Network


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

Hotspot


Dive into the research topics where Dongdong Sun is active.

Publication


Featured researches published by Dongdong Sun.


Chemistry: A European Journal | 2012

Stabilization of G-Quadruplex DNA, Inhibition of Telomerase Activity and Live Cell Imaging Studies of Chiral Ruthenium(II) Complexes

Dongdong Sun; Yanan Liu; Du Liu; Rong Zhang; Xicheng Yang; Jie Liu

Telomerase inhibition is an attractive strategy for cancer chemotherapy. In the current study, we have synthesized and characterized two chiral ruthenium(II) complexes, namely, Λ-[Ru(phen)(2)(p-MOPIP)](2+) and Δ-[Ru(phen)(2)(p-MOPIP)](2+), where phen is 1,10-phenanthroline and p-MOPIP is 2-(4-methoxyphenyl)-imidazo[4,5f][1,10]phenanthroline. The chiral selectivity of the compounds and their ability to discriminate quadruplex DNA were investigated by using UV/Vis, fluorescence spectroscopy, circular dichroism spectroscopy, fluorescence resonance energy transfer melting assay, polymerase chain reaction stop assay and telomerase repeat amplification protocol. The results indicate that the two chiral compounds could induce and stabilize the formation of antiparallel G-quadruplexes of telomeric DNA in the presence or absence of metal cations. We report the remarkable ability of the two complexes Λ-[Ru(phen)(2)(p-MOPIP)](2+) and Δ-[Ru(phen)(2)(p-MOPIP)](2+) to stabilize selectively G-quadruplex DNA; the former is a better G-quadruplex binder than the latter. The anticancer activities of these complexes were evaluated by using the MTT assay. Interestingly, the antiproliferative activity of Λ-[Ru(phen)(2)(p-MOPIP)](2+) was higher than that of Δ-[Ru(phen)(2)(p-MOPIP)](2+), and Λ-[Ru(phen)(2)(p-MOPIP)](2+) showed a significant antitumor activity in HepG2 cells. The status of the nuclei in Λ/Δ-[Ru(phen)(2) (p-MOPIP)](2+)-treated HepG2 cells was investigated by using real-time living cell microscopy to determine the effects of Λ/Δ-[Ru(phen)(2)(p-MOPIP)](2+) on intracellular accumulation. The results show that Λ/Δ-[Ru(phen)(2)(p-MOPIP)](2+) can be taken up by HepG2 cells and can enter into the cytoplasm as well as accumulate in the nuclei; this suggests that the nuclei were the cellular targets of Λ/Δ-[Ru(phen)(2)(p-MOPIP)](2+).


Biomaterials | 2013

The effects of luminescent ruthenium(II) polypyridyl functionalized selenium nanoparticles on bFGF-induced angiogenesis and AKT/ERK signaling

Dongdong Sun; Yanan Liu; Qianqian Yu; Yanhui Zhou; Rong Zhang; Xiaojia Chen; An Hong; Jie Liu

Anti-angiogenesis is an effective strategy for cancer treatment because uncontrolled tumor growth depends on tumor angiogenesis and sufficient blood supply. Thus, blocking angiogenesis could be a strategy to arrest tumor growth. The function and mechanism of luminescent ruthenium-modified selenium nanoparticles (Ru-SeNPs) in angiogenesis have not been elucidated to date. Here, we found that Ru-SeNPs significantly inhibited human umbilical vascular endothelial cell (HUVEC) proliferation, migration and tube formation. Ru-SeNPs was also tested in vivo in the chicken chorioallantoic membrane (CAM) assay and found to inhibit bFGF-treated CAMs development like suramin. Moreover, we showed that Ru-SeNPs inhibited the activations of FGFR1 and its downstream protein kinases, such ErK and AKT. Furthermore, by using fluorescence confocal microscopy and TEM imaging studies, we have demonstrated their cellular uptake and localization within the cytoplasm of HepG2 and HUVEC cells. These findings indicate that Ru-SeNPs inhibits angiogenesis and may be a viable drug candidate in anti-angiogenesis and anticancer therapies.


Acta Biomaterialia | 2015

Multifunctional polyamidoamine-modified selenium nanoparticles dual-delivering siRNA and cisplatin to A549/DDP cells for reversal multidrug resistance.

Wenjing Zheng; Chengwen Cao; Yanan Liu; Qianqian Yu; Chuping Zheng; Dongdong Sun; Xiaofan Ren; Jie Liu

Multidrug resistance (MDR) is a major barrier against effective cancer treatment. Dual-delivering a therapeutic small interfering RNA (siRNA) and chemotherapeutic agents has been developed to reverse drug resistance in tumor cells. In this study, amine-terminated generation 5 polyamidoamine (PAMAM) dendrimers (G5.NH2)-modified selenium nanoparticles (G5@Se NP) were synthesized for the systemic dual-delivery of mdr1 siRNA and cisplatin (cis-diamminedichloroplatinum-(II), DDP), which was demonstrated to enhance siRNA loading, releasing efficiency and gene-silencing efficacy. When the mdr1 siRNA was conjugated with G5@Se NP via electrostatic interaction, a significant down-regulation of P-glycoprotein and multidrug resistance-associated protein expression was observed; G5@Se-DDP-siRNA arrested A549/DDP cells at G1 phase and led to enhanced cytotoxicity in A549/DDP cells through induction of apoptosis involving the AKT and ERK signaling pathways. Interestingly, G5@Se-DDP NP were much less reactive than DDP in the reactions with both MT and GSH, indicating that loading of DDP in a nano-delivery system could effectively prevent cell detoxification. Furthermore, animal studies demonstrated that the new delivery system of G5@Se-DDP-siRNA significantly enhanced the anti-tumor effect on tumor-bearing nude mice, with no appreciable abnormality in the major organs. These results suggest that G5@Se NP could be a potential platform to combine chemotherapy and gene therapy technology in the treatment of human disease.


Biomaterials | 2014

Inhibition of tumor growth and vasculature and fluorescence imaging using functiOnalized ruthenium-thiol protected selenium nanoparticles

Dongdong Sun; Yanan Liu; Qianqian Yu; Xiuying Qin; Licong Yang; Yanhui Zhou; Lanmei Chen; Jie Liu

Here we reported the high tumor targeting efficacy of luminescent Ru(II)-thiols protected selenium nanoparticles (Ru-MUA@Se). We have shown that a dual-target inhibitor Ru-MUA@Se directly suppress the tumor growth but also block blood-vessel growth. We also determined that the nanoparticles entered the cells via clathrin-mediated endocytosis pathway. In a xenograft HepG2 tumor model, we found that Ru-MUA@Se effectively inhibited tumor angiogenesis and suppressed tumor growth with low side effects using metronomic chemotherapy with Ru-MUA@Se. In vivo investigation of nanoparticles on nude mice bearing HepG2 cancer xenografts confirmed that Ru-MUA@Se nanoparticles possessed high tumor-targeted fluorescence imaging, exhibited enhanced antitumor efficacy and decreased systemic toxicity. Moreover, Ru-MUA@Se not only significantly induced dose-dependent disruption of mitochondrial membrane potential in HepG2 cells after 24 h treatment, but it also enhanced reactive oxygen species (ROS) generation. Our results suggest that the potential application of these Ru-MUA@Se nanoparticles in targeting cancer imaging and chemotherapy.


PLOS ONE | 2012

Chiral Ruthenium(II) Polypyridyl Complexes: Stabilization of G-Quadruplex DNA, Inhibition of Telomerase Activity and Cellular Uptake

Qianqian Yu; Yanan Liu; Chuan Wang; Dongdong Sun; Xingcheng Yang; Yanyu Liu; Jie Liu

Two ruthenium(II) complexes, Λ-[Ru(phen)2(p-HPIP)]2+ and Δ-[Ru(phen)2(p-HPIP)]2+, were synthesized and characterized via proton nuclear magnetic resonance spectroscopy, electrospray ionization-mass spectrometry, and circular dichroism spectroscopy. This study aims to clarify the anticancer effect of metal complexes as novel and potent telomerase inhibitors and cellular nucleus target drug. First, the chiral selectivity of the compounds and their ability to stabilize quadruplex DNA were studied via absorption and emission analyses, circular dichroism spectroscopy, fluorescence-resonance energy transfer melting assay, electrophoretic mobility shift assay, and polymerase chain reaction stop assay. The two chiral compounds selectively induced and stabilized the G-quadruplex of telomeric DNA with or without metal cations. These results provide new insights into the development of chiral anticancer agents for G-quadruplex DNA targeting. Telomerase repeat amplification protocol reveals the higher inhibitory activity of Λ-[Ru(phen)2(p-HPIP)]2+ against telomerase, suggesting that Λ-[Ru(phen)2(p-HPIP)]2+ may be a potential telomerase inhibitor for cancer chemotherapy. MTT assay results show that these chiral complexes have significant antitumor activities in HepG2 cells. More interestingly, cellular uptake and laser-scanning confocal microscopic studies reveal the efficient uptake of Λ-[Ru(phen)2(p-HPIP)]2+ by HepG2 cells. This complex then enters the cytoplasm and tends to accumulate in the nucleus. This nuclear penetration of the ruthenium complexes and their subsequent accumulation are associated with the chirality of the isomers as well as with the subtle environment of the ruthenium complexes. Therefore, the nucleus can be the cellular target of chiral ruthenium complexes for anticancer therapy.


ACS Applied Materials & Interfaces | 2014

Epigallocatechin-3-gallate (EGCG)-Stabilized Selenium Nanoparticles Coated with Tet-1 Peptide To Reduce Amyloid-β Aggregation and Cytotoxicity

Jingnan Zhang; Xianbo Zhou; Qianqian Yu; Licong Yang; Dongdong Sun; Yanhui Zhou; Jie Liu

Alzheimers disease (AD), the most common neurodegenerative disease, is caused by an accumulation of amyloid-β (Aβ) plaque deposits in the brains. Evidence is increasingly showing that epigallocatechin-3-gallate (EGCG) can partly protect cells from Aβ-mediated neurotoxicity by inhibiting Aβ aggregation. In order to better understand the process of Aβ aggregation and amyloid fibril disaggregation and reduce the cytotoxicity of EGCG at high doses, we attached EGCG onto the surface of selenium nanoparticles (EGCG@Se). Given the low delivery efficiency of EGCG@Se to the targeted cells and the involvement of selenoprotein in antioxidation and neuroprotection, which are the key factors for preventing the onset and progression of AD, we synthesized EGCG-stabilized selenium nanoparticles coated with Tet-1 peptide (Tet-1-EGCG@Se, a synthetic selenoprotein analogue), considering the affinity of Tet-1 peptide to neurons. We revealed that Tet-1-EGCG@Se can effectively inhibit Aβ fibrillation and disaggregate preformed Aβ fibrils into nontoxic aggregates. In addition, we found that both EGCG@Se and Tet-1-EGCG@Se can label Aβ fibrils with a high affinity, and Tet-1 peptides can significantly enhance the cellular uptake of Tet-1-EGCG@Se in PC12 cells rather than in NIH/3T3 cells.


Acta Biomaterialia | 2016

Investigation of functional selenium nanoparticles as potent antimicrobial agents against superbugs.

Xiaoquan Huang; Xu Chen; Qingchang Chen; Qianqian Yu; Dongdong Sun; Jie Liu

UNLABELLED Developing highly effective antibacterial agents is important for a wide range of applications. However, the emergence of multiple antibiotic-resistant bacteria poses a public health threat. Many developed agents have limited practical application due to chemical instability, low biocompatibility, and poor long-term antibacterial efficiency. In the following study, we synthesize a synergistic nanocomposite by conjugating quercetin (Qu) and acetylcholine (Ach) to the surface of Se nanoparticles (Qu-Ach@SeNPs). Quercetin has been reported to exhibit a wide range of biological activities related to their antibacterial activity and acetylcholine as a neurotransmitter, which can combine with the receptor on the bacterial cell. Arrows indicate NPs and arrowheads indicate compromised cell walls. The study demonstrated how Qu-Ach@SeNPs exhibit a synergistically enhanced antibacterial performance against the multidrug-resistant superbugs (MDRs) compared to Qu@SeNPs and Ach@SeNPs alone. Qu-Ach@SeNPs are effective against MDRs, such as Methicillin-resistant Staphylococcus aureus (MRSA), at a low dose. The mechanistic studies showed that Qu-Ach@SeNPs attach to the bacterial cell wall, causing irreversible damage to the membrane, and thereby achieving a remarkable synergistic antibacterial effect to inhibit MRSA. The findings suggested that the synergistic properties of quercetin and acetylcholine enhance the antibacterial activity of SeNPs. In this way, Qu-Ach@SeNPs comprise a new class of inorganic nano-antibacterial agents that can be used as useful applications in biomedical devices. STATEMENT OF SIGNIFICANCE The Qu-Ach@SeNPs have low cytotoxicity when tested on normal human cells in vitro. Qu-Ach@SeNPs are effective against MDRs, such as Methicillin-resistant S. aureus (MRSA), at a low dose. Importantly, Qu-Ach@SeNPs showed no emergence of resistance. These results suggest that Qu-Ach@SeNPs have excellent antibacterial activities. These agents can serve as good antibacterial agents against superbugs. Our data suggest that these antibacterial agents may have widespread application in the field of medicine for combating infectious diseases caused by MDRs, as well as other infectious diseases.


Journal of Materials Chemistry B | 2014

Se/Ru nanoparticles as inhibitors of metal-induced Aβ aggregation in Alzheimer's disease

Licong Yang; Qingchang Chen; Ying Liu; Jingnan Zhang; Dongdong Sun; Yanhui Zhou; Jie Liu

Amyloid β (Aβ) aggregates are considered as possible targets for therapy of Alzheimers disease (AD). Metal ions play an important role in amyloid aggregation and neurotoxicity in the AD pathogenesis. Disruption of the interactions between these metal ions and peptides holds considerable promise as a therapeutic strategy for AD treatment. In this study, l-Cys-modified Se/Ru nanoparticles (NPs) have been designed as Aβ-binding units to inhibit metal-induced Aβ aggregation. l-Cys was used as both the reducing agent and surface modifier in the formation of SeNPs, RuNPs and Se/RuNPs. We found that RuNPs and Se/RuNPs have a strong affinity toward Aβ species and efficiently suppress extracellular Aβ40 self-assembly and Zn2+-induced fibrillization. Also, Se/RuNPs can suppress the Zn2+-Aβ40 mediated generation of reactive oxygen species (ROS) and their corresponding neurotoxicity in PC12 cells. Intriguingly, SeNPs do not have the same ability as Se/RuNPs. In addition, Se/RuNPs also decrease intracellular Aβ40 fibrillization, but this process does not involve the lysosomal pathway. These results suggest that ruthenium significantly enhances the activity of Se/RuNPs binding to Aβ40. This interaction would block the Zn2+ binding to Aβ40 peptides and lower the concentration of the free monomer, thus decreasing fibrillization. Owing to this, Se/RuNPs may represent a new strategy in AD treatment.


Biometals | 2013

Ruthenium (II) polypyridyl complexes stabilize the bcl-2 promoter quadruplex and induce apoptosis of Hela tumor cells

Chuan Wang; Qianqian Yu; Licong Yang; Yanyu Liu; Dongdong Sun; Yongchao Huang; Yanhui Zhou; Qianling Zhang; Jie Liu

In the present study, the interaction between GC-rich sequence of bcl-2 gene P1 promoter (Pu39) and two ruthenium (II) polypyridyl complexes, [Ru(bpy)2(tip)]2+ (1) and [Ru(phen)2(tip)]2+ (2), was investigated by UV–Visible, fluorescence spectroscopy, circular dichroism, fluorescence resonance energy transfer melting assay and polymerase chain reaction stop assay. Those experimental results indicated that the two complexes can effectively stabilize the G-quadruplex of Pu39. It was found that the complex 2 exhibited greater cytotoxic activity than 1 against human Hela cells and can enter into Hela cells in a short period of time to effectively induce apoptosis of cells. Further experiments found that complexes 1 and 2 had as potent inhibitory effects on ECV-304 cell migration as suramin. Those noteworthy results provide new insights into the development of anticancer agents for targeting G-quadruplex DNA.


Chirality | 2012

Chiral ruthenium complexes induce apoptosis of tumor cell and interact with bovine serum albumin

Fang Yuan; Xiaojia Chen; Yanan Liu; Tingting Zhang; Dongdong Sun; Jie Liu

In this study, two isomeric ruthenium(II) complexes [Ru(bpy)(2)(p-mopip)](2+) (1) and [Ru(bpy)(2)(o-mopip)](2+) (2) (bpy = 2, 2-bipyridine; L: p-mopip = 2-(4-methoxylphenyl) imidazo [4,5-f][1,10]phenanthroline, o-mopip = 2-(2-methoxylphenyl) imidazo[4,5-f][1,10] phenan-throline) contained -OCH(3) at different positions on the phenyl ring and their enantiomers Λ-1, -2 and Δ-1, -2 displayed different properties. The cell viability of these ruthenium(II) complexes was evaluated by MTT, and complex Λ-1 has shown significant higher anticancer potency than Δ-1 against all the cell lines screened. Fluorescence microscopy and flow cytometric analyses demonstrated that complex Λ-1 was able to induce apoptosis. The interactions of complexes Λ-1, 1, and Δ-1 with bovine serum albumin (BSA) were investigated by fluorescence and circular dichroism (CD) measurements. The fluorescence quenching mechanism of BSA by complexes Λ-1, 1, and Δ-1 was determined to be a static process, and the apparent binding constant K(a) values is as follows: Λ-1 >1 > Δ-1. The number of binding sites n for all these complexes was 1. The result of CD showed that the secondary structure of BSA molecules was changed in the presence of the ruthenium(II) complex.

Collaboration


Dive into the Dongdong Sun's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge