Network


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

Hotspot


Dive into the research topics where Maogen Chen is active.

Publication


Featured researches published by Maogen Chen.


Arthritis & Rheumatism | 2013

Adoptive Transfer of Human Gingiva‐Derived Mesenchymal Stem Cells Ameliorates Collagen‐Induced Arthritis via Suppression of Th1 and Th17 Cells and Enhancement of Regulatory T Cell Differentiation

Maogen Chen; Wenru Su; Xiaohong Lin; Zhiyong Guo; Julie Wang; Qunzhou Zhang; David D. Brand; Bernhard Ryffel; Jiefu Huang; Zhongmin Liu; Xiaoshun He; Anh D. Le; Song Guo Zheng

OBJECTIVE Current approaches offer no cures for rheumatoid arthritis (RA). Accumulating evidence has revealed that manipulation of bone marrow-derived mesenchymal stem cells (BM-MSCs) may have the potential to control or even prevent RA, but BM-MSC-based therapy faces many challenges, such as limited cell availability and reduced clinical feasibility. This study in mice with established collagen-induced arthritis (CIA) was undertaken to determine whether substitution of human gingiva-derived mesenchymal stem cells (G-MSCs) would significantly improve the therapeutic effects. METHODS CIA was induced in DBA/1J mice by immunization with type II collagen and Freunds complete adjuvant. G-MSCs were injected intravenously into the mice on day 14 after immunization. In some experiments, intraperitoneal injection of PC61 (anti-CD25 antibody) was used to deplete Treg cells in arthritic mice. RESULTS Infusion of G-MSCs in DBA/1J mice with CIA significantly reduced the severity of arthritis, decreased the histopathology scores, and down-regulated the production of inflammatory cytokines (interferon-γ and interleukin-17A). Infusion of G-MSCs also resulted in increased levels of CD4+CD39+FoxP3+ cells in arthritic mice. These increases were noted early after infusion in the spleens and lymph nodes, and later after infusion in the synovial fluid. The FoxP3+ Treg cells that were increased in frequency mainly consisted of Helios-negative cells. When Treg cells were depleted, infusion of G-MSCs partially interfered with the progression of CIA. Pretreatment of G-MSCs with a CD39 or CD73 inhibitor significantly reversed the protective effect of G-MSCs on CIA. CONCLUSION The role of G-MSCs in controlling the development and severity of CIA mostly depends on CD39/CD73 signals and partially depends on the induction of CD4+CD39+FoxP3+ Treg cells. G-MSCs provide a promising approach for the treatment of autoimmune diseases.


Arthritis & Rheumatism | 2012

Antigen-specific transforming growth factor β-induced Treg cells, but not natural Treg cells, ameliorate autoimmune arthritis in mice by shifting the Th17/Treg cell balance from Th17 predominance to Treg cell predominance.

Ning Kong; Qin Lan; Maogen Chen; Julie Wang; Wei Shi; David A. Horwitz; Valerie Quesniaux; Bernhard Ryffel; Zhongmin Liu; David D. Brand; Hejian Zou; Song Guo Zheng

OBJECTIVE Transferred CD4+CD25+FoxP3+ Treg cells can prevent autoimmune disease, but generally fail to ameliorate established disease. This study was undertaken to compare the effects of antigen-specific Treg cells induced with interleukin-2 (IL-2) and transforming growth factor β (TGFβ) ex vivo (induced Treg [iTreg] cells) to the effects of equivalent expanded thymus-derived natural Treg (nTreg) cells on established collagen-induced arthritis (CIA). METHODS CIA was induced in DBA/1 mice by immunization with type II collagen (CII), and before or shortly after immunization, mice were treated with iTreg or nTreg cells that were generated or expanded in vitro. Clinical scores were determined. Inflammatory responses were determined by measuring the levels of anti-CII antibody in the serum and examining the histologic features of the mouse joints. The Th1/Th17-mediated autoreactive response was evaluated by determining the cytokine profile of the draining lymph node (LN) cells of the mice by flow cytometry. RESULTS Following transfer, nTreg cells exhibited decreased FoxP3 and Bcl-2 expression and decreased suppressive activity, and many converted to Th17 cells. In contrast, transferred iTreg cells were more numerous, retained FoxP3 expression and their suppressive activity in the presence of IL-6, and were resistant to Th17 conversion. Notably, 10 days after the transfer of donor iTreg cells, predominance was shifted from Th17 cells to Treg cells in the draining LNs of recipient mice. CONCLUSION These findings provide evidence that transferred TGFβ-induced iTreg cells are more stable and functional than nTreg cells in mice with established autoimmunity. Moreover, iTreg cells can have tolerogenic effects even in the presence of ongoing inflammation. The therapeutic potential of human iTreg cells in subjects with chronic, immune-mediated inflammatory diseases should be investigated.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Critical role of all-trans retinoic acid in stabilizing human natural regulatory T cells under inflammatory conditions

Ling Lu; Qin Lan; Zhiyuan Li; Xiaohui Zhou; Jian Gu; Qiang Li; Julie Wang; Maogen Chen; Ya Liu; Yi Shen; David D. Brand; Bernhard Ryffel; David A. Horwitz; Francisco P. Quismorio; Zhongmin Liu; Bin Li; Nancy J. Olsen; Song Guo Zheng

Significance Natural regulatory T cells (nTregs) play important roles in preventing autoimmune diseases, but they may be unstable in the presence of inflammation. Here we report that all-trans RA (atRA) but not rapamycin prevents human nTregs from converting to Th1/Th17 cells and sustains their suppressive function in inflammatory environments. Adoptive transfer of nTregs pretreated with atRA enhances their suppressive effects on xenograft-vs.-host diseases. Moreover, we show that atRA suppresses IL-1 receptor upregulation, accelerates IL-6 receptor downregulation, and affects the epigenetic modifications in Foxp3 locus in nTregs following inflammatory stimulation. We suggest that nTregs primed with atRA may represent a novel treatment strategy to control established chronic immune-mediated diseases. Recent studies have demonstrated that thymus-derived naturally occurring CD4+Foxp3+ regulatory T cells (Tregs) in human and mouse may be unstable and dysfunctional in the presence of proinflammatory cytokines. All-trans RA (atRA), the active derivative of vitamin A, has been shown to regulate Treg and T effector cell differentiation. We hypothesize atRA stabilizes human natural Tregs (nTregs) under inflammatory conditions. atRA prevents human nTregs from converting to Th1 and/or Th17 cells and sustains their Foxp3 expression and suppressive function in vitro or in vivo following encounters with IL-1 and IL-6. Interestingly, adoptive transfer of human nTregs pretreated with atRA significantly enhanced their suppressive effects on xenograft-vs.-host diseases (xGVHDs), and atRA- but not rapamycin-pretreated nTregs sustained the functional activity against xGVHD after stimulation with IL-1/IL-6. atRA suppresses IL-1 receptor (IL-1R) up-regulation, accelerates IL-6R down-regulation, and diminishes their signaling events as well as prevents the up-regulation of STIP1 homology and U-Box containing protein 1 on Foxp3+ cells following IL-1/IL-6 stimulation. atRA also increases histone acetylation on Foxp3 gene promoter and CpG demethylation in the region of Foxp3 locus (i.e., Treg-specific demethylated region). These results strongly implicate that nTregs primed with atRA may represent a novel treatment strategy to control established chronic immune-mediated autoimmune and inflammatory diseases.


PLOS ONE | 2011

All-trans retinoic acid promotes TGF-β-induced Tregs via histone modification but not DNA demethylation on Foxp3 gene locus.

Ling Lu; Jilin Ma; Zhiyuan Li; Qin Lan; Maogen Chen; Ya Liu; Zanxian Xia; Julie Wang; Yuan-Ping Han; Wei Shi; Valerie Quesniaux; Bernhard Ryffel; David D. Brand; Bin Li; Zhongmin Liu; Song Guo Zheng

Background It has been documented all-trans retinoic acid (atRA) promotes the development of TGF-β-induced CD4+Foxp3+ regulatory T cells (iTreg) that play a vital role in the prevention of autoimmune responses, however, molecular mechanisms involved remain elusive. Our objective, therefore, was to determine how atRA promotes the differentiation of iTregs. Methodology/Principal Findings Addition of atRA to naïve CD4+CD25− cells stimulated with anti-CD3/CD28 antibodies in the presence of TGF-β not only increased Foxp3+ iTreg differentiation, but maintained Foxp3 expression through apoptosis inhibition. atRA/TGF-β-treated CD4+ cells developed complete anergy and displayed increased suppressive activity. Infusion of atRA/TGF-β-treated CD4+ cells resulted in the greater effects on suppressing symptoms and protecting the survival of chronic GVHD mice with typical lupus-like syndromes than did CD4+ cells treated with TGF-β alone. atRA did not significantly affect the phosphorylation levels of Smad2/3 and still promoted iTreg differentiation in CD4+ cells isolated from Smad3 KO and Smad2 conditional KO mice. Conversely, atRA markedly increased ERK1/2 activation, and blockade of ERK1/2 signaling completely abolished the enhanced effects of atRA on Foxp3 expression. Moreover, atRA significantly increased histone methylation and acetylation within the promoter and conserved non-coding DNA sequence (CNS) elements at the Foxp3 gene locus and the recruitment of phosphor-RNA polymerase II, while DNA methylation in the CNS3 was not significantly altered. Conclusions/Significance We have identified the cellular and molecular mechanism(s) by which atRA promotes the development and maintenance of iTregs. These results will help to enhance the quantity and quality of development of iTregs and may provide novel insights into clinical cell therapy for patients with autoimmune diseases and those needing organ transplantation.


PLOS ONE | 2012

Adoptive Transfer of Induced-Treg Cells Effectively Attenuates Murine Airway Allergic Inflammation

Wei Xu; Qin Lan; Maogen Chen; Hui Chen; Ning Zhu; Xiaohui Zhou; Julie Wang; Huimin Fan; Chun-Song Yan; Jiu-Long Kuang; David Warburton; Dieudonnée Togbe; Bernhard Ryffel; Song Guo Zheng; Wei Shi

Both nature and induced regulatory T (Treg) lymphocytes are potent regulators of autoimmune and allergic disorders. Defects in endogenous Treg cells have been reported in patients with allergic asthma, suggesting that disrupted Treg cell-mediated immunological regulation may play an important role in airway allergic inflammation. In order to determine whether adoptive transfer of induced Treg cells generated in vitro can be used as an effective therapeutic approach to suppress airway allergic inflammation, exogenously induced Treg cells were infused into ovalbumin-sensitized mice prior to or during intranasal ovalbumin challenge. The results showed that adoptive transfer of induced Treg cells prior to allergen challenge markedly reduced airway hyperresponsiveness, eosinophil recruitment, mucus hyper-production, airway remodeling, and IgE levels. This effect was associated with increase of Treg cells (CD4+FoxP3+) and decrease of dendritic cells in the draining lymph nodes, and with reduction of Th1, Th2, and Th17 cell response as compared to the controls. Moreover, adoptive transfer of induced Treg cells during allergen challenge also effectively attenuate airway inflammation and improve airway function, which are comparable to those by natural Treg cell infusion. Therefore, adoptive transfer of in vitro induced Treg cells may be a promising therapeutic approach to prevent and treat severe asthma.


Annals of the Rheumatic Diseases | 2012

Induced T regulatory cells suppress osteoclastogenesis and bone erosion in collagen-induced arthritis better than natural T regulatory cells

Ning Kong; Qin Lan; Maogen Chen; Tina Zheng; Wenru Su; Julie Wang; Ziyan Yang; Ryan Park; Grant Dagliyan; Peter S. Conti; David D. Brand; Zhongmin Liu; Hejian Zou; William Stohl; Song Guo Zheng

Background Although natural regulatory T cells (nTregs) can suppress osteoclastogenesis, the role of TGF-β-induced CD4+Foxp3+ Tregs (iTregs) in osteoclastogenesis remains unknown. Objective To determine the effects of iTregs on osteoclastogenesis in vitro and on bone erosion in vivo in collagen-induced arthritis (CIA). Methods Osteoclastogenesis was induced in bone marrow CD11b+ cells with receptor activator of nuclear factor κ B (NF-κB) ligand (RANKL) and macrophage colony stimulating factor. Graded doses of Tregs were added to inhibit osteoclastogenesis. Transwell and antibody blockade experiments were performed to assess the roles for cell contact and soluble cytokines. NF-κB activation was determined by western blot. iTregs or nTregs were adoptively transferred to mice with CIA to assess in vivo effects on disease incidence and bone erosion, the latter determined by CT scanning. Results Both nTregs and iTregs greatly suppressed osteoclastogenesis in vitro, but only iTregs sustained this effect when interleukin-6 was present. iTregs, but not nTregs, significantly suppressed development of CIA. Bone erosions in iTregs-treated mice were diminished compared with untreated mice or nTregs-treated mice. Treatment with iTregs, but not with nTregs, dramatically decreased NF-κB p65/p50 levels in osteoclasts in vitro and p65/50 and RANKL expression by synovial tissues in vivo. Conclusion iTregs may be therapeutically beneficial in rheumatoid arthritis and related diseases associated with bone erosions.


Cellular & Molecular Immunology | 2012

The development and function of follicular helper T cells in immune responses

Maogen Chen; Zhiyong Guo; Weiqiang Ju; Bernhard Ryffel; Xiaoshun He; Song Guo Zheng

Follicular helper T cells (Tfh) have been referred as a lineage that provides a help for B cells to proliferate and undergo antibody affinity maturation in the germinal center. Evidence has supported that Tfh subset development, like other lineages, is dependent on microenvironment where a particular transcriptional program is initiated. It has been shown that Bcl-6 and IL-21 act as master regulators for the development and function of Tfh cells. Tfh dysregulation is involved in the development of autoimmune pathologies, such as systemic lupus erythematosus, rheumatoid arthritis and other autoimmune diseases. The present review highlights the recent advances in the field of Tfh cells and focus on their development and function.


Journal of Molecular Cell Biology | 2014

Phenotypic and functional characteristic of a newly identified CD8+Foxp3−CD103+ regulatory T cells

Ya Liu; Qin Lan; Ling Lu; Maogen Chen; Zanxian Xia; Jilin Ma; Julie Wang; Huimin Fan; Yi Shen; Bernhard Ryffel; David D. Brand; Francisco P. Quismorio; Zhongmin Liu; David A. Horwitz; A. Xu; Song Guo Zheng

TGF-β and Foxp3 expressions are crucial for the induction and functional activity of CD4(+)Foxp3(+) regulatory T (iTreg) cells. Here, we demonstrate that although TGF-β-primed CD8(+) cells display much lower Foxp3 expression, their suppressive capacity is equivalent to that of CD4(+) iTreg cells, and both Foxp3(-) and Foxp3(+) CD8+ subsets have suppressive activities in vitro and in vivo. CD8(+)Foxp3(-) iTreg cells produce little IFN-γ but almost no IL-2, and display a typical anergic phenotype. Among phenotypic markers expressed in CD8(+)Foxp3(-) cells, we identify CD103 expression particularly crucial for the generation and function of this subset. Moreover, IL-10 and TGF-β signals rather than cytotoxicity mediate the suppressive effect of this novel Treg population. Therefore, TGF-β can induce both CD8(+)Foxp3(-) and CD8(+)Foxp3(+) iTreg subsets, which may represent the unique immunoregulatory means to treat autoimmune and inflammatory diseases.


The Journal of Allergy and Clinical Immunology | 2012

Induced CD4+ forkhead box protein–positive T cells inhibit mast cell function and established contact hypersensitivity through TGF-β1

Wenru Su; Huimin Fan; Maogen Chen; Julie Wang; David D. Brand; Xiaoshun He; Valerie Quesniaux; Bernhard Ryffel; Ling Zhu; Dan Liang; Song Guo Zheng

BACKGROUND Induced CD4(+) forkhead box protein 3-positive regulatory T (iTreg) cells are a promising source for cell-based therapies of established inflammatory and autoimmune diseases. However, their relationship to mast cell (MC) function and MC-driven diseases remains unknown. OBJECTIVE We sought to explore the roles of iTreg cells on MC function and the established MC-driven disease contact hypersensitivity (CHS). METHODS In vitro coculture studies were carried out to investigate the interaction between iTreg cells in murine or human MCs by using both direct cell-cell contact and transwell systems to separate cell-cell contact. In vivo mice iTreg cells were administered to mice with established CHS, and innate immunologic responses, such as MC infiltration and inflammatory cytokine expression at contact sites, were evaluated. RESULTS In vitro coculture under direct cell-cell contact resulted in indirect suppression of IgE-independent activation of MCs by murine or human iTreg cells. Mechanistically, iTreg cells suppressed proinflammatory cytokine levels by modulating nuclear factor κB p65 activation in MCs through T cell-derived TGF-β1. Injection of iTreg cells but not natural CD4(+)CD25(+) regulatory T cells into animals with established CHS resulted in the suppression of infiltration and functions of MCs and also led to decreased production of inflammatory cytokines at allergic contact areas. iTreg cell-mediated immunosuppressive effects were abrogated when iTreg cells were pretreated with TGF-β1 small interfering RNA. CONCLUSIONS Our study demonstrates that iTreg cells suppress MC function and attenuate established MC-driven CHS through TGF-β1-dependent mechanisms.


Cytokine & Growth Factor Reviews | 2014

The function of BAFF on T helper cells in autoimmunity

Maogen Chen; Xiaohong Lin; Ya Liu; Qiang Li; Yiling Deng; Zhongmin Liu; David D. Brand; Zhiyong Guo; Xiaoshun He; Bernhard Ryffel; Song Guo Zheng

B cell-activating factor belonging to the TNF family (BAFF) exerts its pathogenic role in supporting the survival and proliferation of B cells, regulating class switch recombination as well as the selection of autoreactive B cells. Overexpression of BAFF induces a dramatic expansion of activated B cells, particularly marginal zone B cells, as well as hypergammaglobulinemia, autoantibody production and immune complex deposition. However, in addition to its effect on B cells, recent work has also demonstrated that BAFF can promote T cell activation, proliferation and differentiation. In this review, we have discussed the recent progress on the function and role of BAFF on T cells and T cell-mediated diseases.

Collaboration


Dive into the Maogen Chen's collaboration.

Top Co-Authors

Avatar

Song Guo Zheng

Penn State Milton S. Hershey Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiaoshun He

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

David D. Brand

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Julie Wang

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhiyong Guo

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Qin Lan

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Linwei Wu

Sun Yat-sen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge