Shaozhen Xie
University of Rochester
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Shaozhen Xie.
Biochemical and Biophysical Research Communications | 2003
Lin Yang; Liang Wang; Hui Kuan Lin; Pu Yeh Kan; Shaozhen Xie; Meng Yin Tsai; Peng Hui Wang; Yen Ta Chen; Chawnshang Chang
The effects of IL-6 on prostate cancer cells are well documented yet remain controversial. Some reports suggested that IL-6 could promote prostate cancer cell growth, while others showed that IL-6 could repress prostate cancer cell growth. Here, we systemically examined various IL-6 signaling pathways in prostate cancer cells and found that IL-6 could go through at least three distinct pathways to modulate the functions of androgen receptor (AR), a key transcriptional factor to control the prostate cancer growth. Our results show that IL-6 can enhance AR transactivation via either the STAT3 or MAPK pathways. In contrast, IL-6 can suppress AR transactivation via the PI3K-Akt pathway. Co-existence of these various signaling pathways may result in either additive or conflicting effects on AR transactivation. Together, our results indicate that the balance of these various pathways may then determine the overall effect of IL-6 on AR transactivation.
Cancer Research | 2005
Jing Ni; Xingqiao Wen; Jorge L. Yao; Hong-Chiang Chang; Yi Yin; Min Zhang; Shaozhen Xie; Ming Chen; Brenna Simons; Philip Chang; Anthony Di Sant'agnese; Edward M. Messing; Shuyuan Yeh
Epidemiologic studies suggested that vitamin E has a protective effect against prostate cancer. We showed here that tocopherol-associated protein (TAP), a vitamin E-binding protein, promoted vitamin E uptake and facilitated vitamin E antiproliferation effect in prostate cancer cells. Interestingly, without vitamin E treatment, overexpression of TAP in prostate cancer cells significantly suppressed cell growth; knockdown of endogenous TAP by TAP small interfering RNA (siRNA) in nonmalignant prostate HPr-1 cells increased cell growth. Further mechanism dissection studies suggested that the tumor suppressor function of TAP was via down-regulation of phosphoinositide 3-kinase (PI3K)/Akt signaling, but not by modulating cell cycle arrest or androgen receptor signaling. Immunoprecipitation results indicated that TAP inhibited the interaction of PI3K subunits, p110 with p85, and subsequently reduced Akt activity. Constitutively active Akt could negate the TAP-suppressive activity on prostate cancer cell growth. Moreover, stable transfection of TAP in LNCaP cells suppressed LNCaP tumor incidence and growth rate in nude mice. Furthermore, TAP mRNA and protein expression levels were significantly down-regulated in human prostate cancer tissue samples compared with benign prostate tissues as measured by reverse transcription-PCR, in situ hybridization, and immunohistochemistry. Together, our data suggest that TAP not only mediates vitamin E absorption to facilitate vitamin E antiproliferation effect in prostate cancer cells, but also functions like a tumor suppressor gene to control cancer cell viability through a non-vitamin E manner. Therefore, TAP may represent a new prognostic marker for prostate cancer progression.
Proceedings of the National Academy of Sciences of the United States of America | 2009
Shaozhen Xie; Yi-Fen Lee; Eungseok Kim; Lu Min Chen; Jing Ni; Lei Ya Fang; Su Liu; Shin Jen Lin; Jun Ichi Abe; Bradford C. Berk; Feng Ming Ho; Chawnshang Chang
Testicular orphan nuclear receptor 4 (TR4) is an orphan member of the nuclear receptor superfamily with diverse physiological functions. Using TR4 knockout (TR4−/−) mice to study its function in cardiovascular diseases, we found reduced cluster of differentiation (CD)36 expression with reduced foam cell formation in TR4−/− mice. Mechanistic dissection suggests that TR4 induces CD36 protein and mRNA expression via a transcriptional regulation. Interestingly, we found this TR4-mediated CD36 transactivation can be further enhanced by polyunsaturated fatty acids (PUFAs), such as omega-3 and -6 fatty acids, and their metabolites such as 15-hydroxyeico-satetraonic acid (15-HETE) and 13-hydroxy octa-deca dieonic acid (13-HODE) and thiazolidinedione (TZD)-rosiglitazone. Both electrophoretic mobility shift assays (EMSA) and chromatin immunoprecipitation (ChIP) assays demonstrate that TR4 binds to the TR4 response element located on the CD36 5′-promoter region for the induction of CD36 expression. Stably transfected TR4-siRNA or functional TR4 cDNA in the RAW264.7 macrophage cells resulted in either decreased or increased CD36 expression with decreased or increased foam cell formation. Restoring functional CD36 cDNA in the TR4 knockdown macrophage cells reversed the decreased foam cell formation. Together, these results reveal an important signaling pathway controlling CD36-mediated foam cell formation/cardiovascular diseases, and findings that TR4 transactivation can be activated via its ligands/activators, such as PUFA metabolites and TZD, may provide a platform to screen new drug(s) to battle the metabolism syndrome, diabetes, and cardiovascular diseases.
Nature | 2017
Shom Goel; Molly J. DeCristo; April C. Watt; Haley BrinJones; Jaclyn Sceneay; Ben B. Li; Naveed Khan; Jessalyn M. Ubellacker; Shaozhen Xie; Otto Metzger-Filho; Jeremy Hoog; Matthew J. Ellis; Cynthia X. Ma; Susanne Ramm; Ian E. Krop; Thomas M. Roberts; Hye-Jung Kim; Sandra S. McAllister; Jean Zhao
Cyclin-dependent kinases 4 and 6 (CDK4/6) are fundamental drivers of the cell cycle and are required for the initiation and progression of various malignancies. Pharmacological inhibitors of CDK4/6 have shown significant activity against several solid tumours. Their primary mechanism of action is thought to be the inhibition of phosphorylation of the retinoblastoma tumour suppressor, inducing G1 cell cycle arrest in tumour cells. Here we use mouse models of breast carcinoma and other solid tumours to show that selective CDK4/6 inhibitors not only induce tumour cell cycle arrest, but also promote anti-tumour immunity. We confirm this phenomenon through transcriptomic analysis of serial biopsies from a clinical trial of CDK4/6 inhibitor treatment for breast cancer. The enhanced anti-tumour immune response has two underpinnings. First, CDK4/6 inhibitors activate tumour cell expression of endogenous retroviral elements, thus increasing intracellular levels of double-stranded RNA. This in turn stimulates production of type III interferons and hence enhances tumour antigen presentation. Second, CDK4/6 inhibitors markedly suppress the proliferation of regulatory T cells. Mechanistically, the effects of CDK4/6 inhibitors both on tumour cells and on regulatory T cells are associated with reduced activity of the E2F target, DNA methyltransferase 1. Ultimately, these events promote cytotoxic T-cell-mediated clearance of tumour cells, which is further enhanced by the addition of immune checkpoint blockade. Our findings indicate that CDK4/6 inhibitors increase tumour immunogenicity and provide a rationale for new combination regimens comprising CDK4/6 inhibitors and immunotherapies as anti-cancer treatment.
Nature Medicine | 2016
Jing Ni; Shakti Ramkissoon; Shaozhen Xie; Shom Goel; Daniel G. Stover; Hanbing Guo; Victor Luu; Eugenio Marco; Lori A. Ramkissoon; Yun Jee Kang; Marika Hayashi; Quang-Dé Nguyen; Azra H. Ligon; Rose Du; Elizabeth B. Claus; Brian M. Alexander; Guo-Cheng Yuan; Zhigang C. Wang; J. Dirk Iglehart; Ian E. Krop; Thomas M. Roberts; Nan Lin; Keith L. Ligon; Jean Zhao
Brain metastases represent the greatest clinical challenge in treating HER2-positive breast cancer. We report the development of orthotopic patient-derived xenografts (PDXs) of HER2-expressing breast cancer brain metastases (BCBM), and their use for the identification of targeted combination therapies. Combined inhibition of PI3K and mTOR resulted in durable tumor regressions in three of five PDXs, and therapeutic response was correlated with a reduction in the phosphorylation of 4EBP1, an mTORC1 effector. The two nonresponding PDXs showed hypermutated genomes with enrichment of mutations in DNA-repair genes, which suggests an association of genomic instability with therapeutic resistance. These findings suggest that a biomarker-driven clinical trial of PI3K inhibitor in combination with an mTOR inhibitor should be conducted for patients with HER2-positive BCBM.
Journal of Biological Chemistry | 2011
Shaozhen Xie; Jing Ni; Yi-Fen Lee; Su Liu; Gonghui Li; Chih-Rong Shyr; Chawnshang Chang
The nuclear receptor TR4 is a key regulator for many physiological processes, including growth, development, and metabolism. However, how the transcriptional activity of TR4 is regulated in the absence of ligand(s) remains largely unknown. Here we found that an androgen receptor (AR) coactivator, ARA55, might function as a corepressor to suppress TR4 transactivation. Molecular mechanistic dissection with mutation analysis found that ARA55 could enhance TR4 acetylation at the conserved acetylation sites of lysine 175 and lysine 176 in the DNA-binding domain via recruiting proteins with histone acetyl transferase activity, which might then reduce significantly the TR4 DNA binding activity that resulted in the suppression of TR4 transactivation. These results are in contrast to the classic ARA55 coactivator function to enhance AR transactivation partially via increased AR acetylation in the hinge/ligand-binding domain. Together, these results not only provide a novel functional mechanism showing that acetylation of different nuclear receptors at different domains by coregulator may lead to differential receptor transactivation activity but also provide a new way for small molecules to control TR4 transactivation via altering TR4 acetylation levels, and such small molecules may have potential therapeutic applications in the future.
Neuro-oncology | 2017
Jing Ni; Shaozhen Xie; Shakti Ramkissoon; Victor Luu; Yu Sun; Pratiti Bandopadhayay; Rameen Beroukhim; Thomas M. Roberts; Charles D. Stiles; Rosalind A. Segal; Keith L. Ligon; William C. Hahn; Jean Zhao
Background. Astrocytomas are the most common primary human brain tumors. Receptor tyrosine kinases (RTKs), including tyrosine receptor kinase B (TrkB, also known as tropomyosin-related kinase B; encoded by neurotrophic tyrosine kinase receptor type 2 [NTRK2]), are frequently mutated by rearrangement/fusion in high-grade and low-grade astrocytomas. We found that activated TrkB can contribute to the development of astrocytoma and might serve as a therapeutic target in this tumor type. Methods. To identify RTKs capable of inducing astrocytoma formation, a library of human tyrosine kinases was screened for the ability to transform murine Ink4a−/−/Arf−/− astrocytes. Orthotopic allograft studies were conducted to evaluate the effects of RTKs on the development of astrocytoma. Since TrkB was identified as a driver of astrocytoma formation, the effect of the Trk inhibitors AZD1480 and RXDX-101 was assessed in astrocytoma cells expressing activated TrkB. RNA sequencing, real-time PCR, western blotting, and enzyme-linked immunosorbent assays were conducted to characterize NTRK2 in astrocytomas. Results. Activated TrkB cooperated with Ink4a/Arf loss to induce the formation of astrocytomas through a mechanism mediated by activation of signal transducer and activator of transcription 3 (STAT3). TrkB activation positively correlated with Ccl2 expression. TrkB-induced astrocytomas remained dependent on TrkB signaling for survival, highlighting a role of NTRK2 as an addictive oncogene. Furthermore, the QKI-NTRK2 fusion associated with human astrocytoma transformed Ink4a−/−/Arf−/− astrocytes, and this process was also mediated via STAT3 signaling. Conclusions. Our findings provide evidence that constitutively activated NTRK2 alleles, notably the human tumor-associated QKI-NTRK2 fusion, can cooperate with Ink4a/Arf loss to drive astrocytoma formation. Therefore, we propose NTRK2 as a potential therapeutic target in the subset of astrocytoma patients defined by QKI-NTRK2 fusion.
Clinical Cancer Research | 2009
Jing Ni; Tiejun Mai; See-Tong Pang; Imranul Haque; Kaohsing Huang; Matthew A. DiMaggio; Shaozhen Xie; Nadine S. James; Dhanalakshmi Kasi; Sherry R. Chemler; Shuyuan Yeh
Purpose: Among derivatives of α-vitamin E, α-vitamin E succinate (VES), has attracted much attention due to its potent anti–prostate cancer activity in vitro and in vivo. However, the in vivo antitumor activity of VES might be compromised if administrated orally due to the VES hydrolysis by esterases in the gastrointestinal tract. Experimental Design: New nonhydrolyzable VES ether analogues were synthesized and their growth inhibition was screened by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide growth assay. Among them, RRR-α-tocopheryloxybutyl sulfonic acid (VEBSA) was further characterized by terminal deoxyribonucleotidyl transferase–mediated dUTP nick end labeling apoptosis assay, soft agar assay, and in vivo tumor formation. Results: VEBSA has potent antitumor ability, albeit to a lesser extent than VES, in in vitro cultured prostate cancer LNCaP and PC3 cells. Like VES, VEBSA induced apoptosis, repressed androgen receptor protein expression, and enhanced vitamin D receptor expression, suggesting that VEBSA can go through mechanisms similar to those used by VES to inhibit the growth of prostate cancer cells in vitro. However, 6 weeks of oral consumption of VEBSA, but not of VES, reduced the tumor burden in the xenografted prostate tumors in nude mice. Furthermore, oral intake of VEBSA for 20 weeks inhibited prostate tumor growth and progression more efficiently compared with VES in the prostate cancer tumor model of TRAMP mice. Conclusion: Oral consumption of VEBSA allows a greater anticancer activity compared with VES. Chemoprevention prefers the oral consumption of agents; the advantage of VEBSA over VES to be administrated orally will allow VEBSA to serve as an agent for both preventive and therapeutic purposes for prostate cancer.
Archive | 2010
Su Liu; Shaozhen Xie; Yi-Fen Lee; Chawnshang Chang
The human testicular receptor 2 and 4 (TR2 and TR4) are two evolutionarily related orphan nuclear receptors belonging to the same nuclear receptor subfamily (Lee et al. 2002, J Steroid Biochem Mol Biol 81(4–5), 291–308). They regulate gene expression by binding to DNA as homodimers or a heterodimer with each other. TR4 may also cross-talk with other nuclear receptors, to control its target genes. In vitro and in vivo studies have identified several TR4 target genes, including ciliary neurotrophic factor alpha (CNTFRα) (Young et al. 1997, J Biol Chem 272(5), 3109–3116), apolipoprotein E (ApoE) (Kim et al. 2003, J Biol Chem 278(47), 46919–46926) and phosphenolpyruvate carboxykinase (PEPCK) (Liu et al. 2007, Diabetes 56(12), 2901–2909). Recent studies using TR4 knockout (TR4–/–) mice suggested that TR4 may play essential roles in growth, development, and metabolism (Zhang et al. 2007, Mol Endocrinol 21(4), 908–920; Kim et al. 2005, Biochem Biophys Res Commun 328(1), 85–90; Chen et al. 2005, Mol Cell Biol 25(7), 2722–2732; Mu et al. 2004, Mol Cell Biol 24(13), 5887–5899; Collins et al. 2004, Proc Natl Acad Sci U S A 101(42), 15058–15063). Mice with a germline deletion of TR4 are viable but have high early postnatal mortality, growth retardation, and profound reduction in body weight. Further studies showed that TR4 plays essential roles in the development and functioning in the central nervous system (Chen et al. 2005, Mol Cell Biol 25(7), 2722–2732), such as proper myelination and oligodendrocyte differentiation (Zhang et al. 2007, Mol Endocrinol 21(4), 908–920). Studies also showed that TR4 is important for spermatogenesis in male mice (Mu et al. 2004, Mol Cell Biol 24(13), 5887–5899) and folliculogenesis in female mice (Chen et al. 2008, Mol Endocrinol 22, 858–867). In addition, TR4 might be involved in skeletal muscle function and bone remodeling. TR4 and TR2 also regulate embryonic and fetal globin gene transcription (Tanabe et al. 2002, EMBO J 21(13), 3434–3442; Tanabe et al. 2007, EMBO J 26(9), 2295–2306). Surprisingly, mice lacking TR2 are viable and have no serious developmental defects. Thus, TR2 may either not be important in spermatogenesis and testis development, or its roles may be compensated by other closely related proteins such as TR4. Therefore, this chapter will focus on the in vivo roles of TR4.
Cancer Research | 2017
Molly J. DeCristo; Shom Goel; April C. Watt; Haley BrinJones; Jaclyn Sceneay; Ben Li; Jessalyn M. Ubellacker; Shaozhen Xie; Susanne Ramm; Hye-Jung Kim; Sandra S. McAllister; Jean Zhao
The cyclin D:cyclin-dependent kinases 4 and 6 (CDK4/6) axis is one of the most frequently dysregulated pathways in human cancers, and CDK4/6 inhibitors have shown significant activity against a number of solid tumors, including breast cancer. Analogous to clinical experience, we found that the CDK4/6 inhibitor, abemaciclib, caused significant tumor regression in the MMTV-rtTA/tetO-HER2 mouse model of luminal breast cancer. However, as CDK4/6 inhibitors are known to block tumor cell proliferation, but not directly induce tumor cell apoptosis, as we confirmed in our study, the mechanisms by which CDK4/6 inhibition caused tumor regression were not clear. Notably, abemaciclib therapy increased total CD3+ T cells in these tumors, while decreasing the immunosuppressive CD4+ regulatory T cell population. Further investigation revealed that CDK4/6 inhibition directly suppresses regulatory T cells by inhibiting their proliferation without impacting natural regulatory T cell formation in the thymus, the differentiation of inducible regulatory T cells, or regulatory T cell apoptosis. We are currently exploring potential effects of CDK4/6 inhibition on regulatory T cell function and lineage stability. Analysis of CD8+ and CD4+ T cells in abemaciclib-treated tumors revealed a marked reduction in the expression of the inhibitory immune checkpoint receptors PD-1, Tim-3, CTLA-4, and LAG3. Specifically, the fraction of CD8+ T cells expressing known markers of T cell exhaustion (PD-1/Tim-3 double-positivity or high expression of PD-1) was decreased in abemaciclib-treated tumors. Importantly, antibody-mediated depletion of CD8+ T cells established that response of MMTV-rtTA/tetO-HER2 tumors to abemaciclib is dependent on CD8+ T cells. Finally, given the observed immunomodulatory effects of abemaciclib, we sought to determine if abemaciclib treatment would sensitize MMTV-rtTA/tetO-HER2 tumors to immune checkpoint blockade. Pretreatment with abemaciclib followed by combination abemaciclib and α-PDL1 significantly enhanced tumor regression compared to abemaciclib or α-PDL1 alone. Our studies reveal a novel mechanism by which CDK4/6 inhibitors directly elicit an anti-tumor immune response. These results provide strong rationale for further investigations into combining CDK4/6 inhibitors with immune checkpoint blockade in breast cancer. Citation Format: Molly J. DeCristo*, Shom Goel*, April C. Watt, Haley BrinJones, Jaclyn Sceneay, Ben Li, Jessalyn M. Ubellacker, Shaozhen Xie, Susanne Ramm, Hye-Jung Kim, Sandra S. McAllister, Jean J. Zhao. CDK4/6 inhibition directly enhances an anti-tumor immune response in breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1634. doi:10.1158/1538-7445.AM2017-1634