Network


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

Hotspot


Dive into the research topics where Na Man is active.

Publication


Featured researches published by Na Man.


Autophagy | 2009

Autophagy-mediated chemosensitization in cancer cells by fullerene C60 nanocrystal

Qiang Zhang; Wenjun Yang; Na Man; Fang Zheng; Yuanyuan Shen; Kejing Sun; Yang Li; Longping Wen

Autophagy may represent a common cellular response to nanomaterials, and modulation of autophagy holds great promise for improving the efficacy of cancer therapy. Fullerene C60 possesses potent anti-cancer activities, but its considerable toxicity towards normal cells may hinder its practical applications. It has been reported that fullerene C60 induces certain hallmarks of autophagy in cancer cells. Here we show that the water-dispersed nanocrystal of underivatized fullerene C60 (Nano-C60) at non-cytotoxic concentrations caused authentic autophagy and sensitized chemotherapeutic killing of both normal and drug-resistant cancer cells in a reactive oxygen species (ROS)-dependent and photo-enhanced fashion. We further demonstrated that the chemosensitization effect of Nano-C60 was autophagy-mediated and required a functional Atg5, a key gene in the autophagy signaling pathway. Our results revealed a novel biological function for Nano-C60 in enhancing the cytotoxic action of chemotherapeutic agents through autophagy modulation and may point to the potential application of Nano-C60 in adjunct chemotherapy.


Nanotechnology | 2010

C60(Nd) nanoparticles enhance chemotherapeutic susceptibility of cancer cells by modulation of autophagy

Pengfei Wei; Li Zhang; Yang Lu; Na Man; Longping Wen

Autophagy, an evolutionally conserved intracellular process degrading cytoplasmic proteins and organelles for recycling, has become one of the most remarkable strategies applied in cancer research. The fullerene C60 nanoparticle (nC60) has been shown to induce autophagy and sensitize chemotherapeutic killing of cancer cells, but the details still remain unknown. Here we show that a water-dispersed nanoparticle solution of derivatized fullerene C60, C60(Nd) nanoparticles (nC60(Nd)), has greater potential in inducing autophagy and sensitizing chemotherapeutic killing of both normal and drug-resistant cancer cells than nC60 does in an autophagy-dependent fashion. Additionally we further demonstrated that autophagy induced by nC60/C60(Nd) and Rapamycin had completely different roles in cancer chemotherapy. Our results, for the first time, revealed a novel and more potent derivative of the C60 nanoparticle in enhancing the cytotoxicity of chemotherapeutic agents and reducing drug resistance through autophagy modulation, which may ultimately lead to novel therapeutic strategies in cancer therapy.


Cell Death & Differentiation | 2010

Omi/HtrA2 is a positive regulator of autophagy that facilitates the degradation of mutant proteins involved in neurodegenerative diseases

Bin Li; Q Hu; H Wang; Na Man; Haigang Ren; Longping Wen; Nobuyuki Nukina; Erkang Fei; Guanghui Wang

Omi, also known as high temperature requirement factor A2 (HtrA2), is a serine protease that was originally identified as a proapoptotic protein. Like Smac/Diablo, it antagonizes inhibitor of apoptosis proteins when released into the cytosol on apoptotic stimulation. Loss of its protease activity in mnd2 (motor neuron degeneration 2) mice is associated with neurodegeneration. However, the detailed mechanisms by which Omi regulates the pathogenesis of neurodegenerative disease remain largely unknown. We report here that Omi participates in the pivotal cellular degradation process known as autophagy. It activates autophagy through digestion of Hax-1, a Bcl-2 family-related protein that represses autophagy in a Beclin-1 (mammalian homologue of yeast ATG6)-dependent pathway. Moreover, Omi-induced autophagy facilitates the degradation of neurodegenerative proteins such as pathogenic A53T α-synuclein and truncated polyglutamine-expanded huntingtin, as well as the endogenous autophagy substrate p62. Knockdown of Omi decreases the basal level of autophagy and increases the level of the above target proteins. Furthermore, S276C Omi, the protease-defective mutant found in mnd2 mice, fails to regulate autophagy. Increased autophagy substrates and the formation of aggregate structures are observed in the brains of mnd2 mice. These results identify Omi as a novel regulator of autophagy and suggest that Omi might be important in the cellular quality control of proteins involved in neurodegenerative diseases.


Autophagy | 2010

Induction of genuine autophagy by cationic lipids in mammalian cells.

Na Man; Yong Chen; Fang Zheng; Wei Zhou; Longping Wen

Autophagy is a cellular stress response that results in the activation of a lysosomal degradation pathway. In this report, we showed that cationic lipids, a common-used class of transfection reagents, induced genuine autophagy in mammalian cells. Extensive LC3 dot formation was observed by treatment with cationic lipids (with or without DNA), but not neutral lipids, in a HeLa cell line stably expressing GFP-LC3 (HeLa-LC3). Further proofs for autophagy were obtained by the co-localization of the LC3 dots with lysosome-specific staining patterns, observation of LC3-I to LC3-II form conversion and appearance of autophagic vacuoles under TEM. The autophagic flux assay with bafilomycin A1 and degradation of p62/SQSTM1 suggested that the autophagy induced by cationic lipids was primarily due to increased formation of autophagosomes and not decreased turnover. Moreover, cationic lipids induced autophagy in an mTOR-independent manner.


Biomaterials | 2014

Accelerating the clearance of mutant huntingtin protein aggregates through autophagy induction by europium hydroxide nanorods

Pengfei Wei; Li Zhang; Susheel Kumar Nethi; Ayan Kumar Barui; Jun Lin; Wei Zhou; Yi Shen; Na Man; Yunjiao Zhang; Jing Xu; Chitta Ranjan Patra; Longping Wen

Autophagy is one of the well-known pathways to accelerate the clearance of protein aggregates, which contributes to the therapy of neurodegenerative diseases. Although there are numerous reports that demonstrate the induction of autophagy with small molecules including rapamycin, trehalose and lithium, however, there are few reports mentioning the clearance of aggregate-prone proteins through autophagy induction by nanoparticles. In the present article, we have demonstrated that europium hydroxide [Eu(III)(OH)3] nanorods can reduce huntingtin protein aggregation (EGFP-tagged huntingtin protein with 74 polyQ repeats), responsible for neurodegenerative diseases. Again, we have found that these nanorods induce authentic autophagy flux in different cell lines (Neuro 2a, PC12 and HeLa cells) through the expression of higher levels of characteristic autophagy marker protein LC3-II and degradation of selective autophagy substrate/cargo receptor p62/SQSTM1. Furthermore, depression of protein aggregation clearance through the autophagy blockade has also been observed by using specific inhibitors (wortmannin and chloroquine), indicating that autophagy is involved in the degradation of huntingtin protein aggregation. Since [Eu(III)(OH)3] nanorods can enhance the degradation of huntingtin protein aggregation via autophagy induction, we strongly believe that these nanorods would be useful for the development of therapeutic treatment strategies for various neurodegenerative diseases in near future using nanomedicine approach.


International Journal of Cancer | 2011

Autophagy-mediated chemosensitization by cysteamine in cancer cells.

Xiaomei Wan; Fang Zheng; Li Zhang; Yanyan Miao; Na Man; Longping Wen

Cysteamine (CS) has many biomedical and clinical applications because of its excellent water solubility, low cytotoxicity and good biocompatibility. A previous study by Brawer et al. reported the occurrence of many Gomori inclusion bodies in CS‐treated astrocytes, which would suggest the induction of autophagy. Here we provided a comprehensive line of evidence demonstrating that CS caused autophagosome accumulation in cancer cells. CS exerted a biphasic effect on the autophagy process, increasing the formation of autophagosomes in the early phase and blocking the autophagic degradation in a later phase. Furthermore, we showed that CS sensitized doxorubicin‐elicited chemotherapeutic killing in HeLa, B16 melanoma and doxorubicin‐resistant MCF‐7 cells and also enhanced chemotherapeutic efficacy of doxorubicin in a mouse melanoma model. Finally, we demonstrated that the chemosensitizing effect of CS was at least partly dependent on its ability to modulate autophagy. Our results revealed a novel biological function for CS in enhancing the chemotherapeutic effect of doxorubicin through autophagy modulation and pointed to the potential use of CS in adjunct cancer chemotherapy.


Autophagy | 2010

Rare earth oxide nanocrystals as a new class of autophagy inducers.

Na Man; Le Yu; Shu-Hong Yu; Longping Wen

Functional interaction of nanomaterial with autophagy, a fundamental biological process for cellular degradation, is of great interest to nanobiology. Rare earth nanomaterials hold tremendous potential for a variety of diagnostic and therapeutic applications and have also been reported to protect cells against bacterial, viral and oxidative stress. In a brief communication we report that both light and heavy classes of rare earth oxide nanocrystals (REOs) elicit an autophagic response in HeLa cells, a human cancer cell line, in a dose- and time-dependent manner. The autophagy induced by REOs is complete and is accompanied by vacuolization within the cytoplasm. Autophagy induction may help explain some of the biological effects caused by REOs, and at the same time raises a special point of consideration for this type of nanomaterial, with regard to both safety assessment and application exploration.


ACS Nano | 2014

Nanoparticle as Signaling Protein Mimic: Robust Structural and Functional Modulation of CaMKII upon Specific Binding to Fullerene C60 Nanocrystals

Yanyan Miao; Jing Xu; Yi Shen; Liang Chen; Yunpeng Bian; Yi Hu; Wei Zhou; Fang Zheng; Na Man; Yuanyuan Shen; Yunjiao Zhang; Ming Wang; Longping Wen

In a biological environment, nanoparticles encounter and interact with thousands of proteins, forming a protein corona on the surface of the nanoparticles, but these interactions are oftentimes perceived as nonspecific protein adsorption, with protein unfolding and deactivation as the most likely consequences. The potential of a nanoparticle-protein interaction to mimic a protein-protein interaction in a cellular signaling process, characterized by stringent binding specificity and robust functional modulation for the interacting protein, has not been adequately demonstrated. Here, we show that water-suspended fullerene C60 nanocrystals (nano-C60) interact with and modulate the function of the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), a multimeric intracellular serine/threonine kinase central to Ca(2+) signal transduction, in a fashion that rivals the well-documented interaction between the NMDA (N-methyl-d-aspartate) receptor subunit NR2B protein and CaMKII. The stable high-affinity binding of CaMKII to distinct sites on nano-C60, mediated by amino acid residues D246 and K250 within the catalytic domain of CaMKIIα, but not the nonspecific adsorption of CaMKII to diamond nanoparticles, leads to functional consequences reminiscent of the NR2B-CaMKII interaction, including generation of autonomous CaMKII activity after Ca(2+) withdrawal, calmodulin trapping and CaMKII translocation to postsynaptic sites. Our results underscore the critical importance of specific interactions between nanoparticles and cellular signaling proteins, and the ability of nano-C60 to sustain the autonomous kinase activity of CaMKII may have significant implications for both the biosafety and the potential therapeutic applications of fullerene C60.


Advanced Healthcare Materials | 2017

Inhibition of Kupffer Cell Autophagy Abrogates Nanoparticle-Induced Liver Injury

Shasha Zhu; Jiqian Zhang; Li Zhang; Wentao Ma; Na Man; Yiming Liu; Wei Zhou; Jun Lin; Pengfei Wei; Peipei Jin; Yunjiao Zhang; Yi Hu; Erwei Gu; Xianfu Lu; Zhilai Yang; Xuesheng Liu; Li Bai; Longping Wen

The possible adverse effects of engineered nanomaterials on human health raise increasing concern as our research on nanosafety intensifies. Upon entry into a human body, whether intended for a theranostic purpose or through unintended exposure, nanomaterials tend to accumulate in the liver, leading to hepatic damage. A variety of nanoparticles, including rare earth upconversion nanoparticles (UCNs), have been reported to elicit hepatotoxicity, in most cases through inducing immune response or activating reactive oxygen species. Many of these nanoparticles also induce autophagy, and autophagy inhibition has been shown to decrease UCN-induced liver damage. Herein, using UCNs as a model engineered nanomaterial, this study uncovers a critical role for Kupffer cells in nanomaterial-induced liver toxicity, as depletion of Kupffer cells significantly exacerbates UCN-induced liver injury. Furthermore, UCN-induced prodeath autophagy in Kupffer cells, and inhibition of autophagy with 3-MA, a well-established chemical inhibitor of autophagy, enhances Kupffer cell survival and further abrogates UCN-induced liver toxicity. The results reveal the critical importance of Kupffer cell autophagy for nanoparticle-induced liver damage, and inhibition of autophagy may constitute a novel strategy for abrogating nanomaterial-elicited liver toxicity.


Free Radical Research | 2012

Hoechst 33342-induced autophagy protected HeLa cells from caspase-independent cell death with the participation of ROS

Fang Zheng; Wenjun Yang; Kejing Sun; Xiao-Mei Wan; Na Man; Longping Wen

Abstract Autophagy, an evolutionarily-conserved intracellular organelle and protein degradation process, may exhibit drastically different effects on cell survival depending on the particular environmental and culturing conditions. Hoechst 33342 (HO), a fluorescent dye widely used for staining DNA, has been reported to induce apoptosis in mammalian cells. Here we showed that, in addition to caspase-independent cell death, HO also induced autophagy in HeLa cells, as evidenced by the accumulation of autophagosomes, LC3 form conversion and LC3 puncta formation in a cell line stably expressing GFP-LC3. HO treatment led to generation of reactive oxygen species (ROS), and inhibition of ROS with N-acetyl-l-cysteine (NAC) abrogated both autophagy and caspase-independent cell death. Finally, autophagy played a protective role against caspase-independent cell death, as cell death induced by HO was enhanced under pharmacological and siRNA-mediated genetic inhibition of autophagy.

Collaboration


Dive into the Na Man's collaboration.

Top Co-Authors

Avatar

Longping Wen

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Li Zhang

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Fang Zheng

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Wei Zhou

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Pengfei Wei

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Yunjiao Zhang

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Jun Lin

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Le Yu

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Peipei Jin

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Shu-Hong Yu

University of Science and Technology of China

View shared research outputs
Researchain Logo
Decentralizing Knowledge