Network


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

Hotspot


Dive into the research topics where Shiguang Qian is active.

Publication


Featured researches published by Shiguang Qian.


Hepatology | 2004

Inhibition of T‐cell responses by hepatic stellate cells via B7‐H1–mediated T‐cell apoptosis in mice

Ming Chin Yu; Cheng Hsu Chen; Xiaoyan Liang; Lianfu Wang; Chandrashekhar R. Gandhi; John J. Fung; Lina Lu; Shiguang Qian

In the injured liver, hepatic stellate cells (HSCs) secrete many different cytokines, recruit lymphocytes, and thus participate actively in the pathogenesis of liver disease. Little is known of the role of HSCs in immune responses. In this study, HSCs isolated from C57BL/10 (H2b) mice were found to express scant key surface molecules in the quiescent stage. Activated HSCs express major histocompatibility complex class I, costimulatory molecules, and produce a variety of cytokines. Stimulation by interferon γ (IFN‐γ) or activated T cells enhanced expression of these molecules. Interestingly, addition of the activated (but not quiescent) HSCs suppressed thymidine uptake by T cells that were stimulated by alloantigens or by anti‐CD3–mediated T‐cell receptor ligation in a dose‐dependent manner. High cytokine production by the T cells suggests that the inhibition was probably not a result of suppression of their activation. T‐cell division was also found to be normal in a CFSE dilution assay. The HSC‐induced T‐cell hyporesponsiveness was associated with enhanced T‐cell apoptosis. Activation of HSCs was associated with markedly enhanced expression of B7‐H1. Blockade of B7‐H1/PD‐1 ligation significantly reduced HSC immunomodulatory activity, suggesting an important role of B7‐H1. In conclusion, the bidirectional interactions between HSCs and immune cells may contribute to hepatic immune tolerance. (HEPATOLOGY 2004;40:1312–1321.)


Journal of Immunology | 2002

Marked Prolongation of Cardiac Allograft Survival by Dendritic Cells Genetically Engineered with NF-κB Oligodeoxyribonucleotide Decoys and Adenoviral Vectors Encoding CTLA4-Ig

C. Andrew Bonham; Lansha Peng; Xiaoyan Liang; Zongyou Chen; Lianfu Wang; Linlin Ma; Holger Hackstein; Paul D. Robbins; Angus W. Thomson; John J. Fung; Shiguang Qian; Lina Lu

Bone marrow-derived dendritic cells (DCs) can be genetically engineered using adenoviral (Ad) vectors to express immunosuppressive molecules that promote T cell unresponsiveness. The success of these DCs for therapy of allograft rejection has been limited in part by the potential of the adenovirus to promote DC maturation and the inherent ability of the DC to undergo maturation following in vivo administration. DC maturation occurs via NF-κB-dependent mechanisms, which can be blocked by double-stranded “decoy” oligodeoxyribonucleotides (ODNs) containing binding sites for NF-κB. Herein, we describe the combined use of NF-κB ODNs and rAd vectors encoding CTLA4-Ig (Ad CTLA4-Ig) to generate stably immature murine myeloid DCs that secrete the potent costimulation blocking agent. These Ad CTLA4-Ig-transduced ODN DCs exhibit markedly impaired allostimulatory ability and promote apoptosis of activated T cells. Furthermore, administration of Ad CTLA4-Ig ODN-treated donor DCs (C57BL10; B10(H-2b)) before transplant significantly prolongs MHC-mismatched (C3HHeJ; C3H(H-2k)) vascularized heart allograft survival, with long-term (>100 days) donor-specific graft survival in 40% of recipients. The mechanism(s) responsible for DC tolerogenicity, which may involve activation-induced apoptosis of alloreactive T cells, do not lead to skewing of intragraft Th cytokine responses. Use of NF-κB antisense decoys in conjunction with rAd encoding a potent costimulation blocking agent offers promise for therapy of allograft rejection or autoimmune disease with minimization of systemic immunosuppression.


Transplantation | 1996

Systemic administration of cellular interleukin-10 can exacerbate cardiac allograft rejection in mice

Shiguang Qian; Wei Li; Youping Li; Fumin Fu; Lina Lu; John J. Fung; Angus W. Thomson

Cellular interleukin-10 (cIL-10) has been shown to inhibit cytokine production by T helper type 1 (Th1) cells by blocking antigen presenting cell function. This activity has suggested that IL-10 might be useful in the treatment of transplant rejection. Stimulatory effects of IL-10 however, have also been observed both on T and B cell differentiation. In this study, we examined the influence of recombinant (r) mouse (m) IL-10 on heterotopic vascularized heart allograft survival in the B10(H2b)-->C3H(H2k) strain combination that crosses both major histocompatibility complex (MHC) and non-MHC-histocompatibility antigen (non-MHC-HA) barriers. The influence of IL-10 was also examined in the B10.BR (H2k)--> C3H combination with disparity at only non-MHC-HA loci. Postoperative intraperitoneal administration of IL-10 (100 microg/d, days 0-6) significantly accelerated heart graft rejection both in the B10-->C3H (mean survival time [MST] 7.8+/-0.2 days; control MST 10.6+/-0.6 days; P<0.05) and the B10.BR-->C3H combination (MST 14.3+/-0.5 days; control MST 77.7+/-14.4 days). Ex vivo IL-10 perfusion of donor hearts for either 15 min or 2 hr did not affect subsequent graft survival. Immunologic monitoring of transplanted mice revealed that IL-10 treatment (100 microg/d, i.p., days 0-6) increased both the circulating complement-dependent cytotoxic (CDC) antibody titer and splenic anti-donor cytotoxic T lymphocyte (CTL) activity measured up to 3 weeks posttransplant. These findings indicate that post transplant systemic administration of cIL-10 can promote vascularized allograft rejection, and that this may reflect stimulation both of B and T cell alloimmune responses.


Transplantation | 1998

Phenotype, function, and in vivo migration and survival of allogeneic dendritic cell progenitors genetically engineered to express TGF-β

Wei Cheng Lee; Cuiping Zhong; Shiguang Qian; Yonghong Wan; Jack Gauldie; Zhibao Mi; Paul D. Robbins; Angus W. Thomson; Lina Lu

BACKGROUND Administration of donor bone marrow (BM)-derived dendritic cell (DC) progenitors (DCp) that are major histocompatibility complex (MHC) class II+ but costimulatory molecule (CD40, CD80, CD86)-deficient can prolong mouse heart allograft survival This is associated with microchimerism and inhibition of antidonor cytotoxic T lymphocyte (CTL) activity. Genetic modification of these donor antigen-presenting cells to express an immunosuppressive molecule(s) may enhance their in vivo survival and potential tolerogenicity. METHODS The surface phenotype of B10(H-2b) DCp before and after gene transfer using replication-deficient adenoviral (Ad) vectors was determined by monoclonal antibody (mAb) staining and flow cytometry. Transforming growth factor-beta (TGF-beta) production was quantitated by enzyme-linked immunosorbent assay. Allostimulatory activity of the gene-transduced DCp was ascertained by mixed leukocyte reaction (MLR) and CTL induction. To assess their in vivo migratory activity and survival, the transduced cells were injected subcutaneously into one hind footpad of C3H (H-2k) mice. Tissues (draining popliteal lymph nodes [LN], spleens, and thymi) were removed 1, 2, 7, and 14 days later and stained for donor MHC class II using anti-LA(b) mAb in an immunohistochemical procedure. The mean number of IAb+ cells per unit area was determined. RESULTS Transduction with a control Ad vector (Ad-LacZ) at 50 multiplicity of infection slightly increased CD40 and CD86 expression and up-regulated the poor allostimulatory activity of the DCp assessed by MLR and CTL responses. These effects on function were negated in Ad-TGF-beta1-transduced cells. After their injection into mouse footpads, the gene-transduced IAb+ cells were observed in maximal numbers in the popliteal LN at day 1 and in marginal zones and T-dependent areas of spleens (peak at day 7) but were rare in thymi. Transduction with Ad-LacZ reduced the numbers of IAb+ cells identified in both LN and spleens at all time points postinjection, suggesting that the vector alone affected DC life span in allogeneic recipients. TGF-beta1 transgene expression not only fully prevented the reduction in DC induced by Ad transduction alone, but also increased numbers and prolonged the survival of donor cells in the spleen, as shown by a two-to fivefold increase in IAb+ cells at days 2-14 compared with control (Ad-LacZ-transduced) DC. CONCLUSION BM-derived DCp can be transduced efficiently to express TGF-beta1 using an Ad vector. They exhibit very poor allostimulatory activity and similar migration characteristics in vivo to unmodified DCp. Survival of TGF-beta gene-transduced DC, however, is enhanced significantly compared with unmodified and (especially) control Ad-LacZ gene-transduced DC. Genetic engineering of donor DC to express the immunosuppressive molecule TGF-beta promotes their survival in allogeneic hosts and may potentiate their previously reported tolerogenicity.


Gene Therapy | 1999

Adenoviral delivery of CTLA4Ig into myeloid dendritic cells promotes their in vitro tolerogenicity and survival in allogeneic recipients.

Lina Lu; Andrea Gambotto; W C Lee; Shiguang Qian; Bonham Ca; Paul D. Robbins; Angus W. Thomson

Dendritic cells (DC) are highly specialized antigen-presenting cells (APC) that initiate and modulate immune responses. They are essential for naive T cell activation, but may also play roles both in central and peripheral tolerance. Blockade of costimulatory pathways that provide the crucial second signal for lymphocyte activation is one strategy to augment the potential tolerogenicity of DC. Here, in vitro propagated DC were transduced using an adenoviral (Ad) vector to express the gene encoding cytotoxic T lymphocyte antigen 4-immunoglobulin (CTLA4Ig), which blocks interaction of CD80 and CD86 on DC with CD28 on T cells. Supernatants of AdCTLA4Ig-transduced DC strikingly inhibited mixed leukocyte reactions (MLR) induced by non-transduced DC. Whereas transduction of marker genes (LacZ or enhanced green fluorescence protein (EGFP)) did not alter their potent allostimulatory activity, DC transduced with CTLA4Ig exhibited striking reductions in cell surface staining for CD86, but not MHC class II, and were poor stimulators of T cell proliferation and cytotoxic T lymphocyte (CTL) responses. In addition, they induced alloantigen-specific T cell hyporesponsiveness. They were detected, following local injection, in significantly increased numbers in the lymphoid tissue of unmodified allogeneic recipients. This is the first report of the functional properties of DC genetically engineered to express CTLA4Ig.


Journal of Immunology | 2001

Liver-derived DEC205+B220+CD19- dendritic cells regulate T cell responses

Lina Lu; C.A. Bonham; Xiaoyan Liang; Zongyou Chen; Wei Li; Lianfu Wang; Simon C. Watkins; M. Nalesnik; Mark S. Schlissel; A. J. Demestris; John J. Fung; Shiguang Qian

Leukocytes resident in the liver may play a role in immune responses. We describe a cell population propagated from mouse liver nonparenchymal cells in IL-3 and anti-CD40 mAb that exhibits a distinct surface immunophenotype and function in directing differentiation of naive allogeneic T cells. After culture, such cells are DEC-205brightB220+CD11c−CD19−, and negative for T (CD3, CD4, CD8α), NK (NK 1.1) cell markers, and myeloid Ags (CD11b, CD13, CD14). These liver-derived DEC205+B220+ CD19− cells have a morphology and migratory capacity similar to dendritic cells. Interestingly, they possess Ig gene rearrangements, but lack Ig molecule expression on the cell surface. They induce low thymidine uptake of allogeneic T cells in MLR due to extensive apoptosis of activated T cells. T cell proliferation is restored by addition of the common caspase inhibitor peptide, benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (zVAD-fmk). T cells stimulated by liver-derived DEC205+B220+D19− cells release both IL-10 and IFN-γ, small amounts of TGF-β, and no IL-2 or IL-4, a cytokine profile resembling T regulatory type 1 cells. Expression of IL-10 and IFN-γ, but not bioactive IL-12 in liver DEC205+B220+CD19− cells was demonstrated by RNase protection assay. In vivo administration of liver DEC205+B220+CD19− cells significantly prolonged the survival of vascularized cardiac allografts in an alloantigen-specific manner.


Transplantation | 1991

Orthotopic liver transplantation in the mouse.

Shiguang Qian; John J. Fung; Anthony J. Demetris; Suzanne T. Ildstad; Thomas E. Starzl

Heart and kidney transplantation in the mouse has been well established (1, 2), but orthotopic liver transplantation has not been reported. Although the mouse is one-tenth the size of the rat, there are many advantages to using the mouse for immunologic research. The mouse genome has been more thoroughly characterized than the rat or any other species of mammal. The mouse H-2 system also bears a striking resemblance to the human HLA system. In addition, there are numerous genetically defined inbred strains and wealth of monoclonal antibodies that are commercially available for mouse investigations. A key development in rat orthotopic liver transplantation (3) was the introduction by Zimmerman et al. (4) and Kamada and Caine (5), of the cuff technique instead of suture for some of the venous vascular anastomoses. This method shortened the clamping time of the portal vein and increased survival. We have applied this principle to mouse orthotopic liver transplantation. In our pilot studies, more than 40 syngeneic mouse liver replacements were performed before 6 long-term survivors were obtained. The experience reported here is with the next 48 attempts, in which the surgical success rate was 83%. Male inbred syngeneic mice 10–12 weeks old (25–32 g), from Jackson Laboratory, Bar Harbor, ME, were used as size-matched donors and recipients under methoxyflurane (2.2-dichloro-1,1-difluoroethyl methyl ether) anesthesia. The strains used were B6AF1 (27 pairs), C57BL/6 (11 pairs), and BALB/c (10 pairs). Clean but not sterile operative technique was used, and all procedures were performed under the operating microscope with 4–6.4× magnification.


Hepatology | 2011

Hepatic stellate cells regulate immune response by way of induction of myeloid suppressor cells in mice

Hong Shiue Chou; C. Hsieh; Horng Ren Yang; Lianfu Wang; Yusuke Arakawa; Kathleen E. Brown; Qingyu Wu; Feng Lin; Marion G. Peters; John J. Fung; Lina Lu; Shiguang Qian

Although organ transplants have been applied for decades, outcomes of somatic cell transplants remain disappointing, presumably due to lack of appropriate supporting stromal cells. Thus, cotransplantation with liver stromal cells, hepatic stellate cells (HSC), achieves long‐term survival of islet allografts in mice by way of induction of effector T cell apoptosis and generation of regulatory T (Treg) cells. In this study we provide evidence both in vitro and in vivo that HSC can promote generation of myeloid‐derived suppressor cells (MDSC). HSC‐induced MDSC demonstrate potent immune inhibitory activity. Induction of MDSC is dependent on an intact interferon gamma signaling pathway in HSC and is mediated by soluble factors, suggesting that the specific tissue stromal cells, such as HSC, play a crucial role in regulating immune response by way of inflammation‐induced generation of MDSC. Large amounts of MDSC can be propagated in vitro from bone marrow‐derived myeloid precursor cells under the influence of HSC. Conclusion: Cotransplantation with in vitro generated MDSC can effectively protect islet allografts from host immune attack. Local delivery of potent immune suppressor cells for cell transplants holds great clinical application potential. (HEPATOLOGY 2011;)


Journal of Leukocyte Biology | 1999

Genetic engineering of dendritic cells to express immunosuppressive molecules (viral IL-10, TGF-beta, and CTLA4Ig).

Lina Lu; Wei Chen Lee; Takuya Takayama; Shiguang Qian; Andrea Gambotto; Paul D. Robbins; Angus W. Thomson

There is growing evidence that, in addition to their role as initiators of immune responses, dendritic cells (DC) can exhibit tolerogenic properties. Immature DC deficient in cell surface costimulatory/accessory molecules can prolong organ and pancreatic islet allograft survival, whereas in vitro manipulation of DC by exposure to a variety of factors (e.g., viral interleukin‐10; CTLA4Ig) can confer tolerogenic properties on these cells. Genetic engineering of DC to express immunosuppressive molecules is, in theory, an attractive approach to the therapy of allograft rejection and possibly, autoimmune disorders. J. Leukoc. Biol. 66: 293–296; 1999.


Transplantation | 2012

Myeloid-derived suppressor cells protect islet transplants by B7-H1 mediated enhancement of T regulatory cells.

Hong Shiue Chou; C. Hsieh; Ronald Charles; Lianfu Wang; Timothy Wagner; John J. Fung; Shiguang Qian; Lina L. Lu

Background. Side effects of lifetime immunosuppression for cell transplants often outweigh the benefits; therefore, induction of transplant tolerance is needed. We have shown that cotransplantation with myeloid-derived suppressor cells (MDSC) effectively protect islet allografts from rejection without requirement of immunosuppression. This study was to investigate the underlying mechanisms. Methods. MDSC were generated by addition of hepatic stellate cells from various stain mice into dendritic cell (DC) culture. The quality of MDSC was monitored by phenotype and function analyses. MDSC mixed with islet allografts were transplanted into diabetic recipients. T-cell response was analyzed after transplantation by using flow and histochemical analyses, and was compared with islet alone and islet/DC transplant groups. B7-H1 knockout mice were used to determine the role of B7-H1 on MDSC in regulation of T-cell response. Results. Cotransplantation with MDSC (not DC) effectively protected islet allografts without requirement of immunosuppression. This is associated with attenuation of CD8 T cells in the grafts and marked expansion of regulatory T (Treg) cells, which contributed to MDSC-induced T-cell hyporesponsiveness. Antigen-specific Treg cells were prone to accumulate in lymphoid organs close to the grafts. Both in vitro and in vivo data demonstrated that B7-H1 was absolutely required for MDSC to exert immune regulatory activity and induction of Treg cells. Conclusion. The described approach holds great clinical application potential and may overcome the limitation of requiring chronic administration of immunosuppression in cell transplants. Understanding the underlying mechanisms will facilitate the development of this novel therapeutic strategy.

Collaboration


Dive into the Shiguang Qian's collaboration.

Top Co-Authors

Avatar

Lina Lu

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wei Li

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Fumin Fu

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Lianfu Wang

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. Fung

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

A. J. Demetris

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Abdul S. Rao

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge