Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Xian-Zhu Yang is active.

Publication


Featured researches published by Xian-Zhu Yang.


Journal of the American Chemical Society | 2015

Ultrathin Black Phosphorus Nanosheets for Efficient Singlet Oxygen Generation.

Hui Wang; Xian-Zhu Yang; Wei Shao; Shichuan Chen; Junfeng Xie; Xiaodong Zhang; Jun Wang; Yi Xie

Benefiting from its strong oxidizing properties, the singlet oxygen has garnered serious attentions in physical, chemical, as well as biological studies. However, the photosensitizers for the generation of singlet oxygen bear in low quantum yields, lack of long wavelength absorption band, poor biocompatibility, undegradable in living tissues, and so on. Here we first demonstrate the exfoliated black phosphorus nanosheets to be effective photosensitizers for the generation of singlet oxygen with a high quantum yield of about 0.91, rendering their attractive applications in catalysis and photodynamic therapy. Through in vitro and in vivo studies, the water dispersible black phosphorus nanosheets show notable cancer therapy ability. In addition, the photodegradable character of black phosphorus from element to biocompatible phosphorus oxides further highlights its therapeutic potential against cancer. This study will not only expand the breadth of study in black phosphorus but also offer an efficient catalyst and photodynamic therapy agent.


Small | 2013

Polyethylene Glycol and Polyethylenimine Dual‐Functionalized Nano‐Graphene Oxide for Photothermally Enhanced Gene Delivery

Liangzhu Feng; Xian-Zhu Yang; Xiaoze Shi; Xiaofang Tan; Rui Peng; Jun Wang; Zhuang Liu

Graphene oxide (GO) has been extensively explored in nanomedicine for its excellent physiochemical, electrical, and optical properties. Here, polyethylene glycol (PEG) and polyethylenimine (PEI) are covalently conjugated to GO via amide bonds, obtaining a physiologically stable dual-polymer-functionalized nano-GO conjugate (NGO-PEG-PEI) with ultra-small size. Compared with free PEI and the GO-PEI conjugate without PEGylation, NGO-PEG-PEI shows superior gene transfection efficiency without serum interference, as well as reduced cytotoxicity. Utilizing the NIR optical absorbance of NGO, the cellular uptake of NGO-PEG-PEI is shown to be enhanced under a low power NIR laser irradiation, owing to the mild photothermal heating that increases the cell membrane permeability without significantly damaging cells. As the results, remarkably enhanced plasmid DNA transfection efficiencies induced by the NIR laser are achieved using NGO-PEG-PEI as the light-responsive gene carrier. More importantly, it is shown that our NGO-PEG-PEI is able to deliver small interfering RNA (siRNA) into cells under the control of NIR light, resulting in obvious down-regulation of the target gene, Polo-like kinase 1 (Plk1), in the presence of laser irradiation. This study is the first to use photothermally enhanced intracellular trafficking of nanocarriers for light-controllable gene delivery. This work also encourages further explorations of functionalized nano-GO as a photocontrollable nanovector for combined photothermal and gene therapies.


ACS Nano | 2012

Sheddable Ternary Nanoparticles for Tumor Acidity-Targeted siRNA Delivery

Xian-Zhu Yang; Jin-Zhi Du; Shuang Dou; Cheng-Qiong Mao; Hong-Yan Long; Jun Wang

Drug delivery systems for cancer therapy usually need to be sterically stabilized by a poly(ethylene glycol) (PEG) layer during blood circulation to minimize nonspecific interactions with serum components. However, PEGylation significantly reduces cellular uptake of the delivery systems after they accumulate at the tumor site, which markedly impairs the in vivo antitumor efficiency. Here, we develop a ternary small interfering RNA (siRNA) delivery system with tumor acidity-activated sheddable PEG layer to overcome the challenge. The sheddable nanoparticle is fabricated by introducing a tumor acidity-responsive PEGylated anionic polymer to the surface of positively charged polycation/siRNA complexes via electrostatic interaction. We show clear evidence that introducing the PEGylated anionic polymer to the surface of a nanoparticle markedly reduces its nonspecific interactions with protein. We further demonstrate that the nanoparticle is capable of deshielding the PEG layer at the slightly acidic tumor extracellular microenvironment to facilitate the delivery of siRNA to the tumor cells after accumulation at the tumor site. Accordingly, this promotes the RNA-interfering efficiencies and enhances the inhibition of tumor growth. Such delivery system with the ability to deshield the PEG layer at the target tissues has remarkable potential in cancer therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Stimuli-responsive clustered nanoparticles for improved tumor penetration and therapeutic efficacy

Hong-Jun Li; Jin Zhi Du; Xiao Jiao Du; Cong Fei Xu; Chun Yang Sun; Hong-Xia Wang; Zhi Ting Cao; Xian-Zhu Yang; Yan Hua Zhu; Shuming Nie; Jun Wang

Significance Successively overcoming a series of biological barriers that cancer nanotherapeutics would encounter upon intravenous administration is required for achieving positive treatment outcomes. A hurdle to this goal is the inherently unfavorable tumor penetration of nanoparticles due to their relatively large sizes. We developed a stimuli-responsive clustered nanoparticle (iCluster) and justified that its adaptive alterations of physicochemical properties (e.g. size, zeta potential, and drug release rate) in accordance with the endogenous stimuli of the tumor microenvironment made possible the ultimate overcoming of these barriers, especially the bottleneck of tumor penetration. Results in varying intractable tumor models demonstrated significantly improved antitumor efficacy of iCluster than its control groups, demonstrating that overcoming these delivery barriers can be achieved by innovative nanoparticle design. A principal goal of cancer nanomedicine is to deliver therapeutics effectively to cancer cells within solid tumors. However, there are a series of biological barriers that impede nanomedicine from reaching target cells. Here, we report a stimuli-responsive clustered nanoparticle to systematically overcome these multiple barriers by sequentially responding to the endogenous attributes of the tumor microenvironment. The smart polymeric clustered nanoparticle (iCluster) has an initial size of ∼100 nm, which is favorable for long blood circulation and high propensity of extravasation through tumor vascular fenestrations. Once iCluster accumulates at tumor sites, the intrinsic tumor extracellular acidity would trigger the discharge of platinum prodrug-conjugated poly(amidoamine) dendrimers (diameter ∼5 nm). Such a structural alteration greatly facilitates tumor penetration and cell internalization of the therapeutics. The internalized dendrimer prodrugs are further reduced intracellularly to release cisplatin to kill cancer cells. The superior in vivo antitumor activities of iCluster are validated in varying intractable tumor models including poorly permeable pancreatic cancer, drug-resistant cancer, and metastatic cancer, demonstrating its versatility and broad applicability.


Journal of the American Chemical Society | 2012

Lipase-Sensitive Polymeric Triple-Layered Nanogel for “On-Demand” Drug Delivery

Meng-Hua Xiong; Yan Bao; Xian-Zhu Yang; Yu-Cai Wang; Baolin Sun; Jun Wang

We report a new strategy for differential delivery of antimicrobials to bacterial infection sites with a lipase-sensitive polymeric triple-layered nanogel (TLN) as the drug carrier. The TLN was synthesized by a convenient arm-first procedure using an amphiphilic diblock copolymer, namely, monomethoxy poly(ethylene glycol)-b-poly(ε-caprolactone), to initiate the ring-opening polymerization of the difunctional monomer 3-oxapentane-1,5-diyl bis(ethylene phosphate). The hydrophobic poly(ε-caprolactone) (PCL) segments collapsed and surrounded the polyphosphoester core, forming a hydrophobic and compact molecular fence in aqueous solution which prevented antibiotic release from the polyphosphoester core prior to reaching bacterial infection sites. However, once the TLN sensed the lipase-secreting bacteria, the PCL fence of the TLN degraded to release the antibiotic. Using Staphylococcus aureus (S. aureus) as the model bacterium and vancomycin as the model antimicrobial, we demonstrated that the TLN released almost all the encapsulated vancomycin within 24 h only in the presence of S. aureus, significantly inhibiting S. aureus growth. The TLN further delivered the drug into bacteria-infected cells and efficiently released the drug to kill intracellular bacteria. This technique can be generalized to selectively deliver a variety of antibiotics for the treatment of various infections caused by lipase-secreting bacteria and thus provides a new, safe, effective, and universal approach for the treatment of extracellular and intracellular bacterial infections.


Macromolecular Bioscience | 2009

Recent Progress in Polyphosphoesters: From Controlled Synthesis to Biomedical Applications

Yu-Cai Wang; Youyong Yuan; Jin-Zhi Du; Xian-Zhu Yang; Jun Wang

Polyphosphoesters (PPEs) with repeating phosphoester bonds in the backbone are structurally versatile, biocompatible, and biodegradable through hydrolysis as well as enzymatic digestion under physiological conditions. They are appealing for biological applications because of their potential functionality, biocompatibility, and similarity to biomacromolecules such as nucleic acids. The expanding scope of PPEs in materials science, especially as biomaterials, is described in this review. We mainly focus on controlled synthetic methods of PPEs, which provide access to novel and complex polymer structures, especially for block copolymers. The hydrolytic and enzymatic degradation of PPEs, thermoresponsive PPEs, and biomedical applications have also been discussed.


Journal of Controlled Release | 2011

Systemic delivery of siRNA with cationic lipid assisted PEG-PLA nanoparticles for cancer therapy.

Xian-Zhu Yang; Shuang Dou; Tian-Meng Sun; Cheng-Qiong Mao; Hong-Xia Wang; Jun Wang

Delivery of small interfering RNA (siRNA) has been one of the major hurdles for the application of RNA interference in therapeutics. Here, we describe a cationic lipid assisted polymeric nanoparticle system with stealthy property for efficient siRNA encapsulation and delivery, which was fabricated with poly(ethylene glycol)-b-poly(d,l-lactide), siRNA and a cationic lipid, using a double emulsion-solvent evaporation technique. By incorporation of the cationic lipid, the encapsulation efficiency of siRNA into the nanoparticles could be above 90% and the siRNA loading weight ratio was up to 4.47%, while the diameter of the nanoparticles was around 170 to 200nm. The siRNA retained its integrity within the nanoparticles, which were effectively internalized by cancer cells and escaped from the endosome, resulting in significant gene knockdown. This effect was demonstrated by significant down-regulation of luciferase expression in HepG2-luciferase cells which stably express luciferase, and suppression of polo-like kinase 1 (Plk1) expression in HepG2 cells, following delivery of specific siRNAs by the nanoparticles. Furthermore, the nanoparticles carrying siRNA targeting the Plk1 gene were found to induce remarkable apoptosis in both HepG2 and MDA-MB-435s cancer cells. Systemic delivery of specific siRNA by nanoparticles significantly inhibited luciferase expression in an orthotopic murine liver cancer model and suppressed tumor growth in a MDA-MB-435s murine xenograft model, suggesting its therapeutic promise in disease treatment.


Journal of the American Chemical Society | 2015

Tumor Acidity-Sensitive Polymeric Vector for Active Targeted siRNA Delivery.

Chun-Yang Sun; Song Shen; Cong-Fei Xu; Hong-Jun Li; Yang Liu; Zhi-Ting Cao; Xian-Zhu Yang; Jin-Xing Xia; Jun Wang

Although surface PEGylation of siRNA vectors is effective for preventing protein adsorption and thereby helps these vectors to evade the reticuloendothelial system (RES) in vivo, it also suppresses the cellular uptake of these vectors by target cells. This dilemma could be overcome by employing stimuli-responsive shell-detachable nanovectors to achieve enhanced cellular internalization while maintaining prolonged blood circulation. Among the possible stimuli, dysregulated pH in tumor (pHe) is the most universal and practical. However, the design of pHe-sensitive system is problematic because of the subtle differences between the pHe and pH in other tissues. Here, a simple acid-sensitive bridged copolymer is developed and used for tumor-targeted systemic delivery of siRNA. After forming the micelleplex delivery system, the corresponding nanoparticles (Dm-NP) might undergo several modifications as follows: (i) a poly(ethylene glycol) (PEG) corona, which is stable in the circulatory system and protects nanovectors from RES clearance; (ii) a pHe responsive linkage breakage, which induces PEG detachment at tumor sites and thereby facilitates cell targeting; and (iii) a cell-penetration peptide, which is exposed upon the removal of PEG and further enhances cellular uptake. Thus, Dm-NP achieved both prolonged circulation and effective accumulation in tumor cells and resulted in the safe and enhanced inhibition of non-small cell lung cancer growth.


Advanced Materials | 2012

Bacteria‐Responsive Multifunctional Nanogel for Targeted Antibiotic Delivery

Meng-Hua Xiong; Yajuan Li; Yan Bao; Xian-Zhu Yang; Bing Hu; Jun Wang

Bacteria-Responsive Multifunctional Nanogel: We developed a bacteria-responsive multifunctional nanogel for targeted antibiotic delivery, in which bacterial enzymes are utilized to trigger antibiotic release by degrading the polyphosphoester core. The mannosylated nanogel preferentially delivers drugs to macrophages and leads to drug accumulation at bacterial infection sites through macrophage transport. This nanogel provides macrophage targeting and lesion site-activatable drug release properties, which enhances bacterial growth inhibition.


ACS Nano | 2012

Single-step assembly of cationic lipid-polymer hybrid nanoparticles for systemic delivery of siRNA.

Xian-Zhu Yang; Shuang Dou; Yu-Cai Wang; Hong-Yan Long; Meng-Hua Xiong; Cheng-Qiong Mao; Yandan Yao; Jun Wang

The clinical success of therapeutics of small interfering RNA (siRNA) is still hindered by its delivery systems. Cationic polymer or lipid-based vehicles as the major delivery systems of siRNA cannot sufficiently satisfy siRNA therapeutic applications. It is hypothesized that cationic lipid-polymer hybrid nanoparticles may take advantage of both polymeric and lipid-based nanoparticles for siRNA delivery, while diminishing the shortcomings of both. In this study, cationic lipid-polymer hybrid nanoparticles were prepared by a single-step nanoprecipitation of a cationic lipid (N,N-bis(2-hydroxyethyl)-N-methyl-N-(2-cholesteryloxycarbonyl aminoethyl) ammonium bromide, BHEM-Chol) and amphiphilic polymers for systemic delivery of siRNA. The formed hybrid nanoparticles comprised a hydrophobic polylactide core, a hydrophilic poly(ethylene glycol) shell, and a cationic lipid monolayer at the interface of the core and the shell. Such hybrid nanoparticles exhibited excellent stability in serum and showed significantly improved biocompatibility compared to that of pure BHEM-Chol particles. The hybrid nanoparticles were capable of delivering siRNA into BT474 cells and facilitated the escape of loaded siRNA from the endosome into the cytoplasm. The hybrid nanoparticles carrying polo-like kinase 1 (Plk1)-specific siRNA (siPlk1) remarkably and specifically downregulated expression of the oncogene Plk1 and induced cancer cell apoptosis both in vitro and in vivo and significantly suppressed tumor growth following systemic administration. We demonstrate that this system is stable, nontoxic, highly efficient, and easy to scale up, bringing the clinical application of siRNA therapy one important step closer to reality.

Collaboration


Dive into the Xian-Zhu Yang's collaboration.

Top Co-Authors

Avatar

Jun Wang

South China University of Technology

View shared research outputs
Top Co-Authors

Avatar

Chun-Yang Sun

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Wei Tao

Hefei University of Technology

View shared research outputs
Top Co-Authors

Avatar

Yu-Cai Wang

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Hai-Sheng Qian

Hefei University of Technology

View shared research outputs
Top Co-Authors

Avatar

Jin-Zhi Du

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Yinchu Ma

Hefei University of Technology

View shared research outputs
Top Co-Authors

Avatar

Yan-Hua Zhu

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Cheng-Qiong Mao

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Jun-Xia Wang

Hefei University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge