Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Small ; 19(14): e2205630, 2023 04.
Article in English | MEDLINE | ID: mdl-36634975

ABSTRACT

Intravesical instillation is an effective treatment for bladder cancer. However, clinical anticancer agents always suffer rapid excretion by periodic urination, leading to low therapeutic efficacy. Prolonging the retention time of drugs in the bladder is the key challenge for intravesical instillation treatment. Herein, a facile and powerful surface cross-linking-freeze drying strategy is proposed to generate ultra-stable albumin bovine air microbubbles (BSA-MBs) that can float and adhere to the bladder wall to overcome the excretion of urination and exhibit a remarkable property of long-term retention in the bladder. More noteworthy, BSA-MBs are endowed with a specific three-layer structure, namely, the outer membrane, middle drug loading layer and inner air core, which makes them have a low density to easily float and possess a high drug loading capacity. Based on their unique superiorities, the therapeutic potential of doxorubicin (DOX)-loaded BSA-MBs (DOX-MBs) is exemplified by intravesical instillation for bladder cancer. After injection into the bladder, DOX-MBs can remain in the bladder for a long time and sustain the release of DOX in urine, exhibiting potent anticancer efficacy. Consequently, the prolonged retention of BSA-MBs in the bladder renders them as an effective floating drug delivery system for intravesical instillation therapy.


Subject(s)
Antineoplastic Agents , Urinary Bladder Neoplasms , Humans , Animals , Cattle , Administration, Intravesical , Microbubbles , Antineoplastic Agents/therapeutic use , Doxorubicin/chemistry , Urinary Bladder Neoplasms/drug therapy
2.
Opt Express ; 31(20): 32322-32334, 2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37859038

ABSTRACT

In plasmonic Fano resonance, the interaction between a discrete plasmonic mode and a continuum of plasmonic mode gives rise to an asymmetric line shape in the scattering or absorption spectrum, enabling a wide range of applications such as sensing, switching, and slow light devices. Here, we establish a theoretical solution in the framework of temporal coupled-mode theory (TCMT) to study the three-dimensional (3D) and two-dimensional (2D) Fano resonances induced by strong coupling between metal hole (MH) and split ring resonator (SRR) array. We first separately analyze the transmission spectra of the MH array and SRR array under different polarized light excitation. We further investigate the electromagnetic field and charge density distribution corresponding to the resonant modes at the peak or valley wavelength of the transmission spectrum and figure out the electric/magnetic dipole feature of these resonance modes. We then establish a theoretical solution by TCMT for Fano resonances arising from the coupling of these modes. The calculated transmission spectrum is closely matching with the numerically simulated transmission spectrum for these Fano resonances in the MH-SRR array, which effectively elucidates that the asymmetry of the Fano resonances is caused by the coupling between bright and dark plasmonic modes involved in the two structures. Our results can help to understand the profound physics in such coupled plasmonic systems.

3.
Biomacromolecules ; 16(4): 1390-400, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25756930

ABSTRACT

Polycations often suffer from the irreconcilable inconsistency between transfection efficiency and toxicity. Polymers with high molecular weight (MW) and cationic charge feature potent gene delivery capabilities, while in the meantime suffer from strong chemotoxicity, restricted intracellular DNA release, and low stability in vivo. To address these critical challenges, we herein developed pH-responsive, reversibly cross-linked, polyetheleneimine (PEI)-based polyplexes coated with hyaluronic acid (HA) for the effective and targeted gene delivery to cancer cells. Low-MW PEI was cross-linked with the ketal-containing linker, and the obtained high-MW analogue afforded potent gene delivery capabilities during transfection, while rapidly degraded into low-MW segments upon acid treatment in the endosomes, which promoted intracellular DNA release and reduced material toxicity. HA coating of the polyplexes shielded the surface positive charges to enhance their stability under physiological condition and simultaneously reduced the toxicity. Additionally, HA coating allowed active targeting to cancer cells to potentiate the transfection efficiencies in cancer cells in vitro and in vivo. This study therefore provides an effective approach to overcome the efficiency-toxicity inconsistence of nonviral vectors, which contributes insights into the design strategy of effective and safe vectors for cancer gene therapy.


Subject(s)
DNA/administration & dosage , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Neoplasms/therapy , Animals , Cross-Linking Reagents/chemistry , DNA/genetics , DNA/toxicity , Endosomes/metabolism , Genetic Therapy/methods , Genetic Vectors/toxicity , HeLa Cells , Humans , Hyaluronic Acid/chemistry , Male , Mice , Mice, Inbred C57BL , Polyethyleneimine/chemistry
4.
Light Sci Appl ; 12(1): 219, 2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37673900

ABSTRACT

Scanning near-field optical microscopy (SNOM) offers a means to reach a fine spatial resolution down to ~ 10 nm, but unfortunately suffers from low transmission efficiency of optical signal. Here we present design and 3D printing of a fiber-bound polymer-core/gold-shell spiral-grating conical tip that allows for coupling the inner incident optical signal to the outer surface plasmon polariton with high efficiency, which then adiabatically transport, squeeze, and interfere constructively at the tip apex to form a plasmonic superfocusing spot with tiny size and high brightness. Numerical simulations and optical measurements show that this specially designed and fabricated tip has 10% transmission efficiency, ~ 5 nm spatial resolution, 20 dB signal-to-noise ratio, and 7000 pixels per second fast scanning speed. This high-resolution, high throughput, and high contrast SNOM would open up a new frontier of high spatial-temporal resolution detecting, imaging, and monitoring of single-molecule physical, chemical, and biological systems, and deepen our understanding of their basic science in the single-molecule level.

5.
ACS Nano ; 17(1): 263-274, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36354372

ABSTRACT

Microbubbles have been widely used as ultrasound contrast agents in clinical diagnosis. Moreover, most current preparation methods for microbubbles are uncontrollable, and the as-obtained microbubbles are unstable in aqueous solution or under ultrasound. Here, we report a strategy to prepare superiorly stable microbubbles with three-layer structures by the ethanol-water exchange. This versatile method can also be applied to prepare different kinds of protein microbubbles with various sizes for advanced biomedical applications. To demonstrate this, the protein air microbubbles are created, which is stable in water for several days with intact structures and exhibits excellent contrast-enhanced ultrasound imaging. Moreover, the protein air microbubbles can also deliver a mass of drugs while maintaining their stable structures, making them a platform for ultrasound imaging-guided drug delivery. The versatile protein air microbubbles have great potential for the design and application of theranostic platforms.


Subject(s)
Ethanol , Precision Medicine , Microbubbles , Ultrasonography/methods , Drug Delivery Systems/methods , Contrast Media/chemistry
6.
Acta Biomater ; 143: 344-355, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35189380

ABSTRACT

Neutrophils serve as a key contributor to the pathophysiology of myocardial ischemia reperfusion injury (MIRI), because the unregulated activation and infiltration of neutrophils lead to overwhelming inflammation in the myocardium to cause tissue damage. Herein, endothelial cell-targeting and  reactive oxygen species (ROS)-ultrasensitive nanocomplexes (NCs) were developed to mediate efficient co-delivery of VCAM-1 siRNA (siVCAM-1) and dexamethasone (DXM), which cooperatively inhibited neutrophil recruitment by impeding neutrophil migration and adhesion. RPPT was first synthesized via crosslinking of PEI 600 with ditellurium followed by modification with PEG and the endothelial cell-targeting peptide cRGD. RPPT was allowed to envelope the DXM-loaded PLGA nanoparticles and condense the siVCAM-1. After systemic administration in rats experiencing MIRI, the cRGD-modified NCs efficiently targeted and entered the inflamed endothelial cells, wherein RPPT was sensitively degraded by over-produced ROS to trigger intracellular siVCAM-1 release and potentiate the VCAM-1 silencing efficiency. As a consequence of the complementary function of DXM and siVCAM-1, the NCs notably mitigated neutrophil infiltration into ischemic myocardium, provoking potent anti-inflammatory efficacy to reduce MIRI and recover cardiac function. The present study offers an effective approach for the controlled co-delivery of siRNA and drug cargoes, and it also highlights the importance of multi-dimensional manipulation of neutrophils in anti-inflammatory treatment. STATEMENT OF SIGNIFICANCE: The unregulated activation and infiltration of neutrophils lead to overwhelming inflammation in the myocardium after myocardial ischemia reperfusion injury (MIRI). Here, endothelial cell-targeting and ROS-ultrasensitive nanocomplexes (NCs), comprised of PLGA NPs decorated with cRGD-poly(ethylene glycol) (PEG)-modified, ditellurium-crosslinked PEI (RPPT), were developed to mediate efficient co-delivery of VCAM-1 siRNA (siVCAM-1) and dexamethasone (DXM). DXM and siVCAM-1 with complementary functions inhibited both the migration and adhesion of neutrophils, efficiently interventing the neutrophil recruitment and interrupting the self-amplified inflammation cascade in the injured myocardium. The molecular design of RPPT renders an effective example for constructing polymeric materials with high ROS sensitivity, and it resolves the critical dilemma related to polycation-mediated siRNA delivery, such as siRNA encapsulation versus release, and transfection efficiency versus toxicity.


Subject(s)
Myocardial Reperfusion Injury , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Dexamethasone/pharmacology , Endothelial Cells , Inflammation/drug therapy , Inflammation/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
7.
Nano Res ; 15(10): 9125-9134, 2022.
Article in English | MEDLINE | ID: mdl-35915748

ABSTRACT

Myocardial ischemia reperfusion (IR) injury is closely related to the overwhelming inflammation in the myocardium. Herein, cardiomyocyte-targeted nanotherapeutics were developed for the reactive oxygen species (ROS)-ultrasensitive co-delivery of dexamethasone (Dex) and RAGE small interfering RNA (siRAGE) to attenuate myocardial inflammation. PPTP, a ROS-degradable polycation based on PGE2-modified, PEGylated, ditellurium-crosslinked polyethylenimine (PEI) was developed to surface-decorate the Dex-encapsulated mesoporous silica nanoparticles (MSNs), which simultaneously condensed siRAGE and gated the MSNs to prevent the Dex pre-leakage. Upon intravenous injection to IR-injured rats, the nanotherapeutics could be efficiently transported into the inflamed cardiomyocytes via PGE2-assisted recognition of over-expressed E-series of prostaglandin (EP) receptors on the cell membranes. Intracellularly, the over-produced ROS degraded PPTP into small segments, promoting the release of siRAGE and Dex to mediate effective RAGE silencing (72%) and cooperative antiinflammatory effect. As a consequence, the nanotherapeutics notably suppressed the myocardial fibrosis and apoptosis, ultimately recovering the systolic function. Therefore, the current nanotherapeutics represent an effective example for the co-delivery and on-demand release of nucleic acid and chemodrug payloads, and might find promising utilities toward the synergistic management of myocardial inflammation. Electronic Supplementary Material: Supplementary material (experimental methods, RNA and primer sequences, 1H NMR spectra, FTIR spectrum, TEM images, zeta potential, drug loading content, RNA and drug release, cytotoxicity, etc.) is available in the online version of this article at 10.1007/s12274-022-4553-6.

8.
J Mater Chem B ; 9(10): 2407-2416, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33623935

ABSTRACT

Imaging-guided vascular embolization is frequently performed on patients with advanced hepatocellular carcinoma (HCC) to alleviate symptoms and extend their survival time. Current operation procedures are not only painful for patients, but are also inaccurate in tumor targeting after the release of embolic agents from the catheter, leading to injury to healthy tissues simultaneously. In this study, we developed an ultrasound-visualized, site-specific vascular embolization strategy with magnetic protein microcapsules (MPMs). MPMs were fabricated using a rapid emulsification method, giving it a smooth surface and a core-shell structure. Their diameters could be controlled within 10 µm, allowing them to pass through capillaries. The core-shell structure and loading of magnetic Fe3O4 endowed MPMs with good contrast under ultrasound imaging and magnetically inducible targeting properties, as well as aggregation response even under flowing conditions. In vitro cytotoxicity and hemolysis evaluation demonstrated good biocompatibility of the MPMs. Furthermore, mock embolization showed that cell death could be induced by aggregation of the MPMs. Such a combination of real-time monitoring using ultrasound and control on targeted vascular embolization might be a breakthrough in the treatment of advanced HCC.


Subject(s)
Blood Vessels/diagnostic imaging , Embolization, Therapeutic/methods , Ferrosoferric Oxide/chemistry , Proteins/chemistry , Capsules , Hep G2 Cells , Humans , Ultrasonography
9.
Biomater Sci ; 8(14): 3856-3870, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32572407

ABSTRACT

siRNA-mediated RNA interference (RNAi) against inflammation-related genes provides a promising modality for the treatment of myocardial ischemia reperfusion (IR) injury, and its success is critically dependent on the development of efficient yet safe siRNA delivery vehicles. Herein, we developed a bioreducible, branched poly(ß-amino ester) with built-in redox-responsive domains (BPAE-SS) for the effective ICAM-1 siRNA delivery into injured rat cardiac microvascular endothelial cells (RCMECs). The branched BPAE-SS with a multivalent structure afforded potent siRNA binding affinity compared to its linear analogue, while upon internalization into RCMECs it was instantaneously degraded by intracellular glutathione (GSH) into small segments to mediate "on-demand" siRNA release and diminish the toxicity of post-transfection materials. By synchronizingly overcoming these critical barriers, BPAE-SS mediated remarkable ICAM-1 knockdown in IR-injured rats at 400 µg siRNA per kg via single i.v. injection, and subsequently suppressed myocardial inflammation, apoptosis, and fibrosis to recover the cardiac function. This study therefore provides a unique delivery system that can address the multiple critical challenges against non-viral siRNA delivery, and the potent therapeutic efficacy of BPAE-SS-mediated ICAM-1 silencing provides a promising strategy for the anti-inflammatory treatment of myocardial IR injury.


Subject(s)
Myocardial Reperfusion Injury , Animals , Anti-Inflammatory Agents , Endothelial Cells , Esters , Intercellular Adhesion Molecule-1/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/therapy , Polymers , RNA, Small Interfering/genetics , Rats
10.
ACS Appl Mater Interfaces ; 9(28): 23586-23601, 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28657294

ABSTRACT

The development of potent yet nontoxic membrane-penetrating materials is in high demand for effective intracellular gene delivery. We have recently developed α-helical polypeptides which afford potent membrane activities to facilitate intracellular DNA delivery via both endocytosis and the nonendocytic "pore formation" mechanism. Endocytosis will cause endosomal entrapment of the DNA cargo, while excessive "pore formation" would cause appreciable cytotoxicity. Additionally, helical polypeptides with stiff, rodlike structure suffer from low siRNA binding affinity. To address such critical issues, we herein incorporated various aromatic domains (benzyl, naphthyl, biphenyl, anthryl, and pyrenyl) into the side-chain terminals of guanidine-rich, helical polypeptides, wherein the flat-rigid shape, π-electronic structures of aromatic motifs "self-activated" the membrane-penetrating capabilities of polypeptides to promote intracellular gene delivery. Benzyl (Bn)- and naphthyl (Naph)-modified polypeptides demonstrated the highest DNA uptake level that outperformed the unmodified polypeptide, P2, by ∼4 fold. More importantly, compared with P2, Bn- and Naph-modified polypeptides allowed more DNA cargos to be internalized via the nonendocytic pathway, which significantly bypassed the endosomal entrapment and accordingly enhanced the transfection efficiency by up to 42 fold, outperforming PEI 25k as the commercial reagent by 3-4 orders of magnitude. The aromatic modification also improved the siRNA condensation capability of polypeptides, achieving notably enhanced gene-silencing efficiency against tumor necrosis factor-α to treat acute hepatic inflammation. Furthermore, we revealed that aromaticity-augmented membrane activity was accompanied by comparable or even significantly reduced "pore formation" capability, thus leading to diminished cytotoxicity at high concentrations. This study therefore provides a promising approach to manipulate the membrane activities and penetration mechanisms of polycations, which overcomes the multiple critical barriers preventing effective and safe gene delivery.


Subject(s)
Peptides/chemistry , Cations , DNA , Gene Transfer Techniques , RNA, Small Interfering , Transfection
11.
Biomater Sci ; 5(6): 1174-1182, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28513659

ABSTRACT

The transfection performance of polycations is often hampered by various systemic barriers that pose conflicting requirements for material design. Herein, we developed fluorinated, ROS-cleavable polyethylenimine (PEI) for effective and serum-resistant gene delivery to cancer cells, by harmonizing the inconsistency between DNA condensation and release, and the inconsistency between cellular internalization and serum stability. Low-molecular weight (MW) PEI was cross-linked with a diselenide-containing linker and further modified with fluorocarbon chains. The obtained high-MW DSe-PEI-F has potent DNA condensation as well as intracellular DNA delivery capabilities, while in the cytoplasm of cancer cells, it can rapidly degrade into low-MW segments upon ROS treatment to promote DNA release and reduce the material toxicity. As such, DSe-PEI-F showed high transfection efficiencies in cancer cells in the presence of serum, outperforming the commercial reagent PEI 25k by several orders of magnitude. This study thus provides an effective approach to overcome various barriers against non-viral gene delivery, which contributes to the development of a new class of gene vectors with high efficiency and low toxicity.


Subject(s)
Cross-Linking Reagents/metabolism , DNA/administration & dosage , Polyethyleneimine/metabolism , Reactive Oxygen Species/metabolism , Transfection/methods , Cross-Linking Reagents/chemistry , DNA/genetics , Gene Transfer Techniques , Halogenation , HeLa Cells , Humans , Organoselenium Compounds/chemistry , Organoselenium Compounds/metabolism , Polyethyleneimine/chemistry , Serum/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL