Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Angew Chem Int Ed Engl ; 60(43): 23299-23305, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34240523

ABSTRACT

Development of proteolysis targeting chimeras (PROTACs) is emerging as a promising strategy for targeted protein degradation. However, the drug development using the heterobifunctional PROTAC molecules is generally limited by poor membrane permeability, low in vivo efficacy and indiscriminate distribution. Herein an aptamer-PROTAC conjugation approach was developed as a novel strategy to improve the tumor-specific targeting ability and in vivo antitumor potency of conventional PROTACs. As proof of concept, the first aptamer-PROTAC conjugate (APC) was designed by conjugating a BET-targeting PROTAC to the nucleic acid aptamer AS1411 (AS) via a cleavable linker. Compared with the unmodified BET PROTAC, the designed molecule (APR) showed improved tumor targeting ability in a MCF-7 xenograft model, leading to enhanced in vivo BET degradation and antitumor potency and decreased toxicity. Thus, the APC strategy may pave the way for the design of tumor-specific targeting PROTACs and have broad applications in the development of PROTAC-based drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Breast Neoplasms/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Proteolysis/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/toxicity , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Disulfides/chemical synthesis , Disulfides/therapeutic use , Disulfides/toxicity , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/therapeutic use , Heterocyclic Compounds, 3-Ring/toxicity , Humans , Mice , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/toxicity , Proof of Concept Study , Pyrrolidines/chemical synthesis , Pyrrolidines/therapeutic use , Pyrrolidines/toxicity , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
2.
Carbohydr Polym ; 266: 118111, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34044928

ABSTRACT

Herein, a novel targeted delivery system was developed for intracellular co-delivery of doxorubicin (DOX) as a chemotherapeutic drug, antimiR-21 as an oncogenic antagomiR. In this system, DOX was loaded into mesoporous silica nanoparticles (MSNs) and chitosan was applied to cover the surface of MSNs. AS1411 aptamer as targeting nucleolin and antimiR-21 were electrostatically attached onto the surface of the chitosan-coated MSNs and formed the final nanocomplex (AACS nanocomplex). The study of drug release was based on DOX release under pH 7.4 and 5.5. Cellular toxicity and cellular uptake assessments of AACS nanocomplex were carried out in nucleolin positive (C26, MCF-7, and 4T1) and nucleolin negative (CHO) cell lines using MTT assay and flow cytometry analysis, respectively. Also, Anti-tumor efficacy of AACS nanocomplex was evaluated in C26 tumor-bearing mice. Overall, the results show that the combination therapy of DOX and antimiR-21, using AACS nanocomplex, could combat the cancer cell growth rate.


Subject(s)
Antagomirs/therapeutic use , Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Antagomirs/chemistry , Antagomirs/toxicity , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , CHO Cells , Cell Line, Tumor , Chitosan/chemistry , Chitosan/toxicity , Cricetulus , Doxorubicin/chemistry , Drug Carriers/toxicity , Drug Liberation , Humans , Immobilized Nucleic Acids/chemistry , Immobilized Nucleic Acids/toxicity , Mice , MicroRNAs/antagonists & inhibitors , Nanoparticles/toxicity , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/toxicity , Silicon Dioxide/chemistry , Silicon Dioxide/toxicity
3.
ChemistryOpen ; 10(4): 408-413, 2021 04.
Article in English | MEDLINE | ID: mdl-33605540

ABSTRACT

A novel method for the preparation of antitumor drug vehicles has been optimized. Biological materials of chitosan oligosaccharide (CO) and γ-polyglutamic acid (γ-PGA) have previously been employed as modifiers to covalently modify graphene oxide (GO), which in turn loaded doxorubicin (DOX) to obtain a nano drug delivery systems of graphene oxide based composites (GO-CO-γ-PGA-DOX). The system was not equipped with the ability of initiative targeting, thus resulting into toxicity and side effects on normal tissues or organs. In order to further improve the targeting property of the system, the nucleic acid aptamer NH2 -AS1411 (APT) of targeted nucleolin (C23) was used to conjugate on GO-CO-γ-PGA to yield the targeted nano drug delivery system APT-GO-CO-γ-PGA. The structure, composition, dispersion, particle size and morphology properties of the synthesized complex have been studied using multiple characterization methods. Drug loading and release profile data showed that APT-GO-CO-γ-PGA is provided with high drug loading capacity and is capable of controlled and sustained release of DOX. Cell experimental results indicated that since C23 was overexpressed on the surface of Hela cells but not on the surface of Beas-2B cells, APT-GO-CO-γ-PGA-DOX can target Hela cells and make increase toxicity to Hela cells than Beas-2B cells, and the IC50 value of APT-GO-CO-γ-PGA-DOX was 3.23±0.04 µg/mL. All results proved that APT-GO-CO-γ-PGA can deliver antitumor drugs in a targeted manner, and achieve the effect of reducing poison, which indicated that the targeted carrier exhibits a broad application prospect in the field of biomedicine.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Graphite/chemistry , Nanocomposites/chemistry , Oligodeoxyribonucleotides/chemistry , Aptamers, Nucleotide/toxicity , Chitin/analogs & derivatives , Chitin/chemistry , Chitin/toxicity , Chitosan , Drug Carriers/toxicity , Drug Liberation , Graphite/toxicity , HeLa Cells , Humans , Immobilized Nucleic Acids/chemistry , Immobilized Nucleic Acids/toxicity , Nanocomposites/toxicity , Oligodeoxyribonucleotides/toxicity , Oligosaccharides , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/chemistry , Polyglutamic Acid/toxicity
4.
ACS Appl Mater Interfaces ; 12(51): 57362-57372, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33301289

ABSTRACT

The rapid development of CRISPR/Cas9 systems has opened up tantalizing prospects to sensitize cancers to chemotherapy using efficient targeted genome editing, but safety concerns and possible off-target effects of viral vectors remain a major obstacle for clinical application. Thus, the construction of novel nonviral tumor-targeting nanodelivery systems has great potential for the safe application of CRISPR/Cas9 systems for gene-chemo-combination therapy. Here, we report a polyamidoamine-aptamer-coated hollow mesoporous silica nanoparticle for the co-delivery of sorafenib and CRISPR/Cas9. The core-shell nanoparticles had good stability, enabled ultrahigh drug loading, targeted delivery, and controlled-release of the gene-drug combination. The nanocomplex showed >60% EGFR-editing efficiency without off-target effects in all nine similar sites, regulating the EGFR-PI3K-Akt pathway to inhibit angiogenesis, and exhibited a synergistic effect on cell proliferation. Importantly, the co-delivery nanosystem achieved efficient EGFR gene therapy and caused 85% tumor inhibition in a mouse model. Furthermore, the nanocomplex showed high accumulation at the tumor site in vivo and exhibited good safety with no damage to major organs. Due to these properties, the nanocomplex provides a versatile delivery approach for efficient co-loading of gene-drug combinations, allowing for precise gene editing and synergistic inhibition of tumor growth without apparent side effects on normal tissues.


Subject(s)
CRISPR-Cas Systems , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Sorafenib/therapeutic use , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , CRISPR-Associated Protein 9/genetics , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/toxicity , Drug Liberation , Epithelial Cell Adhesion Molecule/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Editing , Genes, erbB-1 , Humans , Mice , Nanoparticles/toxicity , Polyamines/chemistry , Polyamines/toxicity , Porosity , Signal Transduction/drug effects , Silicon Dioxide/toxicity
5.
Angew Chem Int Ed Engl ; 59(31): 12698-12702, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32297692

ABSTRACT

We report the use of DNA origami nanostructures, functionalized with aptamers, as a vehicle for delivering the antibacterial enzyme lysozyme in a specific and efficient manner. We test the system against Gram-positive (Bacillus subtilis) and Gram-negative (Escherichia coli) targets. We use direct stochastic optical reconstruction microscopy (dSTORM) and atomic force microscopy (AFM) to characterize the DNA origami nanostructures and structured illumination microscopy (SIM) to assess the binding of the origami to the bacteria. We show that treatment with lysozyme-functionalized origami slows bacterial growth more effectively than treatment with free lysozyme. Our study introduces DNA origami as a tool in the fight against antibiotic resistance, and our results demonstrate the specificity and efficiency of the nanostructure as a drug delivery vehicle.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA/chemistry , Drug Carriers/chemistry , Muramidase/pharmacology , Nanostructures/chemistry , Animals , Anti-Bacterial Agents/chemistry , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , Bacillus subtilis/chemistry , Bacillus subtilis/drug effects , COS Cells , Chlorocebus aethiops , DNA/toxicity , Drug Carriers/toxicity , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/pharmacology , Escherichia coli/chemistry , Escherichia coli/drug effects , Microbial Sensitivity Tests , Muramidase/chemistry , Nanostructures/toxicity , Nucleic Acid Conformation
6.
Anal Chem ; 91(22): 14681-14690, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31617347

ABSTRACT

Rapid, accurate, and safe screening of foodborne pathogenic bacteria is essential to effectively control and prevent outbreaks of foodborne illness. Fluorescent sensors constructed from carbon dots (CDs) and nanomaterial-based quenchers have provided an innovative method for screening of pathogenic bacteria. Herein, an ultrasensitive magnetic fluorescence aptasensor was designed for separation and detection of Staphylococcus aureus (S. aureus). Multicolor fluorescent CDs with a long fluorescent lifetime (6.73 ns) and high fluorescence stability were synthesized using a facile hydrothermal approach and modified cDNA as a highly sensitive fluorescent probe. CD fluorescence was quenched by Fe3O4 + aptamer via fluorescence resonance energy transfer (FRET). Under optimal conditions, the FRET-based aptasensor can detect S. aureus accompanied by a wide linear range of 50-107 CFU·mL-1 and a detection limit of 8 CFU·mL-1. Compared with other standard methods, this method was faster and more convenient, and the entire test was finished within 30 min. The capability of the aptasensor was simultaneously investigated on food samples. Additionally, the developed CDs exhibited excellent biocompatibility and were thus applied as fluorescent probes for bioimaging both in vitro and in vivo. This new platform provided an excellent application of the CDs for detecting and bioimaging pathogenic bacteria.


Subject(s)
Biosensing Techniques/methods , Fluorescent Dyes/chemistry , Quantum Dots/chemistry , Staphylococcus aureus/isolation & purification , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , Carbon/chemistry , Carbon/toxicity , DNA/chemistry , DNA/toxicity , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/toxicity , Food Contamination/analysis , Fruit and Vegetable Juices/microbiology , Hep G2 Cells , Humans , Limit of Detection , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Mice, Nude , Milk/microbiology , Neoplasms/diagnostic imaging , Optical Imaging/methods , Phenylenediamines/chemistry , Phenylenediamines/toxicity , Quantum Dots/toxicity , Staphylococcus aureus/chemistry
7.
Arch Biochem Biophys ; 671: 143-151, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31283911

ABSTRACT

Aptamer based drug delivery systems are gaining the importance in anticancer therapy due to their targeted drug delivery efficiency without harming the normal cells. The present work formulated the pH-dependent aptamer functionalized polymer-based drug delivery system against human lung cancer. The prepared aptamer functionalized doxorubicin (DOX) loaded poly (D, L-lactic-co-glycolic acid) (PLGA), poly (N-vinylpyrrolidone) (PVP) nanoparticles (APT-DOX-PLGA-PVP NPs) were spherical in shape with an average size of 87.168 nm. The crystallography and presence of the PLGA (poly (D, L-lactic-co-glycolic acid)) and DOX (doxorubicin) in APT-DOX-PLGA-PVP NPs were indicated by the X-ray diffraction (XRD), Fourier transforms infrared spectroscopy (FTIR), and 1H and 13C nuclear magnetic resonance spectrometer (NMR). The pH-dependent aptamer AS1411 based drug release triggered the cancer cell death was evidenced by cytotoxicity assay, flow cytometry, and fluorescent microscopic imaging. In addition, the cellular uptake of the DOX was determined and the apoptosis-related signaling pathway in the A549 cells was studied by Western blot analysis. Further, the in vivo study revealed that mice treated with APT-DOX-PLGA-PVP NPs were significantly recovered from cancer as evident by mice weight and tumor size followed by the histopathological study. It was reported that the APT-DOX-PLGA-PVP NPs induced the apoptosis through the activation of the apoptosis-related proteins. Hence, the present study revealed that the APT-DOX-PLGA-PVP NPs improved the therapeutic efficiency through the nucleolin receptor endocytosis targeted drug release.


Subject(s)
Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/chemistry , Doxorubicin/therapeutic use , Nanoparticles/chemistry , Neoplasms/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , A549 Cells , Animals , Antineoplastic Agents/pharmacokinetics , Aptamers, Nucleotide/toxicity , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/toxicity , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Nanoparticles/toxicity , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer/toxicity , Povidone/chemistry , Povidone/toxicity , Xenograft Model Antitumor Assays
8.
Analyst ; 144(12): 3843-3852, 2019 Jun 21.
Article in English | MEDLINE | ID: mdl-31098604

ABSTRACT

Acute leukemia is a malignant clonal disease of hematopoietic stem cells with a high prevalence and mortality rate. However, there are no efficient tools to facilitate early diagnosis and treatment of leukemia. Therefore, development of new methods for the early diagnosis and prevention of leukemia, especially non-invasive diagnosis at the cellular level, is imperative. Here, a label-free signal-on fluorescence aptasensor based on terbium(iii)-aptamer (Tb3+-apt) was applied for the detection of leukemia. The aptamer sensitizes the fluorescence of Tb3+ and forms the strong fluorescent Tb3+-apt probe. The target cells, the T-cell acute lymphoblastic leukemia cell line (CCRF-CEM) combined with the Tb3+-apt probe to form the Tb3+-apt-CEM complex, were removed by centrifugation, and the supernatant containing a small amount of the Tb3+-apt probe was detected using a fluorescence spectrophotometer. The logarithm of cell concentration showed a good linear relationship (R2 = 0.9881) with the fluorescence signal. The linear range for CCRF-CEM detection was 5-5 × 106 cells per ml, while the detection limit was 5 cells per ml of the binding buffer. Clinical samples were collected from 100 cases, and the specificity and positive rates detected by this method were up to 94% and 90%, respectively. Therefore, a single-stranded DNA-sensitized terbium(iii) luminescence method diagnostic was developed which is rapid, sensitive, and economical and can be used for diagnosis of various types of leukemia at the early stage.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Terbium/chemistry , Adolescent , Adult , Aptamers, Nucleotide/toxicity , Base Sequence , Cell Line, Tumor , Child , Child, Preschool , Female , Fluorescence , Humans , Limit of Detection , Male , Middle Aged , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/blood , Spectrometry, Fluorescence/methods , Terbium/toxicity , Young Adult
9.
Anal Chem ; 91(3): 2425-2430, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30620179

ABSTRACT

Exosomes (Exos) are nanoscale natural vehicles for transporting biomolecules to facilitate cell-to-cell communication, indicating a high potential of them for delivering therapeutics/diagnostics. To improve their delivery capacity, a simple, noninvasive, and efficient strategy for functionalizing Exos with effective targeting ligands as well as elucidation of the cellular uptake mechanism of these functionalized Exos was found be to necessary, but remained a challenge. In this work, we used diacyllipid-aptamer conjugates as the targeting ligand to develop an aptamer-functionalized Exos (Apt-Exos) nanoplatform for cell type-specific delivery of molecular therapeutics. The cellular uptake mechanism of Apt-Exos was investigated in details, and distinct behavior was observed in comparison to free Exos. By combining the excellent molecular recognition capability of aptamers and the superiority of Exos as natural vehicles, Apt-Exos can efficiently deliver molecular drugs/fluorophores to target cancer cells, providing a promising delivery platform for cancer theranostics.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Exosomes/chemistry , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/toxicity , Cell Line, Tumor , Drug Carriers/metabolism , Drug Carriers/toxicity , Drug Liberation , Electroporation , Endocytosis/physiology , Exosomes/metabolism , Humans , Kinetics , Phospholipids/chemistry , Phospholipids/metabolism , Phospholipids/toxicity , Proof of Concept Study
10.
ACS Sens ; 3(12): 2702-2708, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30460840

ABSTRACT

DNA fluorescent probes are versatile tools that are widely used for biological detection in tubes. Using DNA probes in living systems, however, represents a significant challenge because of the endogenous nuclease-induced DNA degradation and strong background fluorescence in complex biological environments. Here, we show that assembling DNA probes into core-satellite gold nanoparticle (AuNP) superstructures could unprecedentedly enhance enzymatic stability and reduce background interference. The embedded DNA probes are protected from interaction with nuclease, eliminating the enzymatic degradation. In the meantime, the AuNP superstructures show extremely high quenching efficiency (>98%) toward the embedded DNA probes, whose fluorescence can be instantly turned on by the target binding, resulting in high signal-to-background ratio. To demonstrate these distinct properties, we made use of the assembled nanoprobes to monitor the ATP levels under different stimuli in living cells. The assembly strategy leads to a new opportunity for accurately sensing targets in living systems.


Subject(s)
DNA Probes/chemistry , DNA/chemistry , Metal Nanoparticles/chemistry , Adenosine Triphosphate/analysis , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , Carbocyanines/chemistry , Carbocyanines/toxicity , Cell Line, Tumor , DNA/toxicity , DNA Probes/toxicity , Deoxyribonuclease I/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/toxicity , Gold/chemistry , Gold/toxicity , Humans , Metal Nanoparticles/toxicity , Mice , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Proof of Concept Study
11.
Anal Chem ; 90(22): 13188-13192, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30380845

ABSTRACT

The efficient detection and in situ monitoring of telomerase activity is of great importance for cancer diagnosis and biomedical research. Here we report for the first time that the development of a novel multivalent self-assembled DNA polymer, constructed through telomerase primer sequence (ITS) triggered hybridization chain assembly using two functional hairpin probes (tumor-trageting aptamer modified H1 and signal probe modified H2), for sensitive detection and imaging of telomerase activity in living cells. After internalizing into the tumor cells by multivalent aptamer targeting, the ITS on DNA polymers can be elongated by intracellular telomerase to generate telomere repeat sequences that are complementary with the signal probe, which can proceed along the DNA polymers, and gradually light up the whole DNA polymers, leading to an enhanced fluorescence signal directly correlated with the activity of telomerase. Our results demonstrated that the developed DNA polymer show excellent performance for specifically detecting telomerase activity in cancer cells, dynamically monitoring the activity change of telomerase in response to telomerase-based drugs, and efficiently distinguishing cancer cells from normal cells. The proposed strategy may afford a valuable tool for the monitoring of telomerase activity in living cells and have great implications for biological and diagnostic applications.


Subject(s)
Aptamers, Nucleotide/chemistry , DNA/chemistry , Polymers/chemistry , Telomerase/analysis , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/toxicity , Carbocyanines/chemistry , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA/toxicity , Enzyme Assays/methods , Fluorescence , Fluorescent Dyes/chemistry , Humans , Limit of Detection , Nucleic Acid Hybridization , Phosphoproteins/metabolism , Polymers/metabolism , Polymers/toxicity , RNA-Binding Proteins/metabolism , Nucleolin
12.
Theranostics ; 8(16): 4332-4344, 2018.
Article in English | MEDLINE | ID: mdl-30214624

ABSTRACT

Photodynamic therapy (PDT) has been applied in clinical cancer treatment. Here we report an aptamer-functionalized nanoscale metal-organic framework for targeted PDT. Our nanosystem can be easily prepared and successfully used for targeted PDT with a significantly enhanced therapeutic efficacy in vitro and in vivo. Methods: By combining the strong binding ability between phosphate-terminated aptamers and Zr-based nanoscale metal-organic frameworks (Zr-NMOFs) and the intercalation of photosensitizer TMPyP4 within the G-quadruplex DNA structure, TMPyP4-G4-aptamer-NMOFs were prepared. The characteristics and photodynamic performance of TMPyP4-G4-aptamer-NMOFs were examined after preparation. Then, we studied their stability, specific recognition ability, and phototoxicity in vitro. For in vivo experiments, the nanosystem was intratumorally injected into a HeLa subcutaneous xenograft tumor mouse model. After irradiation on day 0, mice were further injected with the nanosystem on day 5 and were again subjected to laser irradiation for 30 min. Tumor volumes and body weights of all mice were measured by caliper every 2 days after the treatment. Results: The nanosystem induced 90% cell death of targeted cells. In contrast, the control cells maintained about 40% cell viability at the same concentration of nanosystem. For the in vivo experiments, the nanosystem-treated group maintained more than 76% inhibition within the entire experimental period. Conclusion: We have demonstrated that our smart TMPyP4-G4-sgc8-NMOFs nanosystem can be used for targeted cancer therapy with high efficiency.


Subject(s)
Aptamers, Nucleotide/administration & dosage , G-Quadruplexes , Metal-Organic Frameworks/administration & dosage , Molecular Targeted Therapy/methods , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Animals , Aptamers, Nucleotide/toxicity , Cell Survival/drug effects , Disease Models, Animal , Drug Carriers/administration & dosage , Drug Stability , HeLa Cells , Heterografts , Humans , Metal-Organic Frameworks/toxicity , Mice , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Photosensitizing Agents/toxicity , Porphyrins/toxicity , Treatment Outcome
13.
Chem Commun (Camb) ; 54(81): 11487-11490, 2018 Oct 09.
Article in English | MEDLINE | ID: mdl-30256356

ABSTRACT

Herein, we have developed a novel fluorescence labeling strategy for nucleic acid aptamers based on self-assembling between AGG tri-nucleotide repeats and a pyrene-modified oligonucleotide. This strategy could be an effective tool for developing targeting-imaging systems and biosensor systems to detect target molecules.


Subject(s)
Aptamers, Nucleotide/genetics , DNA/genetics , G-Quadruplexes , Microscopy, Fluorescence/methods , Adenosine Monophosphate/analysis , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , Biosensing Techniques/methods , DNA/chemistry , DNA/toxicity , Fluorescence , Fluorescent Dyes/chemistry , Humans , Inverted Repeat Sequences , MCF-7 Cells , Microscopy, Confocal/methods , Oligodeoxyribonucleotides/chemistry , Trinucleotide Repeats , Uridine/analogs & derivatives , Uridine/chemistry
14.
Arch Biochem Biophys ; 646: 16-23, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29580944

ABSTRACT

Anthrax is caused by Bacillus anthracis, a bacterium that is able to secrete the toxins protective antigen, edema factor and lethal factor. Due to the high level of secretion from the bacteria and its severe virulence, lethal factor (LF) has been sought as a biomarker for detecting bacterial infection and as an effective target to neutralize toxicity. In this study, we found three aptamers, and binding affinity was determined by fluorescently labeled aptamers. One of the aptamers exhibited high affinity, with a Kd value of 11.0 ±â€¯2.7 nM, along with low cross reactivity relative to bovine serum albumin and protective antigen. The therapeutic functionality of the aptamer was examined by assessing the inhibition of LF protease activity against a mitogen-activated protein kinase kinase. The aptamer appears to be an effective inhibitor of LF with an IC50 value of 15 ±â€¯1.5 µM and approximately 85% cell viability, suggesting that this aptamer provides a potential clue for not only development of a sensitive diagnostic device of B. anthracis infection but also the design of novel inhibitors of LF.


Subject(s)
Aptamers, Nucleotide/metabolism , Bacterial Toxins/antagonists & inhibitors , DNA, Single-Stranded/metabolism , Animals , Antigens, Bacterial/metabolism , Aptamers, Nucleotide/toxicity , Bacillus anthracis/chemistry , Bacterial Toxins/metabolism , DNA, Single-Stranded/toxicity , Enzyme-Linked Immunosorbent Assay , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , Mice , Protein Binding , Proteolysis , RAW 264.7 Cells , SELEX Aptamer Technique
15.
Acta Pharmacol Sin ; 39(2): 261-274, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29388568

ABSTRACT

Melanoma is the deadliest type of skin cancer. CD20+ melanoma stem cells (CSCs) are pivotal for metastasis and initiation of melanoma. Therefore, selective elimination of CD20+ melanoma CSCs represents an effective treatment to eradicate melanoma. Salinomycin has emerged as an effective drug toward various CSCs. Due to its poor solubility, its therapeutic efficacy against melanoma CSCs has never been evaluated. In order to target CD20+ melanoma CSCs, we designed salinomycin-loaded lipid-polymer nanoparticles with anti-CD20 aptamers (CD20-SA-NPs). Using a single-step nanoprecipitation method, salinomycin-loaded lipid-polymer nanoparticles (SA-NPs) were prepared, then CD20-SA-NPs were obtained through conjugation of thiolated anti-CD20 aptamers to SA-NPs via a maleimide-thiol reaction. CD20-SA-NPs displayed a small size of 96.3 nm, encapsulation efficiency higher than 60% and sustained drug release ability. The uptake of CD20-SA-NPs by CD20+ melanoma CSCs was significantly higher than that of SA-NPs and salinomycin, leading to greatly enhanced cytotoxic effects in vitro, thus the IC50 values of CD20-SA-NPs were reduced to 5.7 and 2.6 µg/mL in A375 CD+20 cells and WM266-4 CD+ cells, respectively. CD20-SA-NPs showed a selective cytotoxicity toward CD20+ melanoma CSCs, as evidenced by the best therapeutic efficacy in suppressing the formation of tumor spheres and the proportion of CD20+ cells in melanoma cell lines. In mice bearing melanoma xenografts, administration of CD20-SA-NPs (salinomycin 5 mg·kg-1·d-1, iv, for 60 d) showed a superior efficacy in inhibition of melanoma growth compared with SA-NPs and salinomycin. In conclusion, CD20 is a superior target for delivering drugs to melanoma CSCs. CD20-SA-NPs display effective delivery of salinomycin to CD20+ melanoma CSCs and represent a promising treatment for melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Carriers/therapeutic use , Melanoma/drug therapy , Nanoparticles/therapeutic use , Neoplastic Stem Cells/drug effects , Pyrans/therapeutic use , Animals , Antigens, CD20/chemistry , Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/therapeutic use , Aptamers, Nucleotide/toxicity , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/toxicity , Humans , Lecithins/chemistry , Lecithins/metabolism , Lecithins/therapeutic use , Lecithins/toxicity , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/metabolism , Nanoparticles/toxicity , Particle Size , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Polyethylene Glycols/therapeutic use , Polyethylene Glycols/toxicity , Pyrans/pharmacology , Xenograft Model Antitumor Assays
16.
Anal Chem ; 88(19): 9355-9358, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27649276

ABSTRACT

Inorganic nanocrystals, such as quantum dots (QDs), hold great promise as molecular imaging contrast agents because of their superior optical properties. However, the molecular imaging sensitivity of these probes is far from optimized due to the lack of efficient and general method for molecular engineering of nanocrystal into effective bioprobes for signal-amplified imaging. Herein, we develop a strategy to boost the molecular imaging sensitivity of QDs over the limit by copolymerizing QDs and cell-binding aptamers into linear QD-aptamer polymers (QAPs) through DNA-programmed hybridization chain reaction. We show that the cancer cells treated with QAPs exhibit much stronger photoluminescence (PL) signal than those treated with QD-aptamer monomers (QAMs) because of multivalent binding and multi-QD-based signal amplification. The enhanced cell binding and imaging capacity of QAPs significantly improves imaging-based discrimination between different cancer cell types. This approach adds a new dimension for engineering inorganic nanoparticles into effective bioprobes for biomedical applications.


Subject(s)
DNA/chemistry , Neoplasms/diagnostic imaging , Quantum Dots/chemistry , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/toxicity , Biosensing Techniques/methods , Cell Line, Tumor , Humans , Luminescence , Molecular Imaging/methods , Nucleic Acid Hybridization , Polymerization , Quantum Dots/toxicity
17.
Sci Rep ; 6: 26059, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27189805

ABSTRACT

Vascular endothelial growth factor B (VEGF-B) was discovered a long time ago. However, its role in hyperglycemia- and VEGF-A inhibition-induced retinal apoptosis remains unknown thus far. Yet, drugs that can block VEGF-B are being used to treat patients with diabetic retinopathy and other ocular neovascular diseases. It is therefore urgent to have a better understanding of the function of VEGF-B in these pathologies. Here, we report that both streptozotocin (STZ)-induced diabetes in rats and Macugen intravitreal injection in mice leads to retinal apoptosis in retinal ganglion cell and outer nuclear layers respectively. Importantly, VEGF-B treatment by intravitreal injection markedly reduced retinal apoptosis in both models. We further reveal that VEGF-B and its receptors, vascular endothelial growth factor 1 (VEGFR1) and neuropilin 1 (NP1), are abundantly expressed in rat retinae and choroids and are upregulated by high glucose with concomitant activation of Akt and Erk. These data highlight an important function of VEGF-B in protecting retinal cells from apoptosis induced by hyperglycemia and VEGF-A inhibition. VEGF-B may therefore have a therapeutic potential in treating various retinal degenerative diseases, and modulation of VEGF-B activity in the eye needs careful consideration.


Subject(s)
Apoptosis/drug effects , Retinal Diseases/drug therapy , Retinal Ganglion Cells/physiology , Vascular Endothelial Growth Factor B/administration & dosage , Animals , Aptamers, Nucleotide/toxicity , Diabetes Mellitus, Experimental/complications , Mice, Inbred C57BL , Rats , Retinal Diseases/physiopathology , Retinal Ganglion Cells/drug effects , Treatment Outcome
18.
Toxicol Appl Pharmacol ; 287(1): 86-92, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26048585

ABSTRACT

The medical applications of aptamers have recently emerged. We developed an antagonistic thioaptamer (ESTA) against E-selectin. Previously, we showed that a single injection of ESTA at a dose of 100µg inhibits breast cancer metastasis in mice through the functional blockade of E-selectin. In the present study, we evaluated the safety of different doses of intravenously administered ESTA in single-dose acute and repeat-dose subacute studies in ICR mice. Our data indicated that intravenous administration of up to 500µg ESTA did not result in hematologic abnormality in either study. Additionally, intravenous injection of ESTA did not affect the levels of plasma cytokines (IL-1ß, IL-2, IL-4, IL-5, IL-6, IL-10, GM-CSF, IFN-γ, and TNF-α) or complement split products (C3a and C5a) in either study. However, repeated injections of ESTA slightly increased plasma ALT and AST activities, in accordance with the appearance of small necrotic areas in the liver. In conclusion, our data demonstrated that intravenous administration of ESTA does not cause overt hematologic, organs, and immunologic responses under the experimental conditions.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/administration & dosage , E-Selectin/drug effects , Alanine Transaminase/blood , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , Aspartate Aminotransferases/blood , Biomarkers/blood , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Complement C3a/metabolism , Complement C5a/metabolism , Cytokines/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , E-Selectin/metabolism , Female , Injections, Intravenous , Kidney/drug effects , Liver/drug effects , Liver/pathology , Male , Mice, Inbred ICR , Necrosis , Risk Assessment
19.
Nanoscale ; 7(22): 10240-8, 2015 Jun 14.
Article in English | MEDLINE | ID: mdl-25990591

ABSTRACT

Aptamer-conjugated gold nanorods (AuNRs) are excellent candidates for targeted hyperthermia therapy of cancer cells. However, in high concentrations of AuNRs, aptamer conjugation alone fails to result in highly cell-specific AuNRs due to the presence of positively charged cetyltrimethylammonium bromide (CTAB) as a templating surfactant. Besides causing nonspecific electrostatic interactions with the cell surfaces, CTAB can also be cytotoxic, leading to uncontrolled cell death. To avoid the nonspecific interactions and cytotoxicity triggered by CTAB, we report the further biologically inspired modification of aptamer-conjugated AuNRs with bovine serum albumin (BSA) protein. Following this modification, interaction between CTAB and the cell surface was efficiently blocked, thereby dramatically reducing the side effects of CTAB. This approach may provide a general and simple method to avoid one of the most serious issues in biomedical applications of nanomaterials: nonspecific binding of the nanomaterials with biological cells.


Subject(s)
Aptamers, Nucleotide/toxicity , Cetrimonium Compounds/toxicity , Gold/toxicity , Nanotubes/toxicity , Serum Albumin, Bovine/chemistry , Animals , Aptamers, Nucleotide/chemistry , Cattle , Cell Line, Tumor , Cell Survival/drug effects , Cetrimonium , Cetrimonium Compounds/chemistry , Gold/chemistry , Humans , Immobilized Nucleic Acids , Nanotubes/chemistry , Protein Binding
20.
Int J Biol Macromol ; 77: 293-302, 2015.
Article in English | MEDLINE | ID: mdl-25841381

ABSTRACT

Anthrax toxin excreted by Bacillus anthracis is the key causative agent of infectious anthrax disease. In the present study, we targeted the binding of PA to the ATR/TEM8 Von Willebrand factor type A (VWA) domain, which we cloned into Escherichia coli and purified to homogeneity under denaturing conditions. To develop an anthrax toxin inhibitor, we selected and identified short single strand RNA aptamers (approximately 30mer) consisting of different sequences of nucleic acids with a high binding affinity in the 100 nanomolar range against the recombinant ATR/TEM8 VWA domain using systematic evolution of ligands by exponential enrichment (SELEX). Five candidate aptamers were further characterized by several techniques including secondary structural analysis. The inhibitor efficiency (IC50) of one of the aptamers toward anthrax toxin was approximately 5µM in macrophage RAW 264.7 cells, as determined from cytotoxicity analysis by MTT assay. We believe that the candidate aptamers should be useful for blocking the binding of PA to its receptor in order to neutralize anthrax toxin.


Subject(s)
Antigens, Bacterial/metabolism , Aptamers, Nucleotide/metabolism , Bacterial Toxins/metabolism , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , SELEX Aptamer Technique , Amino Acid Sequence , Animals , Aptamers, Nucleotide/toxicity , Humans , Mice , Microfilament Proteins , Molecular Sequence Data , Neoplasm Proteins/chemistry , Protein Structure, Tertiary , RAW 264.7 Cells , Receptors, Cell Surface/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...