Network


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

Hotspot


Dive into the research topics where Jing Jiao is active.

Publication


Featured researches published by Jing Jiao.


The FASEB Journal | 2015

Essential role of mitochondrial energy metabolism in Foxp3+ T-regulatory cell function and allograft survival

Ulf H. Beier; Alessia Angelin; Tatiana Akimova; Liqing Wang; Yujie Liu; Haiyan Xiao; Maya Koike; Saege Hancock; Tricia R. Bhatti; Rongxiang Han; Jing Jiao; Sigrid C. Veasey; Carrie A. Sims; Joseph A. Baur; Douglas C. Wallace; Wayne W. Hancock

Conventional T (Tcon) cells and Foxp3+ T‐regulatory (Treg) cells are thought to have differing metabolic requirements, but little is known of mitochondrial functions within these cell populations in vivo. In murine studies, we found that activation of both Tcon and Treg cells led to myocyte enhancer factor 2 (Mef2)‐induced expression of genes important to oxidative phosphorylation (OXPHOS). Inhibition of OXPHOS impaired both Tcon and Treg cell function compared to wild‐type cells but disproportionally affected Treg cells. Deletion of Pgc1α or Sirt3, which are key regulators of OXPHOS, abrogated Treg‐dependent suppressive function and impaired allograft survival. Mef2 is inhibited by histone/protein deacetylase‐9 (Hdac9), and Hdac9 deletion increased Treg suppressive function. Hdac9‐/‐ Treg showed increased expression of Pgc1α and Sirt3, and improved mitochondrial respiration, compared to wild‐type Treg cells. Our data show that key OXPHOS regulators are required for optimal Treg function and Treg‐dependent allograft acceptance. These findings provide a novel approach to increase Treg function and give insights into the fundamental mechanisms by which mitochondrial energy metabolism regulates immune cell functions in vivo.—Beier, U. H., Angelin, A., Akimova, T., Wang, L., Liu, Y., Xiao, H., Koike, M. A., Hancock, S. A., Bhatti, T. R., Han, R., Jiao, J., Veasey, S. C., Sims, C. A., Baur, J. A., Wallace, D. C., Hancock, W. W. Essential role of mitochondrial energy metabolism in Foxp3+ T‐regulatory cell function and allograft survival. FASEB J. 29, 2315‐2326 (2015). www.fasebj.org


Mucosal Immunology | 2014

Targeting sirtuin-1 alleviates experimental autoimmune colitis by induction of Foxp3 + T-regulatory cells

Tatiana Akimova; Haiyan Xiao; Yujie Liu; Tricia R. Bhatti; Jing Jiao; Evgeniy Eruslanov; Sunil Singhal; Liqing Wang; Rongxiang Han; Keziah Zacharia; Wayne W. Hancock; Ulf H. Beier

Induced Forkhead box P3-positive (Foxp3+) T-regulatory cells (iTregs) are essential to gastrointestinal immune homeostasis, and loss of the ability to develop iTregs may lead to autoimmune colitis. We previously showed a role for sirtuin-1 (Sirt1) in control of Treg function and hypothesized that targeting of Sirt1 might enhance iTreg development and thereby represent a potential therapy for inflammatory bowel disease (IBD). We adoptively transferred CD4+CD25−Foxp3− T effector (TE) cells from wild-type (WT) (C57BL/6) or fl-Sirt1/CD4cre mice into B6/Rag1−/− mice and monitored the mice until they lost 10–15% of their weight. Adoptive transfer of TE cells lacking Sirt1 to B6/Rag1−/− mice resulted in a 2.8-fold increase in iTreg formation compared with mice receiving WT TE cells and correlated with attenuated colitis and reduced weight loss (1.04±1.4% vs. 13.97±2.2%, respectively, P<0.001). In a second model of IBD, we used pharmacologic Sirt1 targeting of mice receiving multiple cycles of dextran sodium sulfate (DSS) in their drinking water, alternated with fresh water. Likewise, WT mice receiving cyclic DSS and a Sirt1 inhibitor, EX-527, had reduced weight loss (5.8±5.9% vs. 13.2±6.9%, respectively, P=0.03) and increased iTreg formation compared with controls. Sirt1 appears a promising target for pharmacologic therapy of IBD as a result of promoting iTreg development.


International Journal of Cancer | 2016

HDAC5 controls the functions of Foxp3(+) T-regulatory and CD8(+) T cells.

Haiyan Xiao; Jing Jiao; Liqing Wang; Shaun O'Brien; Kheng Newick; Liang-Chuan S. Wang; Eva Falkensammer; Yujie Liu; Rongxiang Han; Veena Kapoor; Finn K. Hansen; Thomas Kurz; Wayne W. Hancock; Ulf H. Beier

Histone/protein deacetylases (HDACs) are frequently upregulated in human malignancies and have therefore become therapeutic targets in cancer therapy. However, inhibiting certain HDAC isoforms can have protolerogenic effects on the immune system, which could make it easier for tumor cells to evade the host immune system. Therefore, a better understanding of how each HDAC isoform affects immune biology is needed to develop targeted cancer therapy. Here, we studied the immune phenotype of HDAC5–/– mice on a C57BL/6 background. While HDAC5–/– mice replicate at expected Mendelian ratios and do not develop overt autoimmune disease, their T‐regulatory (Treg) cells show reduced suppressive function in vitro and in vivo. Likewise, CD4+ T‐cells lacking HDAC5 convert poorly to Tregs under appropriately polarizing conditions. To test if this attenuated Treg formation and suppressive function translated into improved anticancer immunity, we inoculated HDAC5–/– mice and littermate controls with a lung adenocarcinoma cell line. Cumulatively, lack of HDAC5 did not lead to better anticancer immunity. We found that CD8+ T cells missing HDAC5 had a reduced ability to produce the cytokine, IFN‐γ, in vitro and in vivo, which may offset the benefit of weakened Treg function and formation. Taken together, targeting HDAC5 weakens suppressive function and de‐novo induction of Tregs, but also reduces the ability of CD8+ T cells to produce IFN‐γ.


Kidney International | 2016

Targeting Sirtuin-1 prolongs murine renal allograft survival and function

Matthew H. Levine; Zhonglin Wang; Haiyan Xiao; Jing Jiao; Liqing Wang; Tricia R. Bhatti; Wayne W. Hancock; Ulf H. Beier

Current immunosuppressive medications used after transplantation have significant toxicities. Foxp3(+) T-regulatory cells can prevent allograft rejection without compromising protective host immunity. Interestingly, inhibiting the class III histone/protein deacetylase Sirtuin-1 can augment Foxp3(+) T-regulatory suppressive function through increasing Foxp3 acetylation. Here we determined whether Sirtuin-1 targeting can stabilize biological allograft function. BALB/c kidney allografts were transplanted into C57BL/6 recipients with a CD4-conditional deletion of Sirtuin-1 (Sirt1(fl/fl)CD4(cre)) or mice treated with a Sirtuin-1-specific inhibitor (EX-527), and the native kidneys removed. Blood chemistries and hematocrit were followed weekly. Sirt1(fl/fl)CD4(cre) recipients showed markedly longer survival and improved kidney function. Sirt1(fl/fl)CD4(cre) recipients exhibited donor-specific tolerance, accepted BALB/c, but rejected third-party C3H cardiac allografts. C57BL/6 recipients of BALB/c renal allografts that were treated with EX-527 showed improved survival and renal function at 1, but not 10 mg/kg/day. Pharmacologic inhibition of Sirtuin-1 also improved renal allograft survival and function with dosing effects having relevance to outcome. Thus, inhibiting Sirtuin-1 can be a useful asset in controlling T-cell-mediated rejection. However, effects on non-T cells that could adversely affect allograft survival and function merit consideration.


Transplantation direct | 2017

The Effects of Tacrolimus on T-Cell Proliferation Are Short-Lived: A Pilot Analysis of Immune Function Testing

Benjamin L. Laskin; Jing Jiao; H. Jorge Baluarte; Sandra Amaral; Susan L. Furth; Tatiana Akimova; Wayne W. Hancock; Matthew H. Levine; Peter P. Reese; Ulf H. Beier

Background Optimal immunosuppression after organ transplant should balance the risks of rejection, infection, and malignancy while minimizing barriers to adherence including frequent or time-sensitive dosing. There is currently no reliable immune function assay to directly measure the degree of immunosuppression after transplantation. Methods We developed an immune function assay to mea//sure T-cell proliferation after exposure to immunosuppression in vivo. We tested the assay in mice, and then piloted the approach using single time point samples, 11 pediatric kidney transplant recipients prescribed tacrolimus, mycophenolate, and prednisone 6 months to 5 years posttransplant, with no history of rejection, opportunistic infection, or cancer. Twelve healthy adults were controls. Results We demonstrated that our assay can quantify suppression of murine T-cell proliferation after tacrolimus treatment in vivo. In humans, we found a mean 25% reduction in CD4 and CD8 T-cell proliferation in pediatric renal transplant recipients on triple immunosuppression compared with adult healthy controls, but the pilot results were not statistically significant nor correlated with serum tacrolimus levels. We observed that cell processing and washing reduced the effects of tacrolimus on T-cell proliferation, as did discontinuation of tacrolimus treatment shortly before sampling. Conclusions T-cell proliferation is currently not suitable to measure immunosuppression because sample processing diminishes observable effects. Future immune function testing should focus on fresh samples with minimal washing steps. Our results also emphasize the importance of adherence to immunosuppressive treatment, because T-cell proliferation recovered substantially after even brief discontinuation of tacrolimus.


Methods of Molecular Biology | 2017

Proximity Ligation Assay to Quantify Foxp3 Acetylation in Regulatory T Cells.

Jing Jiao; Rongxiang Han; Wayne W. Hancock; Ulf H. Beier

Determining protein acetylation by immunoprecipitation and immunoblotting can be challenging, especially if the tissue of interest is low in quantity, and when good quality acetylation site-specific antibodies are not available. Proximity ligation assays allow a sensitive and quantitative method to assess Foxp3 acetylation in regulatory T cells, with as little as 1.5 × 105 cells within two days turnaround time. This method is of potential use in other similar scenarios, when post-translational modifications of a protein of interest need to be determined with only a small amount of sample and in the absence of specific antibodies that can assess the post-translational modification in the protein of interest.


Cell Metabolism | 2017

Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments

Alessia Angelin; Luis Gil-de-Gómez; Satinder Dahiya; Jing Jiao; Lili Guo; Matthew H. Levine; Zhonglin Wang; William J. Quinn; Piotr K. Kopinski; Liqing Wang; Tatiana Akimova; Yujie Liu; Tricia Bhatti; Rongxiang Han; Benjamin L. Laskin; Joseph A. Baur; Ian A. Blair; Douglas C. Wallace; Wayne W. Hancock; Ulf H. Beier


Journal of Immunology | 2016

Foxp3 controls oxidative phosphorylation in regulatory T cells

Ulf H. Beier; Alessia Angelin; Jing Jiao; Haiyan Xiao; Liqing Wang; Satinder Dahiya; Andrew D. Wells; Douglas C. Wallace; Wayne W. Hancock


Journal of Immunology | 2014

Targeting sirtuin-1 alleviates experimental autoimmune colitis by induction of Foxp3+ T-regulatory cells (MUC2P.818)

Tatiana Akimova; Haiyan Xiao; Jing Jiao; Tricia R. Bhatti; Yujie Liu; Liqing Wang; Wayne W. Hancock; Ulf H. Beier


The FASEB Journal | 2018

Loss of HDAC6 alters gut microbiota and worsens obesity

Arnon D. Lieber; Ulf H. Beier; Haiyan Xiao; Benjamin J. Wilkins; Jing Jiao; Xinmin S. Li; Rebecca C. Schugar; Christopher M. Strauch; Zeneng Wang; J. Mark Brown; Stanley L. Hazen; Nicholas A. Bokulich; Kelly V. Ruggles; Tatiana Akimova; Wayne W. Hancock; Martin J. Blaser

Collaboration


Dive into the Jing Jiao's collaboration.

Top Co-Authors

Avatar

Wayne W. Hancock

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ulf H. Beier

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Haiyan Xiao

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Liqing Wang

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Tatiana Akimova

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Rongxiang Han

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Yujie Liu

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Alessia Angelin

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Douglas C. Wallace

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Matthew H. Levine

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge