Network


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

Hotspot


Dive into the research topics where Xiubao Ren is active.

Publication


Featured researches published by Xiubao Ren.


Tumor Biology | 2014

Long noncoding RNA HOTAIR involvement in cancer

Yansheng Wu; Li Zhang; Yang Wang; Hui Li; Xiubao Ren; Feng Wei; Wenwen Yu; Xudong Wang; Lun Zhang; Jinpu Yu; Xishan Hao

Evidences have been provided that long noncoding RNAs (lncRNAs) act as key molecules in epigenetic regulation and are involved in the development process of cancer in recent studies. HOX transcript antisense RNA (HOTAIR), a long intergenic noncoding RNA (lincRNA), functions as a molecular scaffold to link and target two histone modification complexes PRC2 and LSD1, then reprograms chromatin states by couples histone H3K27 methylation and H3K4 demethylation for epigenetic gene silencing to promote cancer metastasis. HOTAIR, regarded as an oncogene, is pervasively overexpressed in most solid cancers and correlated with tumor invasion, progression, metastasis, and poor prognosis, and HOTAIR has been proven to play a critical role in most biological process of cancer and would be a potential new target in cancer therapy.


Cancer Immunology, Immunotherapy | 2013

Enhanced antitumor effects of DC-activated CIKs to chemotherapy treatment in a single cohort of advanced non-small-cell lung cancer patients.

Lili Yang; Baozhu Ren; Hui Li; Jinpu Yu; Shui Cao; Xishan Hao; Xiubao Ren

Cytokine-induced killer (CIK) cells show cytolytic activity against tumor. The purpose of this study was to evaluate the antitumor effect of dendritic cell (DC)-activated CIK cells in vitro and their clinical efficacy of DC-activated CIK cells in combination with chemotherapy (abbreviated below as chemotherapy plus DCxa0+xa0CIK) in patients with advanced non-small-cell lung cancer (NSCLC). A paired study was performed between 61 patients treated with chemotherapy alone (group 1) and 61 patients treated with chemotherapy plus DCxa0+xa0CIK cells (group 2). In group 2, 36 patients with adenocarcinoma and 18 patients with squamous cell carcinoma were analyzed for the survival rate. Compared to unstimulated CIK cells, DC-activated CIK cells significantly enhanced antitumor activity, increased the ratio of CD3+CD56+ cells, promoted cell proliferation and lessened cell apoptosis. In the paired study, the 1- and 2-year overall survival rates in group 2 were 57.2 and 27.0xa0%, which were significantly higher than that of group 1 (37.3 and 10.1xa0%) (Pxa0<xa00.05). There was no significant difference in the survival rate between the adenocarcinoma and squamous carcinoma patients in group 2. The present study suggests that DC-activated CIK cell has enhanced antitumor effects and chemotherapy plus DCxa0+xa0CIK cells improved the clinical outcomes of chemotherapy for advanced NSCLC patients.


Journal of Clinical Immunology | 2014

A randomized phase II study of autologous cytokine-induced killer cells in treatment of hepatocelluar carcinoma

Xiaozhou Yu; Hua Zhao; Liang Liu; Shui Cao; Baozhu Ren; Naining Zhang; Xiumei An; Jinpu Yu; Hui Li; Xiubao Ren

PurposeThis prospective study aims to explore the benefit of cytokine-induced killer cell (CIK) treatment in hepatocellular carcinoma patients, which has not yet been thoroughly studied before.MethodsFrom January 2004 to May 2009, 132 patients who were initially diagnosed with hepatocellular carcinoma of Barcelona Clinic Liver Cancer (BCLC) stage A, B or C, Child–Pugh scores of A or B and without prior treatment were enrolled in the study. Patients were randomly assigned to either arm 1 (nu2009=u200966) to receive CIK treatment plus standard treatment, or arm 2 (nu2009=u200966) to receive standard treatment only. The primary end point was overall survival (OS) and the secondary endpoint was progression-free survival as evaluated by Kaplan–Meier analyses and treatment hazard ratios with the Cox proportional hazards model.ResultsThe 1-year (OS: 74.2xa0% vs. 50.0xa0%, 95xa0% CI: 63.6–84.8xa0% vs. 37.8–62.2, pu2009=u20090.002), 2-year (OS: 53.0xa0% vs. 30.3xa0%, 95xa0% CI: 40.8–65.2xa0% vs. 19.1–41.5xa0%, pu2009=u20090.002), 3-year (OS: 42.4xa0% vs. 24.2xa0%, 95xa0% CI: 30.4–54.4xa0% vs. 13.8–34.6xa0%, pu2009=u20090.005) and median overall and progression-free survivals of arm 1 patients were significantly higher than those of arm 2. Therefore, in patients who are not suitable for surgery, significant benefit is obtained from CIK treatment. The main adverse effects of CIK included fever, allergy and headache pain.ConclusionsHepatocellular carcinoma patients who were not suitable for surgery demonstrate prolonged overall and progression-free survival from CIK treatment.


Human Immunology | 2015

Regulatory B cell: New member of immunosuppressive cell club

Tingting Ding; Fan Yan; Shui Cao; Xiubao Ren

Historically, the pivotal role of B cells or B lymphocytes in immunity has been attributed to the production of antibodies. They were also demonstrated to present antigens to T cells and to secrete cytokines, thereby acting as positive regulators in immune responses. A series of studies on autoimmune diseases, however, led researchers to find a unique subset of B cells, later described as regulatory B cells (Bregs), that has the ability to suppress immune responses. Bregs occur not only in autoimmune diseases, but also in inflammation and transplantation. Furthermore, recently published literatures suggested that Bregs contributed to the growth and metastasis of certain cancers. In this review, we will discuss these unique subsets of B cells in different kinds of disorders, with particular emphasis on the mechanisms of their immunoregulatory role that were collected from mice and humans.


Cancer Immunology, Immunotherapy | 2015

Expression of TNFR2 by regulatory T cells in peripheral blood is correlated with clinical pathology of lung cancer patients.

Fan Yan; Ruijuan Du; Feng Wei; Hua Zhao; Jinpu Yu; Changli Wang; Zhongli Zhan; Tingting Ding; Xiubao Ren; Xin Chen; Hui Li

CD4+FoxP3+ regulatory T cells (Tregs) represent a major cellular mediator of cancer immune evasion. The expression of tumor necrosis factor receptor type II (TNFR2) on Tregs is reported to identify the maximally suppressive Treg population in both mice and human. We therefore investigated the phenotype and function of TNFR2+ Tregs present in the peripheral blood (PB) of 43 lung cancer patients. Further, the association of TNFR2 expression on Tregs with clinicopathological factors was analyzed. The results showed that in the PB of lung cancer patients, Tregs expressed markedly higher levels of TNFR2 than conventional T cells (Tconvs). Expression of TNFR2 appeared to correlate better than CD25+ and CD127− with FoxP3 expression. PB TNFR2+ Tregs in lung cancer patients were more proliferative and expressed higher levels of the immunosuppressive molecule CTLA-4, and consequently more potently suppressed IFNγ production by cocultured CD8 CTLs. More importantly, higher TNFR2 expression levels on Tregs were associated with lymphatic invasion, distant metastasis and more advanced clinical stage of lung cancer patients. Therefore, our study suggests that TNFR2+ Tregs play a role in promoting tumor progressive metastasis and expression of TNFR2 by PB Tregs may prove to be a useful prognostic marker in lung cancer patients.


Cytotherapy | 2008

Beneficial effects of fetal–maternal microchimerism on the activated haplo-identical peripheral blood stem cell treatment for cancer

Jinpu Yu; Xiubao Ren; Shui Cao; Huikai Li; Xishan Hao

Background Previous studies have shown that activated haplo-identical peripheral blood stem cell (haplo-PBSC) treatment exerts an anti-tumor effect on patients with metastatic solid tumors. The purpose of this study was to test the hypothesis that fetal-maternal microchimerism enhances the beneficial effect of the haplo-PBSC treatment for cancer. Methods Twenty-five patients with advanced-stage solid tumors refractory to standard chemotherapy were treated with haplo-PBSC donated by parents or children. Fetal-maternal microchimerism status was determined using nested polymerase chain reaction typing using sequence-specific primers (PCR-SSP). Clinical outcomes, including therapeutic response by measuring tumor size changes using CT scanning and survival times, were evaluated. The donor-recipient mixed lymphocyte response (MLR) was detected using an MTT proliferation assay. Cytokine production was determined using an ELISA method. Results Six patients receiving maternal-child transplants were fetal-maternal microchimerism positive (+). The mean survival time of patients with the microchimerism(+) haplo-PBSC treatment was 30.17+/-5.32 months (median 17 months), which was significantly longer than that of patients with negative (-) microchimerism (mean 16.95+/-3.29 months, median 8 months; P=0.043). The therapeutic response rate was significantly higher in microchimerism(+) patients (83.3%) than that in microchimerism(-) patients (36.8%) (P=0.047). Furthermore, suppression of donor-recipient MLR and increased production of a T-helper type 1 (Th1) type cytokine, interferon (IFN)-gamma, were found in microchimerism(+) patients after haplo-PBSC treatment. Discussion This small study suggests that fetal-maternal microchimerism is associated with a statistically significant improvement in anti-tumor effect of activated haplo-PBSC treatment. Further study is required to elucidate the mechanism for this observation.


Journal of Cancer Research and Clinical Oncology | 2002

Induction of specific CTL by MAGE-3/CEA peptide-pulsed dendritic cells from HLA-A2/A24+ gastrointestinal cancer patients

Xishan Hao; Ying Shao; Xiubao Ren; Hong Liu; Qi Xu; Hui Li; Peng Zhang; Xiumei An; Baozhu Ren

Abstractn Purpose. In this study, we aimed to investigate whether MAGE3/CEA peptide-pulsed dendritic cells could induce specific cytotoxic T lymphocytes (CTL).n Methods. In this pilot study, we selected 25 patients expressing MAGE-3-HLA-A2/A24 or CEA-HLA-A24. Patients dendritic cells (DCs) were expanded in vitro in the presence of recombined-human granular macrophage colony stimulating factor (rhGM-CSF) and recombined-human interleukin 4 (rhIL-4), pulsed with MAGE-3/CEA (HLA-A2/A24) peptide. The cytolytic cells activity, induced by peptide-pulsed DCs and unpurified T cells as effector cells, and with Mel526, 803, Raji, and K562 as target cells, were measured using LDH-releasing assay.n Results. DCs were obtained by in vitro expansion in all cases although DC harvest rates varied among different patients (7.1±3.2%). Compared with T-IL-2 (IL-2-induced T cells), T-DC-P – which resulted from T-IL-2 co-cultured with DCs pulsed by MAGE3 or CEA peptides – exhibited an increase in cytolytic activity against Mel526 (expressing MAGE-3-HLA-A2) and 803 (expressing CEA-HLA-A24) cell lines by about 25–30% (P<0.01). In contrast, there was no significant difference between the activity against Raji and K562 cells, which are negative for both peptides.n Conclusions. This study showed that combined usage of rhGM-CSF and rhIL-4 in vitro could expand DCs, and that the DCs pulsed with specific peptides could induce MAGE- and CEA-specific CTL responses. The DC-based vaccine may provide an important method for the immunological treatment of gastrointestinal cancers.


Protein Expression and Purification | 2008

Expression and purification of recombinant human interleukin-18 protein using a yeast expression system

Lili Yang; Xiumei An; Feng Wei; Hong Liu; Hui Li; Jinpu Yu; Xiubao Ren

Interleukin-18 (IL-18) has been reported to exert significant immunoregulatory effects on inhibiting tumor growth through stimulating natural killer (NK) cell cytotoxicity and promoting production of several cytokines, including interferon-gamma (IFN-gamma) and granulocyte/macrophage colony-stimulating factor (GM-CSF). Therefore, IL-18 might serve as a potential therapeutic target for cancer treatment. However, the resource of this protein limits its availability for the clinical practice. The purpose of this study was to express and purify recombinant human (h) IL-18 protein using a yeast expression system. We reported here that hIL-18 gene was cloned into pPICZaC vector for expressing a recombinant hIL-18 protein using a yeast expression system. The recombinant hIL-18 protein was purified using centrifugal filter devices, hydrophobic chromatography, and anion exchange chromatography. The yield and purity of the recombinant hIL-18 reached 45.1% and 97.6%, respectively. This recombinant hIL-18 was shown to induce IFN-gamma production by human peripheral blood mononuclear cells (PBMCs) and enhance NK cell cytotoxicity synergistically with IL-2. Furthermore, these recombinant hIL-18-induced effects were the same as those by standard hIL-18. Therefore, the yeast expression system used in this study provides a useful method to produce large-scale of hIL-18 for the clinical application.


BMC Surgery | 2015

Functional jejunal interposition, a reconstruction procedure, promotes functional outcomes after total gastrectomy

Xuewei Ding; Fang Yan; Han Liang; Qiang Xue; Kuo Zhang; Hui Li; Xiubao Ren; Xishan Hao

BackgroundFunctional jejunal interposition (FJI) has been applied as a reconstruction procedure to maintain the jejunal continuity and duodenal food passage after total gastrectomy in patients with gastric cancer. The purpose of this study was to evaluate clinical efficacy of the FJI procedure by comparing the functional outcomes of FJI to Roux-en-Y after total gastrectomy in gastric cancer patients, and investigate physiologic mechanisms by which FJI exerts beneficial outcomes in beagles.MethodsPatients with stage I-IV gastric cancer without metastasis and recurrence one year after surgery were enrolled in this retrospective study. Seventy one patients received FJI and seventy nine patients received Roux-en-Y after total gastrectomy. We evaluated the nutritional status at three and twelve months and incidence of complications up to twelve months after surgery. Beagles receiving sham operation, FJI, or Roux-en-Y after total gastrectomy were sacrificed forty eight hours postoperatively. Beagles were gavaged with active carbon for evaluating the intestinal transit rate. Intestinal tissues from the duodenojejunal anastomosis were collected for examining interstitial cells of Cajal (ICC), inflammation, and apoptosis.ResultsCompared to the bodyweight before surgery, the bodyweight loss at three and twelve months after surgery in patients receiving FJI was significant less than that in patients with Roux-en-Y. Patients with the FJI procedure showed significant increase of blood hemoglobin and total protein, compared to those at one month after surgery, and the prognostic nutrition index scores at three and twelve months after surgery. The incidence rates of post-operative complications, including reflux esophagitis, dumping syndrome, and Roux-en-Y syndrome were decreased in patients with FJI. Compared to beagles receiving Roux-en-Y, more ICC in the intestinal submuocsa, less intestinal epithelial cell apoptosis, and decreased inflammation in serosal side of the intestine were found in the FJI group. The intestinal transit rate in FJI group was lower than that in Roux-en Y group, indicating that FJI benefits food storage.ConclusionThe FJI procedure promotes nutritional recovery and decreases post-operative complications in gastric cancer patients after total gastrectomy, which may be through ameliorating intestinal inflammation and damage and reducing ICC loss to preserve food reservoir function and intestinal motility.


Gastroenterology | 2010

M1914 Mechanisms by Which the New Reconstruction Procedure Exerts Beneficial Effects After Total Gastrectomy

Xuewei Ding; Fang Yan; Hui Li; Qiang Xue; Kuo Zhang; Xiubao Ren; Xishan Hao

Background and Aims: Recent studies have shown epsterin-Barr virus (EBV) infection is related to the development of gastric cancer. In particular, latent EBV infection usually established persistently and promoted tumor progression. The zinc finger E-box binding factor 1 (ZEB1) mediated transcriptional silencing of intermediate-early genes and induced EBV infection from lytic to latent stage. We aim to study the effect of ZEB1 on modulating latent-lytic switch in gastric cancer, and to explore the potential of ZEB1 as a novel molecular target for the intervention of EBV associated Gastric Cancer (EBVaGc). Method Gastric cancer cell-lines with latent (YCC10 cell-line) and lytic (AGS-EBV infected cell-line) EBV infection were used in this study. Loss or gain function of ZEB1 was obtained by ZEB1 siRNA knockdown or forced re-expression experiments, respectively. The modulation of the intermediateearly gene and apoptotic mediators were assessed by real time RT-PCR and Western blot. Cell growth was evaluated by cell viability assay and colony formation assay, and cell cycle by flow cytometry analysis. Results: siRNA-mediated knockdown of ZEB1 in latent EBV infected cell-line YCC10 triggered a 3-fold enhanced expression of BZLF1, an intermediateearly gene of EBV, but a 5-fold reduction in expression of EBV nuclear antigen 1 (EBNA1), a gene that plays essential roles in enabling the replication and persistence of EBV genomes in latently infected cells and activating EBV latent gene expression. By doing so, knockdown of ZEB1 in YCC10 significantly inhibited cell viability (100% to 84%, P<0.01), suppressed cell proliferation as evidenced by reduced S phase cells (P=0.005), caused cell cycle arrest in G2M phase (P=0.006) and induced cell apoptosis (4.6±0.15% to 7.1±1.36%, P<0.05), The induction of cell apoptosis is confirmed by the cleavage of caspase-3 and PARP. On the other hand, over-expression of ZEB1 in lytic EBV infected cell line AGS-EBV through transient transfection significantly increased cell viability (P<0.01), induced S phase cell number (32.9±1.17% to 40.3±23.89%, P<0.05) and promoted colony formation (P<0.01). Conclusion: ZEB1 is involved in the latent-lytic switch of EBV associated gastric cancer. Inhibition of ZEB1 may be a promising approach for the treatment of EBVaGc.

Collaboration


Dive into the Xiubao Ren's collaboration.

Top Co-Authors

Avatar

Hui Li

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xishan Hao

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinpu Yu

Tianjin Medical University Cancer Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Feng Wei

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Hong Liu

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Shui Cao

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Baozhu Ren

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Lili Yang

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiumei An

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Fan Yan

Tianjin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge