Network


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

Hotspot


Dive into the research topics where Ende Zhao is active.

Publication


Featured researches published by Ende Zhao.


Journal of Immunology | 2011

IL-17+ Regulatory T Cells in the Microenvironments of Chronic Inflammation and Cancer

Ilona Kryczek; Ke Wu; Ende Zhao; Shuang Wei; Linhua Vatan; Wojciech Szeliga; Emina Huang; Joel K. Greenson; Alfred E. Chang; Jacek Roliński; Piotr Radwan; Jingyuan Fang; Guobin Wang; Weiping Zou

Foxp3+CD4+ regulatory T (Treg) cells inhibit immune responses and temper inflammation. IL-17+CD4+ T (Th17) cells mediate inflammation of autoimmune diseases. A small population of IL-17+Foxp3+CD4+ T cells has been observed in peripheral blood in healthy human beings. However, the biology of IL-17+Foxp3+CD4+ T cells remains poorly understood in humans. We investigated their phenotype, cytokine profile, generation, and pathological relevance in patients with ulcerative colitis. We observed that high levels of IL-17+Foxp3+CD4+ T cells were selectively accumulated in the colitic microenvironment and associated colon carcinoma. The phenotype and cytokine profile of IL-17+Foxp3+CD4+ T cells was overlapping with Th17 and Treg cells. Myeloid APCs, IL-2, and TGF-β are essential for their induction from memory CCR6+ T cells or Treg cells. IL-17+Foxp3+CD4+ T cells functionally suppressed T cell activation and stimulated inflammatory cytokine production in the colitic tissues. Our data indicate that IL-17+Foxp3+ cells may be “inflammatory” Treg cells in the pathological microenvironments. These cells may contribute to the pathogenesis of ulcerative colitis through inducing inflammatory cytokines and inhibiting local T cell immunity, and in turn may mechanistically link human chronic inflammation to tumor development. Our data therefore challenge commonly held beliefs of the anti-inflammatory role of Treg cells and suggest a more complex Treg cell biology, at least in the context of human chronic inflammation and associated carcinoma.


Science Translational Medicine | 2011

Human TH17 cells are long-lived effector memory cells.

Ilona Kryczek; Ende Zhao; Yan Liu; Yin Wang; Linhua Vatan; Wojciech Szeliga; Jeffrey Moyer; Aleksandra Klimczak; Andrzej Lange; Weiping Zou

Human TH17 cells function as long-lived effector memory cells in the context of chronic disease. Forever Seventeen “It” is that quality possessed by some which draws all others with its magnetic force. Novelist Elinor Glyn In teen television drama parlance, T helper 17 (TH17) cells have been immunology’s “it girl” for the past few years. Discovered in 2007, this beguiling subset of TH cells is associated with autoimmune disease and long-term antitumor immunity, and the presence of these cells in tumors positively correlates with patient survival. Although mouse TH17 cells are believed to be short-lived, studies of advanced human cancers suggest that human TH17 cells persist, but little is known about the nature of these cells in the context of human disease. Now, Kryczek et al. define the detailed phenotype and functional aspects of human TH17 cells in diverse diseased human tissues and find that these cells resemble terminally differentiated, long-lived memory T cells with a twist. The authors used a well-defined human system to study the defining features and behavior of TH17 cells in the pathological microenvironments of human graft-versus-host disease, ulcerative colitis, and cancer. The fraction of TH17 cells was increased in these tissues during the chronic phases of the diseases. Furthermore, the TH17 cells not only displayed characteristics in common with terminally differentiated memory T cells, but also sported some genetic and functional signatures of their own. For example, disease-associated human TH17 cells wore the standard phenotypic markers of terminal differentiation and, in adoptive transfer experiments, drove persistent antitumor immunity. However, these versatile human T cells also displayed a high capacity for proliferative self-renewal, cell-type plasticity, and enriched expression of antiapoptotic genes. The cell membrane signaling protein Notch and the transcriptional regulatory protein hypoxia-inducible factor–1α (HIF-1α) were shown to jointly regulate the expression and function of the Bcl-2 family of antiapoptosis proteins. An intricate understanding of human TH17 cell biology in disease contexts should reveal new “it” molecules—therapeutic targets that permit TH17-directed treatment in patients with autoimmune diseases and advanced tumors. T helper 17 (TH17) cells have been shown to contribute to multiple disease systems. However, the functional phenotype and survival pattern of TH17 cells as well as the underlying mechanisms that control TH17 cells have been poorly investigated in humans, significantly hampering the clinical targeting of these cells. Here, we studied human TH17 cells in the pathological microenvironments of graft-versus-host disease, ulcerative colitis, and cancer; TH17 cell numbers were increased in the chronic phase of these diseases. Human TH17 cells phenotypically resembled terminally differentiated memory T cells but were distinct from central memory, exhausted, and senescent T cells. Despite their phenotypic markers of terminal differentiation, TH17 cells mediated and promoted long-term antitumor immunity in in vivo adoptive transfer experiments. Furthermore, TH17 cells had a high capacity for proliferative self-renewal, potent persistence, and apoptotic resistance in vivo, as well as plasticity—converting into other types of TH cells. These cells expressed a relatively specific gene signature including abundant antiapoptotic genes. We found that hypoxia-inducible factor–1α and Notch collaboratively controlled key antiapoptosis Bcl-2 family gene expression and function in TH17 cells. Together, these data indicate that human TH17 cells may be a long-lived proliferating effector memory T cell population with unique genetic and functional characteristics. Targeting TH17-associated signaling pathway would be therapeutically meaningful for treating patients with autoimmune disease and advanced tumor.


Gastroenterology | 2014

Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells.

Shanshan Wan; Ende Zhao; Ilona Kryczek; Linda Vatan; Anna Sadovskaya; Gregory Ludema; Diane M. Simeone; Weiping Zou; Theodore H. Welling

BACKGROUND & AIMS Cancer stem cells (CSCs) can contribute to hepatocellular carcinoma (HCC) progression and recurrence after therapy. The presence of tumor-associated macrophages (TAMs) in patients with HCC is associated with poor outcomes. It is not clear whether TAMs interact with CSCs during HCC development. We investigated whether TAMs affect the activities of CSCs in the microenvironment of human HCCs. METHODS HCCs were collected from 17 patients during surgical resection and single-cell suspensions were analyzed by flow cytometry. CD14(+) TAMs were isolated from the HCC cell suspensions and placed into co-culture with HepG2 or Hep3B cells, and CSC functions were measured. The interleukin 6 (IL6) receptor was blocked with a monoclonal antibody (tocilizumab), and signal transducer and activator of transcription 3 was knocked down with small hairpin RNAs in HepG2 cells. Xenograft tumors were grown in NOD-SCID/Il2Rg(null) mice from human primary HCC cells or HepG2 cells. RESULTS CD44(+) cells from human HCCs and cell lines formed more spheres in culture and more xenograft tumors in mice than CD44(-) cells, indicating that CD44(+) cells are CSCs. Incubation of the CD44(+) cells with TAMs promoted expansion of CD44(+) cells, and increased their sphere formation in culture and formation of xenograft tumors in mice. In human HCC samples, the numbers of TAMs correlated with the numbers of CD44(+) cells. Of all cytokines expressed by TAMs, IL6 was increased at the highest level in human HCC co-cultures, compared with TAMs not undergoing co-culture. IL6 was detected in the microenvironment of HCC samples and induced expansion of CD44(+) cells in culture. Levels of IL6 correlated with stages of human HCCs and detection of CSC markers. Incubation of HCC cell lines with tocilizumab or knockdown of signal transducer and activator of transcription 3 in HCC cells reduced the ability of TAMs to promote sphere formation by CD44+ cells in culture and growth of xenograft tumors in mice. CONCLUSIONS CD44(+) cells isolated from human HCC tissues and cell lines have CSC activities in vitro and form a larger number of xenograft tumors in mice than CD44(-) cells. TAMs produce IL6, which promotes expansion of these CSCs and tumorigenesis. Levels of IL6 in human HCC samples correlate with tumor stage and markers of CSCs. Blockade of IL6 signaling with tocilizumab, a drug approved by the Food and Drug Administration for treatment of rheumatoid arthritis, inhibits TAM-stimulated activity of CD44(+) cells. This drug might be used to treat patients with HCC.


Immunity | 2014

IL-22(+)CD4(+) T cells promote colorectal cancer stemness via STAT3 transcription factor activation and induction of the methyltransferase DOT1L.

Ilona Kryczek; Yanwei Lin; Nisha Nagarsheth; Dongjun Peng; Lili Zhao; Ende Zhao; Linda Vatan; Wojciech Szeliga; Yali Dou; Scott R. Owens; Witold Zgodziński; Marek Majewski; Grzegorz Wallner; Jing-Yuan Fang; Emina Huang; Weiping Zou

Little is known about how the immune system impacts human colorectal cancer invasiveness and stemness. Here we detected interleukin-22 (IL-22) in patient colorectal cancer tissues that was produced predominantly by CD4(+) T cells. In a mouse model, migration of these cells into the colon cancer microenvironment required the chemokine receptor CCR6 and its ligand CCL20. IL-22 acted on cancer cells to promote activation of the transcription factor STAT3 and expression of the histone 3 lysine 79 (H3K79) methytransferase DOT1L. The DOT1L complex induced the core stem cell genes NANOG, SOX2, and Pou5F1, resulting in increased cancer stemness and tumorigenic potential. Furthermore, high DOT1L expression and H3K79me2 in colorectal cancer tissues was a predictor of poor patient survival. Thus, IL-22(+) cells promote colon cancer stemness via regulation of stemness genes that negatively affects patient outcome. Efforts to target this network might be a strategy in treating colorectal cancer patients.


Carcinogenesis | 2011

Th17 cells in cancer: help or hindrance?

Cailin Moira Wilke; Ilona Kryczek; Shuang Wei; Ende Zhao; Ke Wu; Guobin Wang; Weiping Zou

The role of CD4+ T helper (Th) 17 cells in malignancy is currently under debate. However, upon closer scrutiny, it becomes apparent that this discussion includes not only evaluations of Th17 cells but also IL-17+ cells from other immune populations, the cytokine interleukin (IL)-17 itself (both endogenous and exogenous) and IL-23. Further complicating the matter are occasionally conflicting results of studies in humans versus those in mice and contradictory data from immunocompetent versus immunodeficient mice. To better understand the role of Th17 cells in the tumor-bearing host, we focus first upon those studies investigating Th17 cells in patients and then those in mice, all the while keeping in mind that variables such as tumor-initiating agents, a pre-existing inflammatory environment and the immune competence of the host may have direct effects upon this T-cell subset. In this review, we will describe the phenotype of tumor-associated Th17 cells, review those studies that have examined the population directly, and finally, briefly discuss the studies involving Th17-associated signature cytokines.


International Journal of Cancer | 2010

Prognostic significance of regulatory T cells in tumor

Cailin Moira Wilke; Ke Wu; Ende Zhao; Guobin Wang; Weiping Zou

Since entering the immunological stage several decades ago, regulatory T cell biology has been realized as fundamentally important in the prevention of autoimmune conditions, induction of transplant tolerance and the immune response to cancer. The role of regulatory T cells in tumor immunobiology is still being elucidated. Currently, regulatory T cells are implicated in the dampening of antitumor T‐cell responses both through direct and indirect means. A number of investigators have demonstrated that regulatory T cell density and location may serve as independent prognostic factors in several types of cancer and are alternately detrimental or beneficial to patient survival. In this article, we will review the characteristics and functional phenotype of classical regulatory T cells, describe their distribution and quantification in tumor‐bearing hosts and summarize recent studies investigating the prognostic significance of regulatory T cell number and locality in various cancers.


Nature Immunology | 2016

Cancer mediates effector T cell dysfunction by targeting microRNAs and EZH2 via glycolysis restriction

Ende Zhao; Tomasz Maj; Ilona Kryczek; Wei Li; Ke Wu; Lili Zhao; Shuang Wei; Joel Crespo; Shanshan Wan; Linda Vatan; Wojciech Szeliga; Irene Shao; Yin Wang; Yan Liu; Sooryanarayana Varambally; Arul M. Chinnaiyan; Theodore H. Welling; Victor E. Marquez; Jan Kotarski; Hongbo Wang; Zehua Wang; Yi Zhang; Rebecca Liu; Guobin Wang; Weiping Zou

Aerobic glycolysis regulates T cell function. However, whether and how primary cancer alters T cell glycolytic metabolism and affects tumor immunity in cancer patients remains a question. Here we found that ovarian cancers imposed glucose restriction on T cells and dampened their function via maintaining high expression of microRNAs miR-101 and miR-26a, which constrained expression of the methyltransferase EZH2. EZH2 activated the Notch pathway by suppressing Notch repressors Numb and Fbxw7 via trimethylation of histone H3 at Lys27 and, consequently, stimulated T cell polyfunctional cytokine expression and promoted their survival via Bcl-2 signaling. Moreover, small hairpin RNA–mediated knockdown of human EZH2 in T cells elicited poor antitumor immunity. EZH2+CD8+ T cells were associated with improved survival in patients. Together, these data unveil a metabolic target and mechanism of cancer immune evasion.


Cellular & Molecular Immunology | 2012

Bone marrow and the control of immunity

Ende Zhao; Huanbin Xu; Lin Wang; Ilona Kryczek; Ke Wu; Yu Hu; Guobin Wang; Weiping Zou

Bone marrow is thought to be a primary hematopoietic organ. However, accumulated evidences demonstrate that active function and trafficking of immune cells, including regulatory T cells, conventional T cells, B cells, dendritic cells, natural killer T (NKT) cells, neutrophils, myeloid-derived suppressor cells and mesenchymal stem cells, are observed in the bone marrow. Furthermore, bone marrow is a predetermined metastatic location for multiple human tumors. In this review, we discuss the immune network in the bone marrow. We suggest that bone marrow is an immune regulatory organ capable of fine tuning immunity and may be a potential therapeutic target for immunotherapy and immune vaccination.


OncoImmunology | 2012

Regulatory T cells in the bone marrow microenvironment in patients with prostate cancer

Ende Zhao; Lin Wang; Jinlu Dai; Ilona Kryczek; Shuang Wei; Linda Vatan; Saleh Altuwaijri; Tim Sparwasser; Guobin Wang; Evan T. Keller; Weiping Zou

Human prostate cancer frequently metastasizes to bone marrow. What defines the cellular and molecular predilection for prostate cancer to metastasize to bone marrow is not well understood. CD4+CD25+ regulatory T (Treg) cells contribute to self-tolerance and tumor immune pathology. We now show that functional Treg cells are increased in the bone marrow microenvironment in prostate cancer patients with bone metastasis, and that CXCR4/CXCL12 signaling pathway contributes to Treg cell bone marrow trafficking. Treg cells exhibit active cell cycling in the bone marrow, and bone marrow dendritic cells express high levels of receptor activator of NFκB (RANK), and promote Treg cell expansion through RANK and its ligand (RANKL) signals. Furthermore, Treg cells suppress osteoclast differentiation induced by activated T cells and M-CSF, adoptive transferred Treg cells migrate to bone marrow, and increase bone mineral intensity in the xenograft mouse models with human prostate cancer bone marrow inoculation. In vivo Treg cell depletion results in reduced bone density in tumor bearing mice. The data indicates that bone marrow Treg cells may form an immunosuppressive niche to facilitate cancer bone metastasis and contribute to bone deposition, the major bone pathology in prostate cancer patients with bone metastasis. These findings mechanistically explain why Treg cells accumulate in the bone marrow, and demonstrate a previously unappreciated role for Treg cells in patients with prostate cancer. Thus, targeting Treg cells may not only improve anti-tumor immunity, but also ameliorate bone pathology in prostate cancer patients with bone metastasis.


Cancer Research | 2016

PRC2 Epigenetically Silences Th1-Type Chemokines to Suppress Effector T-Cell Trafficking in Colon Cancer

Nisha Nagarsheth; Dongjun Peng; Ilona Kryczek; Ke Wu; Wei Li; Ende Zhao; Lili Zhao; Shuang Wei; Timothy L. Frankel; Linda Vatan; Wojciech Szeliga; Yali Dou; Scott R. Owens; Victor E. Marquez; Kaixiong Tao; Emina Huang; Guobin Wang; Weiping Zou

Infiltration of tumors with effector T cells is positively associated with therapeutic efficacy and patient survival. However, the mechanisms underlying effector T-cell trafficking to the tumor microenvironment remain poorly understood in patients with colon cancer. The polycomb repressive complex 2 (PRC2) is involved in cancer progression, but the regulation of tumor immunity by epigenetic mechanisms has yet to be investigated. In this study, we examined the relationship between the repressive PRC2 machinery and effector T-cell trafficking. We found that PRC2 components and demethylase JMJD3-mediated histone H3 lysine 27 trimethylation (H3K27me3) repress the expression and subsequent production of Th1-type chemokines CXCL9 and CXCL10, mediators of effector T-cell trafficking. Moreover, the expression levels of PRC2 components, including EZH2, SUZ12, and EED, were inversely associated with those of CD4, CD8, and Th1-type chemokines in human colon cancer tissue, and this expression pattern was significantly associated with patient survival. Collectively, our findings reveal that PRC2-mediated epigenetic silencing is not only a crucial oncogenic mechanism, but also a key circuit controlling tumor immunosuppression. Therefore, targeting epigenetic programs may have significant implications for improving the efficacy of current cancer immunotherapies relying on effective T-cell-mediated immunity at the tumor site.

Collaboration


Dive into the Ende Zhao's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weiping Zou

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Guobin Wang

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Linda Vatan

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ke Wu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Shuang Wei

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Kaixiong Tao

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Lili Zhao

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge