Network


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

Hotspot


Dive into the research topics where Xueshu Zhang is active.

Publication


Featured researches published by Xueshu Zhang.


Journal of Immunology | 2005

CD4−8− Dendritic Cells Prime CD4+ T Regulatory 1 Cells to Suppress Antitumor Immunity

Xueshu Zhang; Hui Huang; Jinying Yuan; Deming Sun; Wu-Shiun Hou; John R. Gordon; Jim Xiang

It is clear that dendritic cells (DCs) are essential for priming of T cell responses against tumors. However, the distinct roles DC subsets play in regulation of T cell responses in vivo are largely undefined. In this study, we investigated the capacity of OVA-presenting CD4−8−, CD4+8−, or CD4−8+ DCs (OVA-pulsed DC (DCOVA)) in stimulation of OVA-specific T cell responses. Our data show that each DC subset stimulated proliferation of allogeneic and autologous OVA-specific CD4+ and CD8+ T cells in vitro, but that the CD4−8− DCs did so only weakly. Both CD4+8− and CD4−8+ DCOVA induced strong tumor-specific CD4+ Th1 responses and fully protective CD8+ CTL-mediated antitumor immunity, whereas CD4−8− DCOVA, which were less mature and secreted substantial TGF-β upon coculture with TCR-transgenic OT II CD4+ T cells, induced the development of IL-10-secreting CD4+ T regulatory 1 (Tr1) cells. Transfer of these Tr1 cells, but not T cells from cocultures of CD4−8− DCOVA and IL-10−/− OT II CD4+ T cells, into CD4−8+ DCOVA-immunized animals abrogated otherwise inevitable development of antitumor immunity. Taken together, CD4−8− DCs stimulate development of IL-10-secreting CD4+ Tr1 cells that mediated immune suppression, whereas both CD4+8− and CD4−8+ DCs effectively primed animals for protective CD8+ CTL-mediated antitumor immunity.


Life Sciences | 2002

DNA microarray analysis of the gene expression profiles of naı̈ve versus activated tumor-specific T cells

Xueshu Zhang; Zhuang Chen; Hui Huang; John Gordon; Jim Xiang

T cells are a key element in effective cancer immunity, recognizing MHC-antigen peptide complexes on the surface of antigen presenting cells and translating these signals into cytotoxic effector T cell responses. In this study, we systematically investigated by DNA array analysis the expression profiles of 514 immunologically relevant genes in naïve and SP2/0 tumor-specific activated mouse T cell populations. Our data shows that naïve T cells expressed 37 (i.e., 7.6% of the 514) transcripts with expression level (EL) values of > or =2.0, while the activated T cells expressed 101 such transcripts. The expression levels of 9 (1.75% of 514) of the shared transcripts were equivalent in the two populations of T cells. Ninety-six genes were differently expressed upon T cell activation, with 71 (13.81%) being up-regulated and 25 (4.86%) down-regulated. The list of significantly affected genes includes numerous cytokines and their receptors (e.g., IL-2Ralpha, IL-6Ralpha, IL-7Ralpha, IL-16, IL-17R, TGF-beta), chemokines and chemokine receptors (e.g., RANTES, CCR7, CXCR4), alternate surface proteins (e.g., 4-1BB, GITR, integrins-alphaL and -beta7, L-selectin, CD6, CD45 and EMMPRIN), cytoplasmic signaling intermediates (e,g., GATA-3, 14-3-3-eta, CIS1, SMAD4 and JAK1) and an array of other molecules (e.g., NFkappa-B inducing kinase, LTBP3 and persephin), several of which are associated with Th1 responses, and T cell self-regulation or migration. Taken together, our data contribute to our understanding of the generalized processes that accompany T cell activation and, more specifically, to our understanding of the processes associated with T cell activation during antitumor responses.


Journal of Immunology | 2007

Novel Exosome-Targeted CD4+ T Cell Vaccine Counteracting CD4+ 25+ Regulatory T Cell-Mediated Immune Suppression and Stimulating Efficient Central Memory CD8+ CTL Responses

Siguo Hao; Yongqing Liu; Jinying Yuan; Xueshu Zhang; Tianpei He; Xiaochu Wu; Yangdou Wei; Deming Sun; Jim Xiang

T cell-to-T cell Ag presentation is increasingly attracting attention. In this study, we demonstrated that active CD4+ T (aT) cells with uptake of OVA-pulsed dendritic cell-derived exosome (EXOOVA) express exosomal peptide/MHC class I and costimulatory molecules. These EXOOVA-uptaken (targeted) CD4+ aT cells can stimulate CD8+ T cell proliferation and differentiation into central memory CD8+ CTLs and induce more efficient in vivo antitumor immunity and long-term CD8+ T cell memory responses than OVA-pulsed dendritic cells. They can also counteract CD4+25+ regulatory T cell-mediated suppression of in vitro CD8+ T cell proliferation and in vivo CD8+ CTL responses and antitumor immunity. We further elucidate that the EXOOVA-uptaken (targeted)CD4+ aT cell’s stimulatory effect is mediated via its IL-2 secretion and acquired exosomal CD80 costimulation and is specifically delivered to CD8+ T cells in vivo via acquired exosomal peptide/MHC class I complexes. Therefore, EXO-targeted active CD4+ T cell vaccine may represent a novel and highly effective vaccine strategy for inducing immune responses against not only tumors, but also other infectious diseases.


Cancer Biotherapy and Radiopharmaceuticals | 2002

Advances in Dendritic Cell-Based Vaccine of Cancer

Xueshu Zhang; John Gordon; Jim Xiang

Dendritic cells (DCs) are potent antigen presenting cells that exist in virtually every tissue, and from which they capture antigens and migrate to secondary lymphoid organs where they activate naïve T cells. Although DCs are normally present in extremely small numbers in the circulation, recent advances in DC biology have allowed the development of methods to generate large numbers of these cells in vitro. Because of their immunoregulatory capacity, vaccination with tumor antigen-presenting DCs has been proposed as a treatment modality for cancer. In animal models, vaccination with DCs pulsed with tumor peptides, lysates, or RNA or loaded with apoptotic/necrotic tumor cells could induce significant antitumor CTL responses and antitumor immunity. However, the results from early clinical trails pointed to a need for additional improvement of DC-based vaccines before they could be considered as practical alternatives to the existing cancer treatment strategies. In this regard, subsequent studies have shown that DCs that express transgenes encoding tumor antigens are more potent primers of antitumor immunity both in vitro and in vivo than DCs simply pulsed with tumor peptides. Furthermore, DCs that have been engineered to express certain cytokines or chemokines can display a substantially improved maturation status, capacity to migrate to secondary lymphoid organs in vivo, and abilities to stimulate tumor-specific T cell responses and induce tumor immunity in vivo. In this review we also discuss a number of factors that are important considerations in designing DC vaccine strategies, including (i) the type and concentrations of tumor peptides used for pulsing DCs; (ii) the timing and intervals for DC vaccination/boostable data on DC vaccination portends bright prospects for this approach to tumor immune therapy, either alone or in conjunction with other therapies.


European Respiratory Journal | 2003

Regular salbutamol use increases CXCL8 responses in asthma: relationship to the eosinophil response

John R. Gordon; Veronica A. Swystun; Fang Li; Xueshu Zhang; Beth E. Davis; P. Hull; Donald W. Cockcroft

Regular salbutamol use can exacerbate allergen-induced airway eosinophilia in asthmatics, but its effect on airway eosinophil chemokine responses is unknown. Asthmatic subjects (n=14) were treated for 10 days with placebo or salbutamol in a double-blind, cross-over study, then given same-dose allergen challenges. Their sputa were then analysed 1 and 7 h later for a panel of eosinophil-related cytokines. Eosinophils from five test and three control subjects were tested for expression of CXCL8/interleukin (IL)‐8, and its receptors and responsiveness to CCL11/eotaxin and CXCL8/IL‐8. Sputum CXCL8/IL‐8, but not IL‐5, CCL5/regulated on activation, T‐cell expressed and secreted, CCL7/monocyte chemotactic protein‐3, CCL11/eotaxin, granulocyte-macrophage colony-stimulating factor or tumour necrosis factor levels, were increased (42%) by the salbutamol treatments. The CXCL8/IL‐8 levels correlated with the proportions of sputum eosinophils and these cells, but not other sputum cells, stained strongly for CXCL8/IL‐8. The circulating eosinophils of the tested subjects (n=5) expressed CXCL8/IL‐8 receptors and secreted high levels of this chemokine. Neutralisation of sputum CXCL8/IL‐8 reduced eosinophil chemotactic responses to these samples by 19±5%. These data suggest that regular use of salbutamol can augment airway CXCL8/interleukin‐8 responses to allergen challenge and that this CXCL8/interleukin‐8 could contribute to the airway inflammatory response.


Journal of Leukocyte Biology | 2010

Optimal TLR9 signal converts tolerogenic CD4–8– DCs into immunogenic ones capable of stimulating antitumor immunity via activating CD4+ Th1/Th17 and NK cell responses

Xueshu Zhang; Manjunatha Ankathatti Munegowda; Jinying Yuan; Yi Wei; Jim Xiang

Abstract


Autoimmunity | 2008

Active CD4+ helper T cells directly stimulate CD8+ cytotoxic T lymphocyte responses in wild-type and MHC II gene knockout C57BL/6 mice and transgenic RIP-mOVA mice expressing islet b-cell ovalbumin antigen leading to diabetes

Zhenmin Ye; Khawaja Ashfaque Ahmed; Siguo Hao; Xueshu Zhang; Yufeng Xie; Manju Ankathatti Munegowda; Qinghe Meng; Rajni Chibbar; Jim Xiang

CD4+ helper T (Th) cells play crucial role in priming, expansion and survival of CD8+ cytotoxic T lymphocytes (CTLs). However, how CD4+ Th cells help is delivered to CD8+ T cells in vivo is still unclear. We previously demonstrated that CD4+ Th cells can acquire ovalbumin (OVA) peptide/major histocompatibility complex (pMHC I) and costimulatory CD80 by OVA-pulsed DC (DCOVA) stimulation, and then stimulate OVA-specific CD8+ CTL responses in C57BL/6 mice. In this study, we further investigated CD4+ Th cells effect on stimulation of CD8 CTL responses in major histocompatibility complex (MHC II) gene knockout (KO) mice and transgenic rat insulin promoter (RIP)-mOVA mice with moderate expression of self OVA by using CD4+ Th cells or Th cells with various gene deficiency. We demonstrated that the in vitro DCOVA-activated CD4+ Th cells (3 × 106 cells/mouse) can directly stimulate OVA-specific CD8+ T-cell responses in wild-type C57BL/6 mice and MHC II gene KO mice lacking CD4+ T cells. A large amount of CD4+ Th cells (12 × 106 cells/mouse) can even overcome OVA-specific immune tolerance in transgenic RIP-mOVA mice, leading to CD8+ CTL-mediated mouse pancreatic islet destruction and diabetes. The stimulatory effect of CD4+ Th cells is mediated by its IL-2 secretion and CD40L and CD80 costimulations, and is specifically delivered to OVA-specific CD8+ T cells in vivo via its acquired pMHC I complexes. Therefore, the above elucidated principles for CD4+ Th cells will have substantial implications in autoimmunity and antitumor immunity, and regulatory T-cell-dependent immune suppression.


Biochemical and Biophysical Research Communications | 2009

CD40 ligation converts TGF-β-secreting tolerogenic CD4−8− dendritic cells into IL-12-secreting immunogenic ones

Xueshu Zhang; Ross M. Kedl; Jim Xiang

CD40L, the ligand for CD40 on dendritic cells (DCs), plays an important role in maturation and activation of DCs leading to induction of immune responses. Our previous studies showed that the mouse splenic CD4(-)8(-) DCs are tolerogenic and capable of stimulating suppressive type 1 CD4(+) regulatory T (Tr1) cell responses via TGF-beta secretion. In this study, we investigated whether CD40 ligation is able to convert tolerogenic CD4(-)8(-) DCs into immunogenic ones by in vitro treatment of DCs with anti-CD40 antibody. Our data showed that in vitro CD40 ligation with anti-CD40 antibody converted TGF-beta-secreting tolerogenic CD4(-)8(-) DCs into IL-12-secreting immunogenic ones capable of stimulating type 1 CD4(+) helper T (Th1) and CD8(+) cytotoxic T lymphocyte (CTL) responses leading to induction of antitumor immunity. In addition, in vivo CD40 ligation by intratumoral injection of adenoviral vector AdVCD40L expressing CD40 ligand also induced tumor growth inhibition and regression of established P815 tumors with infiltration of tolerogenic CD4(-)8(-) DCs. Therefore, our data provide new information for and may thus have useful impacts in CD40 ligation-based immunotherapy of cancer.


Cellular & Molecular Immunology | 2008

Acquired pMHC I Complexes Greatly Enhance CD4+ Th Cell's Stimulatory Effect on CD8+ T Cell-Mediated Diabetes in Transgenic RIP-mOVA Mice

Khawaja Ashfaque Ahmed; Yufeng Xie; Xueshu Zhang; Jim Xiang

CD4+ helper T (Th) cells play pivotal roles in induction of CD8+ CTL immunity. However, the mechanism of CD4+ T cell help delivery to CD8+ T cells in vivo is still elusive. In this study, we used ovalbumin (OVA)-pulsed dendritic cells (DCOVA) to activate OT-II mouse CD4+ T cells, and then studied the help effect of these CD4+ T cells on CD8+ cytotoxic T lymphocyte (CTL) responses. We also examined CTL mediated islet β cell destruction which led to diabetes in wild-type C57BL/6 mice and transgenic rat insulin promoter (RIP)-mOVA mice expressing β cell antigen OVA with self OVA-specific tolerance, respectively. In adoptive transfer experiments, we demonstrated that help, in the form of peptide/major histocompatibility complex (pMHC) I acquired from DCOVA by DCOVA activation, was required for induction of OVA-specific CTL responses in C57BL/6 mice. However, in combination with TCR transgenic OT-I mouse CD8+ T cells, the tolerogenic dosage of CD4+ Th cells with acquired pMHC I, but not CD4+ (Kb−/−) Th cells without acquired pMHC I were able to cause diabetes in 8/10 (80%) RIP-mOVA mice. This study thus expands the current knowledge in T cell-mediated autoimmunity and provides insight into the nature of CD4+ T cell-mediated help in CD8+ CTL induction.


Journal of Leukocyte Biology | 2017

Enhanced suppression of polyclonal CD8+25+ regulatory T cells via exosomal arming of antigen-specific peptide/MHC complexes.

Chuanyong Mu; Xueshu Zhang; Lu Wang; Aizhang Xu; Khawaja Ashfaque Ahmed; Xueqin Pang; Rajni Chibbar; Andrew Freywald; Jianan Huang; Yehan Zhu; Jim Xiang

Compared with CD4+25+ regulatory T cells (Tregs), the mechanisms for natural, polyclonal CD8+25+ Treg immune suppression have been significantly less studied. We previously showed that polyclonal T cells can acquire antigen‐specific targeting activity through arming with exosomal peptide‐MHC (pMHC). In this study, we assessed the suppressive effect of CD8+25+ Tregs or CD8+25+ Tregs armed with ovalbumin (OVA)‐specific exosomes on other immune cells and OVA‐specific dendritic cell (DCOVA)‐stimulated antitumor immunity. We demonstrate that CD8+25+ Tregs inhibit T cell proliferation in vitro in a cell contact‐dependent fashion but independent of the expression of immunosuppressive IL‐10, TGF‐β, and CTLA‐4. CD8+25+ Tregs anergize naïve T cells upon stimulation by up‐regulating T cell anergy‐associated Egr2 and down‐regulating IL‐2 production. Tregs also anergize DCs by preventing DC maturation through the down‐regulation of Iab, CD80, CD86, and inflammatory cytokines, leading to defects in T cell stimulation. Moreover, CD8+25+ Tregs inhibit CTLs through inducing CTL death via perforin‐mediated apoptosis and through reducing effector CTL cytotoxic activity via down‐regulating CTL perforin‐production and degranulation. In addition, we show that CD8+25+ Tregs suppress DCOVA‐stimulated CTL responses in priming and effector phases and inhibit immunity against OVA‐expressing CCLOVA lung cancer. Remarkably, polyclonal CD8+25+ Tregs armed with OVA‐specific exosomal pMHC class‐II (pMHC‐II), or pMHC class‐I (pMHC‐I) complexes exert their enhanced inhibition of CTL responses in the priming and the effector phases, respectively. Taken together, our investigation reveals that assigning antigen specificity to nonspecific polyclonal CD8+25+ Tregs for enhanced immune suppression can be achieved through exosomal pMHC arming. This principle may have a great effect on Treg‐mediated immunotherapy of autoimmune diseases.

Collaboration


Dive into the Xueshu Zhang's collaboration.

Top Co-Authors

Avatar

Jim Xiang

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar

John Gordon

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar

Jinying Yuan

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar

Khawaja Ashfaque Ahmed

Vaccine and Infectious Disease Organization

View shared research outputs
Top Co-Authors

Avatar

Siguo Hao

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar

Zhuang Chen

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Huang

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar

Rajni Chibbar

University of Saskatchewan

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge