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










Publication year range
1.
Exploration (Beijing) ; 4(2): 20230029, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38855622

ABSTRACT

The glymphatic system plays a key role in the clearance of waste from the parenchyma, and its dysfunction has been associated with the pathogenesis of Alzheimer's disease (AD). However, questions remain regarding its complete mechanisms. Here, we report that efflux of cerebrospinal fluid (CSF)/interstitial fluid (ISF) solutes occurs through a triphasic process that cannot be explained by the current model, but rather hints at the possibility of other, previously undiscovered routes from paravenous spaces to the blood. Using real-time, in vivo observation of efflux, a novel drainage pathway was discovered, in which CSF molecules enter the bloodstream directly through dynamically assembled, trumpet-shaped pores (basolateral ϕ<8 µm; apical ϕ < 2 µm) on the walls of brain venules. As Zn2+ could facilitate the brain clearance of macromolecular ISF solutes, Zn2+-induced reconstruction of the tight junctions (TJs) in vascular endothelial cells may participate in pore formation. Thus, an updated model for glymphatic clearance of brain metabolites and potential regulation is postulated. In addition, deficient clearance of Aß through these asymmetric venule pores was observed in AD model mice, supporting the notion that impaired brain drainage function contributes to Aß accumulation and pathogenic dilation of the perivascular space in AD.

2.
Acta Pharm Sin B ; 12(6): 2731-2739, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35755292

ABSTRACT

Photothermal therapy has the characteristics of minimal invasiveness, controllability, high efficiency, and strong specificity, which can effectively make up for the toxic side effects and tumor resistance caused by traditional drug treatment. However, due to the limited tissue penetration of infrared light, it is difficult to promote and apply in clinical practice. The eye is the only transparent tissue in human, and infrared light can easily penetrate the eye tissue, so it is expected that photothermal therapy can be used to treat fundus diseases. Here in, a new nano-platform assembled by liposome and indocyanine green (ICG) was used to treat retinoblastoma. ICG was assembled in liposomes to overcome some problems of ICG itself. For example, ICG is easily quenched, self-aggregating and instability. Moreover, liposomes can prevent free ICG from being cleared through the systemic circulation. The construction of the nano-platform not only ensured the stability of ICG in vivo, but also realized imaging-guide photothermal therapy, which created a new strategy for the treatment of retinoblastoma.

3.
Nat Commun ; 12(1): 4964, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400628

ABSTRACT

Immunological adjuvants are essential for successful cancer vaccination. However, traditional adjuvants have some limitations, such as lack of controllability and induction of systemic toxicity, which restrict their broad application. Here, we present a light-activable immunological adjuvant (LIA), which is composed of a hypoxia-responsive amphiphilic dendrimer nanoparticle loaded with chlorin e6. Under irradiation with near-infrared light, the LIA not only induces tumour cell lysis and tumour antigen release, but also promotes the structural transformation of 2-nitroimidazole containing dendrimer to 2-aminoimidazole containing dendrimer which can activate dendritic cells via the Toll-like receptor 7-mediated signaling pathway. The LIA efficiently inhibits both primary and abscopal tumour growth and induces strong antigen-specific immune memory effect to prevent tumour metastasis and recurrence in vivo. Furthermore, LIA localizes the immunological adjuvant effect at the tumour site. We demonstrate this light-activable immunological adjuvant offers a safe and potent platform for in situ cancer vaccination.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/immunology , Dendrimers/pharmacology , Vaccination , Animals , Antigens, Neoplasm , Antitussive Agents , Cell Line, Tumor , Chlorophyllides , Dendritic Cells/immunology , Humans , Hypoxia , Immunotherapy , Light , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells , Nanoparticles/chemistry , Neoplasm Metastasis/prevention & control , Neoplasm Recurrence, Local , Neoplasms/genetics , Neoplasms/prevention & control , Porphyrins , Transcriptome
5.
ACS Nano ; 14(11): 14831-14845, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33084319

ABSTRACT

DNA alkylating agents generally kill tumor cells by covalently binding with DNA to form interstrand or intrastrand cross-links. However, in the case of cisplatin, only a few DNA adducts (<1%) are highly toxic irreparable interstrand cross-links. Furthermore, cisplatin is rapidly detoxified by high levels of intracellular thiols such as glutathione (GSH). Since the discovery of its mechanism of action, people have been looking for ways to directly and efficiently remove intracellular GSH and increase interstrand cross-links to improve drug efficacy and overcome resistance, but there has been little breakthrough. Herein, we hypothesized that the anticancer efficiency of cisplatin can be enhanced through iodo-thiol click chemistry mediated GSH depletion and increased formation of DNA interstrand cross-links via mild hyperthermia triggered by near-infrared (NIR) light. This was achieved by preparing an amphiphilic polymer with platinum(IV) (Pt(IV)) prodrugs and pendant iodine atoms (iodides). The polymer was further used to encapsulate IR780 and assembled into Pt-I-IR780 nanoparticles. Induction of mild hyperthermia (43 °C) at the tumor site by NIR light irradiation had three effects: (1) it accelerated the GSH-mediated reduction of Pt(IV) in the polymer main chain to platinum(II) (Pt(II)); (2) it boosted the iodo-thiol substitution click reaction between GSH and iodide, thereby attenuating the GSH-mediated detoxification of cisplatin; (3) it increased the proportion of highly toxic and irreparable Pt-DNA interstrand cross-links. Therefore, we find that mild hyperthermia induced via NIR irradiation can enhance the killing of cancer cells and reduce the tumor burden, thus delivering efficient chemotherapy.


Subject(s)
Antineoplastic Agents , Cisplatin , Cross-Linking Reagents , DNA Adducts , Glutathione , Hyperthermia, Induced , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA/genetics , Humans
6.
Nat Nanotechnol ; 15(12): 1053-1064, 2020 12.
Article in English | MEDLINE | ID: mdl-33106640

ABSTRACT

Cancer vaccines hold great promise for improved cancer treatment. However, endosomal trapping and low immunogenicity of tumour antigens usually limit the efficiency of vaccination strategies. Here, we present a proton-driven nanotransformer-based vaccine, comprising a polymer-peptide conjugate-based nanotransformer and loaded antigenic peptide. The nanotransformer-based vaccine induces a strong immune response without substantial systemic toxicity. In the acidic endosomal environment, the nanotransformer-based vaccine undergoes a dramatic morphological change from nanospheres (about 100 nanometres in diameter) into nanosheets (several micrometres in length or width), which mechanically disrupts the endosomal membrane and directly delivers the antigenic peptide into the cytoplasm. The re-assembled nanosheets also boost tumour immunity via activation of specific inflammation pathways. The nanotransformer-based vaccine effectively inhibits tumour growth in the B16F10-OVA and human papilloma virus-E6/E7 tumour models in mice. Moreover, combining the nanotransformer-based vaccine with anti-PD-L1 antibodies results in over 83 days of survival and in about half of the mice produces complete tumour regression in the B16F10 model. This proton-driven transformable nanovaccine offers a robust and safe strategy for cancer immunotherapy.


Subject(s)
Antigens/administration & dosage , Cancer Vaccines/administration & dosage , Delayed-Action Preparations/chemistry , Nanospheres/chemistry , Neoplasms/prevention & control , Animals , Antigens/therapeutic use , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Immunotherapy , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Polymers/chemistry , Protons
7.
Nanomedicine ; 29: 102241, 2020 10.
Article in English | MEDLINE | ID: mdl-32565227

ABSTRACT

The damaged DNA strands caused by radiotherapy (RT) can repair by themselves. A gold nanoparticles (GNPs) system with acid-induced aggregation was developed into a dual sensitizer owing to its high radioactive rays attenuation ability and enhanced photothermal heating efficiency after GNPs aggregation to achieve a combination therapy of RT and photothermal therapy (PTT). In this combination therapy, the formed GNP aggregates firstly showed a higher sensitize enhancement ratio (SER) value (1.52). Importantly, the self-repair of damaged DNA strands was inhibited by mild PTT through down-regulating the expression of DNA repair protein, thus resulting in a much higher SER value (1.68). Anti-tumor studies further demonstrated that this combination therapy exhibited ideal anti-tumor efficacy. Furthermore, the imaging signals of GNPs in computed tomography and photoacoustic were significantly improved following the GNPs aggregation. Therefore, a dual sensitizer with multimodal imaging was successfully developed and can be further applied as a new anti-tumor therapy.


Subject(s)
Gene Expression Regulation, Neoplastic/radiation effects , Metal Nanoparticles/therapeutic use , Neoplasms/therapy , Photothermal Therapy , Combined Modality Therapy , DNA Damage/radiation effects , DNA Repair/radiation effects , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/pathology , Radiotherapy
8.
Theranostics ; 10(8): 3793-3815, 2020.
Article in English | MEDLINE | ID: mdl-32206123

ABSTRACT

Magnetic hyperthermia (MH) has been introduced clinically as an alternative approach for the focal treatment of tumors. MH utilizes the heat generated by the magnetic nanoparticles (MNPs) when subjected to an alternating magnetic field (AMF). It has become an important topic in the nanomedical field due to their multitudes of advantages towards effective antitumor therapy such as high biosafety, deep tissue penetration, and targeted selective tumor killing. However, in order for MH to progress and to realize its paramount potential as an alternative choice for cancer treatment, tremendous challenges have to be overcome. Thus, the efficiency of MH therapy needs enhancement. In its recent 60-year of history, the field of MH has focused primarily on heating using MNPs for therapeutic applications. Increasing the thermal conversion efficiency of MNPs is the fundamental strategy for improving therapeutic efficacy. Recently, emerging experimental evidence indicates that MNPs-MH produces nano-scale heat effects without macroscopic temperature rise. A deep understanding of the effect of this localized induction heat for the destruction of subcellular/cellular structures further supports the efficacy of MH in improving therapeutic therapy. In this review, the currently available strategies for improving the antitumor therapeutic efficacy of MNPs-MH will be discussed. Firstly, the recent advancements in engineering MNP size, composition, shape, and surface to significantly improve their energy dissipation rates will be explored. Secondly, the latest studies depicting the effect of local induction heat for selectively disrupting cells/intracellular structures will be examined. Thirdly, strategies to enhance the therapeutics by combining MH therapy with chemotherapy, radiotherapy, immunotherapy, photothermal/photodynamic therapy (PDT), and gene therapy will be reviewed. Lastly, the prospect and significant challenges in MH-based antitumor therapy will be discussed. This review is to provide a comprehensive understanding of MH for improving antitumor therapeutic efficacy, which would be of utmost benefit towards guiding the users and for the future development of MNPs-MH towards successful application in medicine.


Subject(s)
Hyperthermia, Induced/methods , Magnetite Nanoparticles , Neoplasms/therapy , Animals , Chemical Phenomena , Combined Modality Therapy/methods , Drug Therapy, Combination , Humans , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Mice
9.
ACS Nano ; 14(2): 1936-1950, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31961656

ABSTRACT

In this study, a magnetothermodynamic (MTD) therapy is introduced as an efficient systemic cancer treatment, by combining the magnetothermal effect and the reactive oxygen species (ROS)-related immunologic effect, in order to overcome the obstacle of limited therapeutic efficacy in current magnetothermal therapy (MTT). This approach was achieved by the development of an elaborate ferrimagnetic vortex-domain iron oxide nanoring and graphene oxide (FVIOs-GO) hybrid nanoparticle as the efficient MTD agent. Such a FVIOs-GO nanoplatform was shown to have high thermal conversion efficiency, and it was further proved to generate a significantly amplified ROS level under an alternating magnetic field (AMF). Both in vitro and in vivo results revealed that amplified ROS generation was the dominant factor in provoking a strong immune response at a physiological tolerable temperature below 40 °C in a hypoxic tumor microenvironment. This was supported by the exposure of calreticulin (CRT) on 83% of the 4T1 breast cancer cell surface, direct promotion of macrophage polarization to pro-inflammatory M1 phenotypes, and further elevation of tumor-infiltrating T lymphocytes. As a result of the dual action of magnetothermal effect and ROS-related immunologic effect, impressive in vivo systemic therapeutic efficacy was attained at a low dosage of 3 mg Fe/kg with two AMF treatments, as compared to that of MTT (high dosage of 6-18 mg/kg under four to eight AMF treatments). The MTD therapy reported here has highlighted the inadequacy of conventional MTT that solely relies on the heating effect of the MNPs. Thus, by employing a ROS-mediated immunologic effect, future cancer magnetotherapies can be designed with greatly improved antitumor capabilities.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/therapy , Ferric Compounds/pharmacology , Graphite/pharmacology , Nanoparticles/chemistry , Reactive Oxygen Species/immunology , Thermodynamics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Graphite/administration & dosage , Graphite/chemistry , Magnetic Fields , Mice , Mice, Inbred BALB C , Particle Size , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Surface Properties , Tumor Microenvironment/drug effects
10.
Adv Sci (Weinh) ; 6(24): 1902137, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31871871

ABSTRACT

Combined chemo/chemodynamic therapy is a promising strategy to achieve an improved anticancer effect. However, the hypoxic microenvironment and limited amount of H2O2 in most solid tumors severely restrict the efficacy of this treatment. Herein, the construction of a nanocatalytic medicine, CaO2@DOX@ZIF-67, via a bottom-up approach is described. CaO2@DOX@ZIF-67 simultaneously supplies O2 and H2O2 to achieve improved chemo/chemodynamic therapy. In the weakly acidic environment within tumors, CaO2@DOX@ZIF-67 is broken down to rapidly release the Fenton-like catalyst Co2+ and the chemotherapy drug doxorubicin (DOX). The unprotected CaO2 reacts with H2O to generate both O2 and H2O2. The generated O2 relieves the hypoxia in the tumor and further improve the efficacy of DOX. Meanwhile, the generated H2O2 reacts with Co2+ ions to produce highly toxic •OH through a Fenton-like reaction, resulting in improved chemodynamic therapy.

11.
Curr Drug Deliv ; 16(9): 782-792, 2019.
Article in English | MEDLINE | ID: mdl-31530265

ABSTRACT

Bone diseases are a serious problem in modern human life. With the coming acceleration of global population ageing, this problem will become more and more serious. Due to the specific physiological characteristics and local microenvironment of bone tissue, it is difficult to deliver drugs to the lesion site. Therefore, the traditional orthopedic medicine scheme has the disadvantages of high drug frequency, large dose and relatively strong side effects. How to target deliver drugs to the bone tissue or even target cells is the focus of the development of new drugs. Nano drug delivery system with a targeting group can realize precise delivery of orthopedic drugs and effectively reduce the systemic toxicity. In addition, the application of bone tissue engineering scaffolds and biomedical materials to realize in situ drug delivery also are research hotspot. In this article, we briefly review the application of nanotechnology in targeted therapies for bone diseases.


Subject(s)
Bone Diseases/drug therapy , Drug Delivery Systems , Animals , Bone Cements , Humans , Nanotechnology , Tissue Scaffolds
12.
Adv Sci (Weinh) ; 6(8): 1801806, 2019 Apr 17.
Article in English | MEDLINE | ID: mdl-31016110

ABSTRACT

An ideal radiosensitizer holding an enhanced tumor retention can play an incredible role in enhancing tumor radiotherapy. Herein, a strategy of acid-triggered aggregation of small-sized gold nanoparticles (GNPs) system within tumor is proposed and the resulting GNPs aggregates are applied as a radiosensitizer in vitro and in vivo. The GNPs system with the acid-triggered aggregation achieves an enhanced GNPs accumulation and retention in cancer cells and tumors in the form of the resulted GNPs aggregates. As a consequence, the radiosensitization effect shows significant improvement in cancer radiotherapy, which is shown in the studies of DNA breakage and the comet assay, and the sensitizer enhancement ratio (SER) value of the GNPs system (1.730) with MCF-7 cancer cells is much larger than that of the single GNPs (1.16). In vivo antitumor studies reveal that the GNPs system also enhances the sensitivity of MCF-7 tumor xenograft to radiotherapy. Furthermore, the GNPs aggregates improve the signal of small GNPs in vivo photoacoustic imaging. This study provides a new strategy and insights into fabricating nanoaggregates to magnify the radiosensitive efficiency of nanosystems in cancer radiotherapy.

13.
Nat Nanotechnol ; 14(4): 379-387, 2019 04.
Article in English | MEDLINE | ID: mdl-30778211

ABSTRACT

Mitochondrial redox homeostasis, the balance between reactive oxygen species and antioxidants such as glutathione, plays critical roles in many biological processes, including biosynthesis and apoptosis, and thus is a potential target for cancer treatment. Here, we report a mitochondrial oxidative stress amplifier, MitoCAT-g, which consists of carbon-dot-supported atomically dispersed gold (CAT-g) with further surface modifications of triphenylphosphine and cinnamaldehyde. We find that the MitoCAT-g particles specifically target mitochondria and deplete mitochondrial glutathione with atomic economy, thus amplifying the reactive oxygen species damage caused by cinnamaldehyde and finally leading to apoptosis in cancer cells. We show that imaging-guided interventional injection of these particles potently inhibits tumour growth in subcutaneous and orthotopic patient-derived xenograft hepatocellular carcinoma models without adverse effects. Our study demonstrates that MitoCAT-g amplifies the oxidative stress in mitochondria and suppresses tumour growth in vivo, representing a promising agent for anticancer applications.


Subject(s)
Carbon/chemistry , Gold/chemistry , Mitochondria/metabolism , Neoplasms/pathology , Neoplasms/therapy , Oxidative Stress , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Death/drug effects , Cell Line, Tumor , Female , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Mice, Inbred BALB C , Mice, Nude , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
15.
ACS Appl Bio Mater ; 2(1): 362-369, 2019 Jan 22.
Article in English | MEDLINE | ID: mdl-35016359

ABSTRACT

RNA interference (RNAi) is an emerging therapeutic modality for tumors. However, lack of a safe and efficient small interfering RNA (siRNA) delivery system limits its clinical application. Here, we report a bioreducible and less-cationic siRNA delivery carrier by conjugating Zn(II)-dipicolylamine complexes (Zn-DPA) onto hyaluronic acid (HA) via a redox-sensitive disulfide (-SS-) linker. Such polymer conjugates can formulate stable siRNA nanomedicines via coordination between zinc ions of DPA and the anionic phosphate of siRNA. After the conjugates are taken up by cells, intracellular reduction stimulus subsequently triggers the release of siRNAs and elucidates the desired RNAi effect. Our studies showed the formulated siRNA nanomedicines can be efficiently delivered into tumor cells/tissues and mediates less cytotoxicities both in vitro and in vivo. More importantly, when applied in a xenograft glioblastoma tumor model, this siRNA nanomedicine demonstrated significantly enhanced antitumor ability comparing to naked siRNA. This work demonstrates that such bioreducible Zn-DPA-functionalized HA conjugates without using cationic material as a siRNA carrier represents a promising direction for RNAi-based cancer therapy.

16.
Adv Mater ; 31(45): e1804922, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30511746

ABSTRACT

The recent emergence of numerous nanotechnologies is expected to facilitate the development of regenerative medicine, which is a tissue regeneration technique based on the replacement/repair of diseased tissue or organs to restore the function of lost, damaged, and aging cells in the human body. In particular, the unique magnetic properties and specific dimensions of magnetic nanomaterials make them promising innovative components capable of significantly advancing the field of tissue regeneration. Their potential applications in tissue regeneration are the focus here, beginning with the fundamentals of magnetic nanomaterials. How nanomaterials-both those that are intrinsically magnetic and those that respond to an externally applied magnetic field-can enhance the efficiency of tissue regeneration is also described. Applications including magnetically controlled cargo delivery and release, real-time visualization and tracking of transplanted cells, magnetic regulation of cell proliferation/differentiation, and magnetic activation of targeted ion channels and signal pathways involved in regeneration are highlighted, and comments on the perspectives and challenges in magnetic nanomaterial-based tissue regeneration are given.

17.
ACS Nano ; 13(1): 274-283, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30566319

ABSTRACT

Multidrug resistance (MDR) is the key cause that accounts for the failure of clinical cancer chemotherapy. To address the problem, herein, we presented an alternative strategy to conquer drug-resistant breast cancer through the combinatorial delivery of Ca2+ channel siRNA with cytotoxic drugs. Mesoporous silica nanocapsules (MSNCs) with mesoporous and hollow structure were fabricated for co-delivery of T-type Ca2+ channel siRNA and doxorubicin (DOX) with high drug loading efficiency. The DOX/siRNA co-loaded MSNCs showed a synergistic therapeutic effect on drug-resistant breast cancer cells MCF-7/ADR, while had only an additive effect on the drug-sensitive MCF-7 counterpart. It was found that the combination of T-type Ca2+ channel siRNA and DOX had a similar effect on MCF-7 and MCF-7/ADR in the knockdown of overexpressed T-type Ca2+ channels and decrease in cytosolic Ca2+ concentration ([Ca2+]i), but it specifically induced G0/G1 phase cell-cycle arrest and intracellular drug accumulation enhancement in MCF-7/ADR. The in vitro and in vivo results demonstrated that the MSNCs with good biocompatibility had a high efficiency for conquering the drug-resistant breast cancer with the DOX/calcium channel siRNA cocktail co-delivery. It provides a biological target for drug/gene delivery enhanced cancer therapy with nanoformulations.


Subject(s)
Antineoplastic Agents/administration & dosage , Calcium Signaling , Doxorubicin/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Nanocapsules/chemistry , RNAi Therapeutics/methods , Animals , Antineoplastic Agents/therapeutic use , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Female , Humans , MCF-7 Cells , Male , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Nanocapsules/adverse effects , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Silicon Dioxide/chemistry
18.
Biomaterials ; 178: 83-94, 2018 09.
Article in English | MEDLINE | ID: mdl-29913389

ABSTRACT

Improving the supply of O2 and the circulation lifetime of photosensitizers for photodynamic therapy (PDT) in vivo would be a promising approach to eliminate hypoxic tumors. Herein, by taking advantage of the significant gas-adsorption capability of metal-organic frameworks (MOFs), a biomimetic O2-evolving photodynamic therapy (PDT) nanoplatform with long circulating properties was fabricated. Zirconium (IV)-based MOF (UiO-66) was used as a vehicle for O2 storing, then conjugated with indocyanine green (ICG) by coordination reaction, and further coated with red blood cell (RBC) membranes. Upon 808 nm laser irradiation, the initial singlet oxygen (1O2) generated by ICG would decompose RBC membranes. At the same time, The photothermal property of ICG could facilitate the burst release of O2 from UiO-66. Subsequently, the generated O2 could significantly improve the PDT effects on hypoxic tumor. Owing to the advantages of long circulation and O2 self-sufficient, the designed nanotherapeutic agent can improve the efficiency of treatment against hypoxia tumor via PDT. Hence, this study presents a new paradigm for co-delivery of O2 and photosensitizers, and provides a new avenue to eliminate hypoxic tumors.


Subject(s)
Biomimetic Materials/chemistry , Metal-Organic Frameworks/chemistry , Nanoparticles/chemistry , Oxygen/chemistry , Photochemotherapy , Tumor Hypoxia , Animals , Cell Survival/drug effects , Erythrocytes/drug effects , Erythrocytes/metabolism , Humans , Indocyanine Green/pharmacology , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , RAW 264.7 Cells , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Superoxides/chemistry , Tissue Distribution , Tumor Hypoxia/drug effects
19.
Sci China Life Sci ; 61(4): 464-475, 2018 04.
Article in English | MEDLINE | ID: mdl-29623549

ABSTRACT

When orthopedic joints coated by hydroxyapatite (HA) were implanted in the human body, they release wear debris into the surrounding tissues. The generation and accumulation of wear particles will induce aseptic loosening. However, the potential bioeffect and mechanism of HA-coated orthopedic implants on bone cells are poorly understood. In this study, defect-related luminescent bur-like hydroxyapatite (BHA) microspheres with the average diameter of 7-9 µm which are comparable to that of the wear-debris particles from aseptically loosened HA implants or HA debris have been synthesized by hydrothermal synthesis and the MC3T3-E1 cells were set as a cells model to study the potential bioeffect and mechanism of BHA microspheres. The studies demonstrated that BHA microspheres could be taken into MC3T3-E1 cells via endocytosis involved in micropinocytosis- and clathrin-mediated endocytosis process, and exert cytotoxicity effect. BHA microspheres could induce the cell apoptosis by intracellular production of reactive oxygen species (ROS), which led to not only an increase in the permeability of lysosome and release of cathepsins B, but also mitochondrial dysfunction and DNA damage. Our results provide novel evidence to elucidate their toxicity mechanisms and might be helpful for more reasonable applications of HA-based orthopaedic implants in the future.


Subject(s)
Apoptosis/drug effects , Durapatite/toxicity , Lysosomes/metabolism , Microspheres , Mitochondria/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cathepsin B/metabolism , Cell Cycle/drug effects , Cell Line , Cell Survival/drug effects , DNA Damage/drug effects , Durapatite/chemical synthesis , Durapatite/metabolism , Endocytosis , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Mice , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...