Network


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

Hotspot


Dive into the research topics where Cunren Liu is active.

Publication


Featured researches published by Cunren Liu.


Molecular Therapy | 2010

A Novel Nanoparticle Drug Delivery System: The Anti-inflammatory Activity of Curcumin Is Enhanced When Encapsulated in Exosomes

Dongmei Sun; Xiaoying Zhuang; Xiaoyu Xiang; Yuelong Liu; Shuangyin Zhang; Cunren Liu; Stephen Barnes; William E. Grizzle; Donald R. Miller; Huang-Ge Zhang

Monocyte-derived myeloid cells play vital roles in inflammation-related autoimmune/inflammatory diseases and cancers. Here, we report that exosomes can deliver anti-inflammatory agents, such as curcumin, to activated myeloid cells in vivo. This technology provides a means for anti-inflammatory drugs, such as curcumin, to target the inflammatory cells as well as to overcome unwanted off-target effects that limit their utility. Using exosomes as a delivery vehicle, we provide evidence that curcumin delivered by exosomes is more stable and more highly concentrated in the blood. We show that the target specificity is determined by exosomes, and the improvement of curcumin activity is achieved by directing curcumin to inflammatory cells associated with therapeutic, but not toxic, effects. Furthermore, we validate the therapeutic relevance of this technique in a lipopolysaccharide (LPS)-induced septic shock mouse model. We further show that exosomes, but not lipid alone, are required for the enhanced anti-inflammatory activity of curcumin. The specificity of using exosomes as a drug carrier creates opportunities for treatments of many inflammation-related diseases without significant side effects due to innocent bystander or off-target effects.


Journal of Immunology | 2006

Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function.

Cunren Liu; Shaohua Yu; Kurt R. Zinn; Jianhua Wang; Liming Zhang; Yujiang Jia; John C. Kappes; Stephen Barnes; Robert P. Kimberly; William E. Grizzle; Huang-Ge Zhang

Many tumor cells shed specialized membrane vesicles known as exosomes. In this study, we show that pretreatment of mice with exosomes produced by TS/A or 4T.1 murine mammary tumor cells resulted in accelerated growth of implanted tumor cells in both syngeneic BALB/c mice and nude mice. As implanted TS/A tumor cells grew more rapidly in mice that had been depleted of NK cells, we analyzed the effects of the tumor-derived exosomes on NK cells. The tumor-derived exosomes inhibit NK cell cytotoxic activity ex vivo and in vitro as demonstrated by chromium release assays. The treatment of mice with TS/A tumor exosomes also led to a reduction in the percentages of NK cells, as determined by FACS analysis, in the lungs and spleens. Key features of NK cell activity were inhibited, including release of perforin but not granzyme B, as well as the expression of cyclin D3 and activation of the Jak3-mediated pathways. Human tumor cell lines also were found to produce exosomes that were capable of inhibiting IL-2-stimulated NK cell proliferation. Exosomes produced by dendritic cells or B cells did not. The presentation of tumor Ags by exosomes is under consideration as a cancer vaccine strategy; however, we found that pretreatment of mice with tumor exosomes blunted the protective effect of syngeneic dendritic cells pulsed ex vivo with tumor exosomes. We propose that tumor exosomes contribute to the growth of tumors by blocking IL-2-mediated activation of NK cells and their cytotoxic response to tumor cells.


International Journal of Cancer | 2009

Induction of myeloid-derived suppressor cells by tumor exosomes.

Xiaoyu Xiang; Anton Poliakov; Cunren Liu; Yuelong Liu; Zhong-Bin Deng; Jianhua Wang; Ziqiang Cheng; Spandan V. Shah; Gui-Jun Wang; Liming Zhang; William E. Grizzle; Jim Mobley; Huang-Ge Zhang

Myeloid‐derived suppressor cells (MDSCs) promote tumor progression. The mechanisms of MDSC development during tumor growth remain unknown. Tumor exosomes (T‐exosomes) have been implicated to play a role in immune regulation, however the role of exosomes in the induction of MDSCs is unclear. Our previous work demonstrated that exosomes isolated from tumor cells are taken up by bone marrow myeloid cells. Here, we extend those findings showing that exosomes isolated from T‐exosomes switch the differentiation pathway of these myeloid cells to the MDSC pathway (CD11b+Gr‐1+). The resulting cells exhibit MDSC phenotypic and functional characteristics including promotion of tumor growth. Furthermore, we demonstrated that in vivo MDSC mediated promotion of tumor progression is dependent on T‐exosome prostaglandin E2 (PGE2) and TGF‐β molecules. T‐exosomes can induce the accumulation of MDSCs expressing Cox2, IL‐6, VEGF, and arginase‐1. Antibodies against exosomal PGE2 and TGF‐β block the activity of these exosomes on MDSC induction and therefore attenuate MDSC‐mediated tumor‐promoting ability. Exosomal PGE2 and TGF‐β are enriched in T‐exosomes when compared with exosomes isolated from the supernatants of cultured tumor cells (C‐exosomes). The tumor microenvironment has an effect on the potency of T‐exosome mediated induction of MDSCs by regulating the sorting and the amount of exosomal PGE2 and TGF‐β available. Together, these findings lend themselves to developing specific targetable therapeutic strategies to reduce or eliminate MDSC‐induced immunosuppression and hence enhance host antitumor immunotherapy efficacy.


Journal of Immunology | 2007

Tumor Exosomes Inhibit Differentiation of Bone Marrow Dendritic Cells

Shaohua Yu; Cunren Liu; Kaihong Su; Jianhua Wang; Yuelong Liu; Liming Zhang; Chuanyu Li; Yingzi Cong; Robert P. Kimberly; William E. Grizzle; Carla I. Falkson; Huang-Ge Zhang

The production of exosomes by tumor cells has been implicated in tumor-associated immune suppression. In this study, we show that, in mice, exosomes produced by TS/A murine mammary tumor cells target CD11b+ myeloid precursors in the bone marrow (BM) in vivo, and that this is associated with an accumulation of myeloid precursors in the spleen. Moreover, we demonstrate that TS/A exosomes block the differentiation of murine myeloid precursor cells into dendritic cells (DC) in vitro. Addition of tumor exosomes at day 0 led to a significant block of differentiation into DC, whereas addition at later time points was less effective. Similarly, exosomes produced by human breast tumor cells inhibited the differentiation of human monocytes in vitro. The levels of IL-6 and phosphorylated Stat3 were elevated 12 h after the tumor exosome stimulation of murine myeloid precursors, and tumor exosomes were less effective in inhibiting differentiation of BM cells isolated from IL-6 knockout mice. Addition of a rIL-6 to the IL-6 knockout BM cell culture restored the tumor exosome-mediated inhibition of DC differentiation. These data suggest that tumor exosome-mediated induction of IL-6 plays a role in blocking BM DC differentiation.


American Journal of Pathology | 2010

Contribution of MyD88 to the Tumor Exosome-Mediated Induction of Myeloid Derived Suppressor Cells

Yuelong Liu; Xiaoyu Xiang; Xiaoying Zhuang; Shuangyin Zhang; Cunren Liu; Ziqiang Cheng; Sue Michalek; William E. Grizzle; Huang-Ge Zhang

In this study we observed that mice pretreated with tumor exosomes had a significant acceleration of tumor metastasis in the lung. Tumor metastasis correlated significantly with an increase in recruitment of more Myeloid-derived suppressor cells (MDSCs) in the lung of C57BL/6j (B6) mice pretreated with tumor exosomes. These effects were blunted when MyD88 knockout (KO) mice were pretreated with tumor exosomes. MDSCs induced by tumor exosomes and isolated from wild-type B6 mice also more potently inhibited T cell activation and induction of interleukin-6 and tumor necrosis factor-alpha than MDSCs isolated from the lung of MyD88 KO mice. In vitro, addition of tumor exosomes to bone marrow-derived CD11b(+)Gr-1(+) cells isolated from wild-type B6 mice resulted in more cytokine production, including tumor necrosis factor-alpha, interleukin-6, and the chemokine CCL2, than CD11b(+)Gr-1(+) cells isolated from MyD88 KO mice. Moreover, lower levels of CCL2 were observed in the lungs in MyD88 KO mice pretreated with tumor exosomes than that in wild-type mice. Together these data demonstrate a pivotal role for MyD88 in tumor exosome-mediated expansion of MDSCs and tumor metastasis.


Diabetes | 2009

Adipose Tissue Exosome-Like Vesicles Mediate Activation of Macrophage-Induced Insulin Resistance

Zhong-Bin Deng; Anton Poliakov; Robert W. Hardy; Ronald H. Clements; Cunren Liu; Yuelong Liu; Jianhua Wang; Xiaoyu Xiang; Shuangqin Zhang; Xiaoying Zhuang; Spandan V. Shah; Dongmei Sun; Sue Michalek; William E. Grizzle; Timothy W Garvey; Jim Mobley; Huang-Ge Zhang

OBJECTIVE We sought to determine whether exosome-like vesicles (ELVs) released from adipose tissue play a role in activation of macrophages and subsequent development of insulin resistance in a mouse model. RESEARCH DESIGN AND METHODS ELVs released from adipose tissue were purified by sucrose gradient centrifugation and labeled with green fluorescent dye and then intravenously injected into B6 ob/ob mice (obese model) or B6 mice fed a high-fat diet. The effects of injected ELVs on the activation of macrophages were determined through analysis of activation markers by fluorescence-activated cell sorter and induction of inflammatory cytokines using an ELISA. Glucose tolerance and insulin tolerance were also evaluated. Similarly, B6 mice with different gene knockouts including TLR2, TLR4, MyD88, and Toll-interleukin-1 receptor (TIR) domain–containing adaptor protein inducing interferon-β (TRIF) were also used for testing their responses to the injected ELVs. RESULTS ELVs are taken up by peripheral blood monocytes, which then differentiate into activated macrophages with increased secretion of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Injection of obELVs into wild-type C57BL/6 mice results in the development of insulin resistance. When the obELVs were intravenously injected into TLR4 knockout B6 mice, the levels of glucose intolerance and insulin resistance were much lower. RBP4 is enriched in the obELVs. Bone marrow–derived macrophages preincubated with recombinant RBP4 led to attenuation of obELV-mediated induction of IL-6 and TNF-α. CONCLUSIONS ELVs released by adipose tissue can act as a mode of communication between adipose tissues and macrophages. The obELV-mediated induction of TNF-α and IL-6 in macrophages and insulin resistance requires the TLR4/TRIF pathway.


Journal of Immunology | 2006

A Membrane Form of TNF-α Presented by Exosomes Delays T Cell Activation-Induced Cell Death

Huang-Ge Zhang; Cunren Liu; Kaihun Su; Shaohua Yu; Liming Zhang; Shuangqin Zhang; Jianhua Wang; Xu Cao; William E. Grizzle; Robert P. Kimberly

In common with many other cell types, synovial fibroblasts produce exosomes. In this study, we show that the exosomes produced by synovial fibroblasts obtained from individuals with rheumatoid arthritis (RASF), but not exosomes produced by synovial fibroblasts obtained from individuals with osteoarthritis, contain a membrane bound form of TNF-α as demonstrated by colloidal gold immunostaining of TNF-α and confirmed by both Western blot and mass spectrometry. The RASF-derived exosomes, but not exosomes derived from fibroblasts obtained from individuals with osteoarthritis, are cytotoxic for the L929 cell, a TNF-α-sensitive cell line, and stimulate activation of NF-κB and induction of collagenase-1 in RASF. These effects are blocked by addition of soluble TNFR1 (sTNFbp), suggesting that a TNF-α-signaling pathway mediates these biological activities. sTNFbp also reduced the production of exosomes by RASF, suggesting the interruption of a positive amplification loop. Exosomes can transmit signals between cells, and RASF exosomes, effectively taken up by anti-CD3-activated T cells, activated AKT and NF-κB and rendered these activated T cells resistant to apoptosis. Neutralization of exosomal membrane TNF-α by sTNFbp partially reversed this resistance, suggesting that not only TNF-α but also additional exosomal proteins may contribute to the development of apoptosis resistance.


Journal of Immunology | 2008

Thymus Exosomes-Like Particles Induce Regulatory T Cells

Gui-Jun Wang; Yuelong Liu; Aijian Qin; Spandan V. Shah; Zhong-Bin Deng; Xiaoyu Xiang; Ziqiang Cheng; Cunren Liu; Jianhua Wang; Liming Zhang; William E. Grizzle; Huang-Ge Zhang

Exosomes released from different types of cells have been proposed to contribute to intercellular communication. We report that thymic exosome-like particles (ELPs) released from cells of the thymus can induce the development of Foxp3+ regulatory T (Treg) cells in the lung and liver. Thymic ELPs also induce the conversion of thymic CD4+CD25− T cells into Tregs. Tregs induced by thymic ELPs suppress the proliferation of CD4+CD25− T cells in vitro and in vivo. We further show that neutralization of TGF-β in ELPs partially reverses thymic ELP-mediated induction of CD4+Foxp3+ T cells in the lung and liver. This study demonstrates that thymic ELPs participate in the induction of Foxp3+ Tregs. Also, TGF-β of thymic ELPs might be required for the generation of Tregs in the peripheral tissues.


Journal of Clinical Investigation | 2003

CII-DC-AdTRAIL cell gene therapy inhibits infiltration of CII-reactive T cells and CII-induced arthritis

Zhongyu Liu; Xin Xu; Hui-Chen Hsu; Albert Tousson; PingAr Yang; Qi Wu; Cunren Liu; Shaohua Yu; Huang-Ge Zhang; John D. Mountz

Previously, we described an APC-adenovirus (APC-Ad) FasL cell gene therapy method which could be used to deplete autoreactive T cells in vivo. FasL was toxic, however, and controlled regulation of FasL was not achieved. Here we describe an improved approach to delivering TNF-related apoptosis-inducing ligand (TRAIL) in vivo in which collagen II-induced (CII-induced) arthritis-susceptible (CIA-susceptible) DBA/1j mice were treated with CII-pulsed DCs that had been transfected with a novel Ad system. The Ad was engineered to exhibit inducible TRAIL under the control of the doxycycline-inducible (DOX-inducible) tetracycline response element (TRE). Four groups of mice were treated with CII-DC-AdTRAIL+DOX, CII-DC-AdTRAIL (no DOX), CII-DC-AdGFP+DOX, or DC-AdTRAIL+DOX (no CII), beginning 2 weeks after priming with CII in CFA. The incidence of arthritis and infiltration of T cells in the joint was significantly decreased in CII-DC-AdTRAIL+DOX-treated mice. The in vitro splenic T cell proliferative response and induction of IFN-gamma to bovine CII stimulation were also significantly reduced in mice treated with CII-DC-AdTRAIL+DOX. AdTRAIL+DOX was not toxic to DCs or mice but could induce activated T cells to undergo apoptosis in the spleen. Our results suggest that CII-DC-AdTRAIL+DOX cell gene therapy is a safe and effective method for inhibiting the development of CIA.


Hepatology | 2009

Immature myeloid cells induced by a high-fat diet contribute to liver inflammation.

Zhong-Bin Deng; Yuelong Liu; Cunren Liu; Xiaoyu Xiang; Jianhua Wang; Ziqiang Cheng; Spandan V. Shah; Shuangyin Zhang; Liming Zhang; Xiaoying Zhuang; Sue Michalek; William E. Grizzle; Huang-Ge Zhang

Chronic inflammation plays a critical role in promoting obesity‐related disorders, such as fatty liver disease. The inflammatory cells that mediate these effects remain unknown. This study investigated the accumulation of immature myeloid cells in the liver and their role in liver inflammation. We found that the accumulation of immature myeloid cells, i.e., CD11b+Ly6ChiLy6G− cells, in the liver of B6 mice fed a high‐fat diet contribute to liver inflammation. Adoptive transfer of CD11b+Ly6ChiLy6G− cells isolated from the liver of obese B6 mice, but not from lean B6 mice, resulted in liver damage that was evident by an increase in the activity of liver transferases in serum. CD11b+Ly6ChiLy6G− cells isolated from the liver of obese mice are more easily activated by way of Toll‐like receptor (TLR) stimulation resulting in interleukin 12 and other inflammatory cytokine expression in an MyD88‐dependent fashion. TLR7‐activated CD11b+Ly6ChiLy6G− cells also enhance liver natural killer T cell (NKT) death in an Fas‐dependent manner. Experiments using mice depleted of Gr‐1+ immature myeloid cells demonstrated the important role of CD11b+Ly6ChiLy6G− in liver inflammation. Repeated injection of exosome‐like particles causes CD11b+ cell activation and subsequent homing to and accumulation of the cells in the liver. Conclusion: Consumption of a high‐fat diet by B6 mice triggers an accumulation of immature myeloid cells in the liver. The immature myeloid cells release proinflammatory cytokines and induce NKT cell apoptosis. Activation‐induced NKT apoptosis further promotes excessive production of Th‐1 cytokines. This diet‐induced accumulation of immature myeloid cells may contribute to obesity‐related liver disease. (HEPATOLOGY 2009.)

Collaboration


Dive into the Cunren Liu's collaboration.

Top Co-Authors

Avatar

Huang-Ge Zhang

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

William E. Grizzle

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jianhua Wang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Shaohua Yu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Yuelong Liu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Liming Zhang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Xiaoyu Xiang

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

Robert P. Kimberly

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ziqiang Cheng

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge