Network


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

Hotspot


Dive into the research topics where Chengzhen Ren is active.

Publication


Featured researches published by Chengzhen Ren.


Molecular and Cellular Biology | 2003

Caveolin-1 Maintains Activated Akt in Prostate Cancer Cells through Scaffolding Domain Binding Site Interactions with and Inhibition of Serine/Threonine Protein Phosphatases PP1 and PP2A

Likun Li; Cheng Hui Ren; Salahaldin A. Tahir; Chengzhen Ren; Timothy C. Thompson

ABSTRACT Previously it has been reported that caveolin-1 (cav-1) has antiapoptotic activities in prostate cancer cells and functions downstream of androgenic stimulation. In this study, we demonstrate that cav-1 overexpression significantly reduced thapsigargin (Tg)-stimulated apoptosis. Examination of the phosphatidylinositol 3-kinase (PI3-K)/Akt signaling cascade revealed higher activities of PDK1 and Akt but not PI3-K in cav-1-stimulated cells compared to control cells. We subsequently found that cav-1 interacts with and inhibits serine/threonine protein phosphatases PP1 and PP2A through scaffolding domain binding site interactions. Deletion of the cav-1 scaffolding domain significantly reduces phosphorylated Akt and cell viability compared with wild-type cav-1. Analysis of potential substrates for PP1 and PP2A revealed that cav-1-mediated inhibition of PP1 and PP2A leads to increased PDK1, Akt, and ERK1/2 activities. We demonstrate that increased Akt activities are largely responsible for cav-1-mediated cell survival using dominant-negative Akt mutants and specific inhibitors to MEK1/MEK and show that cav-1 increases the half-life of phosphorylated PDK1 and Akt after inhibition of PI3-K by LY294002. We further demonstrate that cav-1-stimulated Akt activities lead to increased phosphorylation of multiple Akt substrates, including GSK3, FKHR, and MDM2. In addition, overexpression of cav-1 significantly increases translocation of phosphorylated androgen receptor to nucleus. Our studies therefore reveal a novel mechanism of Akt activation in prostate cancer and potentially other malignancies.


Nature Medicine | 1998

Suppression of caveolin expression induces androgen sensitivity in metastatic androgen-insensitive mouse prostate cancer cells

Yasutomo Nasu; Terry L. Timme; Guang Yang; Chris H. Bangma; Likun Li; Chengzhen Ren; Sang Hee Park; Marlene DeLeon; Jianxiang Wang; Timothy C. Thompson

Although prostate cancer cells are often initially sensitive to androgen ablation, they eventually lose this response and continue to survive, grow and spread in the absence of androgenic steroids. The mechanism(s) that underlie resistance to androgen ablation therapy remain mostly unknown. We have demonstrated that elevated caveolin protein levels are associated with human prostate cancer progression in pathological specimens. Here we show that suppression of caveolin expression by a stably transfected antisense caveolin-1 cDNA vector converted androgen-insensitive metastatic mouse prostate cancer cells to an androgen-sensitive phenotype. Orthotopically grown tumors and low-density cell cultures derived from antisense caveolin clones had increased apoptosis in the absence of androgenic steroids, whereas similarly grown tumors and cells from vector (control) clones and parental cells were not sensitive to androgens. Studies using a representative antisense caveolin clone showed that selection for androgen resistance in vivo correlated with increased caveolin levels, and that adenovirus-mediated caveolin expression blocked androgen sensitivity. Our results identify a new candidate gene for hormone-resistant prostate cancer in man and indicate that androgen insensitivity can be an inherent property of metastatic prostate cancer.


Oncogene | 2000

Caveolin-1 is regulated by c-myc and suppresses c-myc-induced apoptosis

Terry L. Timme; Alexei Goltsov; Salahaldin A. Tahir; Likun Li; Jianxiang Wang; Chengzhen Ren; Randal N. Johnston; Timothy C. Thompson

Recent data indicating that overexpression of caveolin-1 as well as c-myc are relatively common features of advanced prostate cancer prompted us to test for potential cooperative interactions between caveolin-1 and c-myc that would be consistent with malignant progression. We used the well-characterized Rat1AmycERTM cells to show that the caveolin-1 gene is down-regulated at the level of transcription by c-myc. By maintaining relatively high levels of caveolin-1 with an adenoviral vector or in stably transfected clones we show that caveolin-1 can suppress c-myc-induced apoptosis. Further we established human prostate cancer cell lines with the mycERTM construct and show that clones with increased caveolin-1 are more resistant to myc-induced apoptosis and have increased capacity for growth in soft agar when c-myc is activated.


Human Gene Therapy | 2001

Prostate-Specific Antigen Response and Systemic T Cell Activation After In Situ Gene Therapy in Prostate Cancer Patients Failing Radiotherapy

Brian J. Miles; Moshe Shalev; Estuardo Aguilar-Cordova; Terry L. Timme; Hon-Man Lee; Guang Yang; Howard L. Adler; Kenneth Kernen; Christina K. Pramudji; Takefumi Satoh; Yehoshua Gdor; Chengzhen Ren; Gustavo Ayala; Thomas M. Wheeler; E.B. Butler; Dov Kadmon; Timothy C. Thompson

In an extended phase I/II study we evaluated 36 prostate cancer patients with local recurrence after radiotherapy who received single or repeated cycles of replication-deficient adenoviral vector (ADV)-mediated herpes simplex virus-thymidine kinase (HSV-tk) plus ganciclovir (GCV) in situ gene therapy with respect to serum PSA levels, alterations in immune cells, and numbers of apoptotic cells in needle biopsies. An initial cycle of HSV-tk plus GCV gene therapy caused a significant prolongation of the mean serum PSA-doubling time (PSADT) from 15.9 to 42.5 months (p = 0.0271) and in 28 of the injected patients (77.8%) there was a mean PSA reduction (PSAR) of 28%. It took a mean of 8.5 months for the PSA to return to the initial PSA (TR-PSA) value. A repeated cycle of gene therapy failed to significantly extend PSADT but did result in significant increases in PSAR (29.4%) and TR-PSA (10.5 months). Moderately increased serum adenovirus antibody titers were generally observed 2 weeks after initial vector injection. Also at this time there was a statistically significant increase in the mean percent of CD8(+) T cells positive for the HLA-DR marker of activation in peripheral blood (p = 0.0088). Studies using prostate biopsies obtained at the same time point demonstrated that vector DNA was detectable by PCR in most samples yet all patients remained positive for prostate cancer in at least one biopsy core. Further analysis demonstrated a correlation between the level of CD8(+) cells and the number of apoptotic cells in biopsies containing cancer cells (p = 0.042). We conclude that repeated cycles of in situ HSV-tk plus GCV gene therapy can be administered to prostate cancer patients who failed radiotherapy and have a localized recurrence. Biological responses to this experimental therapy including increases in PSADT, PSAR, and TR-PSA, and activated CD8(+) T cells present in the peripheral blood, were demonstrated. Interestingly, the density of CD8(+) cells in posttreatment biopsies correlated with the number of apoptotic cells.


American Journal of Pathology | 2003

Disruption of the Caveolin-1 Gene Impairs Renal Calcium Reabsorption and Leads to Hypercalciuria and Urolithiasis

Guangwen Cao; Guang Yang; Terry L. Timme; Takashi Saika; Luan D. Truong; Takefumi Satoh; Alexei Goltsov; Sang Hee Park; Taoyan Men; Nobuyuki Kusaka; Weihua Tian; Chengzhen Ren; Hongyu Wang; Dov Kadmon; Wei Wen Cai; A. Craig Chinault; Timothy B. Boone; Allan Bradley; Timothy C. Thompson

Using LoxP/Cre technology, we generated a knockout mouse homozygous for a null mutation in exon 2 of Cav1. In male Cav1-/- animals, we observed a dramatic increase in the incidence of urinary calcium stone formation. In 5-month-old male mice, the incidence of early urinary calculi was 67% in Cav1-/- mice compared to 19% in Cav1+/+ animals. Frank stone formation was observed in 13% of Cav1-/- males but was not seen in Cav1+/+ mice. Urine calcium concentration was significantly higher in Cav1-/- male mice compared to Cav1+/+ mice. In Cav1-/- mice, distal convoluted tubule cells were completely devoid of Cav1 and the localization of plasma membrane calcium ATPase was disrupted. Functional studies confirmed that active calcium absorption was significantly reduced in Cav1-/- compared to Cav1+/+ male mice. These results demonstrate that disruption of the Cav1 gene promotes the progressive steps required for urinary calcium stone formation and establish a new mouse model for urinary stone disease.


The Journal of Urology | 2002

The Role Of Caveolin-1 In Androgen Insensitive Prostate Cancer

Vladimir Mouraviev; Likun Li; Salahaldin A. Tahir; Guang Yang; Terry L. Timme; Alexei Goltsov; Chengzhen Ren; Takefumi Satoh; Thomas M. Wheeler; Michael Ittmann; Brian J. Miles; Robert J. Amato; Dov Kadmon; Timothy C. Thompson

PURPOSE We summarize the literature regarding androgen insensitive prostate cancer and caveolin-1. Caveolin-1 is a major structural component of caveolae, membrane micro-domains known to have important roles in signal transduction and lipid transport. MATERIALS AND METHODS A review of the literature relevant to androgen insensitive caveolin-1 and prostate cancer included the first published report in 1998 through those published in March 2002. RESULTS Caveolin-1 expression is increased in primary and metastatic human prostate cancer with highest levels observed after androgen ablation therapy. Recent studies have documented that caveolin-1 is secreted by prostate cancer cells and can be detected in the serum of men with prostate cancer. CONCLUSIONS The results presented in this review establish that caveolin-1 is an autocrine/paracrine factor associated with androgen insensitive prostate cancer. They show the potential for caveolin-1 as a biomarker therapeutic target for this important malignancy.


Molecular and Cellular Biology | 2002

mRTVP-1, a novel p53 target gene with proapoptotic activities

Chengzhen Ren; Likun Li; Alexei Goltsov; Terry L. Timme; Salahaldin A. Tahir; Jianxiang Wang; Laura Garza; A. Craig Chinault; Timothy C. Thompson

ABSTRACT We identified a novel mouse gene, mRTVP-1, as a p53 target gene using differential display PCR and extensive promoter analysis. The mRTVP-1 protein has 255 amino acids and differs from the human RTVP-1 (hRTVP-1) protein by two short in-frame deletions of two and nine amino acids. RTVP-1 mRNA was induced in multiple cancer cell lines by adenovirus-mediated delivery of p53 and by gamma irradiation or doxorubicin both in the presence and in the absence of endogenous p53. Analysis of RTVP-1 expression in nontransformed and transformed cells further supported p53-independent gene regulation. Using luciferase reporter and electrophoretic mobility shift assays we identified a p53 binding site within intron 1 of the mRTVP-1 gene. Overexpression of mRTVP-1 or hRTVP-1 induced apoptosis in multiple cancer cell lines including prostate cancer cell lines 148-1PA, 178-2BMA, PC-3, TSU-Pr1, and LNCaP, a human lung cancer cell line, H1299, and two isogenic human colon cancer cell lines, HCT116 p53+/+ and HCT116 p53−/−, as demonstrated by annexin V positivity, phase-contrast microscopy, and in selected cases 4′,6′-diamidino-2-phenylindole staining and DNA fragmentation. Deletion of the signal peptide from the N terminus of RTVP-1 reduced its apoptotic activities, suggesting that a secreted and soluble form of RTVP-1 may mediate, in part, its proapoptotic activities.


Cancer Research | 2004

RTVP-1, a Tumor Suppressor Inactivated by Methylation in Prostate Cancer

Chengzhen Ren; Likun Li; Guang Yang; Terry L. Timme; Alexei Goltsov; Chenghui Ren; Xiaorong Ji; Josephine Addai; Hongbin Luo; Michael Ittmann; Timothy C. Thompson

We previously identified and characterized a novel p53-regulated gene in mouse prostate cancer cells that was homologous to a human gene that had been identified in brain cancers and termed RTVP-1 or GLIPR. In this report, we document that the human RTVP-1 gene is also regulated by p53 and induces apoptosis in human prostate cancer cell lines. We show that the expression of the human RTVP-1 gene is down-regulated in human prostate cancer specimens compared with normal human prostate tissue at the mRNA and protein levels. We further document epigenetic changes consistent with RTVP-1 being a tumor suppressor in human prostate cancer.


Cancer Research | 2008

Glioma Pathogenesis-Related Protein 1 Exerts Tumor Suppressor Activities through Proapoptotic Reactive Oxygen Species–c-Jun–NH2 Kinase Signaling

Likun Li; Elmoataz Abdel Fattah; Guangwen Cao; Chengzhen Ren; Guang Yang; Alexei Goltsov; A. Craig Chinault; Wei-Wen Cai; Terry L. Timme; Timothy C. Thompson

Glioma pathogenesis-related protein 1 (GLIPR1), a novel p53 target gene, is down-regulated by methylation in prostate cancer and has p53-dependent and -independent proapoptotic activities in tumor cells. These properties suggest an important tumor suppressor role for GLIPR1, yet direct genetic evidence of a tumor suppressor function for GLIPR1 is lacking and the molecular mechanism(s), through which GLIPR1 exerts its tumor suppressor functions, has not been shown. Here, we report that the expression of GLIPR1 is significantly reduced in human prostate tumor tissues compared with adjacent normal prostate tissues and in multiple human cancer cell lines. Overexpression of GLIPR1 in cancer cells leads to suppression of colony growth and induction of apoptosis. Mice with an inactivated Glipr1 gene had significantly shorter tumor-free survival times than either Glipr1(+/+) or Glipr1(+/-) mice in both p53(+/+) and p53(+/-) genetic backgrounds, owing to their development of a unique array of malignant tumors. Mechanistic analysis indicated that GLIPR1 up-regulation increases the production of reactive oxygen species (ROS) leading to apoptosis through activation of the c-Jun-NH(2) kinase (JNK) signaling cascade. Thus, our results identify GLIPR1 as a proapoptotic tumor suppressor acting through the ROS-JNK pathway and support the therapeutic potential for this protein.


Human Gene Therapy | 2003

Adenoviral Vector-Mediated mRTVP-1 Gene Therapy for Prostate Cancer

Takefumi Satoh; Terry L. Timme; Takashi Saika; Shin Ebara; Guang Yang; Jianxiang Wang; Chengzhen Ren; Nobuyuki Kusaka; Vladimir Mouraviev; Timothy C. Thompson

We previously identified the mouse RTVP-1 (mRTVP-1; related to testes-specific, vespid, and pathogenesis proteins) gene as a direct target of p53 with proapoptotic activities in various cancer cell lines, including prostate cancer. To test the therapeutic potential of mRTVP-1 we constructed an adenoviral vector capable of efficient transduction and expression of mRTVP-1 (AdmRTVP-1) and used this vector in an orthotopic, metastatic mouse model of prostate cancer. A single intratumoral administration of AdmRTVP-1 gene therapy significantly reduced primary tumor wet weight compared with control Adbetagal-injected tumors at two time points after injection with two different vector doses (p < or = 0.01 at 7 and 14 days). Spontaneous metastasis to lung was also significantly reduced (p < or = 0.02). Evaluation of treated tumors revealed increased apoptosis and lower microvessel density counts. In a rat aortic ring sprouting assay, AdmRTVP-1 inhibited endothelial cell sprouting compared with Adbetagal, confirming its antiangiogenic activity. These therapeutic activities were associated with a significant increase in survival from 22.9 to 26.8 days (p = 0.003) in this aggressive model of prostate cancer. Interestingly, there were significant increases in the infiltration of tumor-associated macrophages, dendritic cells, and CD8+ T cells, which persisted at 14 days posttreatment in the AdmRTVP-1-treated tumors compared with Adbetagal control-treated tumors. In addition, significantly increased natural killer and cytotoxic T lymphocyte activities were demonstrated in the mice with AdmRTVP-1-treated tumors. The unique therapeutic properties of AdmRTVP-1 gene therapy demonstrated in this study provide new opportunities for gene and immunotherapy of prostate cancer and potentially other malignancies.

Collaboration


Dive into the Chengzhen Ren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Terry L. Timme

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Guang Yang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Alexei Goltsov

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Likun Li

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianxiang Wang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Chenghui Ren

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Dov Kadmon

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Nobuyuki Kusaka

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge