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










Publication year range
1.
J Hazard Mater ; 477: 135234, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39042990

ABSTRACT

Thorium-232 (Th-232) is a promising fuel for advanced nuclear reactors. However, in case of internal human exposure to Th, there is currently no effective modality for its removal from liver and skeleton or for mitigating its effect. The FDA-approved agent, diethylenetriaminepentaacetate (DTPA), can remove Th and other actinides from blood circulation only. For the first time, a rationally-selected polyherbal hepatoprotective i.e. Liv52® (L52S), was evaluated in-combination with DTPA for its Th decorporation ability in Swiss mice. Inductively-coupled plasma mass spectroscopic analysis showed that oral administration of L52S in conjunction with DTPA significantly decreased Th burden from liver (20 %) and skeleton (33 %) as well as enhanced Th excretion (∼2.5 folds) through urine in comparison to DTPA or L52S alone. The combinatorial therapy was found to be complementary in-action, ameliorating Th-induced tissue damage in liver, spleen, and bone more effectively than monotherapy. Furthermore, markers of liver function (alanine transaminase) and liver inflammation and fibrosis (NF-κB & keratin) further validated the beneficial effect of L52S. The human consumption of L52S for various liver disorders further supports its clinical application for Th decorporation and mitigation of its health effects.

2.
Environ Sci Technol ; 58(28): 12330-12342, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38772857

ABSTRACT

Thorium-232 (Th), the most abundant naturally occurring nuclear fuel, has been identified as a sustainable source of energy. In view of its large-scale utilization and human evidence of lung disorders and carcinogenicity, it is imperative to understand the effect of Th exposure on lung cells. The present study investigated the effect of Th-dioxide (1-100 µg/mL, 24-48 h) on expression of surfactant proteins (SPs) (SP-A, SP-B, SP-C, and SP-D, which are essential to maintain lung's surface tension and host-defense) in human lung cells (WI26 and A549), representative of alveolar cell type-I and type-II, respectively. Results demonstrated the inhibitory effect of Th on transcriptional expression of SP-A, SP-B, and SP-C. However, Th promoted the mRNA expression of SP-D in A549 and reduced its expression in WI26. To a significant extent, the effect of Th on SPs was found to be in accordance with their protein levels. Moreover, Th exposure altered the extracellular release of SP-D/A from A549, which remained unaltered in WI26. Our results suggested the differential role of oxidative stress and ATM and HSP90 signaling in Th-induced alterations of SPs. These effects of Th were found to be consistent in lung tissues of mice exposed to Th aerosols, suggesting a potential role of SPs in Th-associated lung disorders.


Subject(s)
Alveolar Epithelial Cells , Thorium , Humans , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Mice , Animals , A549 Cells , Pulmonary Surfactant-Associated Proteins/metabolism
3.
Biomater Adv ; 161: 213868, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38677038

ABSTRACT

Triple negative breast carcinoma (TNBC) accounts for 15-20 % of all incident breast cancers (BC) and is known to be highly invasive, has fewer treatment options, and tends to have a worse prognosis. However, due to its biological heterogeneity and diverse clinical and epidemiological behaviors, TNBC lacks a tumor-specific targeted therapy. In the present work we have developed a TNBC-specific targeted nano-delivery agent comprising of a cRGD labeled magneto-liposome (T-LMD) co-encapsulated with oleic acid coated iron oxide nanoparticles (MN-OA) and doxorubicin (Dox) in the liposome bilayer and core, respectively. T-LMD was found to show enhanced uptake and induction of ferroptotic cell death in MDA-MB-231, a TNBC model cell line. Additionally, T-LMD induced ferroptosis was found to be accompanied by release of HMGB1, an immunogenic cell death marker, suggesting its immunogenicity for augmenting the activation of anti-tumor immunity in TNBC. The strategic placement of IONPs in the liposome bilayer of T-LMD facilitates the sensitization of MDA-MB-231 cells to undergo ferroptosis; predominantly via the activation of the iron/lipid metabolism pathway, as validated by use of small molecule ferroptosis inhibitor (ferrostatin-1) and iron chelator (deferoxamine). Activation of ferroptotic cell death was also corroborated by ferroptosis specific-ultrastructural alterations in the shape/size of cellular mitochondria and cell ballooning as observed by transmission electron microscopy and bright field imaging, respectively. Thus, our ferroptosis nano-inducer (T-LMD) can efficiently kill TNBC cells via enhanced LPO and ROS generation leading to membrane damage and consequent release of LDH and HMGB1, induce mitochondrial alterations and enhanced DNA double strand breaks. Altogether, our results suggest significant implications of T-LMD for treatment of TNBC.


Subject(s)
Doxorubicin , Ferroptosis , Liposomes , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Ferroptosis/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Liposomes/chemistry , Female , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , HMGB1 Protein/metabolism , Magnetic Iron Oxide Nanoparticles/chemistry
4.
PLoS One ; 18(11): e0281919, 2023.
Article in English | MEDLINE | ID: mdl-37943802

ABSTRACT

The present pilot study aims to investigate the diagnostic and prognostic efficacy of serum HSP90 beta in Head and Neck Squamous Cell Carcinoma (HNSCC) patients subjected to localized hyperthermia therapy (HT). Serum levels of HSP90 beta were measured by ELISA and its diagnostic and prognostic efficacy was determined by receiver operating characteristic curve (ROC) analysis. HNSCC patients showed significantly (P<0.05) higher serum levels of HSP90 beta (65.6±13.08 ng/ml) compared to Healthy Controls (HC: 23.5±3.8 ng/ml). No significant difference was observed in serum HSP90 beta levels between complete responders (CR) and non-responders (NR) in the chemo-radiation therapy (CRT) cohort. However, in CRT+HT cohort, CR showed significantly (P = 0.02) lower serum HSP90 beta levels at 24 h after HT (25.6±9.04 ng/ml) compared to NR (130.5±34.2 ng/ml). Youden's index values between HNSCC versus HC, CR versus NR (CRT) and CR versus NR (CRT+HT) were found to be 0.47, 0.45 and 0.80, respectively. Thus, alterations in the serum HSP90 beta after HT suggest its potential in prognosis of HT response in HNSCC patients. Elevated levels of HSP90 beta may serve as a promising diagnostic serum bio-marker for HNSCC. However, further validation in larger patient samples is needed for clinical translation of HSP90 beta as diagnostic and prognostic biomarker.


Subject(s)
Head and Neck Neoplasms , Hyperthermia, Induced , Humans , Squamous Cell Carcinoma of Head and Neck/therapy , Pilot Projects , Prognosis , Head and Neck Neoplasms/therapy , Biomarkers, Tumor
5.
RSC Adv ; 13(19): 13240-13251, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37123999

ABSTRACT

Deoxyglucose conjugated nanoparticles with persistent luminescence have shown theragnostic potential. In this study, deoxyglucose-conjugated nano-particles with persistent luminescence properties were synthesized, and their theragnostic potential was evaluated in fibrosarcoma cancer cells and a tumor model. The uptake of nano-formulation was found to be higher in mouse fibrosarcoma (WEHI-164) cells cultured in a medium without glucose. Nanoparticles showed a higher killing ability for cancer cells compared to normal cells. A significant accumulation of nanoparticles to the tumor site in mice was evident by the increased tumor/normal leg ratio, resulting in a significant decrease in tumor volume and weight. Histopathological studies showed a significant decrease in the number of dividing mitotic cells but a greater number of apoptotic/necrotic cells in nanoparticle-treated tumor tissues, which was correlated with a lower magnitude of Ki-67 expression (a proliferation marker). Consequently, our results showed the potential of our nano-formulation for cancer theragnosis.

6.
Biomater Adv ; 142: 213147, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36260957

ABSTRACT

The chemo-therapeutic efficacy of Doxorubicin (Dox), a potent anti-cancer drug used in the treatment of several solid tumors, is severely compromised by its cardio-toxicity. To overcome this shortcoming and exploit the utmost theranostic potential of nano-formulations, magnetic nanoparticles co-encapsulated with Dox and indocyanine green (ICG) in a liposomal carrier and tagged with cyclic RGD peptide were rationally designed and synthesized. These magneto-liposomes (T-LMD) showed αvß3-integrin receptor targeting and higher cyto-toxicity in several cancer cell lines (i.e. lung, breast, skin, brain and liver cancer) in combination with or without gamma radiation or magnetic hyperthermia therapy as compared to clinical liposomal nano-formulation of Dox (Lippod™). Mechanism of chemo-radio-sensitization was found to involve activation of JNK mediated pro-apoptotic signaling axis and delayed repair of DNA double strand breaks. Real time imaging of ICG labeled T-LMD suggested ~6-18 fold higher tumor accumulation of T-LMD as compared to off-target organs (kidney, liver, spleen, intestine, lungs and heart) and resulted in its higher combinatorial (chemo-radio-hyperthermia) tumor therapy efficacy as compared to Lippod™. Moreover, T-LMD showed insignificant toxicity to the heart tissue as suggested by serum levels of CK-MB, histo-pathological analysis, anti-oxidant enzyme activities (Catalase and GST) and markers of cardiac fibrosis, suggesting its potential for targeted multi-modal therapy of cancer.


Subject(s)
Magnetite Nanoparticles , Phototherapy , Phototherapy/methods , Precision Medicine , Drug Delivery Systems/methods , Cell Line, Tumor , Doxorubicin/pharmacology , Indocyanine Green , Liposomes , DNA Damage
7.
Environ Pollut ; 281: 116969, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33845224

ABSTRACT

Inhalation represents the most prevalent route of exposure with Thorium-232 compounds (Th-nitrate/Th-dioxide)/Th-containing dust in real occupational scenario. The present study investigated the mechanism of Th response in normal human alveolar epithelial cells (WI26), exposed to Th-nitrate or colloidal Th-dioxide (1-100 µg/ml, 24-72 h). Assessment in terms of changes in cell morphology, cell proliferation (cell count), plasma membrane integrity (lactate dehydrogenase leakage) and mitochondrial metabolic activity (MTT reduction) showed that Th-dioxide was quantitatively more deleterious than Th-nitrate to WI26 cells. TEM and immunofluorescence analysis suggested that Th-dioxide followed a clathrin/caveolin-mediated endocytosis, however, membrane perforation/non-endocytosis seemed to be the mode of Th internalization in cells exposed to Th-nitrate. Th-estimation by ICP-MS showed significantly higher uptake of Th in cells treated with Th-dioxide than with Th-nitrate at a given concentration. Both Th-dioxide and nitrate were found to increase the level of reactive oxygen species, which seemed to be responsible for lipid peroxidation, alteration in mitochondrial membrane potential and DNA-damage. Amongst HSPs, the protein levels of HSP70 and HSP90 were affected differentially by Th-nitrate/dioxide. Specific inhibitors of ATM (KU55933) or HSP90 (17AAG) were found to increase the Th- cytotoxicity suggesting prosurvival role of these signaling molecules in rescuing the cells from Th-toxicity.


Subject(s)
Thorium Dioxide , Cell Count , DNA Damage , Epithelial Cells , Humans , Lung/chemistry , Nitrates/toxicity , Oxidative Stress , Reactive Oxygen Species , Thorium/analysis
8.
Mater Sci Eng C Mater Biol Appl ; 112: 110915, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32409067

ABSTRACT

We have developed surface functionalised Fe3O4 magnetic nanoparticles (MNPs) based system that can be used for tumor-targeted multimodal therapies and MR imaging. Biocompatible, non-essential amino acid (glutamic acid) was introduced onto the surface of Fe3O4 MNPs to provide functional sites for binding of chemotherapeutic drugs. These glutamic acid-coated Fe3O4 MNPs (GAMNPs) exhibit good water-dispersibility, magnetic responsivity and pH dependent charge conversal feature. The magnetic core as well as organic shell of GAMNPs was characterized by XRD, TEM, DLS, FTIR, PPMS and UV-visible spectroscopy and zeta-potential analyzer etc. The broad spectrum anticancer drugs, doxorubicin hydrochloride (DOX) and methotrexate (MTX) were electrostatically and covalently conjugated to the surface of GAMNPs, respectively for combination chemotherapy. These dual drugs loaded system (DOX-MTX-GAMNPs) shows pH dependent release behaviour of both the drugs and enhanced toxicity towards breast cancer cell line (MCF-7) as compared to their individual treatment. Fluorescence microscopy and flow cytometric analyses confirmed the successful uptake of drug loaded system into MCF-7 cell lines. Further MTX being analogue of folic acid, its co-delivery with DOX would help in internalization of both the drugs into MCF-7 cells. These GAMNPs also show good heating efficiency under AC magnetic field (Intrinsic loss power, ILP = 0.95 and 0.73 and 0.48 nHm2/Kg at Fe concentration of 0.5, 1 and 2 mg/ml, respectively) and transverse relaxivity (r2 = 152 mM-1 s-1) indicating their potential capability for hyperthermia therapy and MRI tracking. Furthermore, it has been observed that the combination of chemotherapeutic drugs and hyperthermia leads to an enhancement of cytotoxicity in MCF-7 cells.


Subject(s)
Contrast Media/chemistry , Ferrosoferric Oxide/chemistry , Glutamic Acid/chemistry , Magnetite Nanoparticles/chemistry , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Carriers/chemistry , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Magnetic Resonance Imaging , Methotrexate/chemistry , Methotrexate/metabolism , Methotrexate/pharmacology , Neoplasms/diagnostic imaging , Surface Properties
9.
J Biosci ; 452020.
Article in English | MEDLINE | ID: mdl-33410424

ABSTRACT

Many pathogenic species of bacteria are showing increasing drug resistance against clinically used antibiotics. Molecules structurally distant from known antibiotics and possessing membrane targeting bactericidal activities are more likely to display activity against drug-resistant pathogens. Mitocurcumin (MitoC) is one of such compounds, synthesized by triphenyl-phosphonium conjugation with curcumin, and has been shown recently from our laboratory to have broad-spectrum bactericidal activity (Kumari et al. 2019 Free Radic. Biol. Med. 143, 140-145). Here, we further demonstrate the antibacterial properties of MitoC against resistant strains and also its mechanism of action. It displays efficient bactericidal activity against multidrug-resistant Staphylococcus aureus and Streptococcus pneumoniae (MIC values in the 1.5-12.5 µM range), and coagulase-negative Staphylococci do not show resistance development against MitoC. Liposome based studies and MIC values against TolC deletion mutant (Δ tolC; outer membrane protein) of E. coli suggest extensive membrane damage to be the primary mechanism of bactericidal activity. MitoC did not exhibit toxicity in BALB/c mice with an oral administration of 250 mg/kg body weight and was found to be totally safe without any significant effect on haematological, biochemical parameters and inflammatory responses. Its rapid bactericidal action as assessed by in vitro time-kill assay against B. subtilis, compared to ciprofloxacin, and long half-life in rodent serum, suggest that MitoC could be an excellent lead-molecule against drug-resistant pathogens. The highlights of the study are that mitocurcumin belongs to a structurally new class of bactericidal compounds. It displays activity against MDR strains of pathogenic bacteria and challenging MRSA. Liposome-based studies confirm the membrane damaging property of the molecule. Mitocurcumin does not show resistance development even after 27 bacterial generations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Drug Resistance, Bacterial/genetics , Organophosphorus Compounds/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Infections/microbiology , Curcumin/analogs & derivatives , Curcumin/chemistry , Curcumin/pharmacology , Diarylheptanoids/chemistry , Diarylheptanoids/pharmacology , Drug Resistance, Bacterial/drug effects , Humans , Liposomes/chemistry , Molecular Structure , Organophosphorus Compounds/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity
10.
Eur J Pharm Sci ; 144: 105206, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31870813

ABSTRACT

Nanoparticles coupled with targeting moieties have attracted a great deal of attention for cancer therapy since they can facilitate site-specific delivery of drug and significantly limit the side effects of systemic chemotherapy. In this study, our aim is to develop surface functionalized hydroxyapatite nanoparticles, which could provide binding sites for a cancer cell targeting ligand, folic acid (FA) as well as an anticancer drug, doxorubicin hydrochloride (DOX). In order to attain dual functionalities, hydroxyapatite nanoparticles were functionalized with gelatin molecules. Gelatin, being a protein has both carboxyl and amine moieties, which makes it suitable for binding of DOX and FA. FA was chemically conjugated to the nanoparticles through an EDCNHS coupling reaction. The formation of single-phase hydroxyapatite nanostructure was ascertained by X-ray diffraction studies and the presence of organic moieties on the surface of nanoparticles was evident from Fourier transform infrared spectroscopy, thermogravimetric analysis and U.V.-visible spectroscopy. The FA-conjugated nanoparticles (FA-Gel-HANPs) showed high affinity towards DOX and pH-responsive sustained release of drug with higher release rate under acidic pH conditions, desired for cancer therapy. The FA-Gel-HANPs showed negligible cytotoxicity towards different cell lines (HepG2, WEHI-164, KB, WI-26 VA4 and WRL-68). However, DOX loaded nanoparticles (DOX-FA-Gel-HANPs) exhibited significant toxicity towards these cells, which was however highest in folate receptor (FR)-overexpressing, KB cells. These results were correlated with enhanced cellular uptake of DOX-FA-Gel-HANPs in KB cells in comparison to FR-deficient, WRL-68 cells studied by confocal laser scanning microscopy and flow cytometry. Moreover, cell cycle analysis in KB cells, showed higher sub-G1 population, indicating apoptosis as one of the cell death mechanisms. Overall, this study suggests that DOX-FA-Gel-HANPs could serve as a promising tumor-targeted drug delivery system.


Subject(s)
Cell Line, Tumor/drug effects , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Drug Carriers/pharmacology , Durapatite/chemistry , Nanoparticles/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor/cytology , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Durapatite/chemical synthesis , Folic Acid/chemistry , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , KB Cells , Mice , Neoplasms/drug therapy
11.
ACS Omega ; 4(7): 11728-11736, 2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31460279

ABSTRACT

We report the development of pH-labile ascorbic acid-coated magnetic nanocarriers (AMNCs) for effective delivery of the anticancer drug doxorubicin hydrochloride (DOX) to tumor cells. The uniqueness of this drug delivery system lies in the covalent conjugation of DOX through carbamate and hydrazone bonds, resulting in a slow and sustained drug release profile at different environmental acidities. X-ray diffraction and transmission electron microscopy analyses reveal the formation of crystalline single-phase Fe3O4 nanoparticles with an average size of 10 nm. The changes in the interfacial characteristics of the nanocarriers and the presence of organic coatings are probed by infrared spectroscopy, dynamic light scattering, zeta potential, and thermogravimetric measurements. AMNCs show high colloidal stability in aqueous and cell culture media and possess good magnetic field responsivity and protein resistance characteristics. The drug-loaded nanocarriers exhibited sustained pH-triggered release of drug molecules in acidic mediums, substantial cellular internalization, and significant toxicity toward the proliferation of mouse skin fibrosarcoma (WEHI-164), human breast cancer (MCF-7), and human lung cancer (A549) cells. However, it showed significantly lower toxicity in human normal lung (WI26VA) cells. Overall, these results suggest a pH-sensitive drug release of nanoformulations, which showed selective toxicity to tumor than normal cells.

12.
Biochim Biophys Acta Gen Subj ; 1863(5): 857-869, 2019 05.
Article in English | MEDLINE | ID: mdl-30794824

ABSTRACT

BACKGROUND: Nanoparticle-based therapies have emerged as a promising approach to overcome limitations of conventional chemotherapy. Present study investigates the potential of oleic acid-functionalized iron-oxide nanoparticles (MN-OA) to enhance the radiation response of fibrosarcoma tumor and elucidates its underlying mechanism. METHODS: Various cellular and molecular assays (e.g. MTT, clonogenic, cell cycle analysis, cell death, DNA damage/repair) and tumor growth kinetics were employed to investigate the mechanism of MN-OA induced radio-sensitization. RESULTS: Mouse (WEHI-164) and human (HT-1080) fibrosarcoma cells treated with MN-OA and gamma-radiation (2 Gy) showed a significant decrease in the cell proliferation. Combination treatment showed significant decrease in clonogenic survival of WEHI-164 cells and was found to induce cell cycle arrest, apoptosis and mitotic catastrophe. The mechanism of radio-sensitization was found to involve binding of MN-OA with HSP90, resulting in down-regulation of its client proteins, involved in cell cycle progression (Cyclin B1 and CDC2) and DNA-double strand break repair (e.g. RAD51 and BRCA1). Consistently, longer persistence of DNA damage in cells treated with MN-OA and radiation was observed in the form of γ-H2AX foci. The efficacy and mechanism of MN-OA-induced radio-sensitization was also validated in an immuno-competent murine fibrosarcoma model. CONCLUSION: This study reveals the key role of HSP90 in the mechanism of tumor radio-sensitization by MN-OA. GENERAL SIGNIFICANCE: Present work provides a deeper understanding about the mechanism of MN-OA-induced tumor radiosensitization, highlighting the role of HSP90 protein. In addition to diagnostic and magnetic hyperthermia abilities, present remarkable radiosensitizing activity of MN-OA would further excite the clinicians to test its anti-cancer potential.


Subject(s)
Antineoplastic Agents/pharmacology , Ferric Compounds/pharmacology , Fibrosarcoma/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Nanoparticles/chemistry , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Ferric Compounds/chemistry , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , HSP90 Heat-Shock Proteins/metabolism , Humans , Kinetics , Mice , Radiation-Sensitizing Agents/chemistry
13.
Sci Rep ; 8(1): 14766, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30283083

ABSTRACT

Monodispersed Fe3O4 magnetic nanoparticles (MNPs) having size of 7 nm have been prepared from iron oleate and made water dispersible by functionalization for biomedical applications. Three different reactions employing thioglycolic acid, aspartic acid and aminophosphonate were performed on oleic acid coated Fe3O4. In order to achieve a control on particle size, the pristine nanoparticles were heated in presence of ferric oleate which led to increase in size from 7 to 11 nm. Reaction parameters such as rate of heating, reaction temperature and duration of heating have been studied. Shape of particles was found to change from spherical to cuboid. The cuboid shape in turn enhances magneto-crystalline anisotropy (Ku). Heating efficacy of these nanoparticles for hyperthermia was also evaluated for different shapes and sizes. We demonstrate heat generation from these MNPs for hyperthermia application under alternating current (AC) magnetic field and optimized heating efficiency by controlling morphology of particles. We have also studied intra-cellular uptake and localization of nanoparticles and cytotoxicity under AC magnetic field in human breast carcinoma cell line.


Subject(s)
Breast Neoplasms/therapy , Ferric Compounds/therapeutic use , Hyperthermia, Induced/methods , Magnetite Nanoparticles/therapeutic use , Anisotropy , Breast Neoplasms/pathology , Female , Ferric Compounds/chemistry , Humans , Iron/chemistry , MCF-7 Cells , Magnetic Fields , Magnetite Nanoparticles/chemistry , Temperature , Water/chemistry
14.
Int J Radiat Biol ; 94(7): 628-644, 2018 07.
Article in English | MEDLINE | ID: mdl-29775397

ABSTRACT

PURPOSE: One of the most important implications of 'Radiation Biology' research is to improve cancer radiotherapy with minimum side effects. In this regard, combination of chemotherapy with radiation has significantly improved tumor control as well as overall survival in a variety of cancers. However, this has been achieved at the cost of significant normal tissue toxicity, due to the lack of specificity of chemotherapy. Membrane-localized receptor tyrosine kinases (RTKs) have been found to play a driving role in various hallmarks of cancer. Moreover, an early successful clinical trial using RTK-antagonist (cetuximab) to improve tumor radiosensitivity has led to an advancement in this field of research. However, a comprehensive review integrating these findings of various oncogenic RTKs, from basic radiobiology-to-radiotherapy clinical trials, is lacking in literature. Therefore, the present review analyses relevant in-vitro, in-vivo, preclinical/clinical studies and postulates the concept of 'Radiation Biology of RTKs in Cancer'. CONCLUSIONS: The present review elucidates the effect of IR on various oncogenic RTKs and their mechanisms, downstream signaling, intracellular translocations, their role in the repair of radiation-induced DNA damage and post-irradiation survival. Based on the knowledge derived from RTK biology and the analysis of relevant clinical trials, this review attempts to identify radiobiological considerations, which could be implemented in future trials, combining radiotherapy with RTK-antagonist. Additionally, we identify the radiosensitizing potential of recently developed RTK-targeted nanoformulations. This review would probably change the Radiation Oncologist's view for translation of tumor-specific radiosensitization in clinic.


Subject(s)
Neoplasms/radiotherapy , Protein Kinase Inhibitors/therapeutic use , Radiation Tolerance , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Active Transport, Cell Nucleus , Apoptosis , Chemoradiotherapy , DNA Repair , Endocytosis , ErbB Receptors/antagonists & inhibitors , Humans , Neoplasm Invasiveness , Neoplasms/metabolism , Protein Transport/radiation effects , Receptor, IGF Type 1/antagonists & inhibitors , Tumor Escape
15.
Colloids Surf B Biointerfaces ; 162: 163-171, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29190467

ABSTRACT

Highly water-dispersible surfactant-stabilized Fe3O4 magnetic nanocarriers (SMNCs) were prepared by self-assembly of anionic surfactant, sodium dodecyl sulphate (SDS) on hydrophobic (oleic acid coated) nanoparticles and their biomedical applications were investigated. These nanocarriers have an average size of about 10nm and possess tunable surface charge properties. The formation of an organic coating of SDS was evident from infrared spectroscopy, dynamic light scattering, zeta-potential and thermogravimetric measurements. These nanocarriers were used for loading of both hydrophilic and hydrophobic anticancer agents such as doxorubicin hydrochloride (DOX) and curcumin (CUR), respectively. DOX was conjugated onto the surface of nanocarriers through electrostatic interaction, whereas CUR was encapsulated into the hydrophobic interlayer between oleic acid and SDS. The toxicity and cellular internalization of drug loaded nanocarriers were investigated against WEHI-164 cancer cell line. Specifically, the drug loading, pH sensitive drug release and cellular internalization studies suggested that these nanocarriers are suitable for dual drug delivery. Furthermore, they show good heating ability under AC magnetic field, thus can be used as effective heating source for hyperthermia treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Ferrosoferric Oxide/chemistry , Magnetite Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Curcumin/chemistry , Doxorubicin/chemistry , Drug Combinations , Drug Compounding/methods , Fever/drug therapy , Fibroblasts/drug effects , Fibroblasts/pathology , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Magnetic Fields , Magnetite Nanoparticles/ultrastructure , Mice , Oleic Acid/chemistry , Particle Size , Sodium Dodecyl Sulfate/chemistry , Static Electricity , Surface-Active Agents/chemistry
16.
Dalton Trans ; 45(43): 17401-17408, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27731450

ABSTRACT

The interfacial engineering of magnetic nanoparticles (MNPs) with specific functional groups or targeting ligands is important for their in vivo applications. We report here the preparation and characterization of bifunctional magnetic nanoparticles (BMNPs) which contain a carboxylic moiety for drug binding and an amine moiety for folate mediated drug targeting. BMNPs were prepared by introducing bioactive cysteine molecules onto the surface of undecenoic acid coated Fe3O4 magnetic nanoparticles (UMNPs) via a thiol-ene click reaction and then, folic acid was conjugated with these BMNPs through an EDC-NHS coupling reaction. X-ray diffraction (XRD) and transmission electron microscopy (TEM) analysis indicate the formation of highly crystalline single-phase Fe3O4 nanostructures. The changes in the interfacial characteristics of the nanoparticles and the presence of an organic coating are evident from Fourier transform infrared (FTIR) spectroscopy, dynamic light scattering (DLS), zeta-potential measurement, and thermogravimetric analysis (TGA). These nanocarriers have an average size of 10 nm, and have a pH dependent charge conversional feature and protein resistance characteristic in physiological medium. These nanoparticles also show high loading affinity for an anticancer drug, doxorubicin hydrochloride (DOX) and its pH dependent release. This is highly beneficial for cancer therapy as the relatively low pH in tumors will specifically stimulate the drug release at the site of interest. Furthermore, our fluorescence microscopy and flow cytometry studies confirmed the higher cellular internalization capability of these folic acid conjugated nanoparticles in cancer cells over-expressing folate receptors.


Subject(s)
Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Drug Carriers/chemistry , Folic Acid/chemistry , Magnetite Nanoparticles/chemistry , Antineoplastic Agents/administration & dosage , Cysteine/chemistry , Doxorubicin/administration & dosage , Drug Liberation , Endocytosis , Humans , Hydrogen-Ion Concentration , KB Cells , Microscopy, Electron, Transmission , Spectroscopy, Fourier Transform Infrared , Thermogravimetry , X-Ray Diffraction
17.
PLoS One ; 11(8): e0161662, 2016.
Article in English | MEDLINE | ID: mdl-27561007

ABSTRACT

Even though bystander effects pertaining to radiation risk assessment has been extensively studied, the molecular players of radiation induced bystander effect (RIBE) in the context of cancer radiotherapy are poorly known. In this regard, the present study is aimed to investigate the effect of irradiated tumor cells on the bystander counterparts in mouse fibrosarcoma (WEHI 164 cells) tumor model. Mice co-implanted with WEHI 164 cells γ-irradiated with a lethal dose of 15 Gy and unirradiated (bystander) WEHI 164 cells showed inhibited tumor growth, which was measured in terms of tumor volume and Luc+WEHI 164 cells based bioluminescence in vivo imaging. Histopathological analysis and other assays revealed decreased mitotic index, increased apoptosis and senescence in these tumor tissues. In addition, poor angiogenesis was observed in these tumor tissues, which was further confirmed by fluorescence imaging of tumor vascularisation and CD31 expression by immuno-histochemistry. Interestingly, the growth inhibitory bystander effect was exerted more prominently by soluble factors obtained from the irradiated tumor cells than the cellular fraction. Cytokine profiling of the supernatants obtained from the irradiated tumor cells showed increased levels of VEGF, Rantes, PDGF, GMCSF and IL-2 and decreased levels of IL-6 and SCF. Comparative proteomic analysis of the supernatants from the irradiated tumor cells showed differential expression of total 24 protein spots (21 up- and 3 down-regulated) when compared with the supernatant from the unirradiated control cells. The proteins which showed substantially higher level in the supernatant from the irradiated cells included diphosphate kinase B, heat shock cognate, annexin A1, angiopoietin-2, actin (cytoplasmic 1/2) and stress induced phosphoprotein 1. However, the levels of proteins like annexin A2, protein S100 A4 and cofilin was found to be lower in this supernatant. In conclusion, our results provided deeper insight about the damaging RIBE in an in vivo tumor model, which may have significant implication in improvement of cancer radiotherapy.


Subject(s)
Bystander Effect/radiation effects , Fibrosarcoma/drug therapy , Fibrosarcoma/radiotherapy , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Cellular Senescence , Female , Gamma Rays , Mice , Mice, Inbred BALB C , Mitotic Index , Neoplasm Transplantation , Neovascularization, Pathologic , Proteomics , Radiation Dosage , Radiotherapy , Signal Transduction
18.
Int J Radiat Biol ; 92(4): 173-86, 2016.
Article in English | MEDLINE | ID: mdl-26917443

ABSTRACT

PURPOSE: Radionuclide therapy (RNT) is a rapidly growing area of clinical nuclear medicine, wherein radionuclides are employed to deliver cytotoxic dose of radiation to the diseased cells/tissues. During RNT, radionuclides are either directly administered or delivered through biomolecules targeting the diseased site. RNT has been clinically used for diverse range of diseases including cancer, which is the focus of the review. CONCLUSIONS: The major emphasis in RNT has so far been given towards developing peptides/antibodies and other molecules to conjugate a variety of therapeutic radioisotopes for improved targeting/delivery of radiation dose to the tumor cells. Despite that, many of the RNT approaches have not achieved their desired therapeutic success probably due to poor knowledge about complex and dynamic (i) fate of radiolabeled molecules; (ii) radiation dose delivered; (iii) cellular heterogeneity in tumor mass; and (iv) cellular radiobiological response. Based on understanding gathered during recent years, it may be stated that besides the absorbed dose, the net radiobiological response of tumor/normal cells also determines the clinical response of radiotherapeutic modalities including RNT. The radiosensitivity of tumor/normal cells is governed by radiobiological phenomenon such as radiation-induced bystander effect, genomic instability, adaptive response and low dose hyper-radiosensitivity. These concepts have been well investigated in the context of external beam radiotherapy, but their clinical implications during RNT have received meagre attention. In this direction, a few studies performed using in vitro and in vivo models envisage the possibilities of exploiting the radiobiological knowledge for improved therapeutic outcome of RNT.


Subject(s)
Models, Biological , Neoplasms/physiopathology , Neoplasms/radiotherapy , Radiobiology/methods , Radioisotopes/therapeutic use , Radiotherapy, Computer-Assisted/methods , Apoptosis/radiation effects , Cell Survival/radiation effects , DNA Damage , Dose-Response Relationship, Radiation , Neoplasms/pathology , Radiation Dosage , Radiopharmaceuticals/therapeutic use
19.
Int J Hyperthermia ; 31(8): 909-19, 2015.
Article in English | MEDLINE | ID: mdl-26416812

ABSTRACT

PURPOSE: We have evaluated the hyperthermia efficacy of oleic acid-functionalised Fe(3)O(4) magnetic nanoparticles (MN-OA) under in vivo conditions and elucidated the underlying mechanism of tumour growth inhibition. MATERIALS AND METHODS: The efficacy and mechanism of tumour growth inhibition by MN-OA-mediated magnetic hyperthermia therapy (MHT) was evaluated in a murine fibrosarcoma tumour model (WEHI-164) using techniques such as TUNEL assay, Western blotting (WB), immunofluorescence (IF) staining and histopathological examination. In addition, bio-distribution of MN-OA in tumour/other target organs and its effect on normal organ function were studied by Prussian blue staining and serum biochemical analysis, respectively. RESULTS: MN-OA-induced MHT resulted in significant inhibition of tumour growth as determined by measurement of tumour volume, as well as by in vivo imaging of tumour derived from luciferase-transfected WEHI-164 cells. Histopathology analysis showed presence of severe apoptosis and reduced tumour cells proliferation, which was further confirmed by TUNEL assay, reduced expression of Ki-67 and enhanced level of cleaved caspase-3, in tumours treated with MHT. Moreover, expression of heat stress marker, Hsp90 and its client protein, AKT/PKB was reduced by ∼50 and 80%, respectively, in tumours treated with MHT as studied by WB and IF staining. Serum analysis suggested insignificant toxicity of MN-OA (in terms of liver and kidney function), which was further correlated with minimal accumulation of MN-OA in target organs. CONCLUSIONS: These results suggest the involvement of apoptosis and Hsp90/AKT modulation in MN-OA-mediated MHT-induced tumour growth inhibition.


Subject(s)
Fibrosarcoma/drug therapy , Hyperthermia, Induced , Magnetite Nanoparticles/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Combined Modality Therapy , Female , Fibrosarcoma/metabolism , HSP90 Heat-Shock Proteins/metabolism , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Magnetite Nanoparticles/therapeutic use , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/metabolism , Spleen/metabolism , Tissue Distribution , Tumor Burden/drug effects
20.
Dalton Trans ; 44(33): 14686-96, 2015 Sep 07.
Article in English | MEDLINE | ID: mdl-26215789

ABSTRACT

Magnetic luminescent hybrid nanostructures (MLHN) have received a great deal of attention due to their potential biomedical applications such as thermal therapy, magnetic resonance imaging, drug delivery and intracellular imaging. We report the development of bifunctional Fe3O4 decorated YPO4:Eu hybrid nanostructures by covalent bridging of carboxyl PEGylated Fe3O4 and amine functionalized YPO4:Eu particles. The surface functionalization of individual nanoparticulates as well as their successful conjugation was evident from Fourier transform infrared (FTIR) spectroscopy, dynamic light scattering (DLS), zeta-potential and transmission electron microscopy (TEM) studies. X-ray diffraction (XRD) analysis reveals the formation of highly crystalline hybrid nanostructures. TEM micrographs clearly show the binding/anchoring of 10 nm Fe3O4 nanoparticles onto the surface of 100-150 nm rice grain shaped YPO4:Eu nanostructures. These MLHN show good colloidal stability, magnetic field responsivity and self-heating capacity under an external AC magnetic field. The induction heating studies confirmed localized heating of MLHN under an AC magnetic field with a high specific absorption rate. Photoluminescence spectroscopy and fluorescence microscopy results show optical imaging capability of MLHN. Furthermore, successful internalization of these MLHN in the cells and their cellular imaging ability are confirmed from confocal microscopy imaging. Specifically, the hybrid nanostructure provides an excellent platform to integrate luminescent and magnetic materials into one single entity that can be used as a potential tool for hyperthermia treatment of cancer and cellular imaging.


Subject(s)
Europium/chemistry , Ferrosoferric Oxide/chemistry , Luminescent Agents/chemistry , Nanostructures/chemistry , Phosphates/chemistry , Yttrium/chemistry , Cell Line, Tumor , Europium/therapeutic use , Ferrosoferric Oxide/therapeutic use , Humans , Hyperthermia, Induced , Luminescent Agents/therapeutic use , Magnetic Fields , Magnetics , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Nanostructures/therapeutic use , Nanostructures/ultrastructure , Neoplasms/diagnosis , Neoplasms/therapy , Optical Imaging , Yttrium/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...