Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
2.
Mol Pharm ; 15(8): 3478-3488, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29953815

ABSTRACT

Baicalin (BG) is a natural glycoside with several promising therapeutic and preventive applications. However, its pharmaceutical potential is compromised by its poor water solubility, complex oral absorption kinetics, and low bioavailability. In this work, BG was incorporated in a series of chitosan (Ch)/glycerophosphate (GP)-based thermosensitive hydrogel formulations to improve its water solubility and control its release profile. Molecular interactions between BG and GP were investigated using Fourier transform infrared spectroscopy (FT-IR), and the ability of GP to enhance the water solubility of BG was studied in different release media. Drug-loaded Ch/GP hydrogels were prepared and characterized for their gelation time, swelling ratio, and rheological properties in addition to surface and internal microstructure. Polyethylene glycol (PEG) 6000 and hydroxypropyl methyl cellulose (HPMC) were incorporated in the formulations at different ratios to study their effect on modulating the sol-gel behavior and the in vitro drug release. In vivo pharmacokinetic (PK) studies were carried out using a rabbit model to study the ability of drug-loaded Ch/GP thermosensitive hydrogels to control the absorption rate and improve the bioavailability of BG. Results showed that the solubility of BG was enhanced in the presence of GP, while the incorporation of PEG and/or HPMC had an impact on gelation time, rheological behavior, and rate of drug release in vitro. PK results obtained following buccal application of drug-loaded Ch/GP thermosensitive hydrogels to rabbits showed that the rate of BG absorption was controlled and the in vivo bioavailability was increased by 330% relative to BG aqueous oral suspension. The proposed Ch/GP thermosensitive hydrogel is an easily modifiable delivery platform that is not only capable of improving the solubility and bioavailability of BG following buccal administration but also can be suited for various local and injectable therapeutic applications.


Subject(s)
Drug Carriers/chemistry , Flavonoids/pharmacokinetics , Administration, Buccal , Animals , Biological Availability , Chemistry, Pharmaceutical , Chitosan/chemistry , Drug Liberation , Flavonoids/administration & dosage , Flavonoids/chemistry , Glycerophosphates/chemistry , Hydrogels/chemistry , Male , Models, Animal , Polyethylene Glycols/chemistry , Rabbits , Rheology , Solubility , Spectroscopy, Fourier Transform Infrared , Temperature
3.
Mol Pharm ; 15(1): 247-255, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29232954

ABSTRACT

Atorvastatin calcium (AC)-loaded nanoparticles (NPs) of mean particle diameter <100 nm and narrow distribution were prepared and characterized. Their in vivo PK as well as PD measures following oral administration in different dosage regimens in hyperlipidemic rats were evaluated. The results revealed that the oral bioavailability of two selected AC-NPs formulations was 235% and 169% relative to Lipitor. However, the treatment regimens were not superior in reducing serum total cholesterol (TC), low-density lipoproteins (LDL), and triglycerides (TG) levels compared to Lipitor. Moreover, the AC-NPs treatments were associated with significant adverse effects observed biochemically and histologically. These results were contradictory with those obtained from a previous study in which similarly formulated AC-NPs of mean particle diameter >200 nm were found to be more safe and effective in reducing TC, LDL, and TG levels when administered to hyperlipiemic rats at reduced dosing frequency compared to daily dose of Lipitor despite their lower oral bioavailability. The discrepant correlation between pharmacokinetics (PK) and pharmacodynamics (PD) results was suggested to pertain to the different biodistribution profiles of AC-NPs depending on their sizes. Hereby, we provide a simple approach of particle size modulation to enhance the efficacy and safety of atorvastatin.


Subject(s)
Atorvastatin/chemistry , Atorvastatin/pharmacokinetics , Nanoparticles/chemistry , Administration, Oral , Animals , Atorvastatin/administration & dosage , Cholesterol/blood , Lipoproteins, LDL/blood , Male , Rats , Tissue Distribution , Triglycerides/blood
4.
Ultrason Sonochem ; 31: 206-15, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26964942

ABSTRACT

In this study, we report on the potential use of platinum nanoparticles (Pt-NPs), a superoxide dismutase (SOD)/catalase mimetic antioxidant, in combination with 1MHz ultrasound (US) at an intensity of 0.4 W/cm(2), 10% duty factor, 100 Hz PRF, for 2 min. Apoptosis induction was assessed by DNA fragmentation assay, cell cycle analysis and Annexin V-FITC/PI staining. Cell killing was confirmed by cell counting and microscopic examination. The mitochondrial and Ca(2+)-dependent pathways were investigated. Caspase-8 expression and autophagy-related proteins were detected by spectrophotometry and western blot analysis, respectively. Intracellular reactive oxygen species (ROS) elevation was detected by flow cytometry, while extracellular free radical formation was assessed by electron paramagnetic resonance spin trapping spectrometry. The results showed that Pt-NPs exerted differential effects depending on their internalization. Pt-NPs functioned as potent free radical scavengers when added immediately before sonication while pre-treatment with Pt-NPs suppressed the induction of apoptosis as well as autophagy (AP), and resulted in enhanced cell killing. Dead cells displayed the features of pyknosis. The exact mode of cell death is still unclear. In conclusion, the results indicate that US-induced AP may contribute to cell survival post sonication. To our knowledge this is the first study to discuss autophagy as a pro-survival pathway in the context of US. The combination of Pt-NPs and US might be effective in cancer eradication.


Subject(s)
Apoptosis/drug effects , Lymphoma/pathology , Metal Nanoparticles , Platinum/pharmacology , Humans , Platinum/chemistry , U937 Cells
5.
PLoS One ; 10(8): e0136956, 2015.
Article in English | MEDLINE | ID: mdl-26318000

ABSTRACT

Electron paramagnetic resonance (EPR)-spin trapping and flow cytometry were used to identify free radicals generated using argon-cold atmospheric plasma (Ar-CAP) in aqueous solutions and intracellularly in comparison with those generated by X-irradiation. Ar-CAP was generated using a high-voltage power supply unit with low-frequency excitation. The characteristics of Ar-CAP were estimated by vacuum UV absorption and emission spectra measurements. Hydroxyl (·OH) radicals and hydrogen (H) atoms in aqueous solutions were identified with the spin traps 5,5-dimethyl-1-pyrroline N-oxide (DMPO), 3,3,5,5-tetramethyl-1-pyrroline-N-oxide (M4PO), and phenyl N-t-butylnitrone (PBN). The occurrence of Ar-CAP-induced pyrolysis was evaluated using the spin trap 3,5-dibromo-4-nitrosobenzene sulfonate (DBNBS) in aqueous solutions of DNA constituents, sodium acetate, and L-alanine. Human lymphoma U937 cells were used to study intracellular oxidative stress using five fluorescent probes with different affinities to a number of reactive species. The analysis and quantification of EPR spectra revealed the formation of enormous amounts of ·OH radicals using Ar-CAP compared with that by X-irradiation. Very small amounts of H atoms were detected whereas nitric oxide was not found. The formation of ·OH radicals depended on the type of rare gas used and the yield correlated inversely with ionization energy in the order of krypton > argon = neon > helium. No pyrolysis radicals were detected in aqueous solutions exposed to Ar-CAP. Intracellularly, ·OH, H2O2, which is the recombination product of ·OH, and OCl- were the most likely formed reactive oxygen species after exposure to Ar-CAP. Intracellularly, there was no practical evidence for the formation of NO whereas very small amounts of superoxides were formed. Despite the superiority of Ar-CAP in forming ·OH radicals, the exposure to X-rays proved more lethal. The mechanism of free radical formation in aqueous solutions and an intracellular milieu is discussed.


Subject(s)
Argon/chemistry , Free Radicals/analysis , Intracellular Fluid/chemistry , Plasma Gases/chemistry , Solutions/chemistry , Cell Line , Electron Spin Resonance Spectroscopy/methods , Flow Cytometry/methods , Humans , Intracellular Fluid/radiation effects , Oxidative Stress/radiation effects , Solutions/radiation effects , Spin Trapping/methods , X-Rays
6.
Ultrason Sonochem ; 23: 339-46, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25287395

ABSTRACT

Previously, we have shown that a combination between X-irradiation and low-intensity pulsed ultrasound (US) could synergistically suppress cell survival post exposure (Buldakov et al., 2014). In this study, the cellular effects underlying the enhanced cell killing are investigated. U937 and Molt-4 cell lines were exposed to 1.0 MHz US with 50% duty factor at 0.3 W/cm(2) and pulsed at 1, 5 and 10 Hz immediately after exposure to X-rays at 0, 0.5, 2.5 and 5 Gy. The cells were assayed at different time points to depict the major cellular events that culminated in cell death. For instance, membrane damage and cell lysis were estimated immediately following exposure and 24 h later. Intracellular reactive oxygen species (ROS) were also determined flow cytometrically after treatment. Moreover, the extent of DNA damage and cell cycle progression were determined at 6 and 24 h, respectively. Despite the general trend for synergism, there was a disproportionation of mediating factors depending on the cell type and its specific biological makeup. Immediately, US could induce appreciable necrotic cell death through extensive membrane damage in U937 but induced cell lysis in Molt-4 cells. ROS might have contributed to cell killing in Molt-4 but not in U937 cells. Although both of the physical modalities are significantly DNA-damaging alone, no additional damage was observed in combination. Moreover, override in some arrested cell cycle phases was also observed following combination. Collectively, the interaction between X-rays and US seems to depend mainly on the acoustic environment determined by the setup and this might explain the contradictory data among reports.


Subject(s)
Leukemia/pathology , Ultrasonics , Cell Cycle/radiation effects , Cell Death/radiation effects , Cell Survival/radiation effects , DNA Damage , Humans , Radiation Dosage , U937 Cells , X-Rays
7.
Drug Dev Ind Pharm ; 41(3): 375-81, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24303901

ABSTRACT

The administration of grapefruit juice (GFJ) has been postulated to inhibit the activity of P-glycoprotein (P-gp) transport system and thus can enhance the uptake of substrate drugs. However, for various reasons, the results obtained have been always swaying between confirmation and refutation. This study aims at re-evaluating the effect of lyophilized freshly-prepared grapefruit juice (LGFJ) prepared from the whole peeled fruit on P-gp activity using the model drug doxorubicin (DOX) in-vitro and timolol maleate (TM) in-vivo. Human uterine sarcoma MES-SA/DX5v cells, grown under nanomolar concentration of DOX and highly expressing P-gp, were used as model cells for in-vitro studies whereas white New Zealand male rabbits were used for in-vivo studies. Results showed that the accumulation of DOX in MES-SA/DX5v cells was increased by 18.3 ± 2.0% in presence of LGFJ compared to control experiments. Results from in-vivo absorption studies showed that the relative oral bioavailability of TM ingested with LGFJ was significantly higher by 70% and 43% compared to the oral bioavailability of TM ingested with saline and a commercial GFJ, respectively. This study as such confirms the inhibitory effects of LGFJ on P-gp efflux proteins and highlights the superiority of using lyophilized freshly prepared juices over the commercially available juices in research studies. Also, the results call for further studies to assess the possibility of co-administrating LGFJ with anti-cancer agents to modulate multidrug resistance in their cellular environment or incorporating LGFJ in solid dosage forms to improve oral bioavailability of drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Citrus paradisi , Food-Drug Interactions/physiology , Pharmaceutical Preparations/metabolism , Plant Extracts/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cell Line, Tumor , Doxorubicin/metabolism , Doxorubicin/pharmacology , Freeze Drying/methods , Humans , Male , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rabbits
8.
Apoptosis ; 19(6): 1006-16, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24687228

ABSTRACT

Since polyacrylic acid capped platinum nano-particles (nano-Pts) are known to have a unique ability to quench superoxide (O2(-)) and hydrogen peroxide (H2O2), the anti-oxidant activity of nano-Pts against apoptosis induced by x-irradiation in human lymphoma U937 cells was investigated. DNA fragmentation assay, Annexin V-FITC/PI by flow cytometry and Giemsa staining revealed a significant decrease in apoptosis induced by 10 Gy, when cells were pre-treated with nano-Pts in a dose-dependent manner. Pre-treatment with nano-Pts significantly decreased radiation-induced reactive oxygen species (ROS) production, Fas expression and loss of mitochondrial membrane potential as determined by flow-cytometry. Furthermore, western blot analysis also showed that the expression of cleaved caspase-3, Bid and cytosolic cytochrome-c were significantly reduced in nano-Pts pretreated cells. Due to the catalase mimetic activity of nano-Pts, these results indicate that pre-treatment of U937 cells with nano-Pts significantly protect radiation-induced apoptosis by inhibiting intracellular ROS (mainly H2O2), which plays a key role in the induction of apoptosis, because of no practical observation of intracellular O2(-) formation.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Catalase/chemistry , Metal Nanoparticles , Platinum/pharmacology , Superoxide Dismutase/chemistry , Acrylic Resins , Apoptosis/radiation effects , Humans , Molecular Mimicry , Reactive Oxygen Species/metabolism , U937 Cells
9.
Ultrason Sonochem ; 21(6): 2061-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24657073

ABSTRACT

In recent years, data have been accumulating on the ability of ultrasound to affect at a distance inside the cell. Previous conceptions about therapeutic ultrasound were mainly based on compromising membrane permeability and triggering some biochemical reactions. However, it was shown that ultrasound can access deep to the nuclear territory resulting in enhanced macromolecular localization as well as alterations in gene and protein expression. Recently, we have reported on the occurrence of DNA double-strand breaks in different human cell lines exposed to ultrasound in vitro with some insight into the subsequent DNA damage response and repair pathways. The impact of these observed effects again sways between extremes. It could be advantageous if employed in gene therapy, wound and bone fracture-accelerated healing to promote cellular proliferation, or in cancer eradication if the DNA lesions would culminate in cell death. However, it could be a worrying sign if they were penultimate to further cellular adaptations to stresses and thus shaking the safety of ultrasound application in diagnosis and therapy. In this review, an overview of the rationale of therapeutic ultrasound and the salient knowledge on ultrasound-induced effects on the nucleus and genomic DNA will be presented. The implications of the findings will be discussed hopefully to provide guidance to future ultrasound research.


Subject(s)
Cell Nucleus , DNA , Genome , Ultrasonic Therapy/methods , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA/genetics , Humans
10.
PLoS One ; 8(3): e58325, 2013.
Article in English | MEDLINE | ID: mdl-23505488

ABSTRACT

The inhibition of DNA damage response pathway seems to be an attractive strategy for cancer therapy. It was previously reported that in rodent cells exposed to heat stress, cell growth was promoted by the activity of DNA-dependent protein kinase (DNA-PK), an enzyme involved in DNA non-homologous end joining (NHEJ) required for double-strand break repair. The absence of a functioning DNA-PK was associated with down regulation of heat shock protein 70 (HSP70). The objective of this study is thus to investigate the role of DNA-PK inhibition in heat-induced apoptosis in human cell lines. The inhibitors of phosphorylation of the DNA-PK catalytic subunit (DNA-PKcs) at Ser2056, such as NU7026 and NU7441, were utilized. Furthermore, knock down of DNA-PKcs was carried out using small interfering RNA (siDNA-PKcs). For heat exposure, cells were placed in water bath at 44°C for 60 min. Apoptosis was evaluated after 24 h incubation flow cytometrically. Proteins were extracted after 24 h and analyzed for HSP70 and HSP40 expression by Western blotting. Total RNA was extracted 6 h after treatment and analyzed using a GeneChip® microarray system to identify and select the up-regulated genes (≥1.5 fold). The results showed an enhancement in heat-induced apoptosis in absence of functioning DNA-PKcs. Interestingly, the expression levels of HSP70 and HSP40 were elevated in the absence of DNA-PKcs under heat stress. The results of genetic network analysis showed that HSPs and JUN genes were up-regulated independently of DNA-PKcs in exposed parent and knock out cells. In the presence of functioning DNA-PKcs, there was an observed up-regulation of anti-apoptotic genes, such as NR1D1, whereas in the absence of DNA-PKcs the pro-apoptotic genes, such as EGR2, were preferentially up-regulated. From these findings, we concluded that in human cells, the inactivation of DNA-PKcs can promote heat-induced apoptosis independently of heat-shock proteins.


Subject(s)
Apoptosis , DNA-Activated Protein Kinase/metabolism , Heat-Shock Proteins/metabolism , Hot Temperature , Animals , Apoptosis/drug effects , Apoptosis/genetics , CHO Cells , Cell Line, Tumor , Cricetinae , DNA Damage , DNA-Activated Protein Kinase/genetics , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Gene Silencing , Humans , Protein Kinase Inhibitors/pharmacology , RNA Interference , Reproducibility of Results
11.
PLoS One ; 7(10): e47903, 2012.
Article in English | MEDLINE | ID: mdl-23110127

ABSTRACT

Alkannin is an active constituent from the root extract of Alkanna tinctoria of the Boraginaceae family and it may have utility as a heat shock protein 70 (HSP70) inducer in living organisms. Here, the effects of alkannin-induced HSP70 on ultraviolet (UV) B (40 mJ/cm(2))-induced apoptosis were investigated in human keratinocyte HaCaT cells. Pretreatment of cells with alkannin (1 µM) caused significant inhibition of UVB-induced apoptosis and caspase-3 cleavage. On the other hand, the addition of KNK437 (HSP70 inhibitor) reversed the action of alkannin increasing UVB-induced apoptosis in a dose-dependent manner. In addition, differences in gene expression associated with the suppression of UVB-induced apoptosis in the presence of alkannin were investigated using Gene Chip assay. Our results indicate that alkannin suppresses UVB-induced apoptosis through the induction of HSP70 in human keratinocytes, and therefore, we suggest the usefulness of using alkannin as an antiaging agent.


Subject(s)
Apoptosis/drug effects , HSP70 Heat-Shock Proteins/metabolism , Keratinocytes/drug effects , Naphthoquinones/pharmacology , Ultraviolet Rays/adverse effects , Analysis of Variance , Apoptosis/radiation effects , Benzhydryl Compounds , Blotting, Western , Caspase 3/metabolism , Cell Line , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Humans , Immunohistochemistry , Keratinocytes/radiation effects , Naphthoquinones/metabolism , Oligonucleotide Array Sequence Analysis , Pyrrolidinones , Real-Time Polymerase Chain Reaction
12.
Eur J Pharm Sci ; 47(4): 768-73, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-22921720

ABSTRACT

The capability to controllably disrupt the cell membrane by ultrasound (US), thus facilitating entry of exogenous species, has now reached a state of some maturity. However, a compelling question asks whether there is a residual role for US in enhancing transfection: that is, once the genetic material has been delivered to the cytosol, can US assist in its transport into the nucleus? The present experiment was designed with a view to addressing this question. As such, our experimental setup discriminates between: (i) the precursor cell membrane permealization step, and (ii) any subsequent intracellular trafficking into the nucleus. In this study, calcium phosphate co-precipitates (CaP) were used to internalize plasmid DNA encoding for luciferase (pDNA-Luc) (>90%) in HeLa cells. After 2h incubation with the CaP-pDNA-Luc, cells were washed and insonated for varying durations. The results showed that US can indeed enhance the intracellular trafficking of previously internalized genes when longer insonation periods are implemented, culminating with an increased probability for successful nuclear localization, as inferred from an enhanced luciferase expression. Moreover, the results suggest that the intracellular role of US might be mediated through a pathway that appears not to be limited to destabilizing the endosomal vesicles. The study thus provides new information regarding the intracellular effects of US, and in effect represents a new modality combining US and CaP carriers for improved efficiency in gene delivery.


Subject(s)
Calcium Phosphates/chemistry , DNA/administration & dosage , Transfection/methods , Ultrasonics/methods , Biological Transport , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane Permeability/genetics , Cell Nucleus/metabolism , DNA/genetics , DNA/metabolism , Endosomes/metabolism , Gene Transfer Techniques , HeLa Cells , Humans , Luciferases/metabolism , Plasmids/genetics , Plasmids/metabolism , Transport Vesicles/metabolism
13.
Ultrason Sonochem ; 19(6): 1246-51, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22571845

ABSTRACT

Low-intensity ultrasound (US) has been shown to induce death of cancer cells; however, the underlying mechanism remains unclarified. Here, we provide novel evidence that the inhibition of checkpoint kinase 1 (Chk1) by a selective inhibitor or small interfering RNA (siRNA) enhances US-induced apoptosis in Jurkat cells. Jurkat cells showed insignificant lysis immediately after US at any applied intensity, whereas approximately 70% of the cells were γH2AX-positive 30min after US at 0.4W/cm(2). Regarding DNA damage response (DDR), Chk1, known as a target of ataxia telangiectasia mutated (ATM) and rad3-related (ATR), was phosphorylated in cells after US exposure. An ATM inhibitor showed nearly no effect on Chk1 phosphorylation, whereas chemicals showing the ATR inhibitory effect markedly abrogated the phosphorylation, indicating that Chk1 phosphorylation is preferentially more dependent on ATR than on ATM in cells exposed to US. The pharmacological inhibition of Chk1 promoted caspase-3 cleavage and increased the percentage of cells in SubG1 after US exposure. siRNA targeting Chk1 abrogated approximately 55% of Chk1 expression and also promoted apoptosis, suggesting that Chk1 plays anti-apoptotic roles in response to US. These findings revealed, for the first time, that US activates Chk1 dependently on ATR and the activated Chk1 is involved in apoptosis of cells exposed to US. Moreover, we propose that Chk1 may be a promising target in US-aided therapy.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Leukemia/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Sonication , Tumor Suppressor Proteins/metabolism , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Checkpoint Kinase 1 , DNA Damage , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , HL-60 Cells , Humans , Jurkat Cells , Leukemia/drug therapy , Leukemia/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering , Structure-Activity Relationship , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , U937 Cells
14.
Cancer Lett ; 322(1): 107-12, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22366497

ABSTRACT

Ultrasound (US) has been shown to induce cell death in cancer cells; however, the underlying mechanism remains elusive. Here, we report a set of novel findings on the molecular mechanism. We found that Akt (also known as protein kinase B), a substrate of DNA-dependent protein kinase (DNA-PK), was phosphorylated in U937 cells nullified with p53 or Molt-4 cells artificially abrogated with p53 after US exposure. On the contrary, Akt phosphorylation was transiently down-regulated then recovered in Molt-4 cells harboring wild-type p53 in US-exposed cells, possibly due to a mutual regulation between p53 and Akt. Inhibition of ataxia-telangiectasia mutated (ATM) or DNA-PK revealed that DNA-PK, rather than ATM, was preferentially involved in Akt phosphorylation and cell survival after US-exposure in all cell lines. These results indicate that DNA-PK plays a protective role against US-induced cell death regardless of p53 phenotype. In conclusion, our findings provide the first delineation of the role of DNA-PK in US-induced cell death and suggest that targeting DNA-PK might be a promising strategy to augment cancer eradication by US.


Subject(s)
Apoptosis , DNA-Activated Protein Kinase/physiology , Leukemia/therapy , Ultrasonic Therapy , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/physiology , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Binding Proteins/physiology , Humans , Leukemia/pathology , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , U937 Cells
15.
PLoS One ; 7(1): e29012, 2012.
Article in English | MEDLINE | ID: mdl-22235259

ABSTRACT

Ultrasonic technologies pervade the medical field: as a long established imaging modality in clinical diagnostics; and, with the emergence of targeted high intensity focused ultrasound, as a means of thermally ablating tumours. In parallel, the potential of [non-thermal] intermediate intensity ultrasound as a minimally invasive therapy is also being rigorously assessed. Here, induction of apoptosis in cancer cells has been observed, although definitive identification of the underlying mechanism has thus far remained elusive. A likely candidate process has been suggested to involve sonochemical activity, where reactive oxygen species (ROS) mediate the generation of DNA single-strand breaks. Here however, we provide compelling new evidence that strongly supports a purely mechanical mechanism. Moreover, by a combination of specific assays (neutral comet tail and staining for γH2AX foci formation) we demonstrate for the first time that US exposure at even moderate intensities exhibits genotoxic potential, through its facility to generate DNA damage across multiple cancer lines. Notably, colocalization assays highlight that ionizing radiation and ultrasound have distinctly different signatures to their respective γH2AX foci formation patterns, likely reflecting the different stress distributions that initiated damage formation. Furthermore, parallel immuno-blotting suggests that DNA-PKcs have a preferential role in the repair of ultrasound-induced damage.


Subject(s)
DNA Breaks, Double-Stranded , Mechanical Phenomena , Ultrasonics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromones/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA-Binding Proteins/metabolism , Histones/metabolism , Humans , Morpholines/pharmacology , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Pyrones/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tumor Suppressor Proteins/metabolism
16.
Int J Hyperthermia ; 28(1): 1-8, 2012.
Article in English | MEDLINE | ID: mdl-22235779

ABSTRACT

Environmental stress induces damage that activates an adaptive response in any organism. The cellular stress response is based on the induction of cytoprotective proteins, the so-called stress or heat shock proteins (HSPs). HSPs are known to function as molecular chaperones which are involved in the therapeutic approach of many diseases. Therefore in the current study we searched nontoxic chaperone inducers in chemical compounds isolated from medicinal plants. Screening of 80 compounds for their Hsp70-inducing activity in human lymphoma U937 cells was performed by western blotting. Five compounds showed significant Hsp70 up-regulation among them shikonin was most potent. Shikonin was able to induce Hsp70 at 0.1 µM after 3 h without activation of heat shock transcription factor 1 (HSF-1). It also induces significant reactive oxygen species generation. The expression level of genes responsive to shikonin was studied using global-scale microarrays and computational gene expression analysis tools. Significant increase in the nuclear factor erythroid 2-related factor 2 (Nrf2, NFEL2L2) -mediated oxidative stress response was observed that leads to the activation of HSP. The results of gene chip analysis were further confirmed by real-time qPCR assay. In short, the detailed mechanisms of Hsp70 induction by shikonin is not fully understood, Nrf2 and its target genes might be involved in the Hsp70 up-regulation in U937 cells.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Heat-Shock Proteins/genetics , Naphthoquinones/pharmacology , Plants, Medicinal , Apoptosis/drug effects , Gene Expression Regulation/drug effects , Heat-Shock Proteins/metabolism , Hot Temperature , Humans , Oligonucleotide Array Sequence Analysis , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , U937 Cells
17.
PLoS One ; 7(12): e48291, 2012.
Article in English | MEDLINE | ID: mdl-23284614

ABSTRACT

The acoustic effects in a biological milieu offer several scenarios for the reversal of multidrug resistance. In this study, we have observed higher sensitivity of doxorubicin-resistant uterine sarcoma MES-SA/DX5 cells to ultrasound exposure compared to its parent counterpart MES-SA cells; however, the results showed that the acoustic irradiation was genotoxic and could promote neotic division in exposed cells that was more pronounced in the resistant variant. The neotic progeny, imaged microscopically 24 hr post sonication, could contribute in modulating the final cell survival when an apoptotic dose of doxorubicin was combined with ultrasound applied either simultaneously or sequentially in dual-treatment protocols. Depending on the time and order of application of ultrasound and doxorubicin in combination treatments, there was either desensitization of the parent cells or sensitization of the resistant cells to doxorubicin action.


Subject(s)
Drug Resistance, Neoplasm , Ultrasonics , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Histones/metabolism , Humans , Phosphorylation/drug effects , Time Factors
18.
J Med Ultrason (2001) ; 38(2): 65-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-27278421

ABSTRACT

PURPOSE: Although sanazole has been used as a hypoxic radiosensitizer, we recently reported on its ability to sensitize U937 cells to hyperthermia and X-irradiation under aerobic conditions, enhancing apoptotic cell death following the combined treatment. The current study was undertaken to evaluate the effect of sanazole as a sonosensitizer under previously studied acoustic conditions of different pulse repetition frequencies, using two cell lines representative of solid tumours and haematopoietic cancers. METHODS: Cells were treated with different doses of sanazole. Flow-cytometric analysis and DNA fragmentation assay were carried out at different times, and morphological features were also inspected. For ultrasound treatment, cells were pre-incubated with a non-cytotoxic dose of sanazole for 30 min before exposure. Evaluation of cell killing and a parallel examination of intracellular oxidative stress levels in both cell lines were performed using flow cytometry. RESULTS: Sanazole alone displayed selective cytotoxic effects towards solid tumour-derived cancer cells, resulting in complete cell death after 24 h of treatment, and enhanced the ultrasound-induced cell killing 6 h post-treatment. The enhancement seemed to be mediated by an additive increase in intracellular oxidative stress levels. CONCLUSION: Sanazole seems to be an efficient cytotoxic agent for the treatment of solid tumours and a promising sonosensitizer under aerobic conditions.

19.
Free Radic Res ; 45(3): 326-35, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21047173

ABSTRACT

Platinum nanoparticles (Pt-NPs) are known to possess anti-tumouric activity and the ability to scavenge superoxides and peroxides indicating that they can act as superoxide dismutase (SOD)/catalase mimetics. These potentials seem useful in the protection and/or amelioration of oxidative stress-associated pathologies, but, when they are combined with a therapeutic modality that depends upon the mediation of reactive oxygen species in cell killing induction, the effect of Pt-NPs might be questionable. Here, the effects of polyacrylic acid-capped Pt-NPs (nano-Pts) on hyperthermia (HT)-induced apoptosis and the underlying molecular mechanisms were investigated in human myelomonocytic lymphoma U937 and human cutaneous T-cell lymphoma HH cells. The results showed that the pre-treatment with nano-Pts significantly inhibited HT-induced apoptosis in a dose-dependent manner. Superoxide, but not peroxides, was suppressed to varying extents. All pathways involved in apoptosis execution were also negatively affected. The results reveal that the combination of nano-Pts and HT could result in HT-desensitization.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catalase/metabolism , Hot Temperature , Nanoparticles , Platinum/chemistry , Platinum/pharmacology , Superoxide Dismutase/metabolism , Acrylic Resins , Antineoplastic Agents/chemistry , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , DNA Fragmentation , Flow Cytometry , Free Radical Scavengers/chemistry , Humans , Lymphoma , Peroxides/metabolism , Platinum/therapeutic use , Reactive Oxygen Species/metabolism , Superoxides/metabolism , U937 Cells
20.
Drug Discov Today ; 15(21-22): 892-906, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20727981

ABSTRACT

Intracellular calcium (Ca(2+)) transients have been observed in association with exposure to therapeutic ultrasound and correlated to both early- and late-onset bioeffects. For example, it has been suggested that early 'ultra-short' Ca(2+) transients recorded during sonoporation can mediate Ca(2+)-dependent exocytosis and endocytosis processes as complementary mechanisms for membrane self-sealing. Moreover, apoptosis induction has been reported to occur through a partial mediation of a Ca(2+)-dependent pathway. In this review, we attempt to assemble the salient facts into a cogent whole, with special attention given to the relationships arising through altered Ca(2+) levels, which underscore its crucial role during ultrasonic interactions with biological systems and its consequent implications in the context of therapeutics.


Subject(s)
Apoptosis , Calcium/metabolism , Endocytosis , Exocytosis , Ultrasonic Therapy , Cell Physiological Phenomena , Humans , Membrane Potentials , Permeability , Signal Transduction , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...