Network


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

Hotspot


Dive into the research topics where Fengyang Lei is active.

Publication


Featured researches published by Fengyang Lei.


Cellular Immunology | 2009

T lineage differentiation from induced pluripotent stem cells.

Fengyang Lei; Rizwanul Haque; Lauren Weiler; Kent E. Vrana; Jianxun Song

Induced pluripotent stem (iPS) cells have potential to differentiate into T lymphocytes, however, the actual ability of iPS cells to develop into T lineages is not clear. In this study, we co-cultured iPS cells on OP9 cells expressing the Notch ligand Delta-like 1 (DL1), the iPS cells differentiated into T lymphocytes. In addition, in vitro stimulation of iPS cell-derived T lymphocytes resulted in secretion of IL-2 and IFN-gamma. Moreover, adoptive transfer of iPS cell-derived T lymphocytes into Rag-deficient mice reconstituted their T cell pools. These results indicate that iPS cells are able to follow the normal program of T cell differentiation.


Cancer Research | 2011

In Vivo Programming of Tumor Antigen-Specific T Lymphocytes from Pluripotent Stem Cells to Promote Cancer Immunosurveillance

Fengyang Lei; Baohua Zhao; Rizwanul Haque; Xiaofang Xiong; Lynn R. Budgeon; Neil D. Christensen; Yuzhang Wu; Jianxun Song

Adoptive T-cell immunotherapy has garnered wide attention, but its effective use is limited by the need of multiple ex vivo manipulations and infusions that are complex and expensive. In this study, we show how highly reactive antigen (Ag)-specific CTLs can be generated from induced pluripotent stem (iPS) cells to provide an unlimited source of functional CTLs for adoptive immunotherapy. iPS cell-derived T cells can offer the advantages of avoiding possible immune rejection and circumventing ethical and practical issues associated with other stem cell types. iPS cells can be differentiated into progenitor T cells in vitro by stimulation with the Notch ligand Delta-like 1 (DL1) overexpressed on bone marrow stromal cells, with complete maturation occurring upon adoptive transfer into Rag1-deficient mice. Here, we report that these iPS cells can be differentiated in vivo into functional CTLs after overexpression of MHC I-restricted Ag-specific T-cell receptors (TCR). In this study, we generated murine iPS cells genetically modified with ovalbumin (OVA)-specific and MHC-I restricted TCR (OT-I) by retrovirus-mediated transduction. After their adoptive transfer into recipient mice, the majority of OT-I/iPS cells underwent differentiation into CD8+ CTLs. TCR-transduced iPS cells developed in vivo responded in vitro to peptide stimulation by secreting interleukin 2 and IFN-γ. Most importantly, adoptive transfer of TCR-transduced iPS cells triggered infiltration of OVA-reactive CTLs into tumor tissues and protected animals from tumor challenge. Taken together, our findings offer proof of concept for a potentially more efficient approach to generate Ag-specific T lymphocytes for adoptive immunotherapy.


Journal of Immunology | 2012

Programming of Regulatory T Cells from Pluripotent Stem Cells and Prevention of Autoimmunity

Rizwanul Haque; Fengyang Lei; Xiaofang Xiong; Yanqing Bian; Baohua Zhao; Yuzhang Wu; Jianxun Song

Regulatory T (Treg) cells are being used to treat autoimmunity and prevent organ rejection; however, Treg cell-based therapies have been hampered by the technical limitation in obtaining a high number of functional Treg cells. In this study, we show how to generate functional Treg cells from induced pluripotent stem (iPS) cells and to determine the potential role of such cells for Treg cell-based immunotherapy against autoimmunity in a therapeutic setting. Ligation of a Notch ligand and transduction of the gene Foxp3 induce iPS cells to differentiate into Treg cells. Expression of Foxp3 and coculture on Notch ligand-expressing stromal cells augment expression of CD3, TCR, CD4, CD25, and CTLA-4 on iPS cell-differentiated Treg cells, which are able to secrete TGF-β and IL-10 both in vivo and in vitro. Importantly, adoptive transfer of iPS cell-derived Treg cells expressing large amounts of Foxp3 and Bcl-xL significantly suppresses host immune responses and reduces arthritis development within murine models. These data suggest that Notch signaling and Foxp3 regulate the development and function of Treg cells derived from iPS cells. Our results provide a novel approach for generating potentially therapeutic Treg cells for the treatment of autoimmune diseases.


Frontiers in Oncology | 2014

Utilizing regulatory T cells against rheumatoid arthritis

Mohammad Haque; Kristin Fino; Fengyang Lei; Xiaofang Xiong; Jianxun Song

Regulatory T (Treg) cells are essential for normal immune surveillance systems, and their dysfunction leads to development of diseases, such as autoimmune disorders. CD4+CD25+ Treg cells are well-known suppressive cells, which express the transcription factor Foxp3, are indispensable for the maintenance of immune self-tolerance and homeostasis by suppressing aberrant or excessive immune response. Other Foxp3− Treg cells include Tr1, Th3, CD8+CD28−/−, and Qa1-restricted T cells; however, the contribution of these Treg cells to self-tolerance, immune homeostasis as well as preventing autoimmunity is not well defined. Here, we discuss the phenotypes and function of Foxp3+ Treg cells and the potential use of such Treg cells against rheumatoid arthritis (RA). Of note, even though most expanded populations of Foxp3+ Treg cells exhibit suppressive activity, tissue-associated or antigen-specific Treg cells appear superior in suppressing local autoimmune disorders such as RA. In addition, utilizing tissue-associated Foxp3+ Treg cells from stem cells may stable Foxp3 expression and avoid induction of a potentially detrimental systemic immunosuppression.


Journal of Immunology | 2009

Cooperation between Molecular Targets of Costimulation in Promoting T Cell Persistence and Tumor Regression

Baohua Zhao; Aihua Song; Rizwanul Haque; Fengyang Lei; Lauren Weiler; Xiaofang Xiong; Yuzhang Wu; Michael Croft; Jianxun Song

Costimulation regulates multiple cellular processes of T cells inducing proliferation, expansion, and survival. The molecular targets of costimulation might then be useful to augment T cell activities. Two defined targets of costimulatory signals in primary T cells are the anti-apoptotic bcl-2 family molecule Bcl-xL, and survivin, an inhibitor of apoptosis family member that might regulate both cell division and survival. However, the relative importance of, and relationship between, these molecules in primary T cells is not clear. To understand whether they have overlapping or cooperative functions, we used retrovirus-mediated transduction to introduce Bcl-xL and survivin separately, or together linked by a 2A picornavirus self-cleaving peptide, into Ag-responding CD8+ T cells. We found that CD8+ effector T cells expressing both Bcl-xL and survivin strongly expanded at an early stage and had a long-term survival advantage over cells transduced with either molecule alone. In vivo, with response to tumor-expressed Ag following adoptive T cell transfer, Ag-reactive CD8+ T cells expressing both Bcl-xL and survivin displayed greatly enhanced tumor protective activity compared with CD8+ T cells expressing either molecule introduced separately. These results indicate that Bcl-xL and survivin can critically contribute in a cooperative, nonredundant manner to augment the accumulation and persistence of CD8+ T cells following encounter with Ag. The data provide new insights into why costimulatory signals might need to be sustained over time and suggest a potential novel approach to augment cellular immunotherapy for cancer.


Journal of Visualized Experiments | 2012

Directed Differentiation of Induced Pluripotent Stem Cells towards T Lymphocytes

Fengyang Lei; Rizwanul Haque; Xiaofang Xiong; Jianxun Song

Adoptive cell transfer (ACT) of antigen-specific CD8(+) cytotoxic T lymphocytes (CTLs) is a promising treatment for a variety of malignancies (1). CTLs can recognize malignant cells by interacting tumor antigens with the T cell receptors (TCR), and release cytotoxins as well as cytokines to kill malignant cells. It is known that less-differentiated and central-memory-like (termed highly reactive) CTLs are the optimal population for ACT-based immunotherapy, because these CTLs have a high proliferative potential, are less prone to apoptosis than more differentiated cells and have a higher ability to respond to homeostatic cytokines (2-7). However, due to difficulties in obtaining a high number of such CTLs from patients, there is an urgent need to find a new approach to generate highly reactive Ag-specific CTLs for successful ACT-based therapies. TCR transduction of the self-renewable stem cells for immune reconstitution has a therapeutic potential for the treatment of diseases (8-10). However, the approach to obtain embryonic stem cells (ESCs) from patients is not feasible. Although the use of hematopoietic stem cells (HSCs) for therapeutic purposes has been widely applied in clinic (11-13), HSCs have reduced differentiation and proliferative capacities, and HSCs are difficult to expand in in vitro cell culture (14-16). Recent iPS cell technology and the development of an in vitro system for gene delivery are capable of generating iPS cells from patients without any surgical approach. In addition, like ESCs, iPS cells possess indefinite proliferative capacity in vitro, and have been shown to differentiate into hematopoietic cells. Thus, iPS cells have greater potential to be used in ACT-based immunotherapy compared to ESCs or HSCs. Here, we present methods for the generation of T lymphocytes from iPS cells in vitro, and in vivo programming of antigen-specific CTLs from iPS cells for promoting cancer immune surveillance. Stimulation in vitro with a Notch ligand drives T cell differentiation from iPS cells, and TCR gene transduction results in iPS cells differentiating into antigen-specific T cells in vivo, which prevents tumor growth. Thus, we demonstrate antigen-specific T cell differentiation from iPS cells. Our studies provide a potentially more efficient approach for generating antigen-specific CTLs for ACT-based therapies and facilitate the development of therapeutic strategies for diseases.


PLOS ONE | 2013

Regulation of A1 by OX40 Contributes to CD8+ T Cell Survival and Anti-Tumor Activity

Fengyang Lei; Jianyong Song; Rizwanul Haque; Mohammad Haque; Xiaofang Xiong; Deyu Fang; Michael Croft; Jianxun Song

The TNFR family member OX40 (CD134) is critical for optimal clonal expansion and survival of T cells. However, the intracellular targets of OX40 in CD8 T cells are not fully understood. Here we show that A1, a Bcl-2 family protein, is regulated by OX40 in effector CD8 T cells. In contrast to wild-type T cells, OX40-deficient CD8 T cells failed to maintain A1 expression driven by antigen. Conversely, enforced OX40 stimulation promoted A1 expression. In both situations, the expression of A1 directly correlated with CD8 T cell survival. In addition, exogenous expression of A1 in OX40-deficient CD8 T cells reversed their survival defect in vitro and in vivo. Moreover, forced expression of A1 in CD8 T cells from OX40-deficient mice restored the ability of these T cells to suppress tumor growth in a murine model. These results indicate that OX40 signals regulate CD8 T cell survival at least in part through maintaining expression of the anti-apoptotic molecule A1, and provide new insight into the mechanism by which OX40 may impact anti-tumor immunity.


European Journal of Immunology | 2013

Transgenic expression of survivin compensates for OX40‐deficiency in driving Th2 development and allergic inflammation

Fengyang Lei; Jianyong Song; Rizwanul Haque; Xiaofang Xiong; Deyu Fang; Yuzhang Wu; Susanne M. A. Lens; Michael Croft; Jianxun Song

Survivin, an inhibitor of apoptosis family molecule, has been proposed as a crucial intermediate in the signaling pathways leading to T‐cell development, proliferation, and expansion. However, the importance of survivin to T‐cell‐driven inflammatory responses has not been demonstrated. Here, we show that survivin transgenic mice exhibit an increased antigen‐driven Th2 lung inflammation and that constitutive expression of survivin reversed the defective lung inflammation even in the absence of OX40 costimulation. We found that OX40‐deficient mice were compromised in generating Th2 cells, airway eosinophilia, and IgE responses. In contrast, OX40‐deficient/survivin transgenic mice generated normal Th2 responses and exhibited strong lung inflammation. These results suggest that OX40 costimulation crucially engages survivin during antigen‐mediated Th2 responses. These findings also promote the notion that OX40 costimulation regulates allergic responses or lung inflammation by targeting survivin thereby enhancing T‐cell proliferation and resulting in more differentiated Th2 cells in the allergic inflammatory response.


Endocrine‚ Metabolic & Immune Disorders-Drug Targets | 2011

The Regulation of FoxP3-Expressing Regulatory T Cells

Rizwanul Haque; Fengyang Lei; Xiaofang Xiong; Jianxun Song

Regulatory T (Treg) cells play an important role in the maintenance of self-tolerance and are involved in the prevention of autoimmune diseases and reduce/inhibit the progression of chronic inflammatory diseases. Forkhead box P3 (FoxP3) expression in Treg cells is believed to be a critical factor in the maintenance of Treg cells suppressive function. Multiple mechanisms of action of Treg cells have been proposed, including cell contact-dependent and cytokine-dependent mechanisms. However, no clear picture is available to fully elucidate the regulation of FoxP3 gene expression in Treg cells and how FoxP3-expressing Treg cells mediate the immune response in vivo. This review will discuss the research advancements in Treg cell biology including the transcription factors and signaling pathways involved in the expression of FoxP3 gene as well as the advancements in the understanding of the factors involved in the Treg cell-mediated suppressive mechanisms.


Scientific Reports | 2016

Stem cell-derived tissue-associated regulatory T cells ameliorate the development of autoimmunity.

Mohammad Haque; Jianyong Song; Kristin Fino; Praneet Sandhu; Xinmeng Song; Fengyang Lei; Song Guo Zheng; Bing Ni; Deyu Fang; Jianxun Song

Pluripotent stem cells (PSCs) have the potential to produce almost all of the cells in the body, including regulatory T cells (Tregs). However, the exact conditions required for the development of antigen (Ag)-specific Tregs from PSCs (i.e., PSC-Tregs) are not well delineated. Ag-specific PSC-Tregs can be tissue/organ-associated and migrate to local inflamed tissues/organs to suppress the autoimmune response after adoptive transfer, thereby avoiding potential overall immunosuppression from non-specific Tregs. In this study, we developed a new approach to generate functional Ag-specific Tregs from induced PSCs (iPSCs), i.e., iPSC-Tregs, which had the ability to generate an Ag-specific immunosuppressive response in a murine model of arthritis. We retrovirally transduced murine iPSCs with a construct containing genes of Ag-specific T cell receptor (TCR) and the transcriptional factor FoxP3. We differentiated the iPSCs into Ag-specific iPSC-Tregs using in vitro or in vivo Notch signaling, and demonstrated that adoptive transfer of such Tregs dramatically suppressed autoimmunity in a well-established Ag-induced arthritis model, including the inflammation, joint destruction, cartilage prostaglandin depletion, osteoclast activity, and Th17 production. Our results indicate that PSCs can be used to develop Ag-specific Tregs, which have a therapeutic potential for Treg-based therapies of autoimmune disorders.

Collaboration


Dive into the Fengyang Lei's collaboration.

Top Co-Authors

Avatar

Jianxun Song

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Rizwanul Haque

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Xiaofang Xiong

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Mohammad Haque

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Deyu Fang

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Jianyong Song

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yuzhang Wu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Kristin Fino

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Michael Croft

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Praneet Sandhu

Pennsylvania State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge