Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Phys Chem B ; 128(11): 2773-2781, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38461422

ABSTRACT

1H spin-lattice relaxation experiments have been performed for water and glycerol/water solutions of H2N-Fe3O4 superparamagnetic nanoparticles (NPs) of about 7 nm diameter. The experiments encompass a broad frequency range covering 3 orders of magnitude, from 10 kHz to 10 MHz (referring to 1H resonance frequency), and have been performed in the temperature range from 298 to 313 K, varying the concentration of the superparamagnetic species. This extensive dataset has been used for twofold purposes. The first one is to serve as a challenge for thorough tests of theoretical models describing nuclear relaxation in solutions of superparamagnetic NPs, depending on their magnetic properties and dynamics of the solvent molecules. The challenge is posed by the wish to reproduce the data in a broad range of magnetic fields (not only at high fields) and by the need to explain the differences in the relaxation scenarios for water and glycerol/water solutions by varying only the solvent parameters. The second purpose is to get insights into the magnetic properties (electronic relaxation properties) of the nanoparticles due to their high applicational potential.

2.
Ecotoxicol Environ Saf ; 243: 113967, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-35985197

ABSTRACT

Nanotechnology allows for a greater quality of life, but may also cause environmental and organismic harm. Zinc oxide nanoparticles (ZnONPs) are one of the most commonly used metal oxide nanoparticles for commercial and industrial products. Due to its extensive use in various fields, there has already been much concern raised about the environmental health risks of ZnONPs. Many studies have investigated the toxicological profile of ZnONPs in zebrafish embryonic development; however, the specific characteristics of ZnONPs in zebrafish embryonic/larval developmental damage and their molecular toxic mechanisms of liver development are yet to be fully elucidated. This study aimed to reveal the hazard ranking of different surface modifications of ZnONPs on developing zebrafish and the toxicological mechanisms of these modified ZnONPs in liver tissue. The ~30 nm ZnONPs with amino- (NH2- ZnONPs) or carboxyl- (COOH-ZnONPs) modification were incorporated during the embryonic/larval stage of zebrafish. Severe toxicity was observed in both ZnONP groups, especially NH2-ZnONPs, which presented a higher toxicity in the low concentration groups. After prolonging the exposure time, the long-term toxicity assay showed a greater retardation in body length of zebrafish in the NH2-ZnONP group. Response data from multiple toxicity studies was integrated for the calculation of the EC50 values of bulk ZnO and ZnONPs, and the hazard levels were found to be decreasing in the order of NH2-, COOH-ZnONPs and bulk ZnO. Notably, NH2-ZnONPs induced ROS burden in the developing liver tissue, which activated autophagy-related gene and protein expression and finally induced liver cell apoptosis to reduce liver size. In conclusion, our findings are conducive to understanding the hazard risks of different surface modifications of ZnONPs in aquatic environments and will also be helpful for choosing the type of ZnONPs in future industrial applications.


Subject(s)
Metal Nanoparticles , Nanoparticles , Zinc Oxide , Animals , Larva , Metal Nanoparticles/toxicity , Nanoparticles/toxicity , Quality of Life , Zebrafish , Zinc Oxide/toxicity
3.
Int J Mol Med ; 50(2)2022 08.
Article in English | MEDLINE | ID: mdl-35703361

ABSTRACT

Cancer cells can acquire resistance to targeted therapeutic agents when the designated targets or their downstream signaling molecules develop protein conformational or activity changes. There is an increasing interest in developing poly­pharmacologic anticancer agents to target multiple oncoproteins or signaling pathways in cancer cells. The microRNA 125a­5p (miR­125a­5p) is a tumor suppressor, and its expression has frequently been downregulated in tumors. By contrast, the anti­apoptotic molecule BIRC5/SURVIVIN is highly expressed in tumors but not in the differentiated normal tissues. In the present study, the development of a BIRC5 gene promoter­driven, miR­125a­5p expressing, poly­L­lysine­conjugated magnetite iron poly­pharmacologic nanodrug (pL­MNP­pSur­125a) was reported. The cancer cells self­activating property and the anticancer effects of this nanodrug were examined in both the multidrug efflux protein ABCB1/MDR1­expressing/­non­expressing cancer cells in vitro and in vivo. It was demonstrated that pL­MNP­pSur­125a decreased the expression of ERBB2/HER2, HDAC5, BIRC5, and SP1, which are hot therapeutic targets for cancer in vitro. Notably, pL­MNP­pSur­125a also downregulated the expression of TDO2 in the human KB cervical carcinoma cells. PL­MNP­pSur­125a decreased the viability of various BIRC5­expressing cancer cells, regardless of the tissue origin or the expression of ABCB1, but not of the human BIRC5­non­expressing HMEC­1 endothelial cells. In vivo, pL­MNP­pSur­125a exhibited potent antitumor growth effects, but without inducing liver toxicity, in various zebrafish human­ABCB1­expressing and ABCB1­non­expressing tumor xenograft models. In conclusion, pL­MNP­pSur­125a is an easy­to­prepare and a promising poly­pharmacological anticancer nanodrug that has the potential to manage numerous malignancies, particularly for patients with BIRC5/ABCB1­related drug resistance after prolonged chemotherapeutic treatments.


Subject(s)
Antineoplastic Agents , MicroRNAs , Nanoparticles , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Endothelial Cells , Humans , MicroRNAs/genetics , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Zebrafish/genetics
4.
Nanomaterials (Basel) ; 12(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35215043

ABSTRACT

The global application of engineered nanomaterials and nanoparticles (ENPs) in commercial products, industry, and medical fields has raised some concerns about their safety. These nanoparticles may gain access into rivers and marine environments through industrial or household wastewater discharge and thereby affect the ecosystem. In this study, we investigated the effects of silver nanoparticles (AgNPs) and zinc oxide nanoparticles (ZnONPs) on zebrafish embryos in aquatic environments. We aimed to characterize the AgNP and ZnONP aggregates in natural waters, such as lakes, reservoirs, and rivers, and to determine whether they are toxic to developing zebrafish embryos. Different toxic effects and mechanisms were investigated by measuring the survival rate, hatching rate, body length, reactive oxidative stress (ROS) level, apoptosis, and autophagy. Spiking AgNPs or ZnONPs into natural water samples led to significant acute toxicity to zebrafish embryos, whereas the level of acute toxicity was relatively low when compared to Milli-Q (MQ) water, indicating the interaction and transformation of AgNPs or ZnONPs with complex components in a water environment that led to reduced toxicity. ZnONPs, but not AgNPs, triggered a significant delay of embryo hatching. Zebrafish embryos exposed to filtered natural water spiked with AgNPs or ZnONPs exhibited increased ROS levels, apoptosis, and lysosomal activity, an indicator of autophagy. Since autophagy is considered as an early indicator of ENP interactions with cells and has been recognized as an important mechanism of ENP-induced toxicity, developing a transgenic zebrafish system to detect ENP-induced autophagy may be an ideal strategy for predicting possible ecotoxicity that can be applied in the future for the risk assessment of ENPs.

5.
Part Fibre Toxicol ; 19(1): 6, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35031062

ABSTRACT

BACKGROUND: Silver nanoparticles (AgNPs) are considered a double-edged sword that demonstrates beneficial and harmful effects depending on their dimensions and surface coating types. However, mechanistic understanding of the size- and coating-dependent effects of AgNPs in vitro and in vivo remains elusive. We adopted an in silico decision tree-based knowledge-discovery-in-databases process to prioritize the factors affecting the toxic potential of AgNPs, which included exposure dose, cell type and AgNP type (i.e., size and surface coating), and exposure time. This approach also contributed to effective knowledge integration between cell-based phenomenological observations and in vitro/in vivo mechanistic explorations. RESULTS: The consolidated cell viability assessment results were used to create a tree model for generalizing cytotoxic behavior of the four AgNP types: SCS, LCS, SAS, and LAS. The model ranked the toxicity-related parameters in the following order of importance: exposure dose > cell type > particle size > exposure time ≥ surface coating. Mechanistically, larger AgNPs appeared to provoke greater levels of autophagy in vitro, which occurred during the earlier phase of both subcytotoxic and cytotoxic exposures. Furthermore, apoptosis rather than necrosis majorly accounted for compromised cell survival over the above dosage range. Intriguingly, exposure to non-cytotoxic doses of AgNPs induced G2/M cell cycle arrest and senescence instead. At the organismal level, SCS following a single intraperitoneal injection was found more toxic to BALB/c mice as compared to SAS. Both particles could be deposited in various target organs (e.g., spleen, liver, and kidneys). Morphological observation, along with serum biochemical and histological analyses, indicated that AgNPs could produce pancreatic toxicity, apart from leading to hepatic inflammation. CONCLUSIONS: Our integrated in vitro, in silico, and in vivo study revealed that AgNPs exerted toxicity in dose-, cell/organ type- and particle type-dependent manners. More importantly, a single injection of lethal-dose AgNPs (i.e., SCS and SAS) could incur severe damage to pancreas and raise blood glucose levels at the early phase of exposure.


Subject(s)
Metal Nanoparticles , Silver , Animals , Cell Survival , Knowledge Discovery , Metal Nanoparticles/toxicity , Mice , Particle Size , Silver/toxicity
6.
Int J Nanomedicine ; 16: 2137-2146, 2021.
Article in English | MEDLINE | ID: mdl-33731995

ABSTRACT

PURPOSE: Vitamin D3 is useful for the treatment of peritoneal dialysis (PD)-related peritoneal damage, but its side effects, such as hypercalcemia and vascular calcification, limit its applicability. Thus, we developed vitamin D-loaded magnetic nanoparticles (MNPs) and determined their therapeutic efficacy and side effects in vivo. MATERIALS AND METHODS: Alginate-modified MNPs were combined with 1α, 25 (OH)2D3 to generate vitamin D-loaded nanoparticles. The particles were conjugated with an antibody against peritoneum-glycoprotein M6A (GPM6A). The particles' ability to target the peritoneum was examined following intraperitoneal administration to mice and by monitoring their bio-distribution. We also established a PD animal model to determine the therapeutic and side effects of vitamin D-loaded MNPs in vivo. RESULTS: Vitamin D-loaded MNPs targeted the peritoneum better than vitamin D3, and had the same therapeutic effect as vitamin D3 in ameliorating peritoneal fibrosis and functional deterioration in a PD animal model. Most importantly, the particles reduced the side effects of vitamin D3, such as hypercalcemia and body weight loss, in mice. CONCLUSION: Vitamin D-loaded MNPs could be an ideal future therapeutic option to treat PD-related peritoneal damage.


Subject(s)
Cholecalciferol/administration & dosage , Drug Delivery Systems , Magnetite Nanoparticles/chemistry , Peritoneal Dialysis/adverse effects , Peritoneum/pathology , Alginates/chemistry , Animals , Antibodies/metabolism , Disease Models, Animal , Drug Liberation , Humans , Magnetite Nanoparticles/ultrastructure , Male , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/pathology
7.
Cancers (Basel) ; 13(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546453

ABSTRACT

Cancer response to chemotherapy is regulated not only by intrinsic sensitivity of cancer cells but also by tumor microenvironment. Tumor hypoxia, a condition of low oxygen level in solid tumors, is known to increase the resistance of cancer cells to chemotherapy. Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. Due to lack of target in TNBC, chemotherapy is the only approved systemic treatment. We evaluated the effect of hypoxia on chemotherapy resistance in TNBC in a series of in vitro and in vivo experiments. Furthermore, we synthesized the calcium peroxide-modified magnetic nanoparticles (CaO2-MNPs) with the function of oxygen generation to improve and enhance the therapeutic efficiency of doxorubicin treatment in the hypoxia microenvironment of TNBC. The results of gene set enrichment analysis (GSEA) software showed that the hypoxia and autophagy gene sets are significantly enriched in TNBC patients. We found that the chemical hypoxia stabilized the expression of hypoxia-inducible factor 1α (HIF-1α) protein and increased doxorubicin resistance in TNBC cells. Moreover, hypoxia inhibited the induction of apoptosis and autophagy by doxorubicin. In addition, CaO2-MNPs promoted ubiquitination and protein degradation of HIF-1α. Furthermore, CaO2-MNPs inhibited autophagy and induced apoptosis in TNBC cells. Our animal studies with an orthotopic mouse model showed that CaO2-MNPs in combination with doxorubicin exhibited a stronger tumor-suppressive effect on TNBC, compared to the doxorubicin treatment alone. Our findings suggest that combined with CaO2-MNPs and doxorubicin attenuates HIF-1α expression to improve the efficiency of chemotherapy in TNBC.

8.
Int J Mol Sci ; 21(8)2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32325940

ABSTRACT

As the worldwide application of nanomaterials in commercial products increases every year, various nanoparticles from industry might present possible risks to aquatic systems and human health. Presently, there are many unknowns about the toxic effects of nanomaterials, especially because the unique physicochemical properties of nanomaterials affect functional and toxic reactions. In our research, we sought to identify the targets and mechanisms for the deleterious effects of two different sizes (~10 and ~50 nm) of amine-modified silver nanoparticles (AgNPs) in a zebrafish embryo model. Fluorescently labeled AgNPs were taken up into embryos via the chorion. The larger-sized AgNPs (LAS) were distributed throughout developing zebrafish tissues to a greater extent than small-sized AgNPs (SAS), which led to an enlarged chorion pore size. Time-course survivorship revealed dose- and particle size-responsive effects, and consequently triggered abnormal phenotypes. LAS exposure led to lysosomal activity changes and higher number of apoptotic cells distributed among the developmental organs of the zebrafish embryo. Overall, AgNPs of ~50 nm in diameter exhibited different behavior from the ~10-nm-diameter AgNPs. The specific toxic effects caused by these differences in nanoscale particle size may result from the different mechanisms, which remain to be further investigated in a follow-up study.


Subject(s)
Amines , Chorion/drug effects , Embryo, Nonmammalian/drug effects , Metal Nanoparticles , Silver , Amines/chemistry , Animals , Apoptosis , Chemical Phenomena , Embryonic Development , Lysosomes/metabolism , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/adverse effects , Metal Nanoparticles/chemistry , Particle Size , Silver/chemistry , Toxicity Tests, Acute , Zebrafish
9.
Polymers (Basel) ; 12(12)2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33419339

ABSTRACT

Noninvasive treatments to treat the brain-related disorders have been paying more significant attention and it is an emerging topic. However, overcoming the blood brain barrier (BBB) is a key obstacle to most of the therapeutic drugs to enter into the brain tissue, which significantly results in lower accumulation of therapeutic drugs in the brain. Thus, administering the large quantity/doses of drugs raises more concerns of adverse side effects. Nanoparticle (NP)-mediated drug delivery systems are seen as potential means of enhancing drug transport across the BBB and to targeted brain tissue. These systems offer more accumulation of therapeutic drugs at the tumor site and prolong circulation time in the blood. In this review, we summarize the current knowledge and advancements on various nanoplatforms (NF) and discusses the use of nanoparticles for successful cross of BBB to treat the brain-related disorders such as brain tumors, Alzheimer's disease, Parkinson's disease, and stroke.

10.
Int J Nanomedicine ; 14: 9665-9675, 2019.
Article in English | MEDLINE | ID: mdl-31824158

ABSTRACT

PURPOSE: Vitamin D is a novel potential therapeutic agent for peritoneal dialysis (PD)-related peritoneal fibrosis, but it can induce hypercalcemia and vascular calcification, which limits its applicability. In this study, we create nanotechnology-based drug delivery systems to investigate its therapeutics and side effects. MATERIALS AND METHODS: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [amino-(polyethylene glycol)2000] (DSPE-PEG) and L-α-phosphatidylcholine (PC), which packages with 1α,25(OH)2D3, were used to construct vitamin D nanoliposomes. To confirm the function and safety of vitamin D nanoliposomes, peritoneal mesothelial cells were treated with TGF-ß1 and the reverse was attempted using vitamin D nanoliposomes. Antibodies (Ab) against the peritoneum-glycoprotein M6A (GPM6A) Ab were conjugated with vitamin D nanoliposomes. These particles were implanted into mice by intraperitoneal injection and the animals were monitored for the distribution and side effects induced by vitamin D. RESULTS: Vitamin D nanoliposomes were taken up by the mesothelial cells over time without cell toxicity and it also provided the same therapeutic effect in vitro. In vivo study, fluorescent imaging showed vitamin D nanoliposomes allow specific peritoneum target effect and also ameliorate vitamin D side effect. CONCLUSION: Nanoliposomes vitamin D delivery systems for the prevention of PD-related peritoneal damage may be a potential clinical strategy in the future.


Subject(s)
Nanomedicine , Peritoneal Dialysis/adverse effects , Peritoneum/pathology , Vitamin D/pharmacology , Animals , Antibodies/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Liberation , Epithelial-Mesenchymal Transition/drug effects , Humans , Kinetics , Liposomes , Mice , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Peritoneum/drug effects , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Transforming Growth Factor beta1/metabolism
11.
Aging (Albany NY) ; 11(21): 9875-9892, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31714893

ABSTRACT

Pathophysiological states cause misfolded protein accumulation in the endoplasmic reticulum (ER). Then, ER stress and the unfolded protein response (UPR) are activated. Targeting ER stress may enhance the adaptive UPR and then protect the cell against pathogenic environments. In the present study, we utilized nanotechnology to synthesize thapsigargin nanoparticles (TG NPs) which induced ER stress and the UPR pathway, to study the role of ER stress and autophagy in chronic kidney disease (CKD). We found that the mRNA levels of ER stress- and autophagy-related molecules were elevated in the renal tissue of CKD patients compared to those of healthy individuals. Furthermore, TG NPs induced the UPR pathway and autophagy in HK-2 human kidney tubular epithelial cells. TG NPs protected HK-2 cells against oxidative stress-induced cell death through the activation of Nrf2 and FoxO1. The siRNA-mediated inhibition of Nrf2 or FoxO1 resulted in enhanced oxidative stress-induced cytotoxicity in HK-2 cells. In a mouse model of adenine diet-induced CKD, TG NPs and KIM-1-TG NPs ameliorated renal injury through the stimulation of ER stress and its downstream pathways. Our findings suggest that the induction of ER stress using pharmacological agents may offer a promising therapeutic strategy for preventing or interfering with CKD progression.


Subject(s)
Forkhead Box Protein O1/metabolism , NF-E2-Related Factor 2/metabolism , Nanoparticles/chemistry , Renal Insufficiency, Chronic/drug therapy , Thapsigargin/administration & dosage , Adenine , Animals , Autophagy/drug effects , Autophagy/genetics , Cell Line , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Nanoparticles/therapeutic use , Nanoparticles/ultrastructure , Renal Insufficiency, Chronic/metabolism
12.
Mater Sci Eng C Mater Biol Appl ; 97: 116-123, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30678896

ABSTRACT

Metal oxide nanoparticles are a new class of important materials used in a wide variety of biomedical applications. Bulk zinc oxide (ZnO) particles have been used for temporal or permanent luting cement because of their excellent mechanical strength and biocompatibility. ZnO nanoparticles have distinct optical and antibacterial properties and a high surface-to-volume ratio. We investigated the mechanical and antibacterial properties of luting cement with different ratios of ZnO nanospheres. We showed that luting cement with 5% and 10% ZnO nanospheres was less soluble in low-pH (pH 3) artificial saliva. Antibacterial activity was 40% higher for Streptococcus mutans and 90% higher for Porphyromonas gingivalis when >10% (w/v) of the bulk particles were replaced with ZnO nanospheres in ZnO polycarboxylate cement. ZnO nanospheres were also biocompatible with mammalian cells. Additionally, the compressive strength was 1.2 times greater and the diametral tensile strength was 1.5 times greater for cements with 10% ZnO nanospheres than for conventional ZnO polycarboxylate cement. We propose a new method for improving dental luting cement by integrating it with ZnO nanospheres. This method simultaneously adds their greater antibacterial, mechanical, and acid resistance properties and retains an outstanding degree of biocompatibility.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dental Cements/chemistry , Dental Cements/pharmacology , Nanospheres/chemistry , Zinc Oxide/chemistry , Animals , Anti-Bacterial Agents/chemistry , Dental Implants , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Materials Testing , Mice , Polycarboxylate Cement/chemistry , Polycarboxylate Cement/pharmacology , Porphyromonas gingivalis/drug effects , Saliva/chemistry , Streptococcus mutans/drug effects , Tensile Strength , Zinc Oxide/administration & dosage , Zinc Oxide/pharmacology
13.
Adv Healthc Mater ; 8(5): e1801358, 2019 03.
Article in English | MEDLINE | ID: mdl-30672150

ABSTRACT

There is currently no cure for gene mutation-caused autosomal dominant polycystic kidney disease (ADPKD). Over half of patients with ADPKD eventually develop kidney failure, requiring dialysis or kidney transplantation. Current treatment modalities for ADPKD focus on reducing morbidity and mortality from renal and extrarenal complications of the disease. MicroRNA has been shown to be useful in treating ADPKD. This study combines anti-miRNA plasmids and iron oxide/alginate nanoparticles for conjugation with antikidney antibodies. These nanocomposites can specifically target renal tubular cells, providing a potential treatment for ADPKD. Magnetic resonance imaging and in vivo imaging system results show effective targeting of renal cells. Anti-miRNA plasmids released from the nanocomposites inhibit cell proliferation and cyst formation in the PKD cellular and animal models. The results suggest the novel combination of the anti-miRNA plasmids and nanomaterials provides potential clinical implications for ADPKD treatment.


Subject(s)
MicroRNAs/administration & dosage , Nanocomposites/administration & dosage , Nanocomposites/chemistry , Plasmids/administration & dosage , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/therapy , Animals , Cell Line , Kidney/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Plasmids/genetics
14.
Nanomedicine (Lond) ; 12(22): 2741-2756, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28884615

ABSTRACT

AIM: We used resveratrol (Res)-loaded nanoparticles (Res NPs) as a novel method for improving the pharmacokinetic properties of Res and analyzed the effect of Res NPs in chronic kidney disease (CKD). MATERIALS & METHODS: We coupled anti-kidney injury molecule-1 antibodies to Res NPs and analyzed safety and efficacy. RESULTS: Res NPs had low toxicity and induced autophagy. Res NPs inhibited the NLRP3 inflammasome and IL-1ß secretion. Higher NLRP3 expression levels were observed in peripheral blood monocytic cells of CKD patients than healthy individuals. Treatment with kidney injury molecule-1-Res NPs significantly reduced creatinine and protected against tubulointerstitial injury in a murine model of CKD. CONCLUSION: Res NPs through NLRP3 inflammasome attenuation and autophagy induction may be as a strategy to prevent CKD.


Subject(s)
Hepatitis A Virus Cellular Receptor 1/chemistry , Nanoparticles/chemistry , Renal Insufficiency, Chronic/drug therapy , Stilbenes/administration & dosage , Stilbenes/chemistry , Animals , Antibodies/chemistry , Autophagy/drug effects , Cell Line , Cell Survival , Creatinine/metabolism , Drug Carriers/chemistry , Drug Liberation , Epithelial Cells/cytology , Hepatitis A Virus Cellular Receptor 1/immunology , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Kidney Tubules/cytology , Lactic Acid/chemistry , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephritis, Interstitial/drug therapy , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Resveratrol , Stilbenes/pharmacokinetics
15.
J Nanobiotechnology ; 14(1): 75, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27884158

ABSTRACT

BACKGROUND: Fingermarks are one of the most important and useful forms of physical evidence in forensic investigations. However, latent fingermarks are not directly visible, but can be visualized due to the presence of other residues (such as inorganic salts, proteins, polypeptides, enzymes and human metabolites) which can be detected or recognized through various strategies. Convenient and rapid techniques are still needed to provide obvious contrast between the background and the fingermark ridges and to then visualize latent fingermark with a high degree of selectivity and sensitivity. RESULTS: In this work, lysozyme-binding aptamer-conjugated Au nanoparticles (NPs) are used to recognize and target lysozyme in the fingermark ridges, and Au+-complex solution is used as a growth agent to reduce Au+ from Au+ to Au0 on the surface of the Au NPs. Distinct fingermark patterns were visualized on a range of professional forensic within 3 min; the resulting images could be observed by the naked eye without background interference. The entire processes from fingermark collection to visualization only entails two steps and can be completed in less than 10 min. The proposed method provides cost and time savings over current fingermark visualization methods. CONCLUSIONS: We report a simple, inexpensive, and fast method for the rapid visualization of latent fingermarks on the non-porous substrates using Au seed-mediated enhancement. Au seed-mediated enhancement is used to achieve the rapid visualization of latent fingermarks on non-porous substrates by the naked eye without the use of expensive or sophisticated instruments. The proposed approach offers faster detection and visualization of latent fingermarks than existing methods. The proposed method is expected to increase detection efficiency for latent fingermarks and reduce time requirements and costs for forensic investigations.


Subject(s)
Aptamers, Nucleotide/chemistry , Dermatoglyphics , Forensic Sciences/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Humans , Muramidase
16.
Part Fibre Toxicol ; 13(1): 52, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27678081

ABSTRACT

BACKGROUND: Zinc oxide nanoparticles (ZnO NPs) are used in an increasing number of products, including rubber manufacture, cosmetics, pigments, food additives, medicine, chemical fibers and electronics. However, the molecular mechanisms underlying ZnO NP nephrotoxicity remain unclear. In this study, we evaluated the potential toxicity of ZnO NPs in kidney cells in vitro and in vivo. RESULTS: We found that ZnO NPs were apparently engulfed by the HEK-293 human embryonic kidney cells and then induced reactive oxygen species (ROS) generation. Furthermore, exposure to ZnO NPs led to a reduction in cell viability and induction of apoptosis and autophagy. Interestingly, the ROS-induced hypoxia-inducible factor-1α (HIF-1α) signaling pathway was significantly increased following ZnO NPs exposure. Additionally, connective tissue growth factor (CTGF) and plasminogen activator inhibitor-1 (PAI-1), which are directly regulated by HIF-1 and are involved in the pathogenesis of kidney diseases, displayed significantly increased levels following ZnO NPs exposure in HEK-293 cells. HIF-1α knockdown resulted in significantly decreased levels of autophagy and increased cytotoxicity. Therefore, our results suggest that HIF-1α may have a protective role in adaptation to the toxicity of ZnO NPs in kidney cells. In an animal study, fluorescent ZnO NPs were clearly observed in the liver, lungs, kidneys, spleen and heart. ZnO NPs caused histopathological lesions in the kidney and increase in serum creatinine and blood urea nitrogen (BUN) which indicate possible renal possible damage. Moreover, ZnO NPs enhanced the HIF-1α signaling pathway, apoptosis and autophagy in mouse kidney tissues. CONCLUSIONS: ZnO NPs may cause nephrotoxicity, and the results demonstrate the importance of considering the toxicological hazards of ZnO NP production and application, especially for medicinal use.

17.
Nanoscale ; 8(15): 7866-70, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27035391

ABSTRACT

The blood-brain barrier (BBB) is a physiological structure of the blood vessels in the brain. The BBB efficiently traps most therapeutic drugs in the blood vessels and stops them from entering the brain tissue, resulting in a decreased therapeutic efficiency. In this study, we developed BBB-stealth nanocomposites composed of iron oxide (Fe3O4) nanoparticles (NPs) as a safe nanocarrier for glioblastoma therapy. We showed the antitumor activity of Dox/alg-Fe3O4 NPs using in vitro and in vivo tests. We demonstrated that G23-alg-Fe3O4 NPs crossed the BBB and entered the brain. In situ glioblastoma tumor-bearing mice were used to successfully evaluate the antitumor activity of G23-Dox/alg-Fe3O4 NPs. Magnetic resonance imaging (MRI) and bioluminescence imaging (BLI) confirmed the BBB crossing. The BBB-stealth nanocomposites show great potential for a proof-of-concept clinical trial as a theranostics platform for human brain tumor therapy.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Magnetite Nanoparticles/therapeutic use , Nanocomposites/therapeutic use , Alginates/chemistry , Animals , Antineoplastic Agents/administration & dosage , Brain Neoplasms/diagnostic imaging , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Glioblastoma/diagnostic imaging , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Magnetic Resonance Imaging , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/chemistry , Mice , Nanocomposites/administration & dosage , Nanocomposites/chemistry , Neuroimaging , Theranostic Nanomedicine
18.
ACS Appl Mater Interfaces ; 6(10): 7198-203, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24766528

ABSTRACT

An efficient method for modifying the surface of detonation nanodiamonds (5 and 100 nm) with thiol groups (-SH) by using an organic chemistry strategy is presented herein. Thiolated nanodiamonds were characterized by spectroscopic techniques, and the atomic percentage of sulfur was analyzed by elemental analysis and X-ray photoelectron spectroscopy. The conjugation between thiolated nanodiamonds and gold nanoparticles was elucidated by transmission electron microscopy and UV-vis spectrometry. Moreover, the material did not show significant cytotoxicity to the human lung carcinoma cell line and may prospectively be applied in bioconjugated technology. The new method that we elucidated may significantly improve the approach to surface modification of detonation nanodiamonds and build up a new platform for the application of nanodiamonds.


Subject(s)
Nanodiamonds/chemistry , Sulfhydryl Compounds/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Microscopy, Electron, Transmission , Nanodiamonds/toxicity , Particle Size , Photoelectron Spectroscopy
19.
Curr Med Chem ; 21(29): 3312-22, 2014.
Article in English | MEDLINE | ID: mdl-24606505

ABSTRACT

The increasing morbidity and mortality of infectious diseases is an increasing concern. Despite the continuous development and synthesis of new antimicrobial drugs, microbial pathogens are exhibiting increased multi-drug resistance. Nanomaterials display unique and well-defined physical and chemical properties including a very high surface area to volume ratio, and new approaches for antimicrobial therapies have attempted to combine nanomaterials and current antimicrobial drugs. Magnetic nanoparticles (MNPs) are characterized by biocompatibility, biodegradation, and safety for human ingestion. Iron oxide nanoparticles have been approved for human use by the US Food and Drug Administration (FDA). For biomedicine applications, MNPs require surface modification to become water-soluble and be stable enough to resist the effects of proteins and salts in the physiological environment. MNPs can combine various substrata, such as biomolecules and nanomaterials to generate new antimicrobial agents which combine antibacterial, antiviral, and antifungal properties. This can be accomplished through a series of surface modification methods. Because MNPs have unique superparamagnetic characteristics, they can be controlled and recycled by an external magnetic field.In addition, the antimicrobial activity of MNPs-based nanocomposites is superior to that of metallic nanoparticles. This paper reviews the recent literature on the use of MNP-based nanomaterials in antimicrobial applications in biomedicine. Antimicrobial applications mainly focus on inhibiting and killing bacteria and fungi and viruses inactivation. The synthesis, surface modification, and characteristics related to MNPs will also be briefly addressed.


Subject(s)
Anti-Infective Agents/pharmacology , Biomedical Research , Magnetite Nanoparticles/chemistry , Water/chemistry , Anti-Infective Agents/chemistry , Bacteria/drug effects , Fungi/drug effects , Humans , Microbial Sensitivity Tests , Surface Properties , Viruses/drug effects
20.
Biomaterials ; 35(16): 4706-15, 2014 May.
Article in English | MEDLINE | ID: mdl-24630838

ABSTRACT

With the advancement of nanotechnology, nanomaterials have been comprehensively applied in our modern society. However, the hazardous impacts of nanoscale particles on organisms have not yet been thoroughly clarified. Currently, there exist numerous approaches to perform toxicity tests, but common and reasonable bio-indicators for toxicity evaluations are lacking. In this study, we investigated the effects of silver nanoparticles (AgNPs) on NIH 3T3 cells to explore the potential application of these nanoparticles in consumer products. Our results demonstrated that AgNPs were taken up by NIH 3T3 cells and localized within the intracellular endosomal compartments. Exposure to AgNPs is a potential source of oxidative stress, which leads to the induction of reactive oxygen species (ROS), the up-regulation of Heme oxygenase 1 (HO-1) expression, apoptosis and autophagy. Interestingly, AgNPs induced morphological and biochemical markers of autophagy in NIH 3T3 cells and induced autophagosome formation, as evidenced by transmission electron microscopic analysis, the formation of microtubule-associated protein-1 light chain-3 (LC3) puncta and the expression of LC3-II protein. Thus, autophagy activation may be a key player in the cellular response against nano-toxicity.


Subject(s)
Apoptosis/drug effects , Fibroblasts/drug effects , Metal Nanoparticles/toxicity , Oxidative Stress/drug effects , Silver/toxicity , Animals , Autophagy/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Heme Oxygenase-1/genetics , Mice , NIH 3T3 Cells , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...