Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Chem Rec ; 24(4): e202400010, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38501833

ABSTRACT

Layered double hydroxides (LDH) are a class of functional anionic clays that typically consist of orthorhombic arrays of metal hydroxides with anions sandwiched between the layers. Due to their unique properties, including high chemical stability, good biocompatibility, controlled drug loading, and enhanced drug bioavailability, LDHs have many potential applications in the medical field. Especially in the fields of bioimaging and tumor therapy. This paper reviews the research progress of LDHs and their nanocomposites in the field of tumor imaging and therapy. First, the structure and advantages of LDH are discussed. Then, several commonly used methods for the preparation of LDH are presented, including co-precipitation, hydrothermal and ion exchange methods. Subsequently, recent advances in layered hydroxides and their nanocomposites for cancer imaging and therapy are highlighted. Finally, based on current research, we summaries the prospects and challenges of layered hydroxides and nanocomposites for cancer diagnosis and therapy.


Subject(s)
Nanocomposites , Neoplasms , Humans , Hydroxides/chemistry , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Nanocomposites/therapeutic use , Nanocomposites/chemistry
2.
Small ; 12(21): 2921-31, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27120078

ABSTRACT

In drug delivery systems, pH-sensitive polymers are commonly used as drug carriers, and significant efforts have been devoted to the aspects of controlled delivery and release of drugs. However, few studies address the possible autophagic effects on cells. Here, for the first time, using a fluorescent autophagy-reporting cell line, this study evaluates the autophagy-induced capabilities of four types of pH-sensitive polymeric nanoparticles (NPs) with different physical properties, including size, surface modification, and pH-sensitivity. Based on experimental results, this study concludes that pH-sensitivity is one of the most important factors in autophagy induction. In addition, this study finds that variation of concentration of NPs could cause different autophagic effect, i.e., low concentration of NPs induces autophagy in an mTOR-dependent manner, but high dose of NPs leads to autophagic cell death. Identification of this tunable autophagic effect offers a novel strategy for enhancing therapeutic effect in cancer therapy through modulation of autophagy.


Subject(s)
Autophagy/drug effects , Lysosomes/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Polymers/pharmacology , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Delivery Systems , Humans , Hydrogen-Ion Concentration , MCF-7 Cells
3.
Mol Pharm ; 12(8): 2869-78, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26101892

ABSTRACT

Various nanomaterials have been demonstrated as autophagy inducers owing to their endocytosis cell uptake pathway and impairment of lysosomes. pH-dependent nanomaterials as drug delivery systems that are capable of dissociating in weakly acidic lysosomal environment (pH 4-5) and consequently releasing the payloads into the cytoplasm have been paid extensive attention, but their autophagy-modulating effects are less reported so far. In this study, we report pH-sensitive micelle-like nanoparticles (NPs) that self-assembled from poly(ß-amino ester)s to induce cell autophagy. By encapsulation of gold(I) compounds (Au(I)) into hydrophobic domains of NPs, the resultant Au(I)-loaded NPs (Au(I)⊂NPs) shows synergistic cancer cell killing performance. The Au(I)⊂NPs enter cells through endocytosis pathway and accumulate into acidic lysosomes. Subsequently, the protonation of tertiary amines of poly(ß-amino ester)s triggers the dissociation of micelles, damages the lysosomes, and blocks formation of autolysosomes from fusion of lysosomes with autophagosomes. In addition, Au(I) preferentially inhibits thioredoxin reductase (TrxR) in MCF-7 human breast cancer cells that directly links to up-regulate reactive oxygen species (ROS) and consequently induce autophagy and apoptosis. The blockade of autophagy leads to excessive depletion of cellular organelles and essential proteins and ultimately results in cell death. Therefore, pH-sensitive polymeric nanoparticles with gold(I) compound payloads can synergistically induce cancer cell death through regulation of autophagy. Identification of the pH-sensitive nanomaterials for synergistically inducing cell death through regulation autophagy may open a new avenue for cancer therapy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Drug Delivery Systems , Gold/chemistry , Metal Nanoparticles/chemistry , Polymers/chemistry , Cell Line, Tumor , Drug Carriers/chemistry , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Lysosomes/metabolism , MCF-7 Cells , Micelles , Phagosomes/metabolism , Reactive Oxygen Species/metabolism
4.
Chem Commun (Camb) ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38966911

ABSTRACT

Multifunctional fibers represent a cornerstone of human civilization, playing a pivotal role in numerous aspects of societal development. Natural biomaterials, in contrast to synthetic alternatives, offer environmental sustainability, biocompatibility, and biodegradability. Among these biomaterials, natural silk is favored in biomedical applications and smart fiber technology due to its accessibility, superior mechanical properties, diverse functional groups, controllable structure, and exceptional biocompatibility. This review delves into the intricate structure and properties of natural silk fibers and their extensive applications in biomedicine and smart fiber technology. It highlights the critical significance of silk fibers in the development of multifunctional materials, emphasizing their mechanical strength, biocompatibility, and biodegradability. A detailed analysis of the hierarchical structure of silk fibers elucidates how these structural features contribute to their unique properties. The review also encompasses the biomedical applications of silk fibers, including surgical sutures, tissue engineering, and drug delivery systems, along with recent advancements in smart fiber applications such as sensing, optical technologies, and energy storage. The enhancement of functional properties of silk fibers through chemical or physical modifications is discussed, suggesting broader high-end applications. Additionally, the review addresses current challenges and future directions in the application of silk fibers in biomedicine and smart fiber technologies, underscoring silk's potential in driving contemporary technological innovations. The versatility and sustainability of silk fibers position them as pivotal elements in contemporary materials science and technology, fostering the development of next-generation smart materials.

5.
Adv Mater ; : e2306248, 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37897408

ABSTRACT

Smart nanorobots have emerged as novel drug delivery platforms in nanomedicine, potentially improving anti-cancer efficacy and reducing side effects. In this study, an intelligent tumor microenvironment-responsive nanorobot is developed that effectively delivers CpG payloads to Toll-like receptor 9 (TLR9)-positive tumors to induce autophagy-mediated cell death for immunotherapy. The nanorobots are fabricated by co-self-assembly of two amphiphilic triblock polymer peptides: one containing the matrix metallopeptidase 2 (MMP2)-cleaved GPLGVRGS motif to control the mechanical opening of the nanorobots and provide targeting capability for TLR-9-positive tumors and the other consisting of an arginine-rich GRRRDRGRS sequence that can condense nuclear acid payloads through electrostatic interactions. Using multiple tumor-bearing mouse models, it is investigated whether the intravenous injection of CpG-loaded nanorobots could effectively deliver CpG payloads to TLR-9-positive tumors and elicit anti-tumor immunity through TLR9 signaling and autophagy. Therefore, besides being a commonly used adjuvant for tumor vaccination, CpG-loaded nanorobots can effectively reprogram the tumor immunosuppressive microenvironment and suppress tumor growth and recurrence. This nanorobot-based CpG immunotherapy can be considered a feasible approach to induce anti-tumor immunity, showing great therapeutic potential for the future treatment of TLR9-positive cancers.

6.
J Mater Chem B ; 11(16): 3484-3510, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36988384

ABSTRACT

Messenger RNA (mRNA) has become a key focus in the development of therapeutic agents, showing significant potential in preventing and treating a wide range of diseases. The COVID-19 pandemic in 2020 has accelerated the development of mRNA nucleic therapeutics and attracted significant investment from global biopharmaceutical companies. These therapeutics deliver genetic information into cells without altering the host genome, making them a promising treatment option. However, their clinical applications have been limited by issues such as instability, inefficient in vivo delivery, and low translational efficiency. Recent advances in molecular design and nanotechnology have helped overcome these challenges, and several mRNA formulations have demonstrated promising results in both animal and human testing against infectious diseases and cancer. This review provides an overview of the latest research progress in structural optimization strategies and delivery systems, and discusses key considerations for their future clinical use.


Subject(s)
COVID-19 , Pandemics , Animals , Humans , RNA, Messenger/genetics , RNA, Messenger/therapeutic use , Nanotechnology/methods , Drug Delivery Systems/methods
7.
Biomater Sci ; 10(10): 2550-2556, 2022 May 17.
Article in English | MEDLINE | ID: mdl-35468174

ABSTRACT

Perturbations in mitochondrial membrane stability lead to cytochrome c release and induce caspase-dependent apoptosis. Using synthetic smart chemicals with changeable physicochemical properties to interfere the mitochondrial membrane stability has not yet been reported. Here we show that a thermosensitive anchor-polymer-peptide conjugate (anchor-PPC) destabilizes mitochondrial membranes upon in situ molecule changes from hydrophilic to hydrophobic, which consequently induces apoptosis in a spatiotemporally controlled manner and acts as an antitumor pharmaceutical. The anchor-PPC is composed of a thermosensitive copolymer, a photolabile linker, a hydrophilic HIV Tat-derived peptide both for cell penetration and polymer phase transition temperature (Tt) modulation, and an anchor peptide for intercalating into mitochondrial membranes. The photocontrollable anchor-PPC dehydrates and changes from being hydrophilic to hydrophobic upon photoactivation at body temperature. This cell-penetrable anchor-PPC specifically targets mitochondria and destabilizes mitochondrial membranes upon irradiation, and consequently initiates apoptosis in cells and a complex 3D tumor model. This study provides the first experimental evidence that the synthetic smart chemical can spatiotemporally control the stability of organelle membranes based on its in situ physicochemical property change.


Subject(s)
Mitochondria , Mitochondrial Membranes , Apoptosis , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Peptides/metabolism , Polymers/metabolism
8.
Chem Commun (Camb) ; 58(12): 1982-1985, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35044382

ABSTRACT

A laccase catalyzed colorimetric biosensing approach is promising for the detection of pheochromocytoma biomarkers, yet suffers from the poor stability of enzymes and high cost for production. Here we report for the first time an easy to produce, cheap, stable and reliable laccase-mimicking CuCoFe-LDHzyme, which can catalyze the oxidation of pheochromocytoma biomarkers to form a chromogenic product for smartphone-based colorimetric detection.


Subject(s)
Adrenal Gland Neoplasms/chemistry , Biomarkers, Tumor/analysis , Colorimetry/methods , Hydroxides/chemistry , L-Lactate Dehydrogenase/chemistry , Laccase/chemistry , Molecular Mimicry , Pheochromocytoma/chemistry , Cobalt/chemistry , Copper/chemistry , Humans , Iron/chemistry , Smartphone
9.
Biosens Bioelectron ; 215: 114568, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35850041

ABSTRACT

Accurate and non-invasive monitoring of allograft posttransplant is essential for early detection of acute cellular rejection and determines the long-term survival of the graft. Clinically, tissue biopsy is the most effective approach for diagnosing transplant rejection. Nonetheless, the procedure is invasive and potentially triggers organ failure. This work aims to design and apply GzmB-responsive nanosensors (GBRNs) that can readily size-change in graft tissues. Subsequently, we investigate the activity of serine protease granzyme B by generating a direct colorimetric urinary readout for non-invasive detection of transplant rejection in under 1 h. In preclinical heart graft mice models of transplant rejection, GBRNs were cleaved by GzmB and excreted by the kidneys via accurate nanometre-size glomerular filtration. By exploiting the catalytic activity of ultrasmall gold nanoclusters, GBRNs urinalysis promotes ultrasensitive surveillance of rejection episodes with a receiver operator characteristic curve area under the curve of 0.896 as well as a 95% confidence interval of about 0.7701-1.000. Besides, the catalytic activity of gold nanoclusters in urine can be detected at point-of-care testing to predict the immunity responses in mice with insufficient immunosuppressive therapy. Therefore, this non-invasive, sensitive, and quantitative method is a robust and informative approach for rapid and routine monitoring of transplant allografts without invasive biopsy.


Subject(s)
Biosensing Techniques , Kidney Transplantation , Animals , Biomarkers/urine , Gold , Graft Rejection/diagnosis , Graft Rejection/urine , Kidney Transplantation/adverse effects , Mice , Point-of-Care Systems
10.
Adv Biosyst ; 3(2): e1800232, 2019 02.
Article in English | MEDLINE | ID: mdl-32627370

ABSTRACT

Tumor-associated macrophages (TAMs) are of great interest in cancer immunology as they play an important role in the tumor microenvironment as cancer stromal cells recruited from circulating monocytes. TAMs are closely associated with tumor progression, including initiation, trophic growth, metabolism, angiogenesis, and metastasis; moreover, in clinical practice, their quantity can be related to poor prognosis. Fundamental and translational studies imply that TAMs are one of the most promising targets in tumor therapy. Herein, the biological origination and classification of TAMs, which correspond to their functions and differentiations, are reviewed in detail. In addition, recent basic research and clinical preprocess of TAMs in tumor immunotherapy are also discussed. Finally, the advances in the use of nanotechnology and TAMs for tumor therapy are discussed. This review focuses on the background and status of basic research and clinical significance of TAMs, points out the potential of TAMs in tumor immunological therapy, and clarifies the possibility of translation TAM-targeting therapies in medicine.


Subject(s)
Immunotherapy , Macrophages , Neoplasms , Tumor Microenvironment/immunology , Animals , Humans , Mice , Nanomedicine , Neoplasms/immunology , Neoplasms/therapy
11.
ACS Nano ; 13(7): 7568-7577, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31260255

ABSTRACT

Cellular immunotherapeutics aim to employ immune cells as anticancer agents. Ex vivo engineering of dendritic cells (DCs), the initial role of an immune response, benefits tumor elimination by boosting specific antitumor responses. However, directly activating DCs in vivo is less efficient and therefore quite challenging. Here, we designed a nanoactivator that manufactures DCs through autophagy upregulating in vivo directly, which lead to a high-efficiency antigen presention of DCs and antigen-specific T cells generation. The nanoactivator significantly enhances tumor antigen cross-presentation and subsequent T cell priming. Consequently, in vivo experiments show that the nanoactivators successfully reduce tumor growth and prolong murine survival. Taken together, these results indicate in situ DCs manipulation by autophagy induction is a promising strategy for antigen presentation enhancement and tumor elimination.


Subject(s)
Autophagy/immunology , Dendritic Cells/immunology , Immunotherapy , Melanoma, Experimental/therapy , Nanoparticles/chemistry , Animals , Antigen Presentation/immunology , Cell Line, Tumor , Female , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Particle Size , Surface Properties , T-Lymphocytes/immunology
12.
Nat Commun ; 10(1): 4861, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649241

ABSTRACT

Achieving the activation of drugs within cellular systems may provide targeted therapies. Here we construct a tumour-selective cascade activatable self-detained system (TCASS) and incorporate imaging probes and therapeutics. We show in different mouse models that the TCASS system accumulates in solid tumours. The molecules show enhanced accumulation in tumour regions via the effect of recognition induced self-assembly. Analysis of the molecular penetration in tumour tissue shows that in vivo self-assembly increases the penetration capability compared to typical soft or hard nanomaterials. Importantly, the in vivo self-assembled molecules exhibit a comparable clearance pathway to that of small molecules, which are excreted from organs of the reticuloendothelial system (liver and kidney), while are relatively slowly eliminated from tumour tissues. Finally, this system, combined with the NIR probe, shows high specificity and sensitivity for detecting bladder cancer in isolated intact patient bladders.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Carcinoma, Transitional Cell/diagnostic imaging , Coloring Agents/administration & dosage , Drug Delivery Systems , Protein Engineering/methods , Urinary Bladder Neoplasms/diagnostic imaging , Amino Acid Motifs , Animals , Biological Availability , Carbocyanines/administration & dosage , Cell Line, Tumor , Doxorubicin/administration & dosage , HEK293 Cells , Humans , Kidney/metabolism , Liver/metabolism , Mice , Neoplasm Transplantation , Sensitivity and Specificity , Xenograft Model Antitumor Assays
13.
Biomaterials ; 156: 248-257, 2018 02.
Article in English | MEDLINE | ID: mdl-29216535

ABSTRACT

The blockade of PD-1/PD-L1 interaction by peptide antagonists can unleash and enhance pre-existing anti-cancer immune responses of T cells to eradicate cancer cells. However, low proteolytic stability is the "Achilles' Heel" of peptides. Here, we first report a nanoantagonist with a physiological temperature sensitive nanophase-segregated surface that exhibits significantly enhanced blood circulation, peptide stability and PD-L1 immune checkpoint blockade efficacy. Thermosensitive polymers with different phase transition temperatures (Tt) are used to form the nanophase-segregated surface on an Au nanorod core. Importantly, the nanophase-segregated surface aids the nanoantagonist to resist protein adsorption and enhance the systemic stability of the linked peptides. Finally, the as-designed nanoantagonist effectively blocks PD-1/PD-L1 interaction in vitro and in vivo, enhances the pre-existing CD8+ T cell tumor destruction capability and inhibits tumor growth. This study offers a new strategy for designing nano-formulations for cancer immunotherapy.


Subject(s)
Immunotherapy , Nanoparticles/chemistry , Neoplasms/immunology , Neoplasms/therapy , Adsorption , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Blood Circulation/drug effects , Female , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Peptides/chemistry , Polymers/chemical synthesis , Polymers/chemistry , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Surface Properties , Temperature , Tissue Distribution/drug effects
14.
Nat Commun ; 8(1): 1276, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29097677

ABSTRACT

Topological structures of bio-architectonics and bio-interfaces play major roles in maintaining the normal functions of organs, tissues, extracellular matrix, and cells. In-depth understanding of natural self-assembly mechanisms and mimicking functional structures provide us opportunities to artificially control the natural assemblies and their biofunctions. Here, we report an intracellular enzyme-catalyzed polymerization approach for efficient synthesis of polypeptides and in situ construction of topology-controlled nanostructures. We reveal that the phase behavior and topological structure of polypeptides are encoded in monomeric peptide sequences. Next, we elucidate the relationship between polymerization dynamics and their temperature-dependent topological transition in biological conditions. Importantly, the linearly grown elastin-like polypeptides are biocompatible and aggregate into nanoparticles that exhibit significant molecular accumulation and retention effects. However, 3D gel-like structures with thermo-induced multi-directional traction interfere with cellular fates. These findings allow us to exploit new nanomaterials in living subjects for biomedical applications.


Subject(s)
Biocompatible Materials/chemistry , Elastin/chemistry , Nanoparticles/chemistry , Peptides/chemistry , Polymerization , Temperature , HeLa Cells , Humans , MCF-7 Cells , Nanostructures
15.
ACS Nano ; 11(7): 7301-7311, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28628744

ABSTRACT

Intracellular construction of nanoaggregates from synthetic molecules to mimic natural ordered superstructures has gained increasing attention recently. Here, we develop an endogenous stimuli-induced aggregation (eSIA) approach to construct functional nanoaggregates for sensing and monitoring cellular physiological processes in situ. We design a series of thermosensitive polymer-peptide conjugates (PPCs), which are capable of constructing nanoaggregates in cells based on their isothermal phase transition property. The PPCs are composed of three moieties (i.e., a thermoresponsive polymer backbone, a grafted peptide, and a signal-molecule label). The bioenvironment-associated phase transition behavior of PPCs are carefully studied by consideration of various crucial parameters such as chain length, hydrophilicity, ratio of grafted peptides, and concentration. Intriguingly, under the specific intracellular stimulus, the PPCs are tailored and simultaneously form nanoaggregates exhibiting long-term retention effect, which enables specific identification and quantification of endogenous factors. This general approach is expected for high-performance in situ sensing and dynamic monitoring of disease progression in living subjects.


Subject(s)
Nanostructures/chemistry , Neoplasms/diagnosis , Peptides/chemistry , Phase Transition , Polymers/chemistry , Animals , Antibiotics, Antineoplastic/therapeutic use , Disease Progression , Doxorubicin/therapeutic use , Female , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal/methods , Neoplasms/diagnostic imaging , Neoplasms/pathology , Neoplasms/therapy , Optical Imaging/methods , Prognosis , Protein Aggregates , Temperature
16.
Biomaterials ; 141: 199-209, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28689116

ABSTRACT

Autophagic therapy is regarded as a promising strategy for disease treatment. Appropriate autophagy regulations in vivo play a crucial role in translating this new concept from benchside to bedside. So far, emerging technologies are required to spatially and quantitatively monitor autophagic process in vivo in order to minimize the cytotoxity concerns associated with autophagy-mediated therapy. We successfully demonstrate the "proof-of-concept" study on autophagy-mediated chemotherapy in mice. Here, we describe a photoacoustic (PA) nanoprobe based on "in vivo self-assembly" idea for real-time and quantitative detection of autophagy in mice for the first time. The purpurin-18 (P18) monomer is connected to hydrophilic poly(amidoamine) dendrimer (4th generation) through a peptide (GKGSFGFTG) that can be cleaved by an autophagy-specific enzyme, i.e., ATG4B, consequently resulting in aggregation of P18 and enhanced PA signals. Based on this aggregation-induced "turn-on" PA signals, we noninvasively determine the ATG4B activity for monitoring autophagy of tumor in vivo. According to the results of PA imaging, we could optimize chemotherapy efficacy through precisely modulating autophagy, which thereby decrease systemic toxicity from chemotherapeutics and autophagy inhibitors. We envision it will pave the way for developing autophagy-based treatment of diseases in the future.


Subject(s)
Autophagy/drug effects , Photoacoustic Techniques/methods , Porphyrins/metabolism , Animals , Antineoplastic Agents/pharmacology , Autophagy-Related Proteins/metabolism , Cysteine Endopeptidases/metabolism , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/metabolism , Polyamines/chemistry , Polyamines/metabolism , Porphyrins/chemistry
17.
Biomaterials ; 112: 153-163, 2017 01.
Article in English | MEDLINE | ID: mdl-27768970

ABSTRACT

Immunotherapy has shown promising treatment effects for a variety of cancers. However, the immune treatment efficiency for solid tumors is limited owing to insufficient infiltration of immune cells into solid tumors. The conversion of tumor-supportive macrophages to tumor-suppressive macrophages, inducing the functional reversal of macrophages, is an effective method and contributes to a subsequent antitumor response. The current challenge in the field is the poor distribution and systemic side effects associated with the use of cytokines. As a solution to this issue, we designed and synthesized microenvironment-responsive nanoparticles (P) with IL-12 payload (IL-12⊂P1). These nanoparticles could promote the systemic administration and release of IL-12 in the tumor microenvironment, and the locally responsive property of IL-12⊂P1 could subsequently re-educate tumor-associated macrophages (TAMs). In particular, our results illustrated the great therapeutic effects derived from the functional conversion of macrophages. Our strategy was to design a microenvironment-responsive material for local macrophage modification to overcome the physiological barrier of solid tumors. The shifting of macrophage phenotypes via IL-12⊂P1 achieved immunomodulation in the microenvironment for cancer therapy, with negligible cytotoxicity. We expect that the functional regulation of TAMs by pH-responsive nanomaterials is a promising therapeutic approach for cancer immunotherapy.


Subject(s)
Interleukin-12/administration & dosage , Macrophages/immunology , Nanocapsules/chemistry , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Female , Interleukin-12/chemistry , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , Nanocapsules/administration & dosage , Nanocapsules/ultrastructure , Neoplasms, Experimental/pathology , Polymers/chemistry
18.
ACS Nano ; 11(2): 1826-1839, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28112893

ABSTRACT

Autophagy plays a crucial role in the metabolic process. So far, conventional methods are incapable of rapid, precise, and real-time monitoring of autophagy in living objects. Herein, we describe an in situ intracellular self-assembly strategy for quantitative and temporal determination of autophagy in living objectives. The intelligent building blocks (DPBP) are composed by a bulky dendrimer as a carrier, a bis(pyrene) derivative (BP) as a signal molecule, and a peptide linker as a responsive unit that can be cleaved by an autophagy-specific enzyme, i.e., ATG4B. DPBP maintains the quenched fluorescence with monomeric BP. However, the responsive peptide is specifically tailored upon activation of autophagy, resulting in self-aggregation of BP residues which emit a 30-fold enhanced fluorescence. By measuring the intensity of fluorescent signal, we are able to quantitatively evaluate the autophagic level. In comparison with traditional techniques, such as TEM, Western blot, and GFP-LC3, the reliability and accuracy of this method are finally validated. We believe this in situ intracellular self-assembly strategy provides a rapid, effective, real-time, and quantitative method for monitoring autophagy in living objects, and it will be a useful tool for autophagy-related fundamental and clinical research.


Subject(s)
Autophagy/drug effects , Oligopeptides/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , HeLa Cells , Humans , MCF-7 Cells , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Temperature , Zebrafish
19.
Adv Mater ; 29(34)2017 Sep.
Article in English | MEDLINE | ID: mdl-28714205

ABSTRACT

Posterior capsule opacification (PCO) is the most common complication after cataract surgery. So far, the only method for PCO treatment is the precisely focused laser surgery. However, it causes severe complications such as physical damages and neuron impairments. Here, a nanostructured photothermal ring integrated intraocular lens (Nano-IOLs) is reported, in which the rim of commercially available IOLs (C-IOLs) is decorated with silica coated Au nanorods (Au@SiO2 ), for high-efficient prevention of PCO after cataract surgery. The Nano-IOLs is capable of eliminating the residual lens epithelial cells (LECs) around Nano-IOLs under mild laser treatment and block the formation of disordered LECs fibrosis, which eventually leads to the loss of vision. The Nano-IOLs shows good biocompatibility as well as extraordinary region-confined photothermal effect. In vivo studies reveal that PCO occurrence in rabbit models is about 30%-40% by using Nano-IOLs, which is significantly lower than the control group that treated with C-IOLs (100% PCO occurrence) 30 d postsurgery. To the best of our knowledge, it is the first example to integrate nanotechnology with intraocular implants aiming to clinically relevant PCO. Our findings indicate that spatial controllability of photothermal effect from nanomaterials may provide a unique way to intervene the PCO-induced loss of vision.


Subject(s)
Lenses, Intraocular , Acrylic Resins , Animals , Eye Diseases , Eye, Artificial , Lens Implantation, Intraocular , Rabbits , Silicon Dioxide
20.
ACS Appl Mater Interfaces ; 8(30): 19202-7, 2016 Aug 03.
Article in English | MEDLINE | ID: mdl-27434548

ABSTRACT

Bioinspired design concept has been recognized as one of the most promising strategies for discovering new biomaterials. However, smart biomaterials that are of growing interests in biomedical field need biological processability to meet their emergent applications in vivo. Herein, a new bio-orthogonally deciphered approach has been demonstrated for modulating optical properties of nanomaterials in living systems. The self-assembled nanoemitters based on cyanine-pyrene molecule 1 with inert optical property are designed and prepared. The structure and optical feature of the nanoemitters 1 can be efficiently and reliably modulated by a unique bio-orthogonal mechanism with abundant glutathione (GSH) as an activator. As a result, the self-assembled nanoemitters 1 spontaneously exhibits binary emissions for high-performance tumor imaging in vivo. We believe that this bio-orthogonally deciphered strategy opens a new avenue for designing variable smart biomaterials or devices in biomedical applications.


Subject(s)
Biocompatible Materials , Diagnostic Imaging/methods , Nanostructures , Neoplasms/diagnostic imaging , Diagnostic Imaging/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL