Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Carbohydr Polym ; 336: 122136, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38670762

ABSTRACT

The standard treatment for early-stage breast cancer involves breast-conserving surgery followed by adjuvant radiotherapy. However, approximately 20 % of patients experience distant metastasis, and adjuvant radiotherapy often leads to radiation-induced skin fibrosis (RISF). In this study, we develop an on-site injectable formulation composed of selenocystamine (SeCA) and hyaluronic acid (HyA), referred to as SeCA cross-linked HyA (SCH) agent, and investigate its potential to mitigate metastasis and prevent RISF associated with breast cancer therapy. SCH agents are synthesized using the nanoprecipitation method to modulate cell-cell tight junctions and tissue inflammation. The toxicity assessments reveal that SCH agents with a higher Se content (Se payload 17.4 µg/mL) are well tolerated by L929 cells compared to SeCA (Se payload 3.2 µg/mL). In vitro, SCH agents significantly enhance cell-cell tight junctions and effectively mitigate migration and invasion of breast cancer cells (4T1). In vivo, SCH agents mitigate distant lung metastasis. Furthermore, in animal models, SCH agents reduce RISF and promote wound repair. These findings highlight the potential of SCH agents as a novel therapeutic formulation for effectively mitigating metastasis and reducing RISF. This holds great promise for improving clinical outcomes in breast cancer patients undergoing adjuvant radiotherapy.


Subject(s)
Breast Neoplasms , Fibrosis , Hyaluronic Acid , Hyaluronic Acid/chemistry , Animals , Female , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Mice , Fibrosis/drug therapy , Cell Line, Tumor , Humans , Mice, Inbred BALB C , Cystamine/chemistry , Cystamine/pharmacology , Skin/drug effects , Skin/pathology , Skin/radiation effects , Cell Movement/drug effects , Injections
2.
Phys Med Biol ; 69(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38170992

ABSTRACT

This study developed a prototype for a rotational cone-beam x-ray luminescence computed tomography (CB-XLCT) system, considering its potential application in pre-clinical theranostic imaging. A geometric calibration method applicable to both imaging chains (XL and CT) was also developed to enhance image quality. The results of systematic performance evaluations were presented to assess the feasibility of commercializing XLCT technology. Monte Carlo GATE simulation was performed to determine the optimal imaging conditions for nanophosphor particles (NPs) irradiated by 70 kV x-rays. We acquired a low-dose transmission x-ray tube and designed a prone positioning platform and a rotating gantry, using mice as targets from commercial small animalµ-CT systems. We then employed the image cross-correlation (ICC) automatic geometric calibration method to calibrate XL and CT images. The performance of the system was evaluated through a series of phantom experiments with a linearity of 0.99, and the contrast-to-noise ratio (CNR) between hydroxyl-apatite (HA) and based epoxy resin is 19.5. The XL images of the CB-XLCT prototype achieved a Dice similarity coefficient (DICE) of 0.149 for a distance of 1 mm between the two light sources. Finally, the final XLCT imaging results were demonstrated using the Letter phantoms with NPs. In summary, the CB-XLCT prototype developed in this study showed the potential to achieve high-quality imaging with acceptable radiation doses for small animals. The performance of CT images was comparable to current commercial machines, while the XL images exhibited promising results in phantom imaging, but further efforts are needed for biomedical applications.


Subject(s)
Image Processing, Computer-Assisted , Luminescence , Animals , Mice , X-Rays , Image Processing, Computer-Assisted/methods , Algorithms , Tomography, X-Ray Computed/methods , Cone-Beam Computed Tomography/methods , Phantoms, Imaging
3.
Int J Nanomedicine ; 18: 7677-7693, 2023.
Article in English | MEDLINE | ID: mdl-38111846

ABSTRACT

Purpose: Glioblastoma is a highly aggressive brain tumor with universally poor outcomes. Recent progress in immune checkpoint inhibitors has led to increased interest in their application in glioblastoma. Nonetheless, the unique immune milieu in the brain has posed remarkable challenges to the efficacy of immunotherapy. We aimed to leverage the radiation-induced immunogenic cell death to overcome the immunosuppressive network in glioblastoma. Methods: We developed a novel approach using the gold-core silica-shell nanoparticles (Au@SiO2 NPs) in combination with low-dose radiation to enhance the therapeutic efficacy of the immune checkpoint inhibitor (atezolizumab) in brain tumors. The biocompatibility, immune cell recruitment, and antitumor ability of the combinatorial strategy were determined using in vitro assays and in vivo models. Results: Our approach successfully induced the migration of macrophages towards brain tumors and promoted cancer cell apoptosis. Subcutaneous tumor models demonstrated favorable safety profiles and significantly enhanced anticancer effects. In orthotopic brain tumor models, the multimodal therapy yielded substantial prognostic benefits over any individual modalities, achieving an impressive 40% survival rate. Conclusion: In summary, the combination of Au@SiO2 NPs and low-dose radiation holds the potential to improve the clinical efficacy of immune checkpoint inhibitors. The synergetic strategy modulates tumor microenvironments and enhances systemic antitumor immunity, paving a novel way for glioblastoma treatment.


Subject(s)
Brain Neoplasms , Glioblastoma , Nanoparticles , Humans , Silicon Dioxide/therapeutic use , Glioblastoma/drug therapy , Gold/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Brain Neoplasms/radiotherapy , Brain Neoplasms/drug therapy , Cell Line, Tumor , Tumor Microenvironment
4.
Biomed Phys Eng Express ; 8(6)2022 10 21.
Article in English | MEDLINE | ID: mdl-36223710

ABSTRACT

Reducing the radiation dose will cause severe image noise and artifacts, and degradation of image quality will also affect the accuracy of diagnosis. To find a solution, we comprise a 2D and 3D concatenating convolutional encoder-decoder (CCE-3D) and the structural sensitive loss (SSL), via transfer learning (TL) denoising in the projection domain for low-dose computed tomography (LDCT), radiography, and tomosynthesis. The simulation and real-world practicing results show that many of the figures-of-merit (FOMs) increase in both projections (2-3 times) and CT imaging (1.5-2 times). From the PSNR and structural similarity index of measurement (SSIM), the CCE-3D model is effective in denoising but keeps the shape of the structure. Hence, we have developed a denoising model that can be served as a promising tool to be implemented in the next generation of x-ray radiography, tomosynthesis, and LDCT systems.


Subject(s)
Deep Learning , Cone-Beam Computed Tomography , Tomography, X-Ray Computed/methods , Artifacts , Computer Simulation
5.
ACS Appl Mater Interfaces ; 14(4): 5586-5597, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35050587

ABSTRACT

It is vital to remove residual tumor cells after resection to avoid the recurrence and metastasis of osteosarcoma. In this study, a mineral nanomedicine, europium-doped calcium fluoride (CaF2:Eu) nanoparticles (NPs), is developed to enhance the efficacy of adjuvant radiotherapy (i.e., surgical resection followed by radiotherapy) for tumor cell growth and metastasis of osteosarcoma. In vitro studies show that CaF2:Eu NPs (200 µg/mL) exert osteosarcoma cell (143B)-selective toxicity and migration-inhibiting effects at a Eu dopant amount of 2.95 atomic weight percentage. These effects are further enhanced under X-ray irradiation (6 MeV, 4 Gy). Furthermore, in vivo tests show that intraosseous injection of CaF2:Eu NPs and X-ray irradiation have satisfactory therapeutic efficacy in controlling primary tumor size and inhibiting primary tumor metastasis. Overall, our results suggest that CaF2:Eu NPs with their osteosarcoma cell (143B)-selective toxicity and migration-inhibiting effects combined with radiotherapy might be nanomedicines for treating osteosarcoma after tumor resection.


Subject(s)
Antineoplastic Agents/therapeutic use , Calcium Fluoride/therapeutic use , Europium/therapeutic use , Metal Nanoparticles/therapeutic use , Osteosarcoma/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Calcium Fluoride/chemistry , Calcium Fluoride/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Europium/chemistry , Europium/toxicity , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Mice , Radiotherapy, Adjuvant
6.
Nanomaterials (Basel) ; 11(9)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34578551

ABSTRACT

Radiotherapy (RT), in combination with surgery, is an essential treatment strategy for oral cancer. Although irradiation provides effective control over tumor growth, the surrounding normal tissues are almost inevitably affected. With further understanding of the molecular mechanisms involved in radiation response and recent advances in nanotechnology, using gold nanoparticles as a radiosensitizer provides the preferential sensitization of tumor cells to radiation and minimizes normal tissue damage. Herein, we developed gold nano-sesame-beads (GNSbs), a gold-nanorod-seeded mesoporous silica nanoparticle, as a novel radioenhancer to achieve radiotherapy with a higher therapeutic index. GNSbs in combination with 2 Gy irradiation effectively enhanced the cytotoxic activity CAL-27 cells. The well-designed structure of GNSbs showed preferential cellular uptake by CAL-27 cells at 24 h after incubation. Gold nanorods with high density modified on mesoporous silica nanoparticles resulted in significant reactive oxygen species (ROS) formation after irradiation exposure compared with irradiation alone. Furthermore, GNSbs and irradiation induced more prominent DNA double-strand breaks and G2/M phase arrest in CAL-27 than those in L929. In animal studies, radiotherapy using GNSbs as a radiosensitizer showed significant suppression of tumor growth in an orthotopic model of oral cancer. These results demonstrate that using GNSbs as a radiosensitizer could possess clinical potential for the treatment of oral squamous carcinoma.

7.
Nanomaterials (Basel) ; 11(7)2021 Jun 24.
Article in English | MEDLINE | ID: mdl-34202555

ABSTRACT

Glioblastoma, formerly known as glioblastoma multiforme (GBM), is refractory to existing adjuvant chemotherapy and radiotherapy. We successfully synthesized a complex, Au-OMV, with two specific nanoparticles: gold nanoparticles (AuNPs) and outer-membrane vesicles (OMVs) from E. coli. Au-OMV, when combined with radiotherapy, produced radiosensitizing and immuno-modulatory effects that successfully suppressed tumor growth in both subcutaneous G261 tumor-bearing and in situ (brain) tumor-bearing C57BL/6 mice. Longer survival was also noted with in situ tumor-bearing mice treated with Au-OMV and radiotherapy. The mechanisms for the successful treatment were evaluated. Intracellular reactive oxygen species (ROS) greatly increased in response to Au-OMV in combination with radiotherapy in G261 glioma cells. Furthermore, with a co-culture of G261 glioma cells and RAW 264.7 macrophages, we found that GL261 cell viability was related to chemotaxis of macrophages and TNF-α production.

8.
Acta Biomater ; 125: 300-311, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33609743

ABSTRACT

Although boron neuron capture therapy (BNCT) has enabled the delivery of stronger radiation dose to glioblastoma multiforme (GBM) cells for precision radiotherapy (RT), patients in need are almost unable to access the treatment due to insufficient operating devices. Therefore, we developed targeted sensitization-enhanced radiotherapy (TSER), a strategy that could achieve precision cell-targeted RT using common linear accelerators. TSER, which involves the combination of GoldenDisk (GD; a spherical radioenhancer), 5-aminolevulinic acid (5-ALA), low-intensity ultrasound (US), and low-dose RT, exhibited synergized radiosensitization effects. Both 5-ALA and hyaluronic-acid-immobilized GD can selectively accumulate in GBM to induce chemical and biological enhancement of radiosensitization, resulting in DNA damage, escalation of reactive oxygen species levels, and cell cycle redistribution, in turn sensitizing GBM cells to radiation under US. TSER showed an enhanced therapeutic effect and survival in the treatment of an orthotropic GBM model with only 20% of the radiation dose compared to that of a 10-Gy RT. The strategy with the potential to inhibit GBM progress and rescue the organ at risk using low-dose RT, thereby improving the quality of life of GBM patients, shedding light on achieving cell-targeted RT using universally available linear accelerators. STATEMENT OF SIGNIFICANCE: We invented GoldenDisk (GD), a radioenhancer with hyaluronic-acid (HAc)-coated gold nanoparticle (AuNP)-core/silica shell nanoparticle, to make radiotherapy (RT) safer and smarter. The surface modification of HAc and silica allows GD to target CD44-overexpressed glioblastoma multiforme (GBM) cells and stay structurally stable in cytoplasm throughout the course of RT. By combining GD with low-energy ultrasound and an FDA-approved imaging agent, 5-aminolevulinic acid (5-ALA), GBM cells were sensitized to RT leaving healthy tissues in the vicinity unaffected. The ionized radiation can further be transferred to photoelectronic products with higher cytotoxicity by GD upon collision, achieving higher therapeutic efficacy. With the newly-developed strategy, we are able to achieve low-dose precision RT with the use of only 20% radiation dose.


Subject(s)
Brain Neoplasms , Glioblastoma , Metal Nanoparticles , Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Gold , Humans , Particle Accelerators , Quality of Life
9.
Curr Med Chem ; 28(19): 3877-3889, 2021.
Article in English | MEDLINE | ID: mdl-33213306

ABSTRACT

Osteosarcoma is insensitive to radiation. High-dose radiation is often used as a treatment but causes side effects in patients. Hence, it is important to develop tumor cell-- targeted radiotherapy that could improve radiotherapy efficiency on tumor cells and reduce the toxic effect on normal cells during radiation treatment. In this study, we developed an innovative method for treating osteosarcoma by using a novel radiation-enhancer (i.e., carboxymethyl-hexanoyl chitosan-coated self-assembled Au@Fe3O4 nanoparticles; CSAF NPs). CSAF NPs were employed together with 5-aminolevulinic acid (5- ALA) to achieve tumor cell-targeted radiotherapy. In this study, osteosarcoma cells (MG63) and normal cells (MC3T3-E1) were used for an in vitro investigation, in which reactive oxygen species (ROS) assay, cell viability assay, clonogenic assay, and western blot were used to confirm the treatment efficiency. The ROS assay showed that the combination of CSAF NPs and 5-ALA enhanced radiation-induced ROS production in tumor cells (MG63); however, this was not observed in normal cells (MC3T3-E1). The cell viability ratio of normal cells to tumor cells after treatment with CSAF NPs and 5-ALA reached 2.79. Moreover, the clonogenic assay showed that the radiosensitivity of MG63 cells was increased by the combination use of CSAF NPs and 5-ALA. This was supported by performing a western blot that confirmed the expression of cytochrome c (a marker of cell mitochondria damage) and caspase-3 (a marker of cell apoptosis). The results provide an essential basis for developing tumor-cell targeted radiotherapy by means of low-- dose radiation.


Subject(s)
Osteosarcoma , Aminolevulinic Acid , Apoptosis , Cell Line, Tumor , Cell Survival , Humans , Reactive Oxygen Species
10.
J Mater Chem B ; 8(25): 5460-5471, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32462165

ABSTRACT

Metastasis resulting from circulating tumor cells (CTCs) is associated with 90% of all cancer mortality. To disrupt cancer dissemination, therapeutic targeting of CTCs by extracorporeal photodynamic therapy (PDT) has emerged; however, it still remains impractical due to its limited therapeutic window. Herein, we developed a photosensitive and magnetic targeted core-satellite nanomedicine (TCSN) to augment the light-induced damage to the targeted cells. The magnetic nanocore (MNC) with multiple iron oxide nanoparticles stabilized using thiolated polyvinyl alcohol can magnetize the CTCs to achieve magnetic enrichment under a magnetic field. Multiple gold nanocage (AuNC) satellites were conjugated on the MNC to facilitate bimodal photothermal therapy and PDT. Adjusting the thiol content in the MNC allows manipulating the AuNC density on TCSNs, which has been found to demonstrate a density-dependent bimodal phototherapeutic effect under laser irradiation at 808 and 940 nm. Moreover, with the immobilization of anti-epithelial cell adhesion molecule (anti-EpCAM), TCSN exhibited an enhanced affinity toward EpCAM-expressing 4T1 cells. We demonstrate that TCSN-labeled 4T1 cells can be isolated and photo-eradicated in a microfluidic channel with a dynamic flow. Our studies showed that TCSN with the complementary properties of MNC and AuNCs can largely augment the therapeutic window by magnetic enrichment and bimodal phototherapy, serving as an advanced extracorporeal strategy to remove CTCs.


Subject(s)
Gold/pharmacology , Metal Nanoparticles/chemistry , Neoplastic Cells, Circulating/drug effects , Photochemotherapy , Photosensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Gold/chemistry , Lasers , Magnetic Fields , Mice , Nanomedicine , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Particle Size , Photosensitizing Agents/chemistry , Surface Properties
11.
Nanoscale ; 12(16): 8809-8818, 2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32250377

ABSTRACT

The application of radiotherapy (RT) to treat osteosarcoma (OS) has been limited, but this is starting to change as the ability to target radiation energy to niches improves. Furthermore, lung cancer from highly metastatic OS is a major cause of death, so it is critical to explore new strategies to tackle metastasis. In this study, we designed a nanoscale radiosensitizer by grafting 2-deoxy-d-glucose (2DG) onto graphene quantum dots (GQD) to achieve OS targeting and boost RT efficacy. Combining the use of 2DG-grafted GQDs (2DG-g-GQD) with RT produced a significant increase in oxidative stress response and DNA damage in the 143B OS cell line compared with RT alone. Moreover, 2DG-g-GQDs selectively associated with 143B cells, and demonstrated the inhibition of migration in a scratch assay. We also demonstrated remarkable improvement in their ability to inhibit tumour progression and lung metastasis in an OS xenograft mouse model. Our results show that the use of 2DG-g-GQDs as OS-targeting radiosensitizers improves their therapeutic outcome and exhibits potential for use in low-dose precision RT for OS.


Subject(s)
Deoxyglucose/chemistry , Graphite/chemistry , Osteosarcoma/radiotherapy , Quantum Dots/therapeutic use , Radiation-Sensitizing Agents/chemistry , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , DNA Damage , Deoxyglucose/pharmacokinetics , Deoxyglucose/therapeutic use , Drug Delivery Systems , Glucose/chemistry , Glucose/pharmacokinetics , Glucose/therapeutic use , Graphite/pharmacokinetics , Graphite/therapeutic use , Humans , Mice , Neoplasm Metastasis/prevention & control , Osteosarcoma/metabolism , Osteosarcoma/pathology , Quantum Dots/chemistry , Radiation-Sensitizing Agents/pharmacokinetics , Radiation-Sensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Treatment Outcome
12.
Int J Mol Sci ; 21(4)2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32102332

ABSTRACT

In-stent restenosis is a serious concern for patients treated through the stenting procedure, although this can be solved using drug-eluting stents and/or drug-eluting balloon catheters. However, the chemical agents released from the drug-eluting layer for inhibiting smooth muscle cell (SMC) migration are inevitably associated with damage to vascular endothelial cell (ECs). The present in vitro study used a distinct strategy, in which a smart gene (phEGR1-PKCδ, an engineered plasmid consists of an SMC-specific promoter (human early growth response 1, hEGR1 promoter) ligated with a gene encoding apoptosis-inducing protein (protein kinase C-delta, PKCδ) was incorporated into a novel gene vehicle (Au cluster-incorporated polyethylenimine/carboxymethyl hexanoyl chitosan, PEI-Au/CHC) to form the PEI-Au/CHC/phEGR1-PKCδ complex, which was proposed for the selective inhibition of SMC proliferation. It was found that the cell viability of SMCs receiving the PEI-Au/CHC/phEGR1-PKCδ complex under simulated inflammation conditions was significantly lower than that of the ECs receiving the same treatment. In addition, the PEI-Au/CHC/phEGR1-PKCδ complex did not demonstrate an inhibitory effect on EC proliferation and migration under simulated inflammation conditions. Finally, the PEI-Au/CHC/phEGR1-PKCδ complexes coated onto a balloon catheter used in percutaneous transluminal coronary angioplasty (PTCA) could be transferred to both the ECs and the SMC layer of Sprague Dawley (SD) rat aortas ex vivo. These preliminary in vitro results suggest that the newly developed approach proposed in the present study might be a potential treatment for reducing the incidence rate of in-stent restenosis and late thrombosis in the future.


Subject(s)
Early Growth Response Protein 1/metabolism , Genetic Therapy/methods , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Kinase C-delta/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Apoptosis/genetics , Cell Survival/genetics , Coronary Restenosis/genetics , Coronary Restenosis/therapy , Drug Carriers/chemistry , Drug-Eluting Stents , Early Growth Response Protein 1/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genetic Engineering , Microscopy, Fluorescence , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Nanostructures/chemistry , Protein Kinase C-delta/genetics , Rats, Sprague-Dawley
13.
Biomed Res Int ; 2017: 4732302, 2017.
Article in English | MEDLINE | ID: mdl-28791302

ABSTRACT

This study constructs a standard in vitro laser treatment platform with dental implant thread surface on bacterial adhesion for peri-implantitis at different tooth positions. The standard clinical adult tooth jaw model was scanned to construct the digital model with 6 mm bone loss depth on behalf of serious peri-implantitis at the incisor, first premolar, and first molar. A cylindrical suite connected to the implant and each tooth root in the jaw model was designed as one experimental unit set to allow the suite to be replaced for individual bacterial adhesion. The digital peri-implantitis and suite models were exported to fulfill the physical model using ABS material in a 3D printer. A 3 mm diameter specimen implant on bacterial adhesion against Escherichia coli was performed for gram-negative bacteria. An Er:YAG laser, working with a chisel type glass tip, was moved from the buccal across the implant thread to the lingual for about 30 seconds per sample to verify the in vitro laser treatment platform. The result showed that the sterilization rate can reach 99.3% and the jaw model was not damaged after laser irradiation testing. This study concluded that using integrated image processing, reverse engineering, CAD system, and a 3D printer to construct a peri-implantitis model replacing the implant on bacterial adhesion and acceptable sterilization rate proved the feasibility of the proposed laser treatment platform.


Subject(s)
Bacterial Adhesion , Dental Implants , Lasers, Solid-State/therapeutic use , Peri-Implantitis/microbiology , Peri-Implantitis/surgery , Alveolar Bone Loss/surgery , Bacterial Adhesion/radiation effects , Jaw/pathology , Printing, Three-Dimensional
14.
Acta Biomater ; 47: 159-169, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27713087

ABSTRACT

Many non-antibiotic strategies, such as photocatalysis and photodynamic therapy, have been proposed to inhibit and/or kill bacteria. However, these approaches still have drawbacks such as insufficient bacterial specificity and the limited penetration depth of ultraviolet and near-infrared light. To overcome these limitations, we developed a bacteria-specific anti-bacterial technique via using low-dose X-ray. Graphene oxide quantum dots (GQDs, a multifunctional vehicle) conjugated with vancomycin (Van, a bacteria-targeting ligand) were assembled with Protoporphyrin IX (PpIX, a photo/radiation sensitizer) to yield a novel Van-GQDs/PpIX complex that specifically attached to Escherichia coli and efficiently generated intracellular reactive oxygen species following X-ray activation. Delivery using GQDs increased the PpIX/Van ratio in the target bacterial cell, damaged bacterial cell wall, and enhanced X-ray-induced PpIX activation. Hence, this approach allowed for the use of a low-dose X-ray to efficiently activate the Van-GQDs/PpIX complex to exert its bactericidal effects on Escherichia coli without damaging normal cells. Furthermore, the E. coli did not develop resistance to the proposed approach for at least 7 rounds of repeated administration during one week. Thus, this proposed vehicle exhibiting bacteria-specific X-ray-triggered toxicity is a promising alternative to antibiotics for treating serious bacterial infections occurring in deep-seated tissues/organs (e.g., osteomyelitis and peritonitis). STATEMENTS OF SIGNIFICANCE: Administration of antibiotics is the most common treatment modality for bacterial infections. However, in some cases, patient attributes such as age, health, tolerance to antibiotics do not allow for the use of high-dose antibiotics. In addition, some bacteria develop resistance to antibiotics because of improper and long-term use of these agents. Therefore, non-antibiotic strategies to treat deeply situated bacterial infections, such as osteomyelitis, are urgently needed for avoiding amputation. To date, several non-antibiotic approaches, such as Ag nanoparticles, graphene-based materials, photocatalysis, and photodynamic therapy have been proposed to inhibit and/or kill bacteria. However, the major challenges of photochemical strategies, specificity and limited penetration depth of light source, still remain for treating the deep-seated bacteria. To overcome these problems, we developed a novel nanovehicle that exerted toxic effects specifically on bacteria following activation by a deeply penetrative low-dose X-ray, without damaging normal cells. As such, it realizes a deeply photochemical route for treating the deep-seated bacteria.


Subject(s)
Escherichia coli/radiation effects , Nanoparticles/chemistry , Animals , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Radiation , Graphite/chemistry , Mice , Microbial Viability/drug effects , Quantum Dots/chemistry , Quantum Dots/ultrastructure , Reactive Oxygen Species/metabolism , Vancomycin/pharmacology , X-Rays
15.
Biomed Mater ; 11(3): 035010, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27271471

ABSTRACT

Extensive epidural fibrosis is a common complication following spinal surgery and can cause pain and limited mobility. In the present study, a novel biomimetic approach was developed to prevent postsurgical adhesion of the dura. We aimed to reconstruct epidural fat, which prevents scar-tissue adhesion, through the development of an injectable decellularized adipose matrix (DAM)-containing hyaluronic acid (HA) hydrogel loaded with adipose stromal cells (ASCs). Injectable DAM was prepared from porcine adipose tissue by four freeze-thaw cycles with subsequent pepsin digestion. Residual analyses confirmed the efficacy of detergent-free decellularization, while most sulfated glycosaminoglycans and collagen were preserved. The Transwell migration assay demonstrated the anti-infiltrative property of the DAM-containing HA hydrogel. After 14 d of 3D culture, the DAM-containing HA hydrogel showed inductive potential in the adipogenic differentiation of ASCs. For an in vivo study, the ASC-loaded DAM-containing HA hydrogel (DAM/ASC-incorporated HA hydrogel) was injected into adult laminectomized male rats, and the results were assessed by microscopic histological examination. The in vivo data indicated that HA hydrogel, DAM, and ASCs were all required for the ability of the engineered fat tissue to block the invasion of the fibrous tissue. Our results suggested that this injectable DAM/ASC-incorporated HA hydrogel has potential applications in minimally invasive surgery for soft-tissue reconstruction and epidural fibrosis prevention.


Subject(s)
Adipocytes/cytology , Adipogenesis , Adipose Tissue/cytology , Extracellular Matrix/chemistry , Hyaluronic Acid/chemistry , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Cell Adhesion , Cell Differentiation , Cell Line , Cell Movement , Cells, Cultured , Fibrosis , Hydrogels/chemistry , Laminectomy , Male , Mice , Rats , Rats, Sprague-Dawley , Solubility
16.
J Biomater Appl ; 30(10): 1589-600, 2016 May.
Article in English | MEDLINE | ID: mdl-26935813

ABSTRACT

The formation of fibrous tissue is part of the natural healing response following a laminectomy. Severe scar tissue adhesion, known as epidural fibrosis, is a common cause of failed back surgery syndrome. In this study, by combining the advantages of drug treatment with a physical barrier, an ibuprofen-conjugated crosslinkable polygalacturonic acid and hyaluronic acid hydrogel was developed for epidural fibrosis prevention. Conjugation was confirmed and measured by 1D(1)H NMR spectroscopy.In vitroanalysis showed that the ibuprofen-conjugated polygalacturonic acid-hyaluronic acid hydrogel showed low cytotoxicity. In addition, the conjugated ibuprofen decreased prostaglandin E2production of the lipopolysaccharide-induced RAW264.7 cells. Histological data inin vivostudies indicated that the scar tissue adhesion of laminectomized male adult rats was reduced by the application of our ibuprofen-conjugated polygalacturonic acid-hyaluronic acid hydrogel. Its use also reduced the population of giant cells and collagen deposition of scar tissue without inducing extensive cell recruitment. The results of this study therefore suggest that the local delivery of ibuprofenviaa polygalacturonic acid-hyaluronic acid-based hydrogel reduces the possibility of epidural fibrosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cicatrix/prevention & control , Epidural Space/drug effects , Epidural Space/pathology , Ibuprofen/administration & dosage , Laminectomy/adverse effects , Pectins/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cicatrix/etiology , Cicatrix/pathology , Drug Carriers/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Ibuprofen/therapeutic use , Male , Mice , Postoperative Complications/pathology , Postoperative Complications/prevention & control , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Tissue Adhesions/etiology , Tissue Adhesions/pathology , Tissue Adhesions/prevention & control , Wound Healing/drug effects
17.
Nanoscale ; 8(25): 12648-57, 2016 Jul 07.
Article in English | MEDLINE | ID: mdl-26838477

ABSTRACT

Sonodynamic therapy (SDT), which induces activation of sonosensitizers in cancer cells through ultrasound irradiation, has emerged as an alternative and promising noninvasive therapeutic approach to kill both superficial and deep parts of tumors. In this study, mesoporous silica (MSN) grown on reduced graphene oxide nanosheet (nrGO) capped with Rose Bengal (RB)-PEG-conjugated iron-oxide nanoparticles (IONs), nrGO@MSN-ION-PEG-RB, was strategically designed to have targeted functionality and therapeutic efficacy under magnetic guiding and focused ultrasound (FUS) irradiation, respectively. The singlet oxygen produced by ultrasound-activated RB and the ultrasound-induced heating effect was enhanced by rGO and IONs, which improved the cytotoxic effect in cancer cells. In an animal experiment, we demonstrated that the combination of sonodynamic/hyperthermia therapy with magnetic guidance using this nanocomposite therapeutic agent can produce remarkable efficacious therapy in tumor growth inhibition. Furthermore, the combination effect induced by FUS irradiation produces significant damage to both superficial and deep parts of the targeted tumor.


Subject(s)
Ferric Compounds , Graphite , Hyperthermia, Induced , Nanostructures , Neoplasms, Experimental/therapy , Theranostic Nanomedicine , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms , Silicon Dioxide
18.
J Control Release ; 226: 182-92, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-26892750

ABSTRACT

Radiotherapy, a common cancer treatment, often adversely affects the surrounding healthy tissue and/or cells. Some tumor tissue-focused radiation therapies have been developed to lower radiation-induced lesion formation; however, achieving tumor cell-targeted radiotherapy (i.e., precisely focusing the radiation efficacy to tumor cells) remains a challenge. In the present study, we developed a novel tumor cell-targeted radiotherapy, named targeted sensitization-enhanced radiotherapy (TSER), that exploits tumor-specific folic acid-conjugated carboxymethyl lauryl chitosan/superparamagnetic iron oxide (FA-CLC/SPIO) micelles to effectively deliver chlorin e6 (Ce6, a sonosensitizer) to mitochondria of HeLa cells under magnetic guidance. For the in vitro tests, the sensitization of Ce6 induced by ultrasound, that could weaken the radiation resistant ability of tumor cells, occurred only in Ce6-internalizing tumor cells. Therefore, low-dose X-ray irradiation, that was not harmful to normal cells, could exert high tumor cell-specific killing ability. The ratio of viable normal cells to tumor cells was increased considerably, from 7.8 (at 24h) to 97.1 (at 72h), after they had received TSER treatment. Our data suggest that TSER treatment significantly weakens tumor cells, resulting in decreased viability in vitro as well as decreased in vivo subcutaneous tumor growth in nude mice, while the adverse effects were minimal. Taken together, TSER treatment appears to be an effective, clinically feasible tumor cell-targeted radiotherapy that can solve the problems of traditional radiotherapy and photodynamic therapy.


Subject(s)
Chitosan/analogs & derivatives , Magnets/chemistry , Neoplasms/radiotherapy , Porphyrins/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Animals , Chitosan/chemistry , Chlorophyllides , Drug Delivery Systems , Female , Ferric Compounds/chemistry , Folic Acid/chemistry , HeLa Cells , Humans , Mice, Nude , Micelles , Neoplasms/pathology , Porphyrins/therapeutic use , Radiation-Sensitizing Agents/therapeutic use
19.
J Med Chem ; 58(9): 3704-19, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25933159

ABSTRACT

A new micelle-forming material, folic acid-conjugated carboxymethyl lauryl chitosan (FA-CLC), and superparamagnetic iron oxide (SPIO) nanoparticles were used for preparing an imaging-guided drug vehicle (the FA-CLC/SPIO hybrid micelle) that demonstrates targeted delivery, imaging, and controlled release of hydrophobic agents. We found that the ratio of viable normal cells to tumor cells was increased prominently after delivery of camptothecin (CPT)-loaded FA-CLC/SPIO micelles and therapeutic sonication. In addition, a magnetic field could enhance the tumor-targeting effect of FA-CLC/SPIO micelles. Therefore, after sequential administration of magnetic attraction to CPT-loaded FA-CLC/SPIO micelles, and therapeutic sonication, the in vivo therapeutic efficacy of CPT was markedly enhanced. However, a nonfocused magnetic field could enhance the undesirable accumulation of iron-containing vehicles in the liver if the tumor (i.e., magnetic attraction site) is near the liver. We propose that magnetic attraction must be carefully applied, far from the liver.


Subject(s)
Antineoplastic Agents/administration & dosage , Chitosan/analogs & derivatives , Chitosan/chemistry , Ferric Compounds/chemistry , Folic Acid/analogs & derivatives , Folic Acid/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Carbocyanines , Cell Line, Tumor , Delayed-Action Preparations , Drug Carriers , Female , Fluorescence , Fluorescent Dyes , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Magnetic Fields , Magnets , Mice, Nude , Micelles , Nanoparticles , Sonication
20.
J Mater Sci Mater Med ; 26(4): 168, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25791456

ABSTRACT

Hyaluronic acid-based hydrogels can reduce postoperative adhesion. However, the long-term application of hyaluronic acid is limited by tissue mediated enzymatic degradation. To overcome this limitation, we developed a polygalacturonic acid and hyaluronate composite hydrogel by Schiff's base crosslinking reaction. The polygalacturonic acid and hyaluronate composite hydrogels had short gelation time (less than 15 s) and degraded by less than 50 % in the presence of hyaluronidase for 7 days. Cell adhesion and migration assays showed polygalacturonic acid and hyaluronate composite hydrogels prevented fibroblasts from adhesion and infiltration into the hydrogels. Compared to hyaluronate hydrogels and commercial Medishield™ gels, polygalacturonic acid and hyaluronate composite hydrogel was not totally degraded in vivo after 4 weeks. In the rat laminectomy model, polygalacturonic acid and hyaluronate composite hydrogel also had better adhesion grade and smaller mean area of fibrous tissue formation over the saline control and hyaluronate hydrogel groups. Polygalacturonic acid and hyaluronate composite hydrogel is a system that can be easy to use due to its in situ cross-linkable property and potentially promising for adhesion prevention in spine surgeries.


Subject(s)
Dura Mater/drug effects , Dura Mater/pathology , Hyaluronic Acid/administration & dosage , Hydrogels/administration & dosage , Pectins/administration & dosage , Tissue Adhesions/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Compressive Strength , Hardness , Male , Rats , Rats, Sprague-Dawley , Tissue Adhesions/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...