Network


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

Hotspot


Dive into the research topics where Na Yin is active.

Publication


Featured researches published by Na Yin.


Journal of Experimental Medicine | 2009

The origin and development of nonlymphoid tissue CD103+ DCs

Florent Ginhoux; Kang Liu; Julie Helft; Milena Bogunovic; Melanie Greter; Daigo Hashimoto; Jeremy Price; Na Yin; Jonathan S. Bromberg; Sergio A. Lira; E. Richard Stanley; Michel C. Nussenzweig; Miriam Merad

CD103+ dendritic cells (DCs) in nonlymphoid tissues are specialized in the cross-presentation of cell-associated antigens. However, little is known about the mechanisms that regulate the development of these cells. We show that two populations of CD11c+MHCII+ cells separated on the basis of CD103 and CD11b expression coexist in most nonlymphoid tissues with the exception of the lamina propria. CD103+ DCs are related to lymphoid organ CD8+ DCs in that they are derived exclusively from pre-DCs under the control of fms-like tyrosine kinase 3 (Flt3) ligand, inhibitor of DNA protein 2 (Id2), and IFN regulatory protein 8 (IRF8). In contrast, lamina propria CD103+ DCs express CD11b and develop independently of Id2 and IRF8. The other population of CD11c+MHCII+ cells in tissues, which is CD103−CD11b+, is heterogenous and depends on both Flt3 and MCSF-R. Our results reveal that nonlymphoid tissue CD103+ DCs and lymphoid organ CD8+ DCs derive from the same precursor and follow a related differentiation program.


Immunity | 2009

Regulatory T cells sequentially migrate from inflamed tissues to draining lymph nodes to suppress the alloimmune response.

Nan Zhang; Bernd Schröppel; Girdhari Lal; Claudia Jakubzick; Xia Mao; Dan Chen; Na Yin; Rolf Jessberger; Jordi Ochando; Yaozhong Ding; Jonathan S. Bromberg

To determine the site and mechanism of suppression by regulatory T (Treg) cells, we investigated their migration and function in an islet allograft model. Treg cells first migrated from blood to the inflamed allograft where they were essential for the suppression of alloimmunity. This process was dependent on the chemokine receptors CCR2, CCR4, and CCR5 and P- and E-selectin ligands. In the allograft, Treg cells were activated and subsequently migrated to the draining lymph nodes (dLNs) in a CCR2, CCR5, and CCR7 fashion; this movement was essential for optimal suppression. Treg cells inhibited dendritic cell migration in a TGF-beta and IL-10 dependent fashion and suppressed antigen-specific T effector cell migration, accumulation, and proliferation in dLNs and allografts. These results showed that sequential migration from blood to the target tissue and to dLNs is required for Treg cells to differentiate and execute fully their suppressive function.


Transplantation | 2011

Targeting Lymphangiogenesis After Islet Transplantation Prolongs Islet Allograft Survival

Na Yin; Nan Zhang; Jiangnan Xu; Qixin Shi; Yaozhong Ding; Jonathan S. Bromberg

Background. Lymphatics are important for their conduit functions of transporting antigen, immune cells, and inflammatory mediators to draining lymph nodes and to the general circulation. Lymphangiogenesis is involved in many pathologic processes; however, the roles for lymphatic responses in transplantation have not been thoroughly investigated. Methods. Mice were made diabetic by a single high dose of streptozotocin and then received islet allografts. Animals were treated with three different lymphatic inhibitors. FTY720, an analog of sphingosine 1-phosphate, inhibited lymphocyte migration into afferent and efferent lymphatics. Sunitinib, a kinase inhibitor, blocked several receptors, including vascular endothelial growth factor receptor 3 (VEGFR3), the major growth factor receptor for lymphatic endothelial cells. Anti-VEGFR3 monoclonal antibody specifically inhibited VEGFR3. Diabetes was determined by daily monitoring of blood glucose levels. Inflammation within islet grafts was assessed by immunohistochemistry for insulin, T cells (CD3), and lymphatics (LYVE-1). Results. After transplantation, lymphangiogenesis occurred in islet allografts and in draining lymph nodes. FTY720, sunitinib, and anti-VEGFR3 each inhibited lymphangiogenesis in the islets and significantly prolonged allograft survival. Immunofluorescent staining demonstrated that administration of each of the lymphatic inhibitors resulted in preservation of islets and &bgr;-cells along with a markedly reduced infiltration of T cells into the grafts. Conclusion. Lymphangiogenesis occurs in islet allografts in response to inflammation and plays a key role in the islet inflammation in alloimmunity. Interfering with lymphatic function leads to inhibition of lymphangiogenesis and prolonged or indefinite allograft survival. These observations suggest new therapeutic targets for rejection and tolerance.


American Journal of Transplantation | 2011

Distinct inflammatory signals have physiologically divergent effects on epigenetic regulation of Foxp3 expression and Treg function.

Girdhari Lal; Na Yin; Jiangnan Xu; Marvin Lin; S. Schroppel; Yaozhong Ding; I. Marie; David E. Levy; Jonathan S. Bromberg

Foxp3 expression in regulatory T cells (Treg) is required for their development and suppressive function. How different inflammatory signals affect Foxp3 chromatin structure, expression and Tregs plasticity are not completely known. In the present study, the Toll‐like receptor 2 (TLR2) ligand peptidoglycan inhibited Foxp3 expression in both natural Treg (nTreg) and TGFβ‐driven adaptive Treg (aTreg). Inhibition was independent of paracrine Th1, Th2 and Th17 cytokines. PGN‐induced T cell‐intrinsic TLR2‐Myd88‐dependent IFR1 expression and induced IRF1 bound to IRF1 response elements (IRF‐E) in the Foxp3 promoter and intronic enhancers, and negatively regulated Foxp3 expression. Inflammatory IL‐6 and TLR2 signals induced divergent chromatin changes at the Foxp3 locus and regulated Treg suppressor function, and in an islet transplant model resulted in differences in their ability to prolong graft survival. These findings are important for understanding how different inflammatory signals can affect the transplantation tolerance and immunity.


European Journal of Immunology | 2010

Islet-expressed TLR2 and TLR4 sense injury and mediate early graft failure after transplantation.

Bernd Krüger; Na Yin; Nan Zhang; Anju Yadav; William Coward; Girdhari Lal; Weiping Zang; Peter S. Heeger; Jonathan S. Bromberg; Barbara Murphy; Bernd Schröppel

Although islet transplantation is an effective treatment for Type 1 diabetes, primary engraftment failure contributes to suboptimal outcomes. We tested the hypothesis that islet isolation and transplantation activate innate immunity through TLR expressed on islets. Murine islets constitutively express TLR2 and TLR4, and TLR activation with peptidoglycan or LPS upregulates islet production of cytokines and chemokines. Following transplantation into streptozotocin‐induced diabetic, syngeneic mice, islets exposed to LPS or peptidoglycan had primary graft failure with intra‐ and peri‐islet mononuclear cell inflammation. The use of knockout mice showed that recipient CD8+ T cells caused engraftment failure and did so in the absence of islet‐derived DC. To mimic physiological islet injury, islets were transplanted with exocrine debris. Transplantation of TLR2/4−/− islets reduced proinflammatory cytokine production and improved islet survival. Stressed islets released the alarmin high‐mobility group box protein 1 (HMGB1) and recombinant HMGB1 (rHMGB1) induced NFkB activation. NFkB activation was prevented in the absence of both TLR2 and TLR4. rHMGB1 pretreatment also prevented primary engraftment through a TLR2/4‐dependent pathway. Our results show that islet graft failure can be initiated by TLR2 and TLR4 signaling and suggest that HMGB1 is one likely early mediator. Subsequent downstream signaling results in intra‐islet inflammation followed by T‐cell‐mediated graft destruction.


Diabetes | 2010

Immune Cell–Derived C3 Is Required for Autoimmune Diabetes Induced by Multiple Low Doses of Streptozotocin

Marvin Lin; Na Yin; Barbara Murphy; M. Edward Medof; Stephan Segerer; Peter S. Heeger; Bernd Schröppel

OBJECTIVE The complement system contributes to autoimmune injury, but its involvement in promoting the development of autoimmune diabetes is unknown. In this study, our goal was to ascertain the role of complement C3 in autoimmune diabetes. RESEARCH DESIGN AND METHODS Susceptibility to diabetes development after multiple low-dose streptozotocin treatment in wild-type (WT) and C3-deficient mice was analyzed. Bone marrow chimeras, luminex, and quantitative reverse transcription PCR assays were performed to evaluate the phenotypic and immunologic impact of C3 in the development of this diabetes model. RESULTS Coincident with the induced elevations in blood glucose levels, we documented alternative pathway complement component gene expression within the islets of the diabetic WT mice. When we repeated the experiments with C3-deficient mice, we observed complete resistance to disease, as assessed by the absence of histologic insulitis and the absence of T-cell reactivity to islet antigens. Studies of WT chimeras bearing C3-deficient bone marrow cells showed that bone marrow cell–derived C3, and not serum C3, is involved in the induction of diabetes in this model. CONCLUSIONS The data reveal a key role for immune cell–derived C3 in the pathogenesis of murine multiple low-dose streptozotocin-induced diabetes and support the concept that immune cell mediated diabetes is in part complement-dependent.


PLOS ONE | 2011

Lymphangiogenesis Is Required for Pancreatic Islet Inflammation and Diabetes

Na Yin; Nan Zhang; Girdhari Lal; Jiangnan Xu; Minhong Yan; Yaozhong Ding; Jonathan S. Bromberg

Lymphangiogenesis is a common phenomenon observed during inflammation and engraftment of transplants, but its precise role in the immune response and underlying mechanisms of regulation remain poorly defined. Here we showed that in response to injury and autoimmunity, lymphangiogenesis occurred around islets and played a key role in the islet inflammation in mice. Vascular endothelial growth factors receptor 3 (VEGFR3) is specifically involved in lymphangiogenesis, and blockade of VEGFR3 potently inhibited lymphangiogenesis in both islets and the draining LN during multiple low-dose streptozotocin (MLDS) induced autoimmune insulitis, which resulted in less T cell infiltration, preservation of islets and prevention of the onset of diabetes. In addition to their well-known conduit function, lymphatic endothelial cells (LEC) also produced chemokines in response to inflammation. These LEC attracted two distinct CX3CR1hi and LYVE-1+ macrophage subsets to the inflamed islets and CX3CR1hi cells were influenced by LEC to differentiate into LYVE-1+ cells closely associated with lymphatic vessels. These observations indicate a linkage among lymphangiogenesis and myeloid cell inflammation during insulitis. Thus, inhibition of lymphangiogenesis holds potential for treating insulitis and autoimmune diabetes.


Journal of Immunology | 2012

Functional Specialization of Islet Dendritic Cell Subsets

Na Yin; Jiangnan Xu; Florent Ginhoux; Gwendalyn J. Randolph; Miriam Merad; Yaozhong Ding; Jonathan S. Bromberg

Dendritic cells (DC) play important roles in both tolerance and immunity to β cells in type 1 diabetes. How and why DC can have diverse and opposing functions in islets remains elusive. To answer these questions, islet DC subsets and their specialized functions were characterized. Under both homeostatic and inflammatory conditions, there were two main tissue-resident DC subsets in islets, defined as CD11blo/−CD103+CX3CR1− (CD103+ DC), the majority of which were derived from fms-like tyrosine kinase 3-dependent pre-DC, and CD11b+CD103−CX3CR1+ (CD11b+ DC), the majority of which were derived from monocytes. CD103+ DC were the major migratory DC and cross-presented islet-derived Ag in the pancreatic draining lymph node, although this DC subset displayed limited phagocytic activity. CD11b+ DC were numerically the predominant subset (60–80%) but poorly migrated to the draining lymph node. Although CD11b+ DC had greater phagocytic activity, they poorly presented Ag to T cells. CD11b+ DC increased in numbers and percentage during T cell-mediated insulitis, suggesting that this subset might be involved in the pathogenesis of diabetes. These data elucidate the phenotype and function of homeostatic and inflammatory islet DC, suggesting differential roles in islet immunity.


Journal of Immunology | 2011

Stat4 Is Critical for the Balance between Th17 Cells and Regulatory T Cells in Colitis

Jiangnan Xu; Yu Yang; Guixing Qiu; Girdhari Lal; Na Yin; Zhihong Wu; Jonathan S. Bromberg; Yaozhong Ding

Th17 play a central role in autoimmune inflammatory responses. Th1 are also necessary for autoimmune disease development. The interplay of Th1 signals and how they coordinate with Th17 during inflammatory disease pathogenesis are incompletely understood. In this study, by adding Stat4 deficiency to Stat6/T-bet double knockout, we further dissected the role of Stat4 in Th1 development and colitis induction. We showed that in the absence of the strong Th2 mediator Stat6, neither Stat4 nor T-bet is required for IFN-γ production and Th1 development. However, addition of Stat4 deficiency abolished colitis induced by Stat6/T-bet double-knockout cells, despite Th1 and Th17 responses. The failure of colitis induction by Stat4/Stat6/T-bet triple-knockout cells is largely due to elevated Foxp3+ regulatory T cell (Treg) development. These results highlight the critical role of Stat4 Th1 signals in autoimmune responses in suppressing Foxp3+ Treg responses and altering the balance between Th17 and Tregs to favor autoimmune disease.


Frontiers in Immunology | 2011

Tolerance and lymphoid organ structure and function.

Bryna E. Burrell; Yaozhong Ding; Yumi Nakayama; Kyung Su Park; Jiangnan Xu; Na Yin; Jonathan S. Bromberg

This issue of Frontiers in Immunologic Tolerance explores barriers to tolerance from a variety of views of cells, molecules, and processes of the immune system. Our laboratory has spent over a decade focused on the migration of the cells of the immune system, and dissecting the signals that determine how and where effector and suppressive regulatory T cells traffic from one site to another in order to reject or protect allografts. These studies have led us to a greater appreciation of the anatomic structure of the immune system, and the realization that the path taken by lymphocytes during the course of the immune response to implanted organs determines the final outcome. In particular, the structures, microanatomic domains, and the cells and molecules that lymphocytes encounter during their transit through blood, tissues, lymphatics, and secondary lymphoid organs are powerful determinants for whether tolerance is achieved. Thus, the understanding of complex cellular and molecular processes of tolerance will not come from “96-well plate immunology,” but from an integrated understanding of the temporal and spatial changes that occur during the response to the allograft. The study of the precise positioning and movement of cells in lymphoid organs has been difficult since it is hard to visualize cells within their three-dimensional setting; instead techniques have tended to be dominated by two-dimensional renderings, although advanced confocal and two-photon systems are changing this view. It is difficult to precisely modify key molecules and events in lymphoid organs, so that existing knockouts, transgenics, inhibitors, and activators have global and pleiotropic effects, rather than precise anatomically restricted influences. Lastly, there are no well-defined postal codes or tracking systems for leukocytes, so that while we can usually track cells from point A to point B, it is exponentially more difficult or even impossible to track them to point C and beyond. We believe this represents one of the fundamental barriers to understanding the immune system and devising therapeutic approaches that take into account anatomy and structure as major controlling principles of tolerance.

Collaboration


Dive into the Na Yin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiangnan Xu

University of Maryland

View shared research outputs
Top Co-Authors

Avatar

Bernd Schröppel

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Girdhari Lal

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Barbara Murphy

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Nan Zhang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

M. Edward Medof

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Peter S. Heeger

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge