduced lipid accumulation within the cells, revealed in Oil Red O stained samples by spectrophotometry and colorimetry. Early cell passages were particularly sensitive to the effect: 30.40 ± 5.77% and 47.96 ± 12.47% below controls in the spectrophotometric and colorimetric assays, respectively. Such antiadipogenic effect was accompanied by significant changes in the expression of key effectors/regulators of early adipogenesis: PPARx03B3;, p-ERK1/2 and Sox9, indicating that at least the ERK/PPARx03B3; signaling pathway could be involved in the effect. CONCLUSIONS: These results constitute an experimental support to the hypothesis that power frequency MF can be one of the factors involved in the etiology of SL.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Magnetics , Adipose Tissue/metabolism , Cells, Cultured , Fluorescent Antibody Technique, Indirect , Humans , Lipid Metabolism , MAP Kinase Signaling System , PPAR gamma/metabolism , SOX9 Transcription Factor/metabolism
9.
Cell Physiol Biochem ; 34(5): 1741-55, 2014.
Article in English | MEDLINE | ID: mdl-25427571

ABSTRACT

BACKGROUND/AIMS: Capacitive-resistive electric transfer (CRET) is a non invasive electrothermal therapy that applies electric currents within the 400 kHz - 450 kHz frequency range to the treatment of musculoskeletal lesions. Evidence exists that electric currents and electric or magnetic fields can influence proliferative and/or differentiating processes involved in tissue regeneration. This work investigates proliferative responses potentially underlying CRET effects on tissue repair. METHODS: XTT assay, flow cytometry, immunofluorescence and Western Blot analyses were conducted to asses viability, proliferation and differentiation of adipose-derived stem cells (ADSC) from healthy donors, after short, repeated (5 m On/4 h Off) in vitro stimulation with a 448-kHz electric signal currently used in CRET therapy, applied at a subthermal dose of 50 µA/mm(2) RESULTS: The treatment induced PCNA and ERK1/2 upregulation, together with significant increases in the fractions of ADSC undergoing cycle phases S, G2 and M, and enhanced cell proliferation rate. This proliferative effect did not compromise the multipotential ability of ADSC for subsequent adipogenic, chondrogenic or osteogenic differentiation. CONCLUSIONS: These data identify cellular and molecular phenomena potentially underlying the response to CRET and indicate that CRET-induced lesion repair could be mediated by stimulation of the proliferation of stem cells present in the injured tissues.


Subject(s)
Cell Proliferation/physiology , Mesenchymal Stem Cells/physiology , Adipose Tissue/metabolism , Adipose Tissue/physiology , Adult , Aged , Cell Cycle/physiology , Cell Differentiation/physiology , Cells, Cultured , Electric Stimulation/methods , Electricity , Female , Humans , MAP Kinase Signaling System/physiology , Male , Mesenchymal Stem Cells/metabolism , Stem Cells/metabolism , Stem Cells/physiology , Up-Regulation/physiology
10.
PLoS One ; 9(1): e84636, 2014.
Article in English | MEDLINE | ID: mdl-24416255

ABSTRACT

Capacitive Resistive Electric Transfer (CRET) therapy applies currents of 0.4-0.6 MHz to treatment of inflammatory and musculoskeletal injuries. Previous studies have shown that intermittent exposure to CRET currents at subthermal doses exert cytotoxic or antiproliferative effects in human neuroblastoma or hepatocarcinoma cells, respectively. It has been proposed that such effects would be mediated by cell cycle arrest and by changes in the expression of cyclins and cyclin-dependent kinase inhibitors. The present work focuses on the study of the molecular mechanisms involved in CRET-induced cytostasis and investigates the possibility that the cellular response to the treatment extends to other phenomena, including induction of apoptosis and/or of changes in the differentiation stage of hepatocarcinoma cells. The obtained results show that the reported antiproliferative action of intermittent stimulation (5 m On/4 h Off) with 0.57 MHz, sine wave signal at a current density of 50 µA/mm(2), could be mediated by significant increase of the apoptotic rate as well as significant changes in the expression of proteins p53 and Bcl-2. The results also revealed a significantly decreased expression of alpha-fetoprotein in the treated samples, which, together with an increased concentration of albumin released into the medium by the stimulated cells, can be interpreted as evidence of a transient cytodifferentiating response elicited by the current. The fact that this type of electrical stimulation is capable of promoting both, differentiation and cell cycle arrest in human cancer cells, is of potential interest for a possible extension of the applications of CRET therapy towards the field of oncology.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Differentiation , Electric Stimulation Therapy , Liver Neoplasms/pathology , Temperature , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Cell Proliferation , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Intracellular Space/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Proto-Oncogene Proteins c-bcl-2 , Serum Albumin/metabolism , Tumor Suppressor Protein p53/metabolism , alpha-Fetoproteins/metabolism
11.
Oncol Rep ; 29(3): 885-94, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23292364

ABSTRACT

We previously reported that intermittent exposure to a 50­Hz magnetic field (MF) at 100 µT stimulates cell proliferation in the human neuroblastoma cell line NB69. The present study aimed to investigate whether the magnetic field-induced growth promotion also occurs at a lower magnetic flux density of 10 µT. To this purpose, NB69 cells were subjected for 42 h to intermittent exposure, 3 h on/3 h off, to a 50­Hz MF at a 10 or 100 µT magnetic flux density. The field exposure took place either in the presence or in the absence of the antiproliferative agent retinoic acid. At the end of the treatment and/or incubation period, the cell growth was estimated by hemocytometric counting and spectrophotometric analysis of total protein and DNA contents. Potential changes in DNA synthesis were also assessed through proliferating cell nuclear antigen (PCNA) immunolabeling. The results confirmed previously reported data that a 42-h exposure to a 50­Hz sine wave MF at 100 µT promotes cell growth in the NB69 cell line, and showed that 10 µT induces a similar proliferative response. This effect, which was significantly associated and linearly correlated with PCNA expression, was abolished by the presence of retinoic acid in the culture medium.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Magnetic Fields , Tretinoin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Neuroblastoma , Proliferating Cell Nuclear Antigen/metabolism
12.
Cell Physiol Biochem ; 30(6): 1502-16, 2012.
Article in English | MEDLINE | ID: mdl-23235525

ABSTRACT

BACKGROUND/AIMS: Epidemiological and experimental evidence exists indicating that exposure to weak, extremely low frequency magnetic fields (ELF - MF) could affect cancer progression. It has been proposed that such hypothetical action could be mediated by MF-induced effects on the cellular response to melatonin (MEL), a potentially oncostatic neurohormone. The present study investigates the response of HepG2 cells to intermittent exposure to a 50 Hz, 10 µT MF, in the presence or absence of MEL at physiological (10 nM) or pharmacological doses (1 µM). METHODS: The Trypan blue cell exclusion test, BrdU incorporation and PCNA expression assays were carried out to assess the cellular response in terms of viability and proliferation. In addition, albumin and alpha-fetoprotein, were analyzed as specific hepatocellular differentiation markers. RESULTS: The results indicate that the MF exerts significant cytoproliferative and dedifferentiating effects that can be prevented by 10 nM MEL. Conversely, MEL exerts cytostatic and differentiating effects on HepG2 that are abolished by simultaneous exposure to MF. CONCLUSION: As a whole, these results support the hypothesis that ELF - MF and MEL exert opposite, mutually counteracting effects on cell proliferation and differentiation.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Magnetic Fields , Melatonin/pharmacology , Albumins/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular , Cell Survival , DNA Replication , Hep G2 Cells , Humans , Melatonin/physiology , Proliferating Cell Nuclear Antigen/metabolism , alpha-Fetoproteins/metabolism
13.
Int J Oncol ; 41(4): 1251-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22843038

ABSTRACT

Recently, a number of electric and electrothermal therapies have been applied to the treatment of specific cancer types. However, the cellular and molecular mechanisms involved in the response to such therapies have not been well characterized yet. Capacitive-resistive electric transfer (CRET) therapy uses electric currents at frequencies within the 0.45-0.6 MHz range to induce hyperthermia in target tissues. Preliminary trials in cancer patients have shown consistent signs that CRET could slow down growth of tumor tissues in brain gliomas, without inducing detectable damage in the surrounding healthy tissue. Previous studies by our group have shown that subthermal treatment with 0.57-MHz electric currents can induce a cytostatic, not cytotoxic response in HepG2 human hepatocarcinoma cells; such effect being mediated by cell cycle alterations. In contrast, the study of the response of NB69 human neuroblastoma cells to the same electric treatment revealed consistent indications of cytotoxic effects. The present study extends the knowledge on the response of NB69 cells to the subthermal stimulus, comparing it to that of primary cultures of human peripheral blood mononuclear cells (PBMC) exposed to the same treatment. The results showed no sensitivity of PBMC to the 0.57 MHz subthermal currents and confirmed that the treatment exerts a cytotoxic action in NB69 cells. The data also revealed a previously undetected cytostatic response of the neuroblastoma cell line. CRET currents affected NB69 cell proliferation by significantly reducing the fraction of cells in the phase G2/M of the cell cycle at 12 h of exposure. These data provide new information on the mechanisms of response to CRET therapy, and are consistent with a cytotoxic and/or cytostatic action of the electric treatment, which would affect human cells of tumor origin but not normal cells with a low proliferation rate.


Subject(s)
Cell Proliferation/radiation effects , Leukocytes, Mononuclear/radiation effects , Neuroblastoma/therapy , Radio Waves , Cell Line, Tumor/radiation effects , Electricity , Humans , Leukocytes, Mononuclear/cytology , Neuroblastoma/pathology
14.
Cell Physiol Biochem ; 29(5-6): 675-86, 2012.
Article in English | MEDLINE | ID: mdl-22613968

ABSTRACT

A number of studies have reported that extremely low frequency magnetic fields (ELF-MF) can modulate proliferative processes in vitro; however, the transduction mechanisms implicated in such phenomena remain to be identified. The present study was aimed to determine whether a 50 Hz, 100 µT MF can induce cell proliferation in the human neuroblastoma line NB69, and whether the signaling pathway MAPK-ERK1/2 (Mitogen-Activated Protein Kinase - Extracellular-Signal-Regulated Kinase 1 and 2) is involved in that proliferative response. The cultures were exposed intermittently or continuously to the MF for a 63-hour duration. The continuous treatment did not induce significant changes in cell proliferation. In contrast, intermittent exposure caused statistically significant increase in the percent of cells in phase S of the cell cycle, followed by a significant increase in cell number. The intermittent treatment also induced an early, transient and repetitive activation of ERK1/2 that could be involved in the proliferative effects. In fact, both the proliferative response and the repeated activation of ERK1/2 were blocked by PD98059, the specific inhibitor of MEK (ERK kinases 1 and 2). Taken together, the described results indicate that a 50 Hz, 100 µT MF can stimulate proliferation in NB69 cells by triggering MAPK-ERK1/ 2 signaling at each of the "On" periods of an intermittent exposure.


Subject(s)
Cell Proliferation , MAP Kinase Signaling System , Magnetics , Neuroblastoma/pathology , Blotting, Western , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Immunohistochemistry , Neuroblastoma/enzymology , Phosphorylation
15.
Int J Oncol ; 40(5): 1405-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22293994

ABSTRACT

In vitro exposure to power frequency magnetic fields (MF) has been reported to influence cell proliferation and differentiation. However, the nature of the response of different human cancer cell types to these fields has not been sufficiently characterized. The present work investigates the response of two proliferating human cell lines of neuroblastoma (NB69) and hepatocarcinoma (HepG2) to a 42 h, intermittent treatment with a weak, 100 µT, 50 Hz MF, alone or in combination with 0.5 µM all-trans-retinol (ROL), a retinoid currently applied in oncostatic therapies. In each experimental replicate the cell samples were submitted to one of the following treatment combinations: MF+/ROL+, MF+/ROL-, MF-/ROL+ or MF-/ROL-. The proliferative response was determined by cell counting (Trypan blue exclusion), BrdU incorporation and by spectrophotometric analysis of total protein and DNA content. The results show that when administered separately, the two treatments, MF and ROL, significantly enhanced cell proliferation in both cell lines. In NB69 simultaneous administration of MF and ROL induced an additive effect on cell proliferation, associated to increased DNA content. By contrast, in HepG2 the ROL-induced cell proliferation and increased protein content were partially blocked by simultaneous exposure to MF. Taken together, these data show that both agents, a weak MF and ROL at a low concentration, induce proliferative responses in the two assayed human cell lines. However, significant differences were observed between the responses of the two cellular species to the combined treatment with ROL and MF, indicating that the mechanisms underlying the cellular response to each of the two agents can mutually interact in a manner that is cell type-specific.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Liver Neoplasms/pathology , Magnetic Fields , Neuroblastoma/pathology , Vitamin A/pharmacology , Vitamins/pharmacology , Carcinoma, Hepatocellular/genetics , Combined Modality Therapy , DNA Replication/drug effects , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Neuroblastoma/genetics , Spectrophotometry , Time Factors
16.
Int J Oncol ; 37(6): 1399-405, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21042707

ABSTRACT

The capacitive-resistive electric transfer (CRet) therapy is a non-invasive technique that applies electrical currents of 0.4-0.6 MHz to the treatment of musculoskeletal injuries. Although this therapy has proved effective in clinical studies, its interaction mechanisms at the cellular level still are insufficiently investigated. Results from previous studies have shown that the application of CRet currents at subthermal doses causes alterations in cell cycle progression and decreased proliferation in hepatocarcinoma (HepG2) and neuroblastoma (NB69) human cell lines. The aim of the present study was to investigate the antiproliferative response of HepG2 to CRet currents. The results showed that 24-h intermittent treatment with 50 µA/mm(2) current density induced in HepG2 statistically significant changes in expression and activation of cell cycle control proteins p27Kip1 and cyclins D1, A and B1. The chronology of these changes is coherent with that of the alterations reported in the cell cycle of HepG2 when exposed to the same electric treatment. We propose that the antiproliferative effect exerted by the electric stimulus would be primarily mediated by changes in the expression and activation of proteins intervening in cell cycle regulation, which are among the targets of emerging chemical therapies. The capability to arrest the cell cycle through electrically-induced changes in cell cycle control proteins might open new possibilities in the field of oncology.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cell Cycle Proteins/physiology , Cell Proliferation , Electric Stimulation Therapy , Liver Neoplasms/therapy , Algorithms , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Electric Stimulation Therapy/methods , Electricity , Fluorescence , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Models, Biological , Time Factors
17.
Int J Oncol ; 30(3): 583-92, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17273759

ABSTRACT

Capacitive-resistive electric transfer (CRET) therapy is a non-invasive technique currently applied to the treatment of skin, muscle and tendon injuries that uses 0.45-0.6 MHz electric currents to transdermically and focally increase the internal temperature of targeted tissues. Because CRET electrothermal treatment has been reported to be more effective than other thermal therapies, it has been proposed that the electric stimulus could induce responses in exposed tissues that are cooperative or synergic with the thermal effects of the treatment. Previous studies by our group, investigating the nature of the alleged electric response, have shown that short, repeated stimuli with 0.57-MHz currents at subthermal levels could provoke partial, cytotoxic effects on human neuroblastoma cells in vitro. The aim of the present study was to investigate the response from another human cell type, the human hepatocarcinoma HepG2 line, during and after the exposure to 0.57-MHz CRET currents at subthermal densities. The electric stimuli provoked a decrease in the proliferation rate of the cultures, possibly due to an electrically-induced blocking of the cell cycle in a fraction of the cellular population.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Bromodeoxyuridine/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Electric Conductivity , Electric Stimulation Therapy , Electricity , Electrochemistry/methods , Electromagnetic Fields , Humans , Microscopy, Fluorescence , Proliferating Cell Nuclear Antigen/biosynthesis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...