Network


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

Hotspot


Dive into the research topics where Xiaoyu Xiang is active.

Publication


Featured researches published by Xiaoyu Xiang.


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.


Molecular Therapy | 2011

Treatment of Brain Inflammatory Diseases by Delivering Exosome Encapsulated Anti-inflammatory Drugs From the Nasal Region to the Brain

Xiaoying Zhuang; Xiaoyu Xiang; William E. Grizzle; Dongmei Sun; Shuangqin Zhang; Robert C. Axtell; Songwen Ju; Jiangyao Mu; Lifeng Zhang; Lawrence Steinman; Donald R. Miller; Huang-Ge Zhang

In this study, exosomes used to encapsulate curcumin (Exo-cur) or a signal transducer and activator of transcription 3 (Stat3) inhibitor, i.e., JSI124 (Exo-JSI124) were delivered noninvasively to microglia cells via an intranasal route. The results generated from three inflammation-mediated disease models, i.e., a lipopolysaccharide (LPS)-induced brain inflammation model, experimental autoimmune encephalitis and a GL26 brain tumor model, showed that mice treated intranasally with Exo-cur or Exo-JSI124 are protected from LPS-induced brain inflammation, the progression of myelin oligodendrocyte glycoprotein (MOG) peptide induced experimental autoimmune encephalomyelitis (EAE), and had significantly delayed brain tumor growth in the GL26 tumor model. Intranasal administration of Exo-cur or Exo-JSI124 led to rapid delivery of exosome encapsulated drug to the brain that was selectively taken up by microglial cells, and subsequently induced apoptosis of microglial cells. Our results demonstrate that this strategy may provide a noninvasive and novel therapeutic approach for treating brain inflammatory-related diseases.


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.


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 | 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.


Molecular Therapy | 2013

Grape Exosome-like Nanoparticles Induce Intestinal Stem Cells and Protect Mice From DSS-Induced Colitis

Songwen Ju; Jingyao Mu; Terje Dokland; Xiaoying Zhuang; Qilong Wang; Hong Jiang; Xiaoyu Xiang; Zhong-Bin Deng; Baomei Wang; Lifeng Zhang; Mary R. Roth; Ruth Welti; James A. Mobley; Yan Jun; Donald R. Miller; Huang-Ge Zhang

Food-derived exosome-like nanoparticles pass through the intestinal tract throughout our lives, but little is known about their impact or function. Here, as a proof of concept, we show that the cells targeted by grape exosome-like nanoparticles (GELNs) are intestinal stem cells whose responses underlie the GELN-mediated intestinal tissue remodeling and protection against dextran sulfate sodium (DSS)-induced colitis. This finding is further supported by the fact that coculturing of crypt or sorted Lgr5⁺ stem cells with GELNs markedly improved organoid formation. GELN lipids play a role in induction of Lgr5⁺ stem cells, and the liposome-like nanoparticles (LLNs) assembled with lipids from GELNs are required for in vivo targeting of intestinal stem cells. Blocking β-catenin-mediated signaling pathways of GELN recipient cells attenuates the production of Lgr5⁺ stem cells. Thus, GELNs not only modulate intestinal tissue renewal processes, but can participate in the remodeling of it in response to pathological triggers.


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.)


Oncogene | 2011

miR-155 promotes macroscopic tumor formation yet inhibits tumor dissemination from mammary fat pads to the lung by preventing EMT.

Xiaoyu Xiang; Zhuang X; Songwen Ju; Shuangyin Zhang; Hong Jiang; Jingyao Mu; Lifeng Zhang; Donald R. Miller; William E. Grizzle; Huang-Ge Zhang

A micro-RNA, miR-155, is overexpressed in many types of cancer cells, including breast cancer, and its role(s) in tumor metastasis has been studied on a very limited basis. Tumor metastasis is a multi-step process with the last step in the process being formation of macroscopic tumor in organs distant from the primary tumor site. This step is the least studied. Here, we report that stable expression of miR-155 in 4T1 breast tumor cells reduces significantly the aggressiveness of tumor cell dissemination as a result of preventing epithelial-to-mesenchymal transition (EMT) of tumor cells in vivo. Further, miR-155 directly suppresses the expression of the transcription factor TCF4, which is an important regulator of EMT. However, when tumor cells are injected directly into the bloodstream, miR-155 remarkably promotes macroscopic tumor formation in the lung. Analysis of gene expression profiling identified a group of genes that are associated with promoting macroscopic tumor formation in the lung. Importantly, most of these genes are overexpressed in epithelial cells. Our findings provide new insight into how miR-155 modulates the development of tumor metastasis. This study suggests that the location of tumor cells overexpressing miR-155 is a critical factor: in mammary fat pads miR-155 prevents tumor dissemination; whereas in the lung miR-155 apparently maintains the epithelial phenotype of tumor cells that is critical for macroscopic tumor formation.


American Journal of Pathology | 2010

TLR2-Mediated Expansion of MDSCs Is Dependent on the Source of Tumor Exosomes

Xiaoyu Xiang; Yuelong Liu; Zhuang X; Shuangqin Zhang; Sue Michalek; Douglas D. Taylor; William E. Grizzle; Huang-Ge Zhang

Exosomes released from tumor cells having been shown to induce interleukin-6 release from myeloid-derived suppressor cells in a Toll-like receptor 2/Stat3-dependent manner. In this study, we show that exosomes released from tumor cells re-isolated from syngeneic mice are capable of inducing interleukin-6 in a Toll-like receptor 2-independent manner, whereas the data generated from exosomes of tumor cells having undergone numerous in vitro passages induce interleukin-6 in a Toll-like receptor 2-dependent manner. This discrepancy may be due to the source of tumor cells used to generate the exosomes for this study. These results suggest that exosomes released from tumor cells that are not within a tumor microenvironment may not realistically represent the role of tumor exosomes in vivo. This is an important consideration since frequently passing tumor cells in vivo is an accepted practice for studying tumor exosome-mediated inflammatory responses.

Collaboration


Dive into the Xiaoyu Xiang's collaboration.

Top Co-Authors

Avatar

Huang-Ge Zhang

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhong-Bin Deng

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
Top Co-Authors

Avatar

Lifeng Zhang

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

Hong Jiang

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

Yuelong Liu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jingyao Mu

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

Jun Yan

University of Louisville

View shared research outputs
Researchain Logo
Decentralizing Knowledge