Network


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

Hotspot


Dive into the research topics where Pingyan Cheng is active.

Publication


Featured researches published by Pingyan Cheng.


Journal of Experimental Medicine | 2008

Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein

Pingyan Cheng; Cesar A. Corzo; Noreen Luetteke; Bin Yu; Srinivas Nagaraj; Marylin M. Bui; Myrna L. Ortiz; Wolfgang Nacken; Clemens Sorg; Thomas Vogl; J. Roth; Dmitry I. Gabrilovich

Accumulation of myeloid-derived suppressor cells (MDSCs) associated with inhibition of dendritic cell (DC) differentiation is one of the major immunological abnormalities in cancer and leads to suppression of antitumor immune responses. The molecular mechanism of this phenomenon remains unclear. We report here that STAT3-inducible up-regulation of the myeloid-related protein S100A9 enhances MDSC production in cancer. Mice lacking this protein mounted potent antitumor immune responses and rejected implanted tumors. This effect was reversed by administration of wild-type MDSCs from tumor-bearing mice to S100A9-null mice. Overexpression of S100A9 in cultured embryonic stem cells or transgenic mice inhibited the differentiation of DCs and macrophages and induced accumulation of MDSCs. This study demonstrates that tumor-induced up-regulation of S100A9 protein is critically important for accumulation of MDSCs and reveals a novel molecular mechanism of immunological abnormalities in cancer.


Journal of Experimental Medicine | 2010

HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment.

Cesar A. Corzo; Thomas Condamine; Lily Lu; Matthew J. Cotter; Je In Youn; Pingyan Cheng; Hyun Il Cho; Esteban Celis; David Quiceno; Tapan A. Padhya; Thomas V. McCaffrey; Judith C. McCaffrey; Dmitry I. Gabrilovich

The hypoxic environment of tumors dictates the phenotype of local myeloid-derived suppressor cells (MDSCs) via HIF-1a expression; hypoxia converts splenic MDSCs from specific into nonspecific suppressors.


Journal of Immunology | 2009

Mechanism Regulating Reactive Oxygen Species in Tumor-Induced Myeloid-Derived Suppressor Cells

Cesar A. Corzo; Matthew J. Cotter; Pingyan Cheng; Fendong Cheng; Sergei Kusmartsev; Eduardo M. Sotomayor; Tapan A. Padhya; Thomas V. McCaffrey; Judith C. McCaffrey; Dmitry I. Gabrilovich

Myeloid-derived suppressor cells (MDSC) are a major component of the immune suppressive network described in cancer and many other pathological conditions. Recent studies have demonstrated that one of the major mechanisms of MDSC-induced immune suppression is mediated by reactive oxygen species (ROS). However, the mechanism of this phenomenon remained unknown. In this study, we observed a substantial up-regulation of ROS by MDSC in all of seven different tumor models and in patients with head and neck cancer. The increased ROS production by MDSC is mediated by up-regulated activity of NADPH oxidase (NOX2). MDSC from tumor-bearing mice had significantly higher expression of NOX2 subunits, primarily p47phox and gp91phox, compared with immature myeloid cells from tumor-free mice. Expression of NOX2 subunits in MDSC was controlled by the STAT3 transcription factor. In the absence of NOX2 activity, MDSC lost the ability to suppress T cell responses and quickly differentiated into mature macrophages and dendritic cells. These findings expand our fundamental understanding of the biology of MDSC and may also open new opportunities for therapeutic regulation of these cells in cancer.


Journal of Immunology | 2004

Hyperactivation of STAT3 Is Involved in Abnormal Differentiation of Dendritic Cells in Cancer

Yulia Nefedova; Mei Huang; Sergei Kusmartsev; Raka Bhattacharya; Pingyan Cheng; Raoul Salup; Richard Jove; Dmitry I. Gabrilovich

Abnormal differentiation of myeloid cells is one of the hallmarks of cancer. However, the molecular mechanisms of this process remain elusive. In this study, we investigated the effect of tumor-derived factors on Janus kinase (Jak)/STAT signaling in myeloid cells during their differentiation into dendritic cells. Tumor cell conditioned medium induced activation of Jak2 and STAT3, which was associated with an accumulation of immature myeloid cells. Jak2/STAT3 activity was localized primarily in these myeloid cells, which prevented the differentiation of immature myeloid cells into mature dendritic cells. This differentiation was restored after removal of tumor-derived factors. Inhibition of STAT3 abrogated the negative effects of these factors on myeloid cell differentiation, and overexpression of STAT3 reproduced the effects of tumor-derived factors. Thus, this is a first demonstration that tumor-derived factors may affect myeloid cell differentiation in cancer via constitutive activation of Jak2/STAT3.


Journal of Immunology | 2001

Notch-1 Regulates NF-κB Activity in Hemopoietic Progenitor Cells

Pingyan Cheng; Andrew Zlobin; Veronica Volgina; Sridevi Gottipati; Barbara A. Osborne; Erica J. Simel; Lucio Miele; Dmitry I. Gabrilovich

We investigated the interaction between two elements critical for differentiation of hemopoietic cells, the Notch-1 receptor and the transcription factor NF-κB. These factors were studied in hemopoietic progenitor cells (HPC) using Notch-1 antisense transgenic (Notch-AS-Tg) mice. DNA binding of NF-κB as well as its ability to activate transcription was strongly decreased in HPC from Notch-AS-Tg mice. NF-κB-driven transcriptional activity was completely restored after transduction of the cells with retroviral constructs containing activated Notch-1 gene. HPC from Notch-AS-Tg mice have decreased levels of several members of the NF-κB family, p65, p50, RelB, and c-Rel and this is due to down-regulation of the gene expression. To investigate functional consequences of decreased NF-κB activity in transgenic mice, we studied LPS-induced proliferation of B cells and GM-CSF-dependent differentiation of dendritic cells from HPC. These two processes are known to be closely dependent on NF-κB. B cells from Notch-AS-Tg mice had almost 3-fold lower response to LPS than B cells isolated from control mice. Differentiation of dendritic cells was significantly affected in Notch-AS-Tg mice. However, it was restored by transduction of activated Notch-1 into HPC. Taken together, these data indicate that in HPC NF-κB activity is regulated by Notch-1 via transcriptional control of NF-κB.


Journal of Clinical Investigation | 2011

Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice

Tangying Lu; Rupal Ramakrishnan; Soner Altiok; Je In Youn; Pingyan Cheng; Esteban Celis; Vladimir Pisarev; Simon Sherman; Michael B. Sporn; Dmitry I. Gabrilovich

Cancer immunotherapeutic approaches induce tumor-specific immune responses, in particular CTL responses, in many patients treated. However, such approaches are clinically beneficial to only a few patients. We set out to investigate one possible explanation for the failure of CTLs to eliminate tumors, specifically, the concept that this failure is not dependent on inhibition of T cell function. In a previous study, we found that in mice, myeloid-derived suppressor cells (MDSCs) are a source of the free radical peroxynitrite (PNT). Here, we show that pre-treatment of mouse and human tumor cells with PNT or with MDSCs inhibits binding of processed peptides to tumor cell-associated MHC, and as a result, tumor cells become resistant to antigen-specific CTLs. This effect was abrogated in MDSCs treated with a PNT inhibitor. In a mouse model of tumor-associated inflammation in which the antitumor effects of antigen-specific CTLs are eradicated by expression of IL-1β in the tumor cells, we determined that therapeutic failure was not caused by more profound suppression of CTLs by IL-1β-expressing tumors than tumors not expressing this proinflammatory cytokine. Rather, therapeutic failure was a result of the presence of PNT. Clinical relevance for these data was suggested by the observation that myeloid cells were the predominant source of PNT in human lung, pancreatic, and breast cancer samples. Our data therefore suggest what we believe to be a novel mechanism of MDSC-mediated tumor cell resistance to CTLs.


Nature Immunology | 2013

Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer

Je-In Youn; Vinit Kumar; Michelle Collazo; Yulia Nefedova; Thomas Condamine; Pingyan Cheng; Alejandro Villagra; Scott Antonia; Judith C. McCaffrey; Mayer Fishman; Amod Sarnaik; Pedro Horna; Eduardo M. Sotomayor; Dmitry I. Gabrilovich

Two major populations of myeloid-derived suppressor cells (MDSCs), monocytic MDSCs (M-MDSCs) and polymorphonuclear MDSCs (PMN-MDSCs) regulate immune responses in cancer and other pathologic conditions. Under physiologic conditions, Ly6ChiLy6G− inflammatory monocytes, which are the normal counterpart of M-MDSCs, differentiate into macrophages and dendritic cells. PMN-MDSCs are the predominant group of MDSCs that accumulates in cancer. Here we show that a large proportion of M-MDSCs in tumor-bearing mice acquired phenotypic, morphological and functional features of PMN-MDSCs. Acquisition of this phenotype, but not the functional attributes of PMN-MDSCs, was mediated by transcriptional silencing of the retinoblastoma gene through epigenetic modifications mediated by histone deacetylase 2 (HDAC-2). These data demonstrate a new regulatory mechanism of myeloid cells in cancer.


Journal of Immunology | 2005

Activation of Dendritic Cells via Inhibition of Jak2/STAT3 Signaling

Yulia Nefedova; Pingyan Cheng; Daniele M. Gilkes; Michelle A. Blaskovich; Amer A. Beg; Said M. Sebti; Dmitry I. Gabrilovich

Signaling via Jak2/STAT3 is critically important for normal dendritic cell (DC) differentiation. In addition, we have previously demonstrated that hyperactivation of the Jak2/STAT3 pathway induced by tumor-derived factors (TDF) may be responsible for abnormal DC differentiation in cancer. In this study, using a novel selective inhibitor of Jak2/STAT3, JSI-124, we investigated the mechanism of the Jak2/STAT3 effect on DCs and the possibility of pharmacological regulation of DC differentiation in cancer. Our experiments have demonstrated that JSI-124 overcomes the differentiation block induced by TDF and promotes the differentiation of mature DCs and macrophages. Surprisingly, inhibition of Jak2/STAT3 signaling resulted in dramatic activation of immature DCs generated in the presence of TDF as well as in control medium. This activation manifested in up-regulation of MHC class II, costimulatory molecules, and a dramatic increase in the ability to stimulate allogeneic or Ag-specific T cells. Inhibition of Jak2/STAT3 signaling resulted in activation of the transcription factor NF-κB. This up-regulation was not due to a conventional pathway involving IκBα, but was probably due to a block of the dominant negative effect of STAT3. This indicates that Jak2/STAT3 play an important role in negative regulation of DC activation, and pharmacological inhibition of the Jak2/STAT3 pathway can be used to enhance DC function.


Journal of Clinical Investigation | 2013

Induction of myelodysplasia by myeloid-derived suppressor cells

Xianghong Chen; Erika A. Eksioglu; Junmin Zhou; Ling Zhang; Julie Y. Djeu; Nicole Fortenbery; Pearlie K. Epling-Burnette; Sandra van Bijnen; Harry Dolstra; John P. Cannon; Je-In Youn; Sarah S. Donatelli; Dahui Qin; Theo de Witte; Jianguo Tao; Huaquan Wang; Pingyan Cheng; Dmitry I. Gabrilovich; Alan F. List; Sheng Wei

Myelodysplastic syndromes (MDS) are age-dependent stem cell malignancies that share biological features of activated adaptive immune response and ineffective hematopoiesis. Here we report that myeloid-derived suppressor cells (MDSC), which are classically linked to immunosuppression, inflammation, and cancer, were markedly expanded in the bone marrow of MDS patients and played a pathogenetic role in the development of ineffective hematopoiesis. These clonally distinct MDSC overproduce hematopoietic suppressive cytokines and function as potent apoptotic effectors targeting autologous hematopoietic progenitors. Using multiple transfected cell models, we found that MDSC expansion is driven by the interaction of the proinflammatory molecule S100A9 with CD33. These 2 proteins formed a functional ligand/receptor pair that recruited components to CD33’s immunoreceptor tyrosine-based inhibition motif (ITIM), inducing secretion of the suppressive cytokines IL-10 and TGF-β by immature myeloid cells. S100A9 transgenic mice displayed bone marrow accumulation of MDSC accompanied by development of progressive multilineage cytopenias and cytological dysplasia. Importantly, early forced maturation of MDSC by either all-trans-retinoic acid treatment or active immunoreceptor tyrosine-based activation motif–bearing (ITAM-bearing) adapter protein (DAP12) interruption of CD33 signaling rescued the hematologic phenotype. These findings indicate that primary bone marrow expansion of MDSC driven by the S100A9/CD33 pathway perturbs hematopoiesis and contributes to the development of MDS.


Immunity | 2016

CD45 Phosphatase Inhibits STAT3 Transcription Factor Activity in Myeloid Cells and Promotes Tumor-Associated Macrophage Differentiation.

Vinit Kumar; Pingyan Cheng; Thomas Condamine; Sridevi Mony; Lucia R. Languino; Judith C. McCaffrey; Neil G. Hockstein; Michael J. Guarino; Gregory A. Masters; Emily Penman; Fred Denstman; Xiaowei Xu; Dario C. Altieri; Hong Du; Cong Yan; Dmitry I. Gabrilovich

Recruitment of monocytic myeloid-derived suppressor cells (MDSCs) and differentiation of tumor-associated macrophages (TAMs) are the major factors contributing to tumor progression and metastasis. We demonstrated that differentiation of TAMs in tumor site from monocytic precursors was controlled by downregulation of the activity of the transcription factor STAT3. Decreased STAT3 activity was caused by hypoxia and affected all myeloid cells but was not observed in tumor cells. Upregulation of CD45 tyrosine phosphatase activity in MDSCs exposed to hypoxia in tumor site was responsible for downregulation of STAT3. This effect was mediated by the disruption of CD45 protein dimerization regulated by sialic acid. Thus, STAT3 has a unique function in the tumor environment in controlling the differentiation of MDSC into TAM, and its regulatory pathway could be a potential target for therapy.

Collaboration


Dive into the Pingyan Cheng's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Je-In Youn

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Cesar A. Corzo

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jie Zhou

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Barbara A. Osborne

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge