Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Vasc Interv Radiol ; 34(4): 619-622.e1, 2023 04.
Article in English | MEDLINE | ID: mdl-36596322

ABSTRACT

The purpose of this study was to evaluate the effect of bone radiofrequency (RF) ablation in the spine with and without controlled saline infusion. RF ablation with and without controlled saline infusion was performed in the vertebral bodies of 2 swine with real-time temperature and impedance recordings. Histology and magnetic resonance (MR) imaging results were reviewed to evaluate the ablation zone size, breach of spinal canal, and damage to the spinal cord and nerves. There was no difference in maximum and mean temperatures between controlled saline and noninfusion groups. The impedance and power output were not significantly different between the groups. MR imaging and histopathology demonstrated ablation zones confined within the vertebral bodies. Ablation zone size correlated on MR imaging and histopathology by groups. No ablation effect, breach of posterior cortex, spinal cord injury, or nerve or ganglion injury was observed at any level using MR imaging or histology. Controlled saline infusion does not appear to impact bone RF ablation and, specifically, does not increase the ablation zone size.


Subject(s)
Catheter Ablation , Vertebral Body , Swine , Animals , Spine/surgery , Temperature , Saline Solution , Radio Waves , Catheter Ablation/adverse effects , Catheter Ablation/methods
2.
Int J Oncol ; 58(4)2021 04.
Article in English | MEDLINE | ID: mdl-33655319

ABSTRACT

Following the publication of the above paper, we were contacted by the University of Illinois at Chicago, to request the retraction of the above article. Following a formal institutional investigation, the investigation panel concluded that the images in question had falsifying elements. Regarding the above study, the specific allegations that were investigated were that of falsifying elements of Fig. 1B, bottom panel, columns 2 and 3; Fig. 4A, top panel, columns 4, 5 and 6, and middle panel, columns 1, 2 and 3; and Fig. 7D, row 1, column 1 and row 2, column 1.
Following a review of this paper conducted independently by the Editor of International Journal of Oncology, the Editor concurred with the conclusions of the investigation panel, and therefore the above paper has been retracted from the publication. We also tried to contact the authors, but did not receive a reply. The Editor apologizes to the readership for the inconvenience caused. [the original article was published in International Journal of Oncology 38: 973­983, 2011; DOI: 10.3892/ijo.2011.934]

.

3.
Int J Oncol ; 58(4)2021 04.
Article in English | MEDLINE | ID: mdl-33655324

ABSTRACT

Following the publication of the above paper, we were contacted by the University of Illinois at Chicago, to request the retraction of the above article. Following a formal institutional investigation, the investigation panel concluded that the images in question had falsifying elements. Regarding the above study, the specific allegations that were investigated were that of falsifying elements of Fig. 2A, right panel, row 3, columns 2, 3 and 4 and Fig. 4D, left panel, row 5, columns 1, 2 and 3; Fig. 4A, row 1, columns 2, 3 and 4, and Fig. 4C, row 1, columns 5, 6 and 7; and Fig. 6C, row 1, column 3, and row 2, column 1.
Following a review of this paper conducted independently by the Editor of International Journal of Oncology, the Editor concurred with the conclusions of the investigation panel, and therefore the above paper has been retracted from the publication. We also tried to contact the authors, but did not receive a reply. The Editor apologizes to the readership for the inconvenience caused. [the original article was published in International Journal of Oncology 40: 1615-1624, 2012; DOI: 10.3892/ijo.2011.987].

4.
J Cancer ; 11(23): 6982-6991, 2020.
Article in English | MEDLINE | ID: mdl-33123288

ABSTRACT

Purpose: The aim of this study was to develop and evaluate a liposome formulation that deliver oxaliplatin under magnetic field stimulus in high concentration to alleviate the off-target effects in a rat model of colorectal liver metastases (CRLM). Materials and Methods: Hybrid liposome-magnetic nanoparticles loaded with Cy5.5 dye and oxaliplatin (L-NIR- Fe3O4/OX) were synthesized by using thermal decomposition method. CRLM (CC-531) cell viability was assessed and rats orthotopically implanted with CC-531 cells were treated with L-NIR-Fe3O4/OX or by drug alone via different routes, up to 3 cycles of alternating magnetic field (AMF). Optical and MR imaging was performed to assess the targeted delivery. Biodistribution and histology was performed to determine the distribution of oxaliplatin. Results: L-NIR-Fe3O4/OX presented a significant increase of oxaliplatin release (~18%) and lower cell viability after AMF exposure (p<0.001). Optical imaging showed a significant release of oxaliplatin among mesenteric vein injected (MV) group of animals. MR imaging on MV injected animals showed R2* changes in the tumor regions at the same regions immediately after infusion compared to the surrounding liver (p<0.001). Biodistribution analysis showed significantly higher levels of oxaliplatin in liver tissues compared to lungs (p<0.001) and intestines (p<0.001) in the MV animals that received AMF after L-NIR- Fe3O4/OX administration. Large tumor necrotic zones and significant improvement in the survival rates were noted in the MV animals treated with AMF. Conclusion: AMF triggers site selective delivery of oxaliplatin at high concentrations and improves survival outcomes in colorectal liver metastasis tumor bearing rats.

5.
Theranostics ; 10(12): 5368-5383, 2020.
Article in English | MEDLINE | ID: mdl-32373218

ABSTRACT

We report the impact of notch-DLL4-based hereditary vascular heterogeneities on the enhanced permeation and retention (EPR) effect and plasmonic photothermal therapy response in tumors. Methods: We generated two consomic rat strains with differing DLL4 expression on 3rd chromosome. These strains were based on immunocompromised Salt-sensitive or SSIL2Rγ- (DLL4-high) and SS.BN3IL2Rγ- (DLL4-low) rats with 3rd chromosome substituted from Brown Norway rat. We further constructed three novel SS.BN3IL2Rγ- congenic strains by introgressing varying segments of BN chromosome 3 into the parental SSIL2Rγ- strain to localize the role of SSIL2Rγ- DLL4 on tumor EPR effect with precision. We synthesized multimodal theranostic nanoparticles (TNPs) based on Au-nanorods which provide magnetic resonance imaging (MRI), X-ray, and optical contrasts to assess image guided PTT response and quantify host specific therapy response differences in tumors orthotopically xenografted in DLL4-high and -low strains. We tested recovery of therapy sensitivity of PTT resistant strains by employing anti-DLL4 conjugated TNPs in two triple negative breast cancer tumor xenografts. Results: Host strains with high DLL4 allele demonstrated slightly increased tumor nanoparticle uptake but consistently developed photothermal therapy resistance compared to tumors in host strains with low DLL4 allele. Tumor micro-environment with low DLL4 expression altered the geographic distribution of nanoparticles towards closer proximity with vasculature which improved efficacy of PTT in spite of lower overall TNP uptake. Targeting TNPs to tumor endothelium via anti-DLL4 antibody conjugation improved therapy sensitivity in high DLL4 allele hosts for two triple negative human breast cancer xenografts. Conclusions: Inherited DLL4 expression modulates EPR effects in tumors, and molecular targeting of endothelial DLL4 via nanoparticles is an effective personalized nanomedicine strategy.


Subject(s)
Breast Neoplasms/metabolism , Nanomedicine/methods , Nanoparticles/chemistry , Photothermal Therapy/methods , Tumor Microenvironment/physiology , Animals , Cell Line, Tumor , Female , Humans , Rats , Tumor Microenvironment/genetics
6.
Int J Hyperthermia ; 35(1): 176-182, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30130988

ABSTRACT

Complete ablation of liver tumors is vital for minimizing the risk of local tumor recurrence. Accurately identifying the hallmarks of tissue necrosis during thermal ablative therapies may significantly increase the efficacy of ablation, while minimizing unnecessary damage to the surrounding normal tissues or critical structures. Light propagation in biological tissues is sensitive to the tissue microstructure and chromophore concentrations. In our previous studies, we found that the wavelength (λ) averaged liver tissue absorption coefficient (µa) and reduced scattering coefficient (µs') change significantly upon heating which may be used for assessment of tissue damage during thermal ablation of solid tumors. Here, we seek to demonstrate the use of an integrated fiber-optic probe for continuous monitoring of the local tissue temperature (T), µa(λ) and µs'(λ) during thermal ablation of ex vivo porcine livers. The wavelength-averaged (435-630 nm) tissue absorption and scattering (µa and µs' ) increased rapidly at 45 °C and plateaued at 67 °C. The mean µa and µs' for liver tissue at 37 °C (n = 10) were 8.5 ± 3.7 and 2.8 ± 1.1 cm-1, respectively. The relative changes in µa and µs' at 37, 55, and 65 °C were significantly different (p < .02) from each other. A relationship between the relative changes in µa and µs' and the degree of tissue damage estimated using the temperature-based Arrhenius model for porcine liver tissues was established and studied.


Subject(s)
Carcinoma, Hepatocellular/surgery , Catheter Ablation/methods , Liver Neoplasms/surgery , Liver/pathology , Animals , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Swine
7.
J Vasc Interv Radiol ; 29(12): 1764-1769, 2018 12.
Article in English | MEDLINE | ID: mdl-30316676

ABSTRACT

PURPOSE: To investigate the differences in immune responses between cryoablation and irreversible electroporation (IRE) in a preclinical mouse model. MATERIAL AND METHODS: A mouse pancreatic cancer cell line (PANC-2) was implanted in the bilateral flanks of mice, and tumor-bearing mice were divided into 6 groups. One of the tumors was ablated either with contact cryoablation using an argon-cooled cryoablation probe for 1 minute at 5% power or by IRE for a total of 64 100-µs-duration, 1250-V/cm2 pulses with 100-ms spacing. The contralateral tumors in the same animal served as controls. At immediate, 6, 12, and 24 hours after ablation, the tumors were processed for immunostaining with F480 (macrophages), CD3 (T cells), and CD-56 (natural killer cells) antibodies. RESULTS: CD3 staining demonstrated significantly more T cells in the IRE group than in the cryoablation group at 6 hours (45 vs 16; P = .027), 12 hours (67 vs 33; P = .020), and 24 hours (161 vs 94; p = .003), with almost a 2-fold increase at every time point. Although the mean number of natural killer cells in the treated tumors was higher, no significant differences were observed between the 2 groups at any of the time points. A significant difference was observed in F480 positivity between the cryoablation group and the IRE group at 12 hours (210 vs 356; P = .0004) and 24 hours (220 vs 328; P = .04), respectively. CONCLUSIONS: In a mouse model of pancreatic cancer, IRE evokes a more robust infiltration of macrophages and T cells than cryoablation within 24 hours.


Subject(s)
Cryosurgery , Electroporation , Neoplasms, Experimental/therapy , Pancreatic Neoplasms/therapy , Animals , Antigens, Differentiation/metabolism , CD3 Complex/metabolism , CD56 Antigen/metabolism , Cell Line, Tumor , Cryosurgery/adverse effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Time Factors
9.
ACS Nano ; 12(7): 6597-6611, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29969226

ABSTRACT

We report sub-100 nm optical/magnetic resonance (MR)/X-ray contrast-bearing theranostic nanoparticles (TNPs) for interventional image-guided photothermal therapy (PTT) of solid tumors. TNPs were composed of Au@Gd2O3:Ln (Ln = Yb/Er) with X-ray contrast (∼486 HU; 1014 NPs/mL, 0.167 nM) and MR contrast (∼1.1 × 108 mM-1 S-1 at 9.4 T field strength). Although TNPs are deposited in tumors following systemic administration via enhanced permeation and retention effect, the delivered dose to tumors is typically low; this can adversely impact the efficacy of PTT. To overcome this limitation, we investigated the feasibility of site-selective hepatic image-guided delivery of TNPs in rats bearing colorectal liver metastasis (CRLM). The mesenteric vein of tumor-bearing rats was catheterized, and TNPs were infused into the liver by accessing the portal vein for site-selective delivery. The uptake of TNPs with hepatic delivery was compared with systemic administration. MR imaging confirmed that delivery via the hepatic portal vein can double the CRLM tumor-to-liver contrast compared with systemic administration. Photothermal ablation was performed by inserting a 100 µm fiber-optic carrying 808 nm light via a JB1, 3-French catheter for 3 min under DynaCT image guidance. Histological analysis revealed that the thermal damage was largely confined to the tumor region with minimal damage to the adjacent liver tissue. Transmission electron microscopy imaging validated the stability of core-shell structure of TNPs in vivo pre- and post-PTT. TNPs comprising Gd-shell-coated Au nanorods can be effectively employed for the site-directed PTT of CRLM by leveraging interventional radiology methods.


Subject(s)
Colorectal Neoplasms/pathology , Gadolinium/therapeutic use , Gold/therapeutic use , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Nanoparticles/therapeutic use , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Contrast Media/therapeutic use , Gadolinium/administration & dosage , Gadolinium/pharmacokinetics , Gold/administration & dosage , Gold/pharmacokinetics , Humans , Hyperthermia, Induced/methods , Liver/blood supply , Liver/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Magnetic Resonance Imaging/methods , Nanoparticles/administration & dosage , Phototherapy/methods , Radiology, Interventional/methods , Rats , Rats, Wistar
10.
Biomed Opt Express ; 9(2): 543-556, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29552392

ABSTRACT

Vascular supply is a critical component of the tumor microenvironment (TME) and is essential for tumor growth and metastasis, yet the endogenous genetic modifiers that impact vascular function in the TME are largely unknown. To identify the host TME modifiers of tumor vascular function, we combined a novel genetic mapping strategy [Consomic Xenograft Model] with near-infrared (NIR) fluorescence imaging and multiparametric analysis of pharmacokinetic modeling. To detect vascular flow, an intensified cooled camera based dynamic NIR imaging system with 785 nm laser diode based excitation was used to image the whole-body fluorescence emission of intravenously injected indocyanine green dye. Principal component analysis was used to extract the spatial segmentation information for the lungs, liver, and tumor regions-of-interest. Vascular function was then quantified by pK modeling of the imaging data, which revealed significantly altered tissue perfusion and vascular permeability that were caused by host genetic modifiers in the TME. Collectively, these data demonstrate that NIR fluorescent imaging can be used as a non-invasive means for characterizing host TME modifiers of vascular function that have been linked with tumor risk, progression, and response to therapy.

11.
Radiology ; 285(3): 809-819, 2017 12.
Article in English | MEDLINE | ID: mdl-28707960

ABSTRACT

Purpose To demonstrate that anti-MG1 conjugated hybrid magnetic gold nanoparticles (HNPs) act as a catalyst during photothermal ablation (PTA) of colorectal liver metastases, and thus increase ablation zones. Materials and Methods All experiments were performed with approval of the institutional animal care and use committee. Therapeutic and diagnostic multifunctional HNPs conjugated with anti-MG1 monoclonal antibodies were synthesized, and the coupling efficiency was determined. Livers of 19 Wistar rats were implanted with 5 × 106 rat colorectal liver metastasis cell line cells. The rats were divided into three groups according to injection: anti-MG1-coupled HNPs (n = 6), HNPs only (n = 6), and cells only (control group, n = 7). Voxel-wise R2 and R2* magnetic resonance (MR) imaging measurements were obtained before, immediately after, and 24 hours after injection. PTA was then performed with a fiber-coupled near-infrared (808 nm) diode laser with laser power of 0.56 W/cm2 for 3 minutes, while temperature changes were measured. Tumors were assessed for necrosis with hematoxylin-eosin staining. Organs were analyzed with inductively coupled plasma mass spectrometry to assess biodistribution. Therapeutic efficacy and tumor necrosis area were compared by using a one-way analysis of variance with post hoc analysis for statistically significant differences. Results The coupling efficiency was 22 µg/mg (55%). Significant differences were found between preinfusion and 24-hour postinfusion measurements of both T2 (repeated measures analysis of variance, P = .025) and T2* (P < .001). Significant differences also existed for T2* measurements between the anti-MG1 HNP and HNP-only groups (P = .034). Mean temperature ± standard deviation with PTA in the anti-MG1-coated HNP, HNP, and control groups was 50.2°C ± 7.8, 51°C ± 4.4, and 39.5°C ± 2.0, respectively. Inductively coupled plasma mass spectrometry revealed significant tumor targeting and splenic sequestration. Mean percentages of tumor necrosis in the anti-MG1-coated HNP, HNP, and control groups were 38% ± 29, 14% ± 17, and 7% ± 8, respectively (P = .043). Conclusion Targeted monoclonal antibody-conjugated HNPs can serve as a catalyst for photothermal ablation of colorectal liver metastases by increasing ablation zones. © RSNA, 2017.


Subject(s)
Colorectal Neoplasms/therapy , Gold/therapeutic use , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Low-Level Light Therapy/methods , Magnetite Nanoparticles/therapeutic use , Nanoconjugates/therapeutic use , Animals , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Hyperthermia, Induced/methods , Liver Neoplasms/immunology , Mucin-5B/immunology , Rats , Rats, Wistar , Treatment Outcome
12.
PLoS One ; 11(5): e0155334, 2016.
Article in English | MEDLINE | ID: mdl-27171151

ABSTRACT

PURPOSE: Surgical resection of colorectal liver metastases is not achievable in more than 70% of the cases. Although the liver directed therapies have become a part of the stand of care, lack of a preclinical model impedes the assessment of toxicity and therapeutic benefits attributed several candidate drugs or treatment regimens that can be designed. In the present study we aim develop and characterize a rat colorectal liver metastasis model. MATERIALS AND METHODS: Growth characteristics of CC-531 cells were determined in vitro followed by subcapsular liver implantation in syngeneic WAG/Rij rats. Tumor growth progression was followed over 3 weeks by ultrasound (US) and magnetic resonance imaging (MRI). Growth characteristics were also assessed by histopathology and immunohistochemistry in harvested tumor tissues. RESULTS: The doubling time of CC-531 cells was found be under 24hrs and all the implanted rats grew tumors. US imaging showed hypoechoic masses and MRI showed contrast enhancement representing complex tumor microenvironments. Hematoxylin and Eosin staining confirmed tumor growth and uniform CD31 staining in tumor confirmed even vessel density. CONCLUSION: CC-531 can be used as a metastatic rat tumor colorectal liver metastases model with well-defined characteristics that can be readily followed by imaging whilst having a therapeutic window for interventions.


Subject(s)
Colorectal Neoplasms/pathology , Disease Models, Animal , Liver Neoplasms/secondary , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/diagnostic imaging , Electroporation , Immunohistochemistry , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/pathology , Magnetic Resonance Imaging , Rats
13.
Mol Cancer Ther ; 13(5): 1309-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24623737

ABSTRACT

Urokinase-type plasminogen activator receptor (uPAR) is overexpressed in the tumor-stromal invasive microenvironment in many human cancers, including medulloblastoma. The role of uPAR in tumor progression and angiogenesis has been well characterized. Previously, in medulloblastoma cells, we showed that ionizing radiation (IR)-induced uPAR is a potent activator of cancer stem cell (CSC)-like properties and is associated with various transcription factors that are involved during embryonic development and cancer. In the present study, we show that uPAR protein acts as a cytoplasmic sequestration factor for a novel basic helix-loop-helix transcription factor, Hand-1. The Hand-1 protein plays an essential role in the differentiation of trophoblast giant cells and cardiac morphogenesis, and yet its precise cellular function and its contribution to cancer remain mostly unknown. We also observed that the Hand-1 protein is upregulated in uPAR short hairpin RNA-treated medulloblastoma cells and accompanies sustained cell growth and angiogenesis. Furthermore, IR-induced uPAR overexpression negatively regulates Hand-1 activity and results in the stabilization of angiogenesis-promoting molecules, including hypoxia-inducible factor-1α. Finally, uPAR overexpression and its association with Hand-1 after IR treatment indicate that uPAR is capable of regulating Hand-1 and that uPAR has a role in the process of IR-induced tumor angiogenesis.


Subject(s)
Active Transport, Cell Nucleus , Basic Helix-Loop-Helix Transcription Factors/metabolism , Medulloblastoma/metabolism , Radiation Tolerance , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cytoplasm/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Neovascularization, Pathologic/genetics , Protein Binding , Protein Transport , Radiation, Ionizing , Receptors, Urokinase Plasminogen Activator/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Burden
14.
Neoplasia ; 15(2): 192-203, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23441133

ABSTRACT

Our previous studies have shown the role of radiation-induced urokinase plasminogen activator (uPA) expression in the progression of meningioma. In the present study, we investigated whether modulation of DNA methylation profiles could regulate uPA expression. Initially, radiation treatment was found to induce hypomethylation in meningioma cells with a decrease in DNA (cytosine-5)-methyltransferase 1 (DNMT1) and methyl-CpG binding domain protein (MBD) expression. However, oxidative damage by H(2)O(2) or pretreatment of irradiated cells with N-acetyl cysteine (NAC) did not show any influence on these proteins, thereby indicating a radiation-specific change in the methylation patterns among meningioma cells. Further, we identified that hypomethylation is coupled to an increase in uPA expression in these cells. Azacytidine treatment induced a dose-dependent surge of uPA expression, whereas pre-treatment with sodium butyrate inhibited radiation-induced uPA expression, which complemented our prior results. Methylation-specific polymerase chain reaction on bisulfite-treated genomic DNA revealed a diminished methylation of uPA promoter in irradiated cells. Transfection with small hairpin RNA (shRNA)-expressing plasmids targeting CpG islands of the uPA promoter showed a marked decline in uPA expression with subsequent decrease in invasion and proliferation of meningioma cells. Further, radiation treatment was found to recruit SP1 transcription factor, which was abrogated by shRNA treatment. Analysis on signaling events demonstrated the activation of MAP kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) in radiation-treated cells, while U0126 (MEK/ERK inhibitor) blocked hypomethylation, recruitment of SP1, and uPA expression. In agreement with our in vitro data, low DNMT1 levels and high uPA were found in intracranial tumors treated with radiation compared to untreated tumors. In conclusion, our data suggest that radiation-mediated hypomethylation triggers uPA expression in meningioma cells.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation/genetics , Meningioma/genetics , Urokinase-Type Plasminogen Activator/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/radiation effects , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Immunoglobulins/metabolism , Meningioma/pathology , Oxidative Stress/radiation effects , Promoter Regions, Genetic/radiation effects , Signal Transduction/radiation effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transcriptional Activation/radiation effects , Urokinase-Type Plasminogen Activator/metabolism
15.
Int J Oncol ; 42(1): 188-96, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23123816

ABSTRACT

Our previous studies showed that overexpression of secreted protein acidic and rich in cysteine (SPARC) induced autophagy-mediated apoptosis in PNET cells. In the present study, we attempted to elucidate the molecular mechanisms and signaling cascades associated with SPARC overexpression in combination with radiation therapy that eventually leads to autophagy-mediated apoptosis in neuroblastoma. SPARC expression in SK-N-AS and NB-1691 cells demonstrated the activation of caspase 3, cleavage of PARP and induction of apoptosis. The experiments to unravel the mechanisms associated with autophagy-apoptosis illustrated that SPARC overexpression triggered endoplasmic reticulum (ER) stress and thereby unfolded protein response (UPR). This was apparent with the activation of stress receptors, inositol-requiring enzyme (IRE 1α), RNA-dependent protein kinase (PKR)-like ER kinase (PERK) and BiP. This study further demonstrated the induction of transcription factor CHOP as a result of IRE-JNK activation in response to increased SPARC levels. Inhibition of ER stress and JNK activation led to inhibition of autophagy-mediated apoptosis. Further, the apparent expression of ER stress molecules among the orthotopic tumors treated by SPARC overexpression plasmids substantiated our in vitro observations. Taken together, these results illustrate the critical role of ER stress in regulating autophagy-mediated apoptosis in SPARC-overexpressed neuroblastoma cells and radiation treatment.


Subject(s)
Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Neuroblastoma/pathology , Osteonectin/metabolism , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Combined Modality Therapy , Flow Cytometry , Humans , Immunoenzyme Techniques , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Neuroblastoma/metabolism , Neuroblastoma/therapy , Osteonectin/genetics , Phosphorylation , RNA, Messenger/genetics , Radiation, Ionizing , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Cells, Cultured , Unfolded Protein Response/genetics
16.
Angiogenesis ; 16(1): 85-100, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22956186

ABSTRACT

Despite existing aggressive treatment modalities, the prognosis for advanced stage neuroblastoma remains poor with significant long-term illness in disease survivors. Advance stage disease features are associated with tumor vascularity, and as such, angiogenesis inhibitors may prove useful along with current therapies. The matricellular protein, secreted protein acidic and rich in cysteine (SPARC), is known to inhibit proliferation and migration of endothelial cells stimulated by growth factors. Here, we sought to determine the effect of SPARC on neuroblastoma tumor cell-induced angiogenesis and to decipher the molecular mechanisms involved in angiogenesis inhibition. Conditioned medium from SPARC-overexpressed neuroblastoma cells (pSPARC-CM) inhibited endothelial tube formation, cell proliferation, induced programmed cell death and suppressed expression of pro-angiogenic molecules such as VEGF, FGF, PDGF, and MMP-9 in endothelial cells. Further analyses revealed that pSPARC-CM-suppressed expression of growth factors was mediated by inhibition of the Notch signaling pathway, and cells cultured on conditioned medium from tumor cells that overexpress both Notch intracellular domain (NICD-CM) and SPARC resumed the pSPARC-CM-suppressed capillary tube formation and growth factor expression in vitro. Further, SPARC overexpression in neuroblastoma cells inhibited neo-vascularization in vivo in a mouse dorsal air sac model. Furthermore, SPARC overexpression-induced endothelial cell death was observed by co-localization studies with TUNEL assay and an endothelial marker, CD31, in xenograft tumor sections from SPARC-overexpressed mice. Our data collectively suggest that SPARC overexpression induces endothelial cell apoptosis and inhibits angiogenesis both in vitro and in vivo.


Subject(s)
Neovascularization, Pathologic/metabolism , Neuroblastoma/blood supply , Neuroblastoma/metabolism , Osteonectin/metabolism , Receptors, Notch/metabolism , Signal Transduction , Angiogenesis Inducing Agents/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Mice , Mice, Nude , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Biotechnol Appl Biochem ; 59(6): 437-44, 2012.
Article in English | MEDLINE | ID: mdl-23586952

ABSTRACT

The present study aimed for an enhanced induction strategy combined with high-level production of a capture antigen of hepatitis C virus (HCV) for use in diagnosis of HCV infection. We have expressed the synthetic gene encoding for HCV multiepitope protein in pET-28a(+) vector and investigated its production in Escherichia coli BL21(DE3) cells using high-cell-density fed-batch cultivation. A maximum cell dry mass of 30 g/L was obtained, and the culture was induced with 1, 5, and 10 mM isopropyl ß-D-1-thiogalactopyranoside (IPTG) for ∼4 H at 30°C; a maximum protein production of 1.5 g/L was observed in the case of induction with 10 mM IPTG. The enhanced induction strategy resulted in a ∼15-fold increase as compared to 1 mM IPTG. The protein was purified using a simple immobilized metal affinity chromatography procedure, yielding 16.6 mg/g dry cell weight of pure protein with more than 99% purity. Further, the protein was evaluated for its diagnostic potential by using the commercially available HCV Seroconversion Panel, Worldwide HCV Performance Panel, and Viral Coinfection Panel. The protein showed high sensitivity and specificity, which was comparable to the best performing commercially available enzyme immunoassay (EIA) kits.


Subject(s)
Batch Cell Culture Techniques , Escherichia coli/growth & development , Escherichia coli/genetics , Genetic Engineering/methods , Hepacivirus/genetics , Hepacivirus/isolation & purification , Viral Proteins/genetics , Amino Acid Sequence , Antigens, Viral/immunology , Epitopes/immunology , Escherichia coli/cytology , Gene Expression , Hepacivirus/immunology , Molecular Sequence Data , Protein Refolding , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Proteins/isolation & purification
18.
Cancer Lett ; 313(1): 64-75, 2011 Dec 26.
Article in English | MEDLINE | ID: mdl-21945852

ABSTRACT

In continuation to our studies on radioresistance in meningioma, here we show that radiation treatment (7Gy) induces G2/M cell cycle arrest in meningioma cells. Phosphorylation of Chk2, Cdc25c and Cdc2 were found to be key events since interference with Chk2 activation and cyclin B1/Cdc2 interaction led to permanent arrest followed by apoptosis. Irradiated cells showed recovery and formed aggressive intracranial tumors with rapid spread and morbidity. Nevertheless, knock down of uPAR with or without radiation induced permanent arrest in G2/M phase and subsequent apoptosis in vitro and in vivo. In conclusion, our data suggest that combination treatment with radiation and uPAR knock down or other inhibitors resulting in non-reversible G2/M arrest may be beneficial in the management of meningiomas.


Subject(s)
Cell Cycle Checkpoints/physiology , Cell Division/physiology , G2 Phase/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Blotting, Western , Brain/metabolism , Brain/pathology , Brain/radiation effects , CDC2 Protein Kinase , Carbanilides/pharmacology , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/radiation effects , Cell Division/genetics , Cell Division/radiation effects , Cell Line, Tumor , Checkpoint Kinase 2 , Cyclin B/genetics , Cyclin B/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin-Dependent Kinases , G2 Phase/genetics , G2 Phase/radiation effects , Humans , Meningioma/genetics , Meningioma/pathology , Meningioma/therapy , Mice , Mice, Nude , Phosphorylation/drug effects , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden , Xenograft Model Antitumor Assays/methods , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
19.
Int J Oncol ; 38(6): 1615-24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21455571

ABSTRACT

Cell motility is influenced by the microenvironment, signal transduction and cytoskeleton rearrangement. Cancer cells become resistant to these control mechanisms and gain the ability to move throughout the body and invade healthy tissues, which leads to metastatic disease. Integrins respond to context-dependent cues and promote cell migration and survival in cancer cells. In the present study, we analyzed the role of integrins in radiation-induced migration of meningioma cells. Migration and cell proliferation assays revealed that radiation treatment (7 Gy) significantly increased migration and decreased proliferation in two cell lines, IOMM-Lee and CH-157-MN. α3 and ß1 integrins were overexpressed at both the protein and transcript levels after radiation treatment and a function-blocking α3ß1 antibody inhibited the radiation-induced migration. Immunofluorescence studies illustrated the localization of α3 integrin and F-actin at the migration front of irradiated cells. Further, an increase in phosphorylation of FAK and ERK was observed, while both FAK phosphorylation inhibitor and FAK shRNA inhibited ERK phosphorylation and downregulated uPA and vinculin. In addition to the co-localization of FAK and ERK at the migration front, these FAK-inhibition results link the downstream effects of ERK to FAK. Correspondingly, U0126 quenched ERK phosphorylation and reduced the expression of molecules involved in migration. Furthermore, brain sections of the animals implanted with tumors followed by radiation treatment showed elevated levels of α3 integrin and active ERK. Taken together, our results show that radiation treatment enhances the migration of meningioma cells with the involvement of α3ß1 integrin-mediated signaling via FAK and ERK.


Subject(s)
Cell Movement/radiation effects , Integrin alpha3beta1/metabolism , Actins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Integrin alpha3beta1/genetics , Intracellular Space/metabolism , Meningioma/radiotherapy , Mice , Mice, Nude , Protein Transport , Radiation, Ionizing , Signal Transduction/radiation effects , Xenograft Model Antitumor Assays , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
20.
Cell Signal ; 23(8): 1299-310, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21426933

ABSTRACT

Chemokines play a vital role in recruiting various cell types in the process of tissue repair. Radiation, a major therapeutic modality in cancer treatment, has been described to induce inflammatory response that might lead to the expression of several chemokines. In the present study, we investigated the mechanism of monocyte chemoattractant protein-1 (MCP-1) induction by radiation in meningioma cell lines and the paracrine effect on human microvascular endothelial cells (HMEC). After radiation, meningioma cell lines (IOMM Lee and SF-3061) showed an increased expression of MCP-1. In addition, irradiated meningioma cancer cell conditioned medium (CM) showed an increased ability to attract HMEC and to stimulate MCP-1-induced protein (MCPIP), VEGF and angiogenin expression in HMEC. This chemotactic activity and angiogenic stimulator effect on HMEC were almost abrogated by depleting MCP-1 from the irradiated cancer cell CM. Further, inhibition of either ERK activation/expression or NF-κB nuclear translocation hindered radiation-induced MCP-1 expression in both meningioma cell lines. Further, supplementing cancer cells with exogenous ATF-uPA (with and without radiation) activated ERK phosphorylation, nuclear translocation of the NF-κB p65 sub-unit (Rel-A), and MCP-1 expression. Downregulation of uPA and uPAR, simultaneously by transfecting the cancer cells with bi-cistronic siRNA-expressing plasmid (pU) inhibited radiation-induced ERK activation, nuclear translocation of Rel-A, NF-κB DNA binding activity, and MCP-1 expression. In addition, pU-transfected cancer cells (with or without radiation) reduced radiation-induced MCP-1 and blocked the recruitment of other cell types during the inflammatory process induced by radiation both in in vitro and in vivo conditions.


Subject(s)
Chemokine CCL2/metabolism , Endothelial Cells/metabolism , Meningeal Neoplasms/metabolism , Meningioma/metabolism , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Cell Line, Tumor , Endothelial Cells/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Meningeal Neoplasms/radiotherapy , Meningioma/radiotherapy , NF-kappa B/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Radiation, Ionizing , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Ribonuclease, Pancreatic/metabolism , Ribonucleases , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...