Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Am J Cancer Res ; 13(10): 4918-4930, 2023.
Article in English | MEDLINE | ID: mdl-37970336

ABSTRACT

Radiation therapy is one of the most commonly used cancer treatments. However, it has important concerns such as damage to normal tissues around cancers and radioresistance. To overcome these problems, combination therapy using radiosensitizer and radiotherapy will be a good alternative. The present study investigated the effects of AZD7648 on overcoming radioresistance as well as radiosensitizing in Hep3B xenografts and cells. The results showed that AZD7648 enhanced ionizing radiation (IR)-induced tumor growth not only in radiosensitive but also radioresistant tumors. In particular, the combination of AZD7648 with radiation reduced the expression of hypoxia induce factor-1α (HIF-1α) in radioresistant tumors. In vitro studies, AZD7648 plus IR increased IR-induced G2/M arrest and regulated cell cycle checkpoints such as cyclinB1, p-cdc2 in normoxia but not in hypoxia. AZD7648 induced more radiation-mediated ROS than radiation only under normoxia, but these ROS were not altered by AZD7648 under hypoxia. Interestingly, AZD7648 downregulated HIF-1α expression level under CoCl2-treated hypoxic condition but not in normoxic condition. In conclusion, AZD7648 synergistically increased radiosensitivity through accumulating IR-induced G2/M arrest and further improved radioresistance via regulation of HIF-1α. The present data suggest that AZD7648 may be a strong radiosensitizer in radioresistant as well as radiosensitive cancers.

2.
Neurobiol Aging ; 132: 175-184, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37837733

ABSTRACT

The anti-inflammatory efficacy of radiation therapy (RT) with single fractions below 1.0 Gy has been demonstrated in Alzheimer's disease mouse models. As neuroinflammation is also a major pathological feature of Parkinson's disease (PD), RT may also be effective in PD treatment. Therefore, this study aimed to investigate the anti-inflammatory effect of low-moderate dose RT (LMDRT, 0.6 Gy/single dose, for 5 days) exposure in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 30 mg/kg, intraperitoneally, for 5 consecutive days)-induced PD mouse model. Importantly, LMDRT reduced the levels of glial fibrillary acidic protein and intercellular adhesion molecule-1 (CD54) in the striatum region, which increased following MPTP administration. LMDRT also modulated inflammatory gene expression patterns in the substantia nigra region of the MPTP-treated mice. However, LMDRT had no direct effects on the severe loss of dopaminergic neurons and impaired motor behavior in the rotarod test. These results indicate that LMDRT has anti-inflammatory effects by modulating neuroinflammatory factors, including glial fibrillary acidic protein and intercellular adhesion molecule-1, but showed no behavioral improvements or neuroprotection in the MPTP-induced mouse model of PD.


Subject(s)
Brain , Glial Fibrillary Acidic Protein , Parkinson Disease , Animals , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use , Brain/metabolism , Brain/radiation effects , Disease Models, Animal , Dopaminergic Neurons/pathology , Glial Fibrillary Acidic Protein/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/pharmacology , Intercellular Adhesion Molecule-1/therapeutic use , Mice, Inbred C57BL , Parkinson Disease/metabolism , Parkinson Disease/radiotherapy , Substantia Nigra/metabolism
3.
Am J Cancer Res ; 13(12): 5887-5900, 2023.
Article in English | MEDLINE | ID: mdl-38187039

ABSTRACT

Inhibition of DNA-dependent protein kinase (DNA-PK) in the non-homologous end-joining repair pathway reportedly increases the radiation sensitivity of cancer cells. We have recently reported that BR101801, a novel triple inhibitor of PI3K-gamma (γ), delta (δ), and DNA-PK, functions as an efficient sensitizer of radiation-induced DNA damage in various human solid cancer cells and a xenograft mouse model. Given that the p53 tumor suppressor gene plays an important role in radiotherapeutic efficacy, in the current study, we focused on the impact of the p53 status on BR101801-induced radiosensitization using isogenic HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cell lines. In vitro, HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cells were pretreated with 1 µM BR101801 for 24 h before exposure to ionizing radiation (IR), followed by assays to analyze colony formation, DNA damage, cell cycle changes, senescence, autophagy, apoptosis, and DNA damage response-related proteins. Xenograft mouse models were constructed to examine the potential synergistic effects of BR101801 (50 mg/kg, orally administered once daily) and fractionated IR (2 Gy × 3 days) on tumor growth inhibition in vivo. BR101801 inhibited cell proliferation and prolonged DNA damage in both HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cells. Combined treatment with BR101801 and IR robustly induced G2/M phase cell cycle arrest, apoptosis, and cellular senescence in HCT116 p53-/- cells when compared with treatment with IR alone. Furthermore, BR101801 synergistically inhibited tumor growth in the HCT116 p53-/- xenograft mouse model. BR101801 enhanced the radiosensitivity of HCT116 human colorectal cancer cells regardless of their p53 status. Moreover, BR101801 exerted robust synergistic effects on IR-induced cell cycle arrest, apoptosis, and tumor growth inhibition, even in radioresistant HCT116 p53-/- cells. Overall, these findings provide a scientific rationale for combining BR101801 with IR as a new therapeutic strategy to overcome radioresistance induced by p53 deficiency.

4.
Am J Cancer Res ; 11(11): 5440-5451, 2021.
Article in English | MEDLINE | ID: mdl-34873471

ABSTRACT

DNA-dependent protein kinase (DNA-PK), an essential component of the non-homologous end-joining (NHEJ) repair pathway, plays an important role in DNA damage repair (DDR). Therefore, DNA-PK inhibition is a promising approach for overcoming radiotherapy or chemotherapy resistance in cancers. In this study, we demonstrated that BR101801, a potent DNA-PK inhibitor, acted as an effective radiosensitizer in various human solid cancer cells and an in vivo xenograft model. Overall, BR101801 strongly elevated ionizing radiation (IR)-induced genomic instability via induction of cell cycle G2/M arrest, autophagic cell death, and impairment of DDR pathway in human solid cancer cells. Interestingly, BR101801 inhibited not only phosphorylation of DNA-PK catalytic subunit in NHEJ factors but also BRCA2 protein level in homologous recombination (HR) factors. In addition, combination BR101801 and IR suppressed tumor growth compared with IR alone by reducing phosphorylation of DNA-PK in human solid cancer xenografts. Our findings suggested that BR101801 is a selective DNA-PK inhibitor with a synergistic radiosensitizing effect in human solid cancers, providing evidence for clinical applications.

5.
Am J Cancer Res ; 11(8): 3935-3945, 2021.
Article in English | MEDLINE | ID: mdl-34522459

ABSTRACT

The Human Epidermal Growth Receptor 2, or the HER2 is one of the highest expressed negative receptor that constitutes approximately 15-20% of malevolent breast cancerous tumors among women. The prevalence of HER2 has untimely and unfavorable consequences on breast cancer, and its underlying carcinomous cell processes, structures, and growth. Trastuzumab (TRZ), a humanized antibody that is rooted in relatively recent foundations, has been found operational in its construction of treatments against HER2-positive breast cancer. This drug is combined with radiotherapy or chemotherapy to deregulate HER2 genes in the body. However, patients who suffer from evolved tumors in advanced stages of cancer exhibit a good amount of tolerance towards singularly used TRZ treatment. Inversely, the factorization of Tumor Testing Fields (TTFields or TTFs) into cancer therapy revives the functions of a TRZ treatment plan, by sensitizing the HER2 genes to the drug. In turn, this facilitates TRZ to continue limiting cancerous cell multiplication and toxicity levels within the treatment. This research evaluates the aspects and effects of this pairing, both in vivo and in vitro through BT474 cells. The TTFields conduct an electromagnetic boundary, which generates sine-wave radiations to manipulate the HER2 gene structure. The methods followed in the research also examines the gene cell cultures and their viability through solutions like Tryptophan blue, or the Crystal violet which may or may not deliver certain testmants to the experiment. The Western Blot Test and the IHC confirm the presence of antibodies and negative receptors in the BT474 cells. These procedures contribute to the formulation of a treatment plan that overcomes the TRZ-resistant nature of the tumor, which is essentially the aim of the research. Thus, the paper substantiates that a healthy combination of TTF's with TRZ can enhance the penetration of TRZ after inducing apoptosis due to TTFields therapy. The success of a TTField in undertaking this pursuit makes room for more utilization of it in future cancerous treatment ventures.

7.
Int J Mol Sci ; 21(3)2020 Jan 29.
Article in English | MEDLINE | ID: mdl-32013255

ABSTRACT

The tropical basidiomycete fungus Phellinus linteus (Mesima) exhibits anti-tumor, anti-angiogenic, and immunomodulatory properties in various cancers including prostate, colon, and lung cancer along with melanoma by, for example, inducing apoptosis or cell cycle arrest. However, whether medina also facilitates treatment of hepatocellular carcinoma (HCC), the third global cause of cancer deaths, remains unknown. Here, we examined its potential as a radiosensitizer in HCC radiotherapy using human HCC Hep3B and HepG2 cell lines and xenograft tumors. Mesima pretreatment significantly enhanced HCC cell radiosensitivity in vitro and the combination of mesima + radiation treatment significantly reduced xenograft tumor growth and size in vivo compared to those with single treatments. Mechanistically, mesima significantly enhanced radiotherapy efficiency by inhibiting tumor cell survival through inducing apoptosis (assessed via annexin V), impairing cell cycle regulation (shown by flow cytometry), and reducing radiation-induced DNA damage repair (measured via γ-H2AX foci). Combination treatment also facilitated autophagic cell death beyond that from single treatments (assessed by quantifying stained acidic vesicular organelles), and diminished tumor cell metastatic potentials (shown by wound and Transwell assays). These findings support the synergistic anti-tumor effects of mesima combined with radiation and suggest scientific evidence for mesima as a radiosensitizer in HCC.


Subject(s)
Autophagic Cell Death , Basidiomycota/chemistry , Carcinoma, Hepatocellular , Liver Neoplasms , Radiation-Sensitizing Agents/pharmacology , Animals , Autophagic Cell Death/drug effects , Autophagic Cell Death/radiation effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/radiotherapy , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/radiotherapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/biosynthesis , Radiation-Sensitizing Agents/chemistry , Xenograft Model Antitumor Assays
8.
Cancers (Basel) ; 11(11)2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31752184

ABSTRACT

Osteosarcoma (OS) originates from osteoid bone tissues and is prone to metastasis, resulting in a high mortality rate. Although several treatments are available for OS, an effective cure does not exist for most patients with advanced OS. Zoledronic acid (ZOL) is a third-generation bisphosphonate that inhibits osteoclast-mediated bone resorption and has shown efficacy in treating bone metastases in patients with various types of solid tumors. Here, we sought to clarify the mechanisms through which ZOL inhibits OS cell proliferation. ZOL treatment inhibited OS cell proliferation, viability, and colony formation. Autophagy inhibition by RNA interference against Beclin-1 or ATG5 inhibited ZOL-induced OS cell death. ZOL induced autophagy by repressing the protein kinase B/mammalian target of rapamycin/p70S6 kinase pathway and extracellular signal-regulated kinase signaling-dependent autophagy in OS cell lines and patient-derived OS cells. Microarrays of miRNA showed that ZOL increased the levels of miR-212-3p, which is known to play an important role in autophagy, in OS in vitro and in vivo systems. Collectively, our data provided mechanistic insight into how increased miR-212-3p through ZOL treatment induces autophagy synergistically in OS cells, providing a preclinical rationale for conducting a broad-scale clinical evaluation of ZOL + miR-212-3p in treating OS.

9.
Oncogene ; 38(39): 6630-6646, 2019 09.
Article in English | MEDLINE | ID: mdl-31375748

ABSTRACT

Tumor-treating fields (TTFs) - a type of electromagnetic field-based therapy using low-intensity electrical fields - has recently been characterized as a potential anticancer therapy for glioblastoma multiforme (GBM). However, the molecular mechanisms involved remain poorly understood. Our results show that the activation of autophagy contributes to the TTF-induced anti-GBM activity in vitro or in vivo and GBM patient stem cells or primary in vivo culture systems. TTF-treatment upregulated several autophagy-related genes (~2-fold) and induced cytomorphological changes. TTF-induced autophagy in GBM was associated with decreased Akt2 expression, not Akt1 or Akt3, via the mTOR/p70S6K pathway. An Affymetrix GeneChip miRNA 4.0 Array analysis revealed that TTFs altered the expression of many microRNAs (miRNAs). TTF-induced autophagy upregulated miR-29b, which subsequently suppressed the Akt signaling pathway. A luciferase reporter assay confirmed that TTFs induced miR-29b to target Akt2, negatively affecting Akt2 expression thereby triggering autophagy. TTF-induced autophagy suppressed tumor growth in GBM mouse models subjected to TTFs as determined by positron emission tomography and computed tomography (PET-CT). GBM patient stem cells and a primary in vivo culture system with high Akt2 levels also showed TTF-induced inhibition. Taken together, our results identified autophagy as a critical cell death pathway triggered by TTFs in GBM and indicate that TTF is a potential treatment option for GBM.


Subject(s)
Autophagy/radiation effects , Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , MicroRNAs/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/radiation effects , Animals , Apoptosis/radiation effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/radiation effects , Electromagnetic Fields , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Proto-Oncogene Proteins c-akt/metabolism , Xenograft Model Antitumor Assays
10.
Technol Cancer Res Treat ; 18: 1533033819845008, 2019 01 01.
Article in English | MEDLINE | ID: mdl-31072204

ABSTRACT

This study aimed to evaluate the biological effectiveness of cancer therapy with tumor treating fields using a fractionated treatment scheme that was originally designed for radiotherapy. Discontinuous fractional tumor treating fields of an intensity of 0.9 to 1.2 V/cm and a frequency of 150 KHz were applied to U373 cancer cells and IEC6 normal cells for 3 days, with durations of 3, 6, 12, or 24 h/d. As the treatment duration of the tumor treating fields increased from 3 to 24 h/d, the relative tumor cell (U373) number (% of control) reduced in proportion to the treatment duration. Compared to a 25% cell number reduction (75% of control) for the group of 6 h/d treatment at 1.2 V/cm, only 5% (70% of control) and 8% (67% of control) of additional reductions were observed for the group of 12 and 24 h/d treatment, respectively. This experimental result indicates that the dependence on treatment duration in tumor cell inhibition was weakened distinctly at treatment duration over 6 h/d. For normal cells (IEC6), the relative cell number corresponding to the treatment time of the tumor treating fields at 1.2 V/cm of electric field strength was not decreased much for the treatment times of 3, 6, and 12 h/d, revealing 93.3%, 90.0%, and 89.3% relative cell numbers, respectively, but it suddenly decreased to ∼73% for the 24 h/d treatment. Our results showed that the effects of tumor treating fields on tumor cells were higher than on normal cells for treatment duration of 3 to 12 h/d, but the difference became minimal for treatment duration of 24 h/d. The fractionated scheme, using tumor treating fields, reduced the treatment time while maintaining efficacy, suggesting that this method may be clinically applicable for cancer treatment.


Subject(s)
Dose Fractionation, Radiation , Head and Neck Neoplasms/radiotherapy , Apoptosis/radiation effects , Biomarkers , Cell Cycle/radiation effects , Cell Line, Tumor , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/etiology , Head and Neck Neoplasms/metabolism , Humans , Immunohistochemistry , Radiotherapy Planning, Computer-Assisted , Treatment Outcome
11.
Cell Death Discov ; 4: 46, 2018.
Article in English | MEDLINE | ID: mdl-30302280

ABSTRACT

Tumor treating fields (TTFs) are a newly developed cancer therapy technology using an alternating electric field that may be a possible candidate for overcoming the limitations of conventional treatment methods currently used in cancer treatment. Although clinical results using TTFs appear promising, concerns regarding side effects must be clarified to demonstrate the effectiveness of this treatment method. To investigate the side effects of TTF treatment, the damage to normal cell lines and normal tissue of a mouse model was compared with the damage to tumor cells and tumors in a mouse model after TTF treatment. No serious damage was found in the normal cells and normal tissues of the mouse model, suggesting that the side effects of TTF treatment may not be serious. Our evidence based on in vitro and in vivo experiments suggests that TTF may cause selective damage to cancer cells, further demonstrating the potential of TTF as an attractive alternative to conventional cancer treatment modalities.

12.
Neurosci Lett ; 666: 64-69, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29273398

ABSTRACT

Given the increased public concern about the deleterious biological consequences of radiofrequency electromagnetic fields (RF-EMFs), the involvement of RF-EMFs in neurodegenerative diseases, especially Alzheimer's disease (AD), has received increased consideration. To investigate the effect of long-term RF-EMF exposure on AD progression, we exposed 5xFAD mice to 1950 MHz RF-EMF at a specific absorption rate of 5.0 W/kg for 2 h/day and 5 days/week for 8 months. Behavioral changes were assessed by an open field test and an object recognition memory task after RF exposure was terminated. In addition, cerebral glucose metabolism was analyzed in the brains of the 5xFAD mice using 18F-deoxyglucose positron emission tomography. The hyperactivity-like and anxiolytic behaviors of the 5xFAD mice in open field tests were rescued by RF exposure. Furthermore, long-term RF-EMF exposure improved the cognitive deficits of 5xFAD mice that were observed in the object recognition memory test. Consistent with the behavioral changes, glucose metabolism in the hippocampus and amygdala regions of the brains of 5xFAD mice following RF exposure was significantly increased compared to glucose metabolism in the brains of sham-exposed mice. These data suggest that long-term exposure to RF-EMF might exert beneficial effects on AD in 5xFAD mice.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Behavior, Animal/drug effects , Brain/physiopathology , Glucose/metabolism , Time , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Electromagnetic Fields , Female , Memory Disorders/metabolism , Mice , Mice, Transgenic , Positron-Emission Tomography/methods
13.
Int J Hyperthermia ; 34(3): 276-283, 2018 05.
Article in English | MEDLINE | ID: mdl-28659004

ABSTRACT

PURPOSE: Mild temperature hyperthermia (MTH) increases blood flow and oxygenation in tumours. On the other hand, high-dose-per-fraction irradiation damages blood vessels, decreases blood flow and increases hypoxia in tumours. The radiation-induced hypoxia in tumours activates hypoxia-inducible factor-1α (HIF-1α) and its target genes, such as vascular endothelial growth factor (VEGF), promoting revascularization and recurrence. In the present study, we examined the hypothesis that MTH inhibits radiation-induced upregulation of HIF-1α and its target genes by increasing tumour oxygenation. MATERIALS AND METHODS: FSaII fibrosarcoma tumours grown subcutaneously in the legs of C3H mice were used. Tumours were irradiated with 15 Gy using a 60Co irradiator or heated at 41 °C for 30 min using an Oncothermia heating unit. Blood perfusion and hypoxia in tumours were assessed with Hoechst 33342 and pimonidazole staining, respectively. Expression levels of HIF-1α and VEGF were determined using immunohistochemical techniques. Apoptosis of tumour cells was quantitated via TUNEL staining and the effects of treatments on tumour growth rate were assessed by measuring tumour diameters. RESULTS: Irradiation of FSaII tumours with a single dose of 15 Gy led to significantly decreased blood perfusion, increased hypoxia and upregulation of HIF-1α and VEGF. On the other hand, MTH at 41 °C for 30 min increased blood perfusion and tumour oxygenation, thereby suppressing radiation-induced HIF-1α and VEGF in tumours, leading to enhanced apoptosis of tumour cells and tumour growth delay. CONCLUSION: MTH enhances the anti-tumour effect of high-dose irradiation, at least partly by inhibiting radiation-induced upregulation of HIF-1α.


Subject(s)
Hyperthermia, Induced/methods , Hypoxia-Inducible Factor 1, alpha Subunit/therapeutic use , Neoplasms/radiotherapy , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Mice
14.
Int J Oncol ; 52(1): 211-218, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115437

ABSTRACT

Simvastatin exhibits anticancer activities, but its molecular mechanisms and radiosensitizing effects relative to p53 status remain unclear. In this study, we investigated whether the combination of simvastatin and ionizing radiation (IR) would enhance the antitumor effects of IR alone in HCT116 p53+/+ and p53­/- colon cancer cells. Using colony formation assays and a xenograft mouse model, we found that simvastatin potently stimulated radiosensitization of HCT116 p53­/- cells and xenograft tumors. The combination of simvastatin with IR decreased G2/M arrest and delayed the repair of IR-induced DNA damage; however, no differences between the HCT116 p53+/+ and p53­/- cells were evident. A further analysis revealed that simvastatin exhibited a novel function, namely, MDM2 suppression, regardless of p53 status. Interestingly, simvastatin induced radiosensitization by enhancing MDM2 suppression and elevating IR-induced p­ATM foci formation compared with IR alone in HCT116 p53­/- cells. Furthermore, simvastatin caused accumulations of the FOXO3a, E-cadherin, and p21 tumor suppressor proteins, which are downstream factors of MDM2, in HCT116 p53­/- cells. In conclusion, simvastatin enhanced radiosensitivity by inducing MDM2 inhibition and increasing tumor suppressor protein levels in radioresistant HCT116 p53­/- cells and xenografts. Overall, our novel findings suggest a scientific rationale for the clinical use of simvastatin as an MDM2 inhibitor and radiosensitizer for p53­deficient colorectal tumor treatments.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Simvastatin/pharmacology , Tumor Suppressor Protein p53/deficiency , Animals , Colonic Neoplasms/metabolism , HCT116 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Radiation Tolerance/drug effects , Random Allocation , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
15.
Biosci Rep ; 37(6)2017 Nov 22.
Article in English | MEDLINE | ID: mdl-29070518

ABSTRACT

Protein phosphatase 2A (PP2A) is a ubiquitous multifunctional enzyme usually known as a tumor suppressor. Recent studies have reported that although inhibition of PP2A leads to acceleration of cell growth, it also induces damaged cells to pass through the cell cycle and renders them sensitive to radiotherapy. Here, we investigated the radiosensitizing effects of digoxin as a PP2A inhibitor in two non-small-cell lung cancer (NSCLC) cell types (H460 and A549) with differential sensitivity to radiation. Digoxin inhibited the proliferation of H460 and A549 cells in a dose-dependent fashion and was especially effective on radioresistant A549 cells. Interestingly, the radiosensitizing effect of digoxin was only present in the radioresistant A549 cells and xenografts. The combination of digoxin and ionizing radiation (IR) significantly reduced clonogenic survival and xenograft tumor growth (P<0.001), compared with IR alone. Digoxin suppressed PP2A protein expression and prevented IR-induced PP2A expression in A549 cells. Digoxin treatment combined with IR allowed the damaged cell to progress through the cell cycle via suppression of cell cycle-related proteins (p53, cyclin D1, cyclin B1, CDK4, and p-cdc2). Moreover, digoxin enhanced IR-induced DNA damage through reduction in levels of repair proteins and elevation of p-ATM foci formation up to 24 h (P<0.001). In conclusion, digoxin has a novel function as a PP2A inhibitor, and combined with IR produces a synergistic effect on radiosensitizing cells, thereby indicating a potentially promising therapeutic approach to radioresistant lung cancer treatment.


Subject(s)
Digoxin/pharmacology , Protein Phosphatase 2/genetics , Radiation-Sensitizing Agents/pharmacology , A549 Cells , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Gene Expression/drug effects , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Protein Phosphatase 2/metabolism , Radiation Tolerance/drug effects , Xenograft Model Antitumor Assays
16.
Int J Oncol ; 51(4): 1124-1134, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849129

ABSTRACT

Osteosarcoma (OS) is a malignant tumor of the bone derived from primitive transformed cells of the mesenchymal origin. Local low-linear energy transfer (LET) radiotherapy has limited benefits on OS owing to its radioresistance. Thus, this study aimed to investigate the effects of high-LET radiation on human OS. Therefore, the human OS cell lines, U2O2 and KHOS/NP, were examined in vitro, or an orthotopic mouse xenograft model was studied in vivo after treatment with low-LET (gamma-ray) and high-LET (neutron) radiation. Notably, OS cells were significantly more sensitive to high-LET radiation in vitro and in the orthotopic xenograft tumor model. Specifically, neutron radiation treatment increased the relative percentage of apoptotic sub-G1 phase cells via caspase-3/9 activation; increased intracellular reactive oxygen species, autophagy, and DNA damage; and decreased invasion and migration. Similarly, the mean size of gamma-irradiated (8 Gy) orthotopic KHOS/NP OS was 195 mm3 at 6 weeks after gamma-irradiation (8 Gy), but it was only 150 mm3 in mice treated with high-LET neutron radiotherapy. Significantly, our results provide a rationale for the use of high-LET radiotherapy to treat patients with OS.


Subject(s)
Bone Neoplasms/radiotherapy , Caspase 3/metabolism , Caspase 9/metabolism , Osteosarcoma/radiotherapy , Animals , Apoptosis/radiation effects , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Movement/radiation effects , DNA Damage , Enzyme Activation/radiation effects , Female , Gamma Rays/therapeutic use , Humans , Linear Energy Transfer , Mice , Mice, Inbred BALB C , Mice, Nude , Neutrons/therapeutic use , Osteosarcoma/enzymology , Osteosarcoma/genetics , Osteosarcoma/pathology , Random Allocation , Xenograft Model Antitumor Assays
17.
Oncol Rep ; 38(3): 1597-1604, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714027

ABSTRACT

Worldwide, colorectal cancer is the third most common cancer in men and the second most common in women. As conventional colorectal cancer therapies result in various side effects, there is a need for adjuvant therapy that can enhance the conventional therapies without complications. In this study, we investigated the anticancer effects of combined mixture of the several medicinal mushrooms and Panax ginseng root extracts (also called Amex7) as an adjuvant compound in the treatment of human colorectal cancer. We observed the in vivo inhibitory effect of Amex7 (1.25, 6.25, and 12.5 ml/kg, oral administration, twice daily) on tumor growth in a mouse model xenografted with HT-29 human colorectal cancer cells. In vitro, at 6, 12, and 24 h after 4% Amex7 treatment, we analyzed cell cycle by flow cytometry and the expression levels of cell cycle progression, apoptosis, and DNA damage repair-related proteins using immunoblotting and immunofluorescence staining in HT-29 cell line. As a result, Amex7 significantly suppressed tumor growth in HT-29 human colorectal cancer cells and xenografts. In vitro, Amex7 induced G2/M arrest through the regulation of cell cycle proteins and cell death by apoptosis and autophagy. Additionally, Amex7 consistently induced DNA damage and delayed the repair of Amex7-induced DNA damage by reducing the level of HR repair proteins. In conclusion, Amex7 enhanced anticancer effects through the induction of G2/M arrest and cell death, including apoptosis and autophagy. Furthermore, Amex7 impaired DNA damage repair. The present study provides a scientific rationale for the clinical use of a combined mixture of medicinal mushrooms and P. ginseng root extracts as an adjuvant treatment in human colorectal cancer.


Subject(s)
Agaricales/chemistry , Colorectal Neoplasms/drug therapy , Panax/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Drug Synergism , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude
18.
Oncotarget ; 8(68): 112390-112400, 2017 Dec 22.
Article in English | MEDLINE | ID: mdl-29348833

ABSTRACT

The purpose of this study was to investigate the potential of gold nanoparticles as radiosensitizer for use in neutron therapy against hepatocellular carcinoma. The hepatocellular carcinoma cells lines Huh7 and HepG2 were irradiated with γ and neutron radiation in the presence or absence of gold nanoparticles. Effects were evaluated by transmission electron microscopy, cell survival, cell cycle, DNA damage, migration, and invasiveness. Gold nanoparticles significantly enhanced the radiosensitivity of Huh7 and HepG2 cells to γ-rays by 1.41- and 1.16-fold, respectively, and by 1.80- and 1.35-fold to neutron radiation, which has high linear energy transfer. Accordingly, exposure to neutron radiation in the presence of gold nanoparticles induced cell cycle arrest, DNA damage, and cell death to a significantly higher extent, and suppressed cell migration and invasiveness more robustly. These effects are presumably due to the ability of gold nanoparticles to amplify the effective dose from neutron radiation more efficiently. The data suggest that gold nanoparticles may be clinically useful in combination therapy against hepatocellular carcinoma by enhancing the toxicity of radiation with high linear energy transfer.

19.
Oncotarget ; 7(38): 62267-62279, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27556699

ABSTRACT

Alternating electric fields at an intermediate frequency (100~300 kHz), referred to as tumour-treating fields (TTF), are believed to interrupt the process of mitosis via apoptosis and to act as an inhibitor of cell proliferation. Although the existence of an antimitotic effect of TTF is widely known, the proposed apoptotic mechanism of TTF on cell function and the efficacy of TTF are controversial issues among medical experts. To resolve these controversial issues, a better understanding of the underlying molecular mechanisms of TTF on cell function and the differences between the effects of TTF alone and in combination with other treatment techniques is essential. Here, we report experimental evidence of TTF-induced apoptosis and the synergistic antimitotic effect of TTF in combination with ionizing radiation (IR). For these experiments, two human Glioblastoma multiforme (GBM) cells (U373 and U87) were treated either with TTF alone or with TTF followed by ionizing radiation (IR). Cell apoptosis, DNA damage, and mitotic abnormalities were quantified after the application of TTF, and their percentages were markedly increased when TTF was combined with IR. Our experimental results also suggested that TTF combined with IR synergistically suppressed both cell migration and invasion, based on the inhibition of MMP-9 and vimentin.


Subject(s)
Apoptosis/radiation effects , Brain Neoplasms/therapy , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Electric Stimulation Therapy/methods , Glioblastoma/therapy , Mitosis/radiation effects , Radiotherapy/methods , Cell Line, Tumor , Combined Modality Therapy/methods , Humans , Immunohistochemistry , Matrix Metalloproteinase 9/radiation effects , Vimentin/radiation effects
20.
Oncol Rep ; 36(2): 763-70, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27373272

ABSTRACT

Colorectal cancer is one of the most common cancers worldwide. Previous studies suggest that chemoradiotherapy is more effective for the treatment of colorectal cancer than is radiotherapy or chemotherapy alone. To enhance the radiosensitivity of tumor cells, several investigators have used targeted therapeutic agents that act as radiosensitizers. In the present study, we provide a scientific rationale for the clinical application of SU5416, an inhibitor of vascular endothelial growth factor receptor-2, as a radiosensitizer for colorectal cancer. Two human colorectal adenocarcinoma cell lines, HCT116 and HT-29, were treated with SU5416 and radiation alone or radiation followed by SU5416. In vitro tests were performed using colony forming assays, flow cytometric analysis, immunohistochemistry, senescence-associated ß-galactosidase, tumor cell motility and invasion assays. The combination of radiation and SU5416 synergistically inhibited cell survival and induced apoptosis through reactive oxygen species, enhanced IR-induced premature senescence, and inhibited DNA repair activity, cell migration and invasion. Collectively, our results favor the use of SU5416 and radiotherapy as a combination therapy for the treatment of colon cancer and it can be combined successfully with a radiation regimen to potentiate its antitumor and antimetastatic activities for future clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Indoles/pharmacology , Pyrroles/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Receptors, Vascular Endothelial Growth Factor/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aging/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Repair/drug effects , HCT116 Cells , HT29 Cells , Humans , Neoplasm Invasiveness/pathology , Reactive Oxygen Species/metabolism , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...