Tianyi Liu
Chinese PLA General Hospital
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Tianyi Liu.
Stem cell reports | 2016
Shuichi Kitayama; Rong Zhang; Tianyi Liu; Norihiro Ueda; Shoichi Iriguchi; Yutaka Yasui; Yohei Kawai; Minako Tatsumi; Norihito Hirai; Yasutaka Mizoro; Tatsuaki Iwama; Akira Watanabe; Mahito Nakanishi; Kiyotaka Kuzushima; Yasushi Uemura; Shin Kaneko
Vα24 invariant natural killer T (iNKT) cells are a subset of T lymphocytes implicated in the regulation of broad immune responses. They recognize lipid antigens presented by CD1d on antigen-presenting cells and induce both innate and adaptive immune responses, which enhance effective immunity against cancer. Conversely, reduced iNKT cell numbers and function have been observed in many patients with cancer. To recover these numbers, we reprogrammed human iNKT cells to pluripotency and then re-differentiated them into regenerated iNKT cells in vitro through an IL-7/IL-15-based optimized cytokine combination. The re-differentiated iNKT cells showed proliferation and IFN-γ production in response to α-galactosylceramide, induced dendritic cell maturation and downstream activation of both cytotoxic T lymphocytes and NK cells, and exhibited NKG2D- and DNAM-1-mediated NK cell-like cytotoxicity against cancer cell lines. The immunological features of re-differentiated iNKT cells and their unlimited availability from induced pluripotent stem cells offer a potentially effective immunotherapy against cancer.
Oncotarget | 2018
Hongyu Liu; Ling Chen; Yaojun Peng; Songyan Yu; Jialin Liu; Liangliang Wu; Lijun Zhang; Qiyan Wu; Xin Chang; Xinguang Yu; Tianyi Liu
Exosomes are vesicles that can be secreted by many types of cell and released into the extracellular space. Studies have found that tumor derived exosomes (TEXs) can promote tumor growth and metastasis, as well as inhibit immune response through transferring their genetic information to the recipient cells. Given their functions in tumor progression, TEXs are considered as promising biomarkers for early detection of human malignancy. Dendritic cells (DCs), a type of antigen presenting cells, can induce tumor-specific T cell immune responses in carcinogenesis. Growing evidences have demonstrated that the matured DCs induced by TEXs exhibit enhanced anti-tumor effects that may be applied for cancer immunotherapy. Thus in this review, according to the previous studies, we summarized the effects of DCs loaded with TEXs in cancer immunotherapy.
Neuroscience | 2017
Fei Shi; Hongchuan Guo; Rong Zhang; Hongyu Liu; Liangliang Wu; Qiyan Wu; Jialin Liu; Tianyi Liu; Qiuhang Zhang
Glioblastoma multiforme (GBM) is among the most lethal of all human tumors. It is the most frequently occurring malignant primary brain tumor in adults. The current standard of care (SOC) for GBM is initial surgical resection followed by treatment with a combination of temozolomide (TMZ) and ionizing radiation (IR). However, GBM has a dismal prognosis, and survivors have compromised quality of life owing to the adverse effects of radiation. GBM is characterized by overt activity of the phosphoinositide 3-kinase (PI3K) signaling pathway. GDC-0941 is a highly specific PI3K inhibitor with promising anti-tumor activity in human solid tumors. It is being evaluated in Phase II clinical trials for the treatment of breast and non-squamous cell lung cancer. We hypothesized that GDC-0941 may act as an antitumor agent and potentiate the effects of TMZ and IR. In this study, GDC-0941 alone induced cytotoxicity and pro-apoptotic effects. Moreover, combined with the standard GBM therapy (TMZ and IR), it suppressed cell viability, showed enhanced pro-apoptotic effects, augmented autophagy response, and attenuated migratory/invasive capacity in three glioma cell lines. Protein microarray analyses showed that treatment with TMZ+GDC-0941+IR induced higher levels of p53 and glycogen synthase kinase 3-beta (GSK3-β) expression in SHG44GBM cells than those induced by other treatments. This was verified in all cell lines by western blot analysis. Furthermore, the combination of TMZ and GDC-0941 with or without IR reduced the levels of p-AKT and O6-methylguanine DNA methyltransferase (MGMT) in T98G cells. The results of this study suggest that the combination of TMZ, IR, and GDC-0941 is a promising choice for future treatments of GBM.
Oncotarget | 2018
Fei Shi; Jinying Zhang; Hongyu Liu; Liangliang Wu; Hongyu Jiang; Qiyan Wu; Tianyi Liu; Meiqing Lou; Hao Wu
Glioblastomas (GBMs) are among the most malignant of all human tumors and have poor prognosis. The current standard of care (SOC) includes maximal surgical tumor resection followed by adjuvant temozolomide (TMZ) and concomitant radiotherapy (RT). However, even with this treatment, the 5-year survival rate is less than 10%, and thus, follow-up treatment is required to improve efficacy. In GBMs as well as many other solid cancers, PI3K/mTOR signaling is overactivated. Therefore, multiple tumor-based PI3K inhibitors have been studied in various cancers. In the current study, we investigated the effect of the dual PI3K/mTOR inhibitor dactolisib on TMZ+RT treatment in three human GBM cell lines and a orthotopic xenograft model. Dactolisib alone induced cytotoxicity and pro-apoptotic effects, which act as antitumor factors. Combined with SOC treatment, dactolisib inhibited cell viability, induced enhanced pro-apoptotic effect, and attenuated migration/invasion in all three cell lines, thereby enhancing the SOC therapeutic effect. Protein microarray analysis showed that A172 cells treated with TMZ+RT+dactolisib had higher p27 and lower Bcl-2 expression than other groups. Moreover, in the xenograft model, oral dactolisib combined with TMZ+RT inhibited tumor growth and prolonged survival. Thus, SOC combined with dactolisib shows potent anti-tumor activity and has promising potential for solid tumor treatment.
Cellular & Molecular Immunology | 2018
Norihiro Ueda; Rong Zhang; Minako Tatsumi; Tianyi Liu; Shuichi Kitayama; Yutaka Yasui; Shiori Sugai; Tatsuaki Iwama; Satoru Senju; Seiji Okada; Tetsuya Nakatsura; Kiyotaka Kuzushima; Hitoshi Kiyoi; Tomoki Naoe; Shin Kaneko; Yasushi Uemura
The advent of tyrosine kinase inhibitor (TKI) therapy markedly improved the outcome of patients with chronic-phase chronic myeloid leukemia (CML). However, the poor prognosis of patients with advanced-phase CML and the lifelong dependency on TKIs are remaining challenges; therefore, an effective therapeutic has been sought. The BCR–ABL p210 fusion protein’s junction region represents a leukemia-specific neoantigen and is thus an attractive target for antigen-specific T-cell immunotherapy. BCR–ABL p210 fusion-region-specific CD4+ T-helper (Th) cells possess antileukemic potential, but their function remains unclear. In this study, we established a BCR–ABL p210 b3a2 fusion-region-specific CD4+ Th-cell clone (b3a2-specific Th clone) and examined its dendritic cell (DC)-mediated antileukemic potential. The b3a2-specific Th clone recognized the b3a2 peptide in the context of HLA-DRB1*09:01 and exhibited a Th1 profile. Activation of this clone through T-cell antigen receptor stimulation triggered DC maturation, as indicated by upregulated production of CD86 and IL-12p70 by DCs, which depended on CD40 ligation by CD40L expressed on b3a2-specific Th cells. Moreover, in the presence of HLA-A*24:02-restricted Wilms tumor 1 (WT1)235–243 peptide, DCs conditioned by b3a2-specific Th cells efficiently stimulated the primary expansion of WTI-specific cytotoxic T lymphocytes (CTLs). The expanded CTLs were cytotoxic toward WT1235–243-peptide-loaded HLA-A*24:02-positive cell lines and exerted a potent antileukemic effect in vivo. However, the b3a2-specific Th-clone-mediated antileukemic CTL responses were strongly inhibited by both TKIs and interferon-α. Our findings indicate a crucial role of b3a2-specific Th cells in leukemia antigen-specific CTL-mediated immunity and provide an experimental basis for establishing novel CML immunotherapies.
Stem cell reports | 2018
Norihiro Ueda; Yasushi Uemura; Rong Zhang; Shuichi Kitayama; Shoichi Iriguchi; Yohei Kawai; Yutaka Yasui; Minako Tatsumi; Tatsuki Ueda; Tianyi Liu; Yasutaka Mizoro; Chihiro Okada; Akira Watanabe; Mahito Nakanishi; Satoru Senju; Yasuharu Nishimura; Kiyotaka Kuzushima; Hitoshi Kiyoi; Tomoki Naoe; Shin Kaneko
Summary CD4+ T helper (Th) cell activation is essential for inducing cytotoxic T lymphocyte (CTL) responses against malignancy. We reprogrammed a Th clone specific for chronic myelogenous leukemia (CML)-derived b3a2 peptide to pluripotency and re-differentiated the cells into original TCR-expressing T-lineage cells (iPS-T cells) with gene expression patterns resembling those of group 1 innate lymphoid cells. CD4 gene transduction into iPS-T cells enhanced b3a2 peptide-specific responses via b3a2 peptide-specific TCR. iPS-T cells upregulated CD40 ligand (CD40L) expression in response to interleukin-2 and interleukin-15. In the presence of Wilms tumor 1 (WT1) peptide, antigen-specific dendritic cells (DCs) conditioned by CD4-modified CD40Lhigh iPS-T cells stimulated WT1-specific CTL priming, which eliminated WT1 peptide-expressing CML cells in vitro and in vivo. Thus, CD4 modification of CD40Lhigh iPS-T cells generates innate lymphoid helper-like cells inducing bcr-abl-specific TCR signaling that mediates effectiveanti-leukemic CTL responses via DC maturation, showing potential for adjuvant immunotherapy against leukemia.
Oncotarget | 2018
Fei Shi; Jinying Zhang; Hongyu Liu; Liangliang Wu; Hongyu Jiang; Qiyan Wu; Tianyi Liu; Meiqing Lou; Hao Wu
[This corrects the article DOI: 10.18632/oncotarget.23091.].
Oncology Letters | 2018
Liangliang Wu; Hongyu Liu; Hongchuan Guo; Qiong Wu; Songyan Yu; Yuanyuan Qin; Gang Wang; Qiyan Wu; Rong Zhang; Lingxiong Wang; Lijun Zhang; Chunxi Liu; Shunchang Jiao; Tianyi Liu
Myeloid-derived suppressor cells (MDSCs) serve an immunosuppressive role in human tumors. Human Lin-/low human leukocyte antigen-antigen D related (HLA-DR-) cluster of differentiation (CD)-11b+CD33+ MDSCs are closely linked with tumor staging, progression, clinical therapeutic efficacy and prognosis for various types of tumors. The present study employed multiparametric flow cytometry to measure the proportion of Lin-/lowHLA-DR-CD11b+CD33+ MDSCs in the peripheral blood of 105 cervical cancer patients and 50 healthy subjects. The level of MDSC was higher in tumor patients than in the normal control group and this was closely associated with clinical staging. Further analysis of tumor-infiltrating MDSCs was performed in 22 patients. The MDSC proportions in tumor tissue were significantly higher than those in the corresponding adjacent tissue. The phenotypic characteristics of Lin-/lowHLA-DR-CD11b+CD33+ MDSCs were then evaluated and the results revealed that they express high CD13 and CD39, and low CD115, CD117, CD124 and programmed cell death ligand 1; they were also devoid of CD14, CD15 and CD66b. MDSCs and T-cells from peripheral blood were sorted by flow cytometry for co-culture experiments. Lin-/lowHLA-DR-CD11b+CD33+ MDSCs from patients significantly inhibited the proliferation of CD4 and CD8 T-cells. Furthermore, functional analysis verified that MDSCs from cervical cancer patients could inhibit interleukin-2 and interferon-γ production from T-cells. In addition, the associations between peripheral circulating MDSCs and tumor infiltrating MDSCs, and tumor relapse and metastasis were analyzed. The number of peripheral MDSCs and MDSCs in tumor tissue were observed to be associated with relapse-free survival. Thus, MDSCs in the peripheral blood and tumors of cervical cancer patients have a significant immunosuppressive effect, and are associated with cervical cancer staging and metastasis. These results suggest that targeting MDSCs may increase antitumor immunity and increase the efficacy of cervical cancer therapies.
Cellular & Molecular Immunology | 2010
Hongchuan Guo; Tianyi Liu; Yasushi Uemura; Shunchang Jiao; Deqing Wang; Zilin Lin; Yayoi Narita; Motoharu Suzuki; Narumi Hirosawa; Yasuko Ichihara; Osamu Ishihara; Hirosato Kikuchi; Yasushi Sakamoto; Satoru Senju; Qiuhang Zhang; Feng Ling
Cancer Letters | 2017
Hongyu Liu; Ling Chen; Jialin Liu; Hengxing Meng; Rong Zhang; Lin Ma; Liangliang Wu; Songyan Yu; Fei Shi; Ying Li; Lijun Zhang; Lingxiong Wang; Shiyu Feng; Qi Zhang; Yaojun Peng; Qiyan Wu; Chunxi Liu; Xin Chang; Lin Yang; Yasushi Uemura; Xinguang Yu; Tianyi Liu