Network


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

Hotspot


Dive into the research topics where Baoying Liu is active.

Publication


Featured researches published by Baoying Liu.


Journal of Immunology | 2005

Cutting Edge: In Vivo Blockade of Human IL-2 Receptor Induces Expansion of CD56bright Regulatory NK Cells in Patients with Active Uveitis

Zhuqing Li; Wee Kiak Lim; Sankaranarayana P. Mahesh; Baoying Liu; Robert B. Nussenblatt

In vivo blockade of the human IL-2R by mAb has been used for immunosuppression in transplantation, therapy for leukemia, and autoimmune diseases. In this study, we report that administration of a humanized IL-2R blocking Ab induced a 4- to 20-fold expansion of CD56bright regulatory NK cells in uveitis patients over time. The induced CD56bright regulatory NK cells from patients exhibited similar phenotype as those naturally occurring CD56bright cells. Patients with active uveitis had a significantly lower level of CD56bright NK cells compared with normal donors (p < 0.01). In addition, the induced CD56bright cells could secrete large amounts of IL-10 whereas CD56dim NK cells could not, suggesting that the induction of the CD56bright cells may have a beneficial effect on the remission of active uveitis. Our observation may have implications to IL-2R blockade therapy and for the potential role of CD56bright regulatory NK cells in autoimmune diseases.


Journal of Translational Medicine | 2011

Complement component C5a Promotes Expression of IL-22 and IL-17 from Human T cells and its Implication in Age-related Macular Degeneration

Baoying Liu; Lai Wei; Catherine B. Meyerle; Jingsheng Tuo; H. Nida Sen; Zhiyu Li; Sagarika Chakrabarty; Elvira Agrón; Chi-Chao Chan; Michael L. Klein; Emily H Chew; Frederick L. Ferris; Robert B. Nussenblatt

BackgroundAge related macular degeneration (AMD) is the leading cause of irreversible blindness in elderly populations worldwide. Inflammation, among many factors, has been suggested to play an important role in AMD pathogenesis. Recent studies have demonstrated a strong genetic association between AMD and complement factor H (CFH), the down-regulatory factor of complement activation. Elevated levels of complement activating molecules including complement component 5a (C5a) have been found in the serum of AMD patients. Our aim is to study whether C5a can impact human T cells and its implication in AMD.MethodsHuman peripheral blood mononuclear cells (PBMCs) were isolated from the blood of exudative form of AMD patients using a Ficoll gradient centrifugation protocol. Intracellular staining and enzyme-linked immunosorbent assays were used to measure protein expression. Apoptotic cells were detected by staining of cells with the annexin-V and TUNEL technology and analyzed by a FACS Caliber flow cytometer. SNP genotyping was analyzed by TaqMan genotyping assay using the Real-time PCR system 7500.ResultsWe show that C5a promotes interleukin (IL)-22 and IL-17 expression by human CD4+ T cells. This effect is dependent on B7, IL-1β and IL-6 expression from monocytes. We have also found that C5a could protect human CD4+ cells from undergoing apoptosis. Importantly, consistent with a role of C5a in promoting IL-22 and IL-17 expression, significant elevation in IL-22 and IL-17 levels was found in AMD patients as compared to non-AMD controls.ConclusionsOur results support the notion that C5a may be one of the factors contributing to the elevated serum IL-22 and IL-17 levels in AMD patients. The possible involvement of IL-22 and IL-17 in the inflammation that contributes to AMD may herald a new approach to treat AMD.


Cell Reports | 2012

Hypomethylation of the IL17RC promoter associates with age-related macular degeneration.

Lai Wei; Baoying Liu; Jingsheng Tuo; Defen Shen; Ping Chen; Zhiyu Li; X. Liu; Jia Ni; Pradeep K. Dagur; H. Nida Sen; Shayma Jawad; Diamond Ling; Stanley Park; Sagarika Chakrabarty; Catherine B. Meyerle; Elvira Agrón; Frederick L. Ferris; Emily Y. Chew; J. Philip McCoy; Emily D. Blum; Peter J. Francis; Michael L. Klein; Robyn H. Guymer; Paul N. Baird; Chi-Chao Chan; Robert B. Nussenblatt

Age-related macular degeneration (AMD) is the leading cause of irreversible blindness in the elderly population worldwide. Although recent studies have demonstrated strong genetic associations between AMD and SNPs in a number of genes, other modes of regulation are also likely to play a role in the etiology of this disease. We identified a significantly decreased level of methylation on the IL17RC promoter in AMD patients. Furthermore, we showed that hypomethylation of the IL17RC promoter in AMD patients led to an elevated expression of its protein and messenger RNA in peripheral blood as well as in the affected retina and choroid, suggesting that the DNA methylation pattern and expression of IL17RC may potentially serve as a biomarker for the diagnosis of AMD and likely plays a role in disease pathogenesis.


Retina-the Journal of Retinal and Vitreous Diseases | 2010

A RANDOMIZED PILOT STUDY OF SYSTEMIC IMMUNOSUPPRESSION IN THE TREATMENT OF AGE-RELATED MACULAR DEGENERATION WITH CHOROIDAL NEOVASCULARIZATION

Robert B. Nussenblatt; Gordon Byrnes; H. Nida Sen; Steven Yeh; Lisa J. Faia; Catherine B. Meyerle; Keith K. Wroblewski; Zhuqing Li; Baoying Liu; Emily Y. Chew; Patti Sherry; Penelope L. Friedman; Fred Gill; Frederick L. Ferris

Background: Age-related macular degeneration remains the leading cause of irreversible blindness in the United States and the developed world. Intravitreal injections of anti-vascular endothelial growth factor (VEGF) medications have become standard of care for the treatment of the wet form of the disease. Recent reports have demonstrated an association with various immune factors. We aimed to investigate the effect of immunosuppressive therapy in the clinical course of the wet form of the disease. We compared anti-VEGF therapy plus one of three systemic immunosuppressive therapies versus anti-VEGF therapy alone for recurrent choroidal neovascularization associated with age-related macular degeneration. Methods: This was a pilot, Phase I/II, prospective, randomized, unmasked, single-center trial. Patients with subretinal exudation secondary to recurrent choroidal neovascularization associated with age-related macular degeneration were included in the study. Patients were randomized to 1 of 3 systemic arms immunosuppressive agents (daclizumab, rapamycin, or infliximab) for 6 months plus intraocular anti-VEGF therapy if indicated, compared with a group who received only anti-VEGF therapy if indicated. Results: The number of anti-VEGF injections per group, visual acuity, retinal thickness, and safety measures were assessed in all groups. Thirteen patients were randomized; comparing anti-VEGF injections before and during the study, a decrease in the number of injections from 0.73 injections per month to 0.42 for daclizumab and from 0.67 to 0.34 for sirolimus was seen, while no apparent decrease was seen for either infliximab or observation. Visual acuities were maintained in all groups. Conclusion: These preliminary data suggest that some immunosuppressive agents given systemically can alter the clinical course of the wet form of the disease and support the notion that more definitive clinical trials of immune mediation of age-related macular degeneration are indicated.


Journal of Immunology | 2008

Gene Expression Profiling in Autoimmune Noninfectious Uveitis Disease

Zhuqing Li; Baoying Liu; Arvydas Maminishkis; Sankaranarayana P. Mahesh; Yeh S; Lew J; Lim Wk; Sen Hn; Clarke G; Ronald R. Buggage; Miller Ss; Robert B. Nussenblatt

Noninfectious uveitis is a predominantly T cell-mediated autoimmune, intraocular inflammatory disease. To characterize the gene expression profile from patients with noninfectious uveitis, PBMCs were isolated from 50 patients with clinically characterized noninfectious uveitis syndrome. A pathway-specific cDNA microarray was used for gene expression profiling and real-time PCR array for further confirmation. Sixty-seven inflammation- and autoimmune-associated genes were found differentially expressed in uveitis patients, with 28 of those genes being validated by real-time PCR. Several genes previously unknown for autoimmune uveitis, including IL-22, IL-19, IL-20, and IL-25/IL-17E, were found to be highly expressed among uveitis patients compared with the normal subjects with IL-22 expression highly variable among the patients. Furthermore, we show that IL-22 can affect primary human retinal pigment epithelial cells by decreasing total tissue resistance and inducing apoptosis possibly by decreasing phospho-Bad level. In addition, the microarray data identified a possible uveitis-associated gene expression pattern, showed distinct gene expression profiles in patients during periods of clinical activity and quiescence, and demonstrated similar expression patterns in related patients with similar clinical phenotypes. Our data provide the first evidence that a subset of IL-10 family genes are implicated in noninfectious uveitis and that IL-22 can affect human retinal pigment epithelial cells. The results may facilitate further understanding of the molecular mechanisms of autoimmune uveitis and other autoimmune originated inflammatory diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2010

PDGF-CC blockade inhibits pathological angiogenesis by acting on multiple cellular and molecular targets

Xu Hou; Anil Kumar; Chunsik Lee; Bin Wang; Pachiappan Arjunan; Lijin Dong; Arvydas Maminishkis; Zhongshu Tang; Yang Li; Fan Zhang; Shi-Zhuang Zhang; Piotr Wardega; Sagarika Chakrabarty; Baoying Liu; Zhijian Wu; Peter Colosi; Robert N. Fariss; Johan Lennartsson; Robert B. Nussenblatt; J. Silvio Gutkind; Yihai Cao; Xuri Li

The importance of identifying VEGF-independent pathways in pathological angiogenesis is increasingly recognized as a result of the emerging drug resistance to anti-VEGF therapies. PDGF-CC is the third member of the PDGF family discovered after more than two decades of studies on PDGF-AA and PDGF-BB. The biological function of PDGF-CC and the underlying cellular and molecular mechanisms remain largely unexplored. Here, using different animal models, we report that PDGF-CC inhibition by neutralizing antibody, shRNA, or genetic deletion suppressed both choroidal and retinal neovascularization. Importantly, we revealed that PDGF-CC targeting acted not only on multiple cell types important for pathological angiogenesis, such as vascular mural and endothelial cells, macrophages, choroidal fibroblasts and retinal pigment epithelial cells, but also on the expression of other important angiogenic genes, such as PDGF-BB and PDGF receptors. At a molecular level, we found that PDGF-CC regulated glycogen synthase kinase (GSK)–3β phosphorylation and expression both in vitro and in vivo. Activation of GSK3β impaired PDGF-CC–induced angiogenesis, and inhibition of GSK3β abolished the antiangiogenic effect of PDGF-CC blockade. Thus, we identified PDGF-CC as an important candidate target gene for antiangiogenic therapy, and PDGF-CC inhibition may be of therapeutic value in treating neovascular diseases.


Journal of Biological Chemistry | 2008

Glucocorticoid-induced tumor necrosis factor receptor negatively regulates activation of human primary natural killer (NK) cells by blocking proliferative signals and increasing NK cell apoptosis.

Baoying Liu; Zhuqing Li; Sankaranarayana P. Mahesh; Seth M. Pantanelli; Frank S. Hwang; Willie O. Siu; Robert B. Nussenblatt

Glucocorticoid-induced tumor necrosis factor receptor (GITR), found constitutively expressed on human primary natural killer (NK) cells at low levels was up-regulated upon stimulation by either Toll-like receptor ligand or NK cell growth factor, interleukin (IL)-15. cDNA microarray analysis showed that engagement of GITR primarily suppressed the activation of NF-KB pathway of NK cells and up-regulated anti-inflammatory genes heme oxygenase-1 and IL-10. Further analysis revealed that GITR activation suppressed NK cell proliferation in response to IL-15. GITR activation also suppressed proinflammatory cytokine secretion and increased NK cell apoptosis. GITR activation resulted in blocked phosphorylation of Stat5 and Akt, which may have contributed to the observed antiproliferative effect of GITR on NK cells. Increased apoptosis was independent of the Fas-FasL pathway, but Bcl-XL and phospho-Bad protein expressions were diminished, suggesting involvement of the mitochondrial apoptosis pathway. The results suggest that although GITR is an activation marker for NK cells similar to that for T cells, GITR serves as a negative regulator for NK cell activation. Our studies demonstrate a novel physiological role of GITR on NK cells.


European Journal of Immunology | 2006

Expression of GITR ligand abrogates immunosuppressive function of ocular tissue and differentially modulates inflammatory cytokines and chemokines

Sankaranarayana P. Mahesh; Zhuqing Li; Baoying Liu; Robert N. Fariss; Robert B. Nussenblatt

The glucocorticoid‐induced TNF‐related receptor ligand (GITRL) was previously shown to be constitutively expressed at low levels in human eye, including retinal pigment epithelial (RPE) cells. By expressing enhanced yellow fluorescent protein‐tagged human GITRL in human RPE cells, we investigated the significance of expression of GITRL on human ocular tissue. Confocal immunofluorescence microscopy and flow cytometry confirmed the surface expression of GITRL on RPE cells. However, a soluble form of GITRL was also detected. Remarkably, expression of GITRL on the RPE cells abrogated RPE‐mediated immunosuppression of CD3+ T cells, implicated as a possible mechanism for ocular immune privilege. This abrogation of immunosuppression by GITRL‐RPE was dependent on GITR‐GITRL interaction and could not be mimicked by anti‐CD28 antibody. Analysis of cytokine profiles revealed high level of TGF‐beta during the immunosuppression by RPE cells while expression of GITRL abrogated the RPE cell‐induced TGF‐beta secretion. Expression of GITRL also stimulates secretion of an array of proinflammatory cytokines/chemokines from T cells. GITR‐GITRL interaction provides a unique proinflammatory costimulation that may signal through a different pathway than that of CD28‐B7 costimulation. This study implicated that GITRL could be a potential candidate for regulation of the ocular immune privilege and the balance between immune privilege and inflammation.


Investigative Ophthalmology & Visual Science | 2009

Biologically active fibronectin fragments stimulate release of MCP-1 and catabolic cytokines from murine retinal pigment epithelium.

Bobbie Ann Austin; Baoying Liu; Zhuqing Li; Robert B. Nussenblatt

PURPOSE High-temperature requirement serine protease (HTRA1) was identified as a candidate age-related macular degeneration gene in multiple genetic studies in humans. To date, no functional studies have shown a mechanism for HTRA1 to instigate ocular tissue abnormalities. In the present study, the authors focused on a substrate of HTRA1, fibronectin, because fibronectin fragments (Fnfs) stimulate biochemical events in other age-related degenerative diseases that are analogous to changes associated with age-related macular degeneration (AMD). The purpose of the study was to determine whether Fnfs stimulate the release of proinflammatory and catabolic cytokines from murine retinal pigment epithelium (RPE). METHODS Fibronectin was purified from murine serum by gelatin cross-linked agarose chromatography and subsequently was enzymatically digested with alpha-chymotrypsin. The bioactivity of Fnfs was verified by measuring levels of IL-6 and TNF-alpha in Fnf-exposed murine splenocytes. To analyze the effect of Fnfs on RPE, cytokine and chemokine levels in RPE culture supernatants were assayed by ELISA. RESULTS IL-6 and TNF-alpha proinflammatory cytokines were released from primary murine splenocytes in proportion to the dose and length of Fnf treatment, indicating that alpha-chymotryptic digests of fibronectin are biologically active. Fnf treatment of murine RPE cells stimulated the release of microgram and nanogram levels of IL-6, MMP-3, MMP-9, and MCP-1, whereas only picogram levels were detected in untreated cells. CONCLUSIONS Fnfs stimulate the release of proinflammatory cytokines, matrix metalloproteinases, and monocyte chemoattractant protein from murine RPE cells. This observation indicated that Fnfs could contribute to ocular abnormalities by promoting inflammation, catabolism, and monocyte chemoattraction.


Investigative Ophthalmology & Visual Science | 2009

LX211 (voclosporin) suppresses experimental uveitis and inhibits human T cells.

Matthew A. Cunningham; Bobbie Ann Austin; Zhuqing Li; Baoying Liu; Steven Yeh; Chi-Chao Chan; Eddy Anglade; Poonam Velagaleti; Robert B. Nussenblatt

PURPOSE To test the therapeutic effectiveness of voclosporin against experimental autoimmune uveoretinitis (EAU) in rats and to evaluate its effect on human T cells. METHODS EAU was induced by immunization with a uveitogenic protein. Voclosporin administration, by subcutaneous injection, began on day (d) 0 or d7 after immunization. Treatment effectiveness was evaluated in vivo using clinical EAU scoring (d7-d13) and histopathologic evaluation of enucleated eyes after experimental termination. Rodent lymphocytes were harvested from lymph nodes on d14 for antigen-specific proliferation assays. The effect of voclosporin on human T-cell proliferation and cytokine secretion was examined in vitro. RESULTS Voclosporin prevented EAU development in rats receiving medium and high preventive doses, whereas high-dose voclosporin administration effectively treated EAU. Lymphocytes from animals treated with voclosporin had decreased antigen-specific proliferation in vitro compared with lymphocytes from untreated animals. No evidence of abnormal ocular histopathology was found in the eyes from animals that received high doses of therapeutic voclosporin. Using human T cells, voclosporin inhibited human T-cell proliferation up to 100-fold. Furthermore, voclosporin treatment of human T cells significantly reduced pan T-cell effector responses. CONCLUSIONS Voclosporin effectively suppressed uveoretinitis in an animal model that imitates the human inflammatory ocular disease by inhibiting lymphocyte proliferation. In addition, voclosporin effectively inhibited human T-cell proliferation and function in vitro. The authors report the first evidence supporting the application of voclosporin to treat intraocular inflammation.

Collaboration


Dive into the Baoying Liu's collaboration.

Top Co-Authors

Avatar

Robert B. Nussenblatt

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhiyu Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhuqing Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

H. Nida Sen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lai Wei

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shayma Jawad

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chi-Chao Chan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ping Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Diamond Ling

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge