Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 142(36): 15320-15330, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32820914

ABSTRACT

Current chemodynamic therapy (CDT) primarily relies on the delivery of transition metal ions with Fenton activity to trigger hydroxyl radical production from hydrogen peroxide. However, administration of an excess amount of exogenous Fenton-type heavy metals may cause potential adverse effects to human health, including acute and chronic damages. Here, we present a new CDT strategy that uses intracellular labile iron pool (LIP) as the endogenous source of Fenton-reactive metals for eliciting free radical generation, and the discovery of hydroperoxides (R'OOH) as an optimal LIP-mediated chemodynamic agent against cancer. By simulating the metabolic fates of peroxo compounds within cells, R'OOH was found to have excellent free radical-producing ability in the presence of labile iron(II) and to suffer only moderate elimination by glutathione/glutathione peroxidase, which contributes to its superior chemodynamic efficacy. The LIP-initiated nontoxic-to-toxic transition of R'OOH, together with increased LIP levels in tumor cells, enabled efficient and specific CDT of cancer. Moreover, pH/labile iron(II) cascade-responsive nanomedicines comprising encapsulated methyl linoleate hydroperoxide and LIP-increasing agent in pH-sensitive polymer particles were fabricated to realize enhanced CDT. This work not only paves the way to using endogenous Fenton-type metals for cancer therapy but also offers a paradigm for the exploration of high-performance chemodynamic agents activated by intracellular LIP.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/therapy , Ferrous Compounds/metabolism , Free Radicals/metabolism , Glioblastoma/therapy , Peroxides/pharmacology , Photothermal Therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Ferrous Compounds/chemistry , Free Radicals/chemistry , Glioblastoma/pathology , Humans , Optical Imaging , Peroxides/chemistry , Peroxides/metabolism
2.
Theranostics ; 9(24): 7200-7209, 2019.
Article in English | MEDLINE | ID: mdl-31695762

ABSTRACT

Reactive oxygen species (ROS)-generating anticancer agents can act through two different mechanisms: (i) elevation of endogenous ROS production in mitochondria, or (ii) formation/delivery of exogenous ROS within cells. However, there is a lack of research on the development of ROS-generating nanosystems that combine endogenous and exogenous ROS to enhance oxidative stress-mediated cancer cell death. Methods: A ROS-generating agent based on polymer-modified zinc peroxide nanoparticles (ZnO2 NPs) was presented, which simultaneously delivered exogenous H2O2 and Zn2+ capable of amplifying endogenous ROS production for synergistic cancer therapy. Results: After internalization into tumor cells, ZnO2 NPs underwent decomposition in response to mild acidic pH, resulting in controlled release of H2O2 and Zn2+. Intriguingly, Zn2+ could increase the production of mitochondrial O2·- and H2O2 by inhibiting the electron transport chain, and thus exerted anticancer effect in a synergistic manner with the exogenously released H2O2 to promote cancer cell killing. Furthermore, ZnO2 NPs were doped with manganese via cation exchange, making them an activatable magnetic resonance imaging contrast agent. Conclusion: This study establishes a ZnO2-based theranostic nanoplatform which achieves enhanced oxidative damage to cancer cells by a two-pronged approach of combining endogenous and exogenous ROS.


Subject(s)
Nanoparticles/chemistry , Neoplasms/therapy , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Zinc Oxide/pharmacology , Zinc/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dynamic Light Scattering , Humans , Hydrogen-Ion Concentration , Manganese/chemistry , Mice , Nanoparticles/ultrastructure , Povidone , Spectrum Analysis, Raman
3.
Chem Commun (Camb) ; 55(73): 10884-10887, 2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31436764

ABSTRACT

The development of high-kinetic catalysts for the hydrogen evolution reaction (HER) in a neutral electrolyte is of great importance but unfortunately remains a challenge so far. Herein, we report hybrids with abundant Ru-S-Sb bonds and engineered ultrathin antimonene (Ru-S-Sb/antimonene) as highly kinetic, active, stable electrocatalysts for the HER in an aqueous neutral electrolyte. Experiments and density functional theory (DFT) calculations reveal that Ru-S-Sb bonds coupling with antimonene synergistically work to promote HER activity. The present study brings us one step closer to understand the structure-composition-property relationships and practical electrolytic H2 production.

4.
J Am Chem Soc ; 141(25): 9937-9945, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31199131

ABSTRACT

Chemodynamic therapy (CDT) employs Fenton catalysts to kill cancer cells by converting intracellular H2O2 into hydroxyl radical (•OH), but endogenous H2O2 is insufficient to achieve satisfactory anticancer efficacy. Despite tremendous efforts, engineering CDT agents with specific and efficient H2O2 self-supplying ability remains a great challenge. Here, we report the fabrication of copper peroxide (CP) nanodot, which is the first example of a Fenton-type metal peroxide nanomaterial, and its use as an activatable agent for enhanced CDT by self-supplying H2O2. The CP nanodots were prepared through coordination of H2O2 to Cu2+ with the aid of hydroxide ion, which could be reversed by acid treatment. After endocytosis into tumor cells, acidic environment of endo/lysosomes accelerated the dissociation of CP nanodots, allowing simultaneous release of Fenton catalytic Cu2+ and H2O2 accompanied by a Fenton-type reaction between them. The resulting •OH induced lysosomal membrane permeabilization through lipid peroxidation and thus caused cell death via a lysosome-associated pathway. In addition to pH-dependent •OH generation property, CP nanodots with small particle size showed high tumor accumulation after intravenous administration, which enabled effective tumor growth inhibition with minimal side effects in vivo. Our work not only provides the first paradigm for fabricating Fenton-type metal peroxide nanomaterials, but also presents a new strategy to improve CDT efficacy.


Subject(s)
Antineoplastic Agents/therapeutic use , Copper/chemistry , Hydrogen Peroxide/metabolism , Neoplasms/drug therapy , Quantum Dots/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Hydroxyl Radical/metabolism , Lysosomes/drug effects , Mice , Quantum Dots/chemistry , Xenograft Model Antitumor Assays
5.
Biosens Bioelectron ; 131: 274-279, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30849727

ABSTRACT

Nucleic acid nanoswitches have a status that cannot be ignored in the field of biosensing due to the excellent biocompatibility and flexibility of design. In our current research, we have constructed a new electrochemical platform based on self-assembled pH-sensitive continuous circular DNA nanoswitch for miRNA-21 detection. We elaborately designed an inside ring probe (IRP) which could form a circle when complemented with an outside ring probe (ORP). Under the weakly acidic condition, IRPs and ORPs are self-assembled into continuous annular DNA, meanwhile, the nanoswitch is activated. However, if it is not a weakly acidic environment with a pH equal to 6, these circles are separated and the nanoswitch cannot be triggered. Therefore, the biosensor doesn't work. Only when the pH is 6, can the nanoswitch be activated. Consequently, a large number of RuHex will accumulate on the continuous annular DNA, which leads to highly sensitive detection of miRNA-21, with concentration ranged from 10-15 to 10-8 M and limit of detection down to 0.84 fM. More importantly, this nanoswitch-based biosensor can directly detect the target microRNA in human serum without pretreatment. Therefore, the proposed novel electrochemical DNA nanoswitch will have broad application prospects in biomarker detection and clinical diagnosis.


Subject(s)
Biosensing Techniques , DNA, Circular/chemistry , Electrochemical Techniques , MicroRNAs/isolation & purification , Humans , Hydrogen-Ion Concentration , Limit of Detection , MicroRNAs/chemistry
6.
Adv Sci (Weinh) ; 5(12): 1801201, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30581711

ABSTRACT

The exploitation of smart nanoagents based drug delivery systems (DDSs) has proven to be a promising strategy for fighting cancers. Hitherto, such nanoagents still face challenges associated with their complicated synthesis, insufficient drug release in tumors, and low cancer cell chemosensitivity. Here, the engineering of an adenosine triphosphate (ATP)-activatable nanoagent is demonstrated based on self-assembled quantum dots-phenolic nanoclusters to circumvent such challenges. The smart nanoagent constructed through a one-step assembly not only has high drug loading and low cytotoxicity to normal cells, but also enables ATP-activated disassembly and controlled drug delivery in cancer cells. Particularly, the nanoagent can induce cell ATP depletion and increase cell chemosensitivity for significantly enhanced cancer chemotherapy. Systematic in vitro and in vivo studies further reveal the capabilities of the nanoagent for intracellular ATP imaging, high tumor accumulation, and eventual body clearance. As a result, the presented multifunctional smart nanoagent shows enhanced antitumor efficacy by simultaneous ATP-responsive chemodrug release and cancer cell sensitization. These findings offer new insights toward the design of smart nanoagents for improved cancer therapeutics.

7.
Mikrochim Acta ; 186(1): 7, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30535761

ABSTRACT

A one-step reduction method was used for the preparation of stable graphitic carbon nitride-gold nanoparticles (g-C3N4-Au) nanocomposites from ultrathin g-C3N4 nanosheets and chloroauric acid by using NaBH4 as a reducing agent under ultrasonication. The nanocomposites were characterized by transmission electron microscopy, X-ray photoelectron spectroscopy, UV-Vis absorption and fluorescence spectroscopy etc. The results revealed that the gold nanoparticles (AuNPs) are uniformly formed on the g-C3N4 nanosheets. It is found that the peroxidase-like catalytic activity of this nanocomposite for the oxidation of 3,3',5,5'-tetramethylbenzidine by H2O2 to form a blue-colored product is strongly enhanced in the presence of Hg(II). Based on this phenomenon, a sensitive "turn-on" colorimetric assay for Hg(II) was developed that works at physiological pH values. Under optimal conditions, the absorption signal at 652 nm increases linearly with Hg(II) concentration in the range from 5 to 500 nM. A detection limit as low as 3.0 nM was achieved. This assay has excellent selectivity over other metal ions. It was successfully applied to the determination of Hg(II) in real water samples. The method is cost-effective, rapid, and allows for visual detection. Graphical abstract The nanocomposite composed of graphitic carbon nitride (g-C3N4) and gold nanoparticles (g-C3N4-AuNPs) can catalyze tetramethylbenzidine (TMB) oxidation by H2O2 to produce light-blue product (oxTMB). The peroxidase-like activity of g-C3N4-AuNPs can be greatly enhanced by Hg2+, thus increases the amount of the blue product formed.


Subject(s)
Colorimetry/methods , Gold , Mercury/analysis , Nanocomposites/chemistry , Nanoparticles/chemistry , Nitriles/chemistry , Colorimetry/standards , Graphite/chemistry , Microscopy , Molecular Mimicry , Peroxidase , Spectrum Analysis
8.
J Am Chem Soc ; 140(48): 16589-16595, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30407002

ABSTRACT

The visualization of glycosylation states of specific proteins in vivo is of great importance for uncovering their roles in disease development. However, the ubiquity of glycosylation makes probing the glycans on a certain protein as difficult as looking for a needle in a haystack. Herein, we demonstrate a proximity-induced hybridization chain reaction (HCR) strategy for amplified visualization of protein-specific glycosylation. The strategy relies on designing two kinds of DNA probes, glycan conversion probes and protein recognition probes, which are attached to glycans and target proteins, respectively. Upon sequential binding to the targets, the proximity-induced hybridization between two probes occurs, which leads to the structure-switching of protein recognition probes, followed by triggering of HCR assembly. This strategy has been used to visualize tyrosine-protein kinase 7-specific sialic acid in living CEM cells and zebrafish and to monitor its variation during drug treatment. It provides a potential tool for investigating protein-specific glycosylation and researching the relation between dynamic glycans state and disease process.


Subject(s)
DNA Probes/chemistry , DNA/chemistry , Glycoproteins/analysis , Animals , Azides/metabolism , Carbocyanines/chemistry , Cell Line, Tumor , DNA/genetics , DNA Probes/genetics , Fluorescence , Fluorescent Dyes/chemistry , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycosylation/drug effects , Hexosamines/metabolism , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Nucleic Acid Hybridization , Polysaccharides/chemistry , Tunicamycin/pharmacology , Zebrafish
9.
Anal Chem ; 90(24): 14433-14438, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30444610

ABSTRACT

On cell-membrane surfaces, receptor-protein dimers play fundamental roles in many signaling pathways that are crucial for normal biological processes and cancer development. Efficient and sensitive analysis of receptor dimers in the native environment is highly desirable. Herein, we present a strategy for amplified imaging of receptor dimers in zebrafish and living cells that relies on aptamer recognition and proximity-induced hybridization chain reaction. Taking advantage of specific aptamer recognition and enzyme-free signal amplification, this strategy is successfully applied to the visualization of c-Met-receptor dimers in an HGF-independent or -dependent manner. Therefore, the developed imaging strategy paves the way for further investigation of the dimerization or oligomerization states of cell-surface receptors and their corresponding activation processes in zebrafish and living cells.


Subject(s)
Aptamers, Nucleotide/metabolism , Nucleic Acid Hybridization , Protein Multimerization , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism , Animals , Cell Line, Tumor , Cell Survival , Humans , Molecular Imaging , Protein Structure, Quaternary , Zebrafish
10.
Biosens Bioelectron ; 120: 137-143, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30195087

ABSTRACT

Cholesterol is an essential compound for maintaining cellular homeostasis and human healthy. Sensitive detection of cholesterol and efficient elimination of excess cholesterol have become the essential manipulations in clinical diagnosis and health management. To date, it is still quite challenging that cholesterol detection and elimination tasks are carried out simultaneously. In this study, bifunctional magnetic nanoparticles (Fe3O4@PDA-PBA-CD) are designed and fabricated to overcome this difficulty. Taking advantages of competitive host-guest interaction and magnetic separation, highly efficient, reusable and simultaneous cholesterol detection and elimination can be achieved. The limit of detection is determined to be 4.3 nM, which is comparable or even lower than existing methods. The distinguished performance may attribute to the high loading efficiency and magnetic enrichment of nanoparticles. Besides, this efficient strategy is resistant to interfering substances, thus realizing sensitive cholesterol detection in real sample. Simultaneously, the bifunctional magnetic nanoparticles also have up to 95% cholesterol elimination efficiency, which is higher than previous reported methods. Furthermore, the nanoparticles are turned out to be reusable within 5 times without noticeable loss in cholesterol elimination efficiency. Therefore, the bifunctional magnetic nanoparticles fabricated here could hold great potential for simultaneous cholesterol detection and elimination in practical applications.


Subject(s)
Biosensing Techniques/methods , Cholesterol/blood , Cholesterol/isolation & purification , Magnetite Nanoparticles/chemistry , Biosensing Techniques/instrumentation , Humans , Limit of Detection
11.
ACS Appl Mater Interfaces ; 10(41): 35495-35502, 2018 Oct 17.
Article in English | MEDLINE | ID: mdl-30251823

ABSTRACT

Black phosphorus (BP) nanosheets with unique biocompatibility and superior optical performance have attracted enormous attention in material science. However, their instability and poor solution-processability severely limit their clinical applications. In this work, we demonstrate the use of silk fibroin (SF) as an exfoliating agent to produce thin-layer BP nanosheets with long-term stability and facile solution-processability. Presence of SF prevents rapid oxidation and degradation of the resultant BP nanosheets, enhancing their performance in physiological environment. The SF-modified BP nanosheets exhibit subtle solution-processability and are fabricated into various BP-based material formats. As superior photothermal agents, BP-based wound dressings effectively prevent bacterial infection and promote wound repair. Therefore, this work opens new avenues for unlocking current challenges of BP nanosheet applications for practical biomedical purposes.


Subject(s)
Bacterial Infections/drug therapy , Fibroins , Nanocomposites , Phosphorus , Wound Healing/drug effects , Wound Infection/drug therapy , Animals , Cell Line , Fibroins/chemistry , Fibroins/pharmacology , Humans , Mice , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Phosphorus/chemistry , Phosphorus/pharmacology
12.
Chemistry ; 24(60): 15988-15992, 2018 Oct 26.
Article in English | MEDLINE | ID: mdl-30155946

ABSTRACT

Optical manipulation appears to be a powerful tool for spatiotemporally controlling a variety of cellular functions. Herein, a photocontrolled DNA assembly approach is described which enables light-induced activation of cellular signal transduction by triggering protein dimerization (c-Met signalling in this case). Three kinds of DNA probes are designed, including a pair of receptor recognition probes with adaptors and a blocker probe with a photocleavable linker (PC-linker). By implementing PC-linkers in blocker probes, the designed DNA probes response to light irradiation, which then induces the assembly of receptor recognition probes through adaptor complementing. Consequently, light-mediated DNA assembly promotes the dimerization of c-Met receptors, resulting in activation of c-Met signalling. It is demonstrated that the proposed photocontrolled DNA assembly approach is effective for regulating c-Met signalling and modulating cellular behaviours, such as cell proliferation and migration. Therefore, this simple approach may offer a promising strategy to manipulate cell signalling pathways precisely in living cells.


Subject(s)
DNA Probes/chemistry , Proto-Oncogene Proteins c-met/metabolism , Aptamers, Nucleotide/chemistry , Carbocyanines/chemistry , Cell Line, Tumor , Cell Membrane , Cell Movement , Cell Proliferation , Cell Survival , Fluorescent Dyes/chemistry , Humans , Light , Photochemical Processes , Protein Multimerization , Signal Transduction
13.
ACS Appl Mater Interfaces ; 10(38): 32579-32587, 2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30156821

ABSTRACT

DNA nanostructures with controllable motions and functions have been used as flexible scaffolds to precisely and spatially organize molecular reactions at the nanoscale. The construction of dynamic DNA nanostructures with site-specifically incorporated functional elements is a critical step toward building nanomachines. Artificial self-assembled DNA nanostructures have also been developed to mimic key biological processes like various small biomolecule- and protein-based functional biochemistry pathways. Here, we report a self-assembled dynamic trident-shaped DNA (TS DNA) nanoactuator, in which biomolecules can be tethered to the three "arms" of the TS DNA nanoactuator. The TS DNA nanoactuator is implemented as the mechanical scaffold for the reconfiguration of fluorescent/quenching molecules and the assembly of gold nanoparticles, which exhibit controlled spatial separation. Furthermore, two enzymes (glucose oxidase and horseradish peroxidase) are attached to the two outer arms of the TS DNA nanoactuator, which show an enhanced cascade reaction efficiency compared to free enzymes. The efficiency of the two-enzyme cascade reaction can be spatially regulated by switching the TS DNA nanoactuator between opened, semiopened, and closed states through adding the "thermodynamic drivers" (fuels or antifuels). This is the first report to precisely modulate the relative position of coupled enzyme with multiple states and only based on one dynamic DNA scaffold. The present TS DNA nanoactuator with multistage conformational transition functionality could be applied as a potential platform to precisely and dynamically control the multienzyme pathways and would broaden the scope of DNA nanostructures in single-molecule biology applications.


Subject(s)
DNA/chemistry , Nanostructures/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Molecular Structure
14.
Chem Commun (Camb) ; 54(62): 8579-8582, 2018 Aug 11.
Article in English | MEDLINE | ID: mdl-30019046

ABSTRACT

We report the synthesis of two-dimensional Te nanosheets through a facile liquid exfoliation method. The as-synthesized Te nanosheets can produce reactive oxygen species under light irradiation and show high photoacoustic imaging performance due to their strong near-infrared absorbance, and can be engineered as a nanoplatform for simultaneous photoacoustic imaging and photodynamic therapy.

15.
Talanta ; 188: 66-73, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30029428

ABSTRACT

Osteosarcoma (OS) is one of most malignant bone tumors in early adolescence, which is a highly metastatic cancer and pulmonary metastasis is the most common cause of death. Thus, the development of efficient approaches to discover potential compounds that target metastasis of OS remains a topic of considerable interest. In this study, subtractive Cell-SELEX was performed to screen OS metastasis specific DNA aptamers by using cell lines with similar tumorigenic potentials but opposite metastatic aggressiveness (highly metastatic 143B cells and non-metastatic U-2 OS cells as the target and negative cells, respectively). This in vitro selection generated an ssDNA aptamer LP-16 that exhibited high binding affinity to 143B cells with an equilibrium dissociation constant (Kd) of 56.73 ±â€¯7.750 nM. However, the aptamer LP-16 did not bind to the non-metastatic U-2 OS and normal hFOB 1.19 cells. We further preliminarily presumed the target molecules of aptamer LP-16 was a membrane protein on the cell surface by proteinase treatment. Furthermore, both in vivo fluorescence imaging and clinical tissue imaging also clearly demonstrated that LP-16 could achieve prominently targeting efficiency. Therefore, the ssDNA aptamer LP-16 generated here could be a promising molecular probe for OS metastasis diagnosis. We have developed subtractive Cell-SELEX to screen osteosarcoma metastasis specific DNA aptamers by using cell lines with similar tumorigenic potentials but opposite metastatic aggressiveness (highly metastatic 143B cells and non-metastatic U-2 OS cells as the target and negative cells, respectively).


Subject(s)
Aptamers, Nucleotide/metabolism , DNA, Single-Stranded/metabolism , Molecular Probes/metabolism , Neoplasm Metastasis/diagnostic imaging , Osteosarcoma/diagnostic imaging , SELEX Aptamer Technique/methods , Animals , Carbocyanines/chemistry , Carbocyanines/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Mice, Inbred BALB C , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Probes/chemistry , Osteosarcoma/classification , Osteosarcoma/metabolism
16.
Anal Chem ; 90(11): 6992-6997, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29757612

ABSTRACT

Autofluorescence background in complex biological samples is a major challenge in achieving high sensitivity of fluorescence immunoassays (FIA). Here we report an X-ray luminescence-based immunoassay for high-sensitivity detection of biomarkers using X-ray scintillating nanotags. Due to the weak scattering and absorption of most biological chromophores by X-ray excitation, a low-dose X-ray source can be used to produce intense scintillating luminescence from the nanotags for autofluorescence-free biosensing. To demonstrate this concept, we designed and synthesized NaGdF4:Tb@NaYF4 core/shell nanoparticles as kind of high-efficiency X-ray scintillating nanotags, which are able to convert high-energy X-ray photons to visible light without autofluorescence in biological samples. Notably, strong X-ray absorption and minimized surface quenching arising from the heavy Gd3+/Tb3+ atoms and core/shell architecture of the nanoparticles were found to be critically important for high-efficiency X-ray excited luminescence for high-sensitivity biosensing. Our method allows for sensing alpha-fetoprotein (AFP) biomarkers with a detection limit down to 0.25 ng/mL. Moreover, the as-described X-ray luminescence immunoassay exhibited an excellent biological specificity, high stability, and sample recovery, implying an opportunity for applications in complex biological samples. Consequently, our method can be readily extended for multiplexing sensing and medical diagnosis.


Subject(s)
Immunoassay , Nanoparticles/chemistry , X-Rays , alpha-Fetoproteins/analysis , Animals , Biomarkers/blood , Cells, Cultured , Fluorescence , Humans , Injections, Subcutaneous , Luminescent Measurements , Mice , Mice, Nude , Nanoparticles/administration & dosage , Neoplasms, Experimental/diagnostic imaging , Optical Imaging , Particle Size , Surface Properties
17.
ACS Chem Biol ; 13(7): 1890-1896, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29595947

ABSTRACT

Emergence of antibiotic bacterial resistance has caused serious clinical issues worldwide due to increasingly difficult treatment. Development of a specific approach for selective visualization of resistant bacteria will be highly significant for clinical investigations to promote timely diagnosis and treatment of bacterial infections. In this article, we present an effective method that not only is able to selectively recognize drug resistant AmpC ß-lactamases enzyme but, more importantly, is able to interact with bacterial cell wall components, resulting in a desired localization effect on the bacterial surface. A unique and specific enzyme-responsive cephalosporin probe (DFD-1) has been developed for the selective recognition of resistance bacteria AmpC ß-lactamase, by employing fluorescence resonance energy transfer with an "off-on" bioimaging. To achieve the desired localization, a lipid-azide conjugate (LA-12) was utilized to facilitate its penetration into the bacterial surface, followed by copper-free click chemistry. This enables the probe DFD-1 to be anchored onto the cell surface. In the presence of AmpC enzymes, the cephalosporin ß-lactam ring on DFD-1 will be hydrolyzed, leading to the quencher release, thus generating fluorescence for real-time resistant bacterial screening. More importantly, the bulky dibenzocyclooctyne group in DFD-1 allowed selective recognition toward the AmpC bacterial enzyme instead of its counterpart ( e.g., TEM-1 ß-lactamase). Both live cell imaging and cell cytometry assays showed the great selectivity of DFD-1 to drug resistant bacterial pathogens containing the AmpC enzyme with significant fluorescence enhancement (∼67-fold). This probe presented promising capability to selectively localize and screen for AmpC resistance bacteria, providing great promise for clinical microbiological applications.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/analysis , Cephalosporins/chemistry , Fluorescent Dyes/chemistry , beta-Lactamases/analysis , Bacterial Proteins/metabolism , Cephalosporins/chemical synthesis , Cephalosporins/metabolism , Drug Resistance, Bacterial , Enterobacter cloacae/enzymology , Enterococcus faecium/enzymology , Fluorescence , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemical synthesis , Hydrolysis , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Microscopy, Confocal , Pseudomonas aeruginosa/enzymology , Pseudomonas putida/enzymology , Staphylococcus aureus/enzymology , beta-Lactamases/metabolism
18.
Angew Chem Int Ed Engl ; 57(18): 4902-4906, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29488312

ABSTRACT

Chemodynamic therapy (CDT) utilizes iron-initiated Fenton chemistry to destroy tumor cells by converting endogenous H2 O2 into the highly toxic hydroxyl radical (. OH). There is a paucity of Fenton-like metal-based CDT agents. Intracellular glutathione (GSH) with . OH scavenging ability greatly reduces CDT efficacy. A self-reinforcing CDT nanoagent based on MnO2 is reported that has both Fenton-like Mn2+ delivery and GSH depletion properties. In the presence of HCO3- , which is abundant in the physiological medium, Mn2+ exerts Fenton-like activity to generate . OH from H2 O2 . Upon uptake of MnO2 -coated mesoporous silica nanoparticles (MS@MnO2 NPs) by cancer cells, the MnO2 shell undergoes a redox reaction with GSH to form glutathione disulfide and Mn2+ , resulting in GSH depletion-enhanced CDT. This, together with the GSH-activated MRI contrast effect and dissociation of MnO2 , allows MS@MnO2 NPs to achieve MRI-monitored chemo-chemodynamic combination therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Glutathione/chemistry , Manganese Compounds/pharmacology , Nanoparticles/chemistry , Oxides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Ions/chemistry , Manganese Compounds/administration & dosage , Manganese Compounds/chemistry , Mice , Nanoparticles/administration & dosage , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Oxides/administration & dosage , Oxides/chemistry , Particle Size , Structure-Activity Relationship , Surface Properties
19.
Chem Commun (Camb) ; 54(25): 3142-3145, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29527603

ABSTRACT

Gene therapy with small interfering RNA (siRNA) has been proved to be a promising technology to treat various diseases by hampering the production of target proteins. However, developing a delivery system that has high efficiency in transporting siRNA without obvious side effects remains a challenge. Herein, we designed a new survivin siRNA delivery system based on polyethyleneimine functionalized black phosphorus (BP) nanosheets which could suppress tumor growth by silencing survivin expression. Combined with the photothermal properties of the BP nanosheets, the presented delivery system shows excellent therapy efficiency for tumors. Therefore, the BP-based delivery system would be a promising tool for future clinical applications.


Subject(s)
Drug Delivery Systems , Gene Silencing/drug effects , Genetic Therapy , Nanostructures/chemistry , Phosphorus/chemistry , Phototherapy , Polyethyleneimine/chemistry , RNA, Small Interfering/pharmacology , Animals , Cell Proliferation/drug effects , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , RNA, Small Interfering/administration & dosage
20.
J Am Chem Soc ; 140(12): 4186-4190, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29522674

ABSTRACT

Herein, we report a nongenetic and real-time approach for imaging protein dimerization on living cell surfaces by aptamer recognition and proximity-induced DNA assembly. We use the aptamer specific for the receptor monomer as a recognition probe. When receptor dimerization occurs, the dimeric receptors bring two aptamer probes into close proximity, thereby triggering dynamic DNA assembly. The proposed approach was successfully applied to visualize dimerization of Met receptor and transforming growth factor-ß type II receptor. This approach allows us to image the two states (monomer/dimer) of a receptor protein on living cell surfaces in real time, opening a universal method for further investigation of protein dimerization and the corresponding activation processes in signal transduction.


Subject(s)
Aptamers, Nucleotide/metabolism , DNA/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Aptamers, Nucleotide/chemistry , Cell Line, Tumor , DNA/chemistry , Dimerization , Humans , Microscopy, Confocal , Proto-Oncogene Proteins c-met/chemistry , Receptors, Transforming Growth Factor beta/chemistry , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL