Network


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

Hotspot


Dive into the research topics where Xiu-Rong Ren is active.

Publication


Featured researches published by Xiu-Rong Ren.


Biochemistry | 2009

The Anti-Helminthic Niclosamide Inhibits Wnt/Frizzled1 Signaling

Minyong Chen; Jiangbo Wang; Jiuyi Lu; Michael C. Bond; Xiu-Rong Ren; H. Kim Lyerly; Larry S. Barak; Wei Chen

Wnt proteins bind to seven-transmembrane Frizzled receptors to mediate the important developmental, morphogenetic, and stem cell related tissue-regenerative effects of Wnt signaling. Dysregulated Wnt signaling is associated with many cancers. Currently, there are no drug candidates or even tool compounds that modulate Wnt-mediated receptor trafficking, and subsequent Wnt signaling. We examined libraries of FDA-approved drugs for their utility as Frizzled internalization modulators, employing a primary imaged-based GFP fluorescence assay that uses Frizzled1 endocytosis as the readout. We now report that the anti-helminthic niclosamide, a drug used for the treatment of tapeworm, promotes Frizzled1 endocytosis, downregulates Dishevelled-2 protein, and inhibits Wnt3A-stimulated beta-catenin stabilization and LEF/TCF reporter activity. Additionally, following niclosamide-mediated internalization, the Frizzled1 receptor colocalizes in vesicles containing transferrin and agonist-activated beta(2)-adrenergic receptor. Therefore, niclosamide may serve as a negative modulator of Wnt/Frizzled1 signaling by depleting upstream signaling molecules (i.e., Frizzled and Dishevelled) and moreover may provide a valuable means of studying the physiological consequences of Wnt signaling.


Journal of Translational Medicine | 2012

Phase I clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibtion

Erika Paige Hamilton; Kimberly L. Blackwell; Amy Hobeika; Timothy M. Clay; Gloria Broadwater; Xiu-Rong Ren; Wei Chen; Henry Castro; Frederic Lehmann; Neil L. Spector; Junping Wei; Takuya Osada; H. Kim Lyerly

BackgroundPatients with HER2-overexpressing metastatic breast cancer, despite initially benefiting from the monoclonal antibody trastuzumab and the EGFR/HER2 tyrosine kinase inhibitor lapatinib, will eventually have progressive disease. HER2-based vaccines induce polyclonal antibody responses against HER2 that demonstrate enhanced anti-tumor activity when combined with lapatinib in murine models. We wished to test the clinical safety, immunogenicity, and activity of a HER2-based cancer vaccine, when combined with lapatinib.MethodsWe immunized women (n = 12) with metastatic, trastuzumab-refractory, HER2-overexpressing breast cancer with dHER2, a recombinant protein consisting of extracellular domain (ECD) and a portion of the intracellular domain (ICD) of HER2 combined with the adjuvant AS15, containing MPL, QS21, CpG and liposome. Lapatinib (1250 mg/day) was administered concurrently. Peripheral blood antibody and T cell responses were measured.ResultsThis regimen was well tolerated, with no cardiotoxicity. Anti-HER2-specific antibody was induced in all patients whereas HER2-specific T cells were detected in one patient. Preliminary analyses of patient serum demonstrated downstream signaling inhibition in HER2 expressing tumor cells. The median time to progression was 55 days, with the majority of patients progressing prior to induction of peak anti-HER2 immune responses; however, 300-day overall survival was 92% (95% CI: 77-100%).ConclusionsdHER2 combined with lapatinib was safe and immunogenic with promising long term survival in those with HER2-overexpressing breast cancers refractory to trastuzumab. Further studies to define the anticancer activity of the antibodies induced by HER2 vaccines along with lapatinib are underway.Trial registryClinicalTrials.gov NCT00952692


Hepatology | 2010

Activation of Rac1 promotes hedgehog-mediated acquisition of the myofibroblastic phenotype in rat and human hepatic stellate cells.

Steve S. Choi; Rafal P. Witek; Liu Yang; Alessia Omenetti; Wing-Kin Syn; Cynthia A. Moylan; Youngmi Jung; Gamze Karaca; Vanessa Teaberry; Thiago A. Pereira; Jiangbo Wang; Xiu-Rong Ren; Anna Mae Diehl

Hepatic accumulation of myofibroblastic hepatic stellate cells (MF‐HSCs) is pivotal in the pathogenesis of cirrhosis. Two events are necessary for MF‐HSCs to accumulate in damaged livers: transition of resident, quiescent hepatic stellate cells (Q‐HSCs) to MF‐HSCs and expansion of MF‐HSC numbers through increased proliferation and/or reduced apoptosis. In this study, we identified two novel mediators of MF‐HSC accumulation: Ras‐related C3 botulinum toxin substrate 1 (Rac1) and Hedgehog (Hh). It is unclear whether Rac1 and Hh interact to regulate the accumulation of MF‐HSCs. We evaluated the hypothesis that Rac1 promotes activation of the Hh pathway, thereby stimulating signals that promote transition of Q‐HSCs into MF‐HSCs and enhance the viability of MF‐HSCs. Using both in vitro and in vivo model systems, Rac1 activity was manipulated through adenoviral vector‐mediated delivery of constitutively active or dominant‐negative rac1. Rac1‐transgenic mice with targeted myofibroblast expression of a mutated human rac1 transgene that produces constitutively active Rac1 were also examined. Results in all models demonstrated that activating Rac1 in HSC enhanced Hh signaling, promoted acquisition/maintenance of the MF‐HSC phenotype, increased MF‐HSC viability, and exacerbated fibrogenesis. Conversely, inhibiting Rac1 with dominant‐negative rac1 reversed these effects in all systems examined. Pharmacologic manipulation of Hh signaling demonstrated that profibrogenic actions of Rac1 were mediated by its ability to activate Hh pathway‐dependent mechanisms that stimulated myofibroblastic transition of HSCs and enhanced MF‐HSC viability. Conclusion: These findings demonstrate that interactions between Rac1 and the Hh pathway control the size of MF‐HSC populations and have important implications for the pathogenesis of cirrhosis. HEPATOLOGY 2010


Breast Cancer Research | 2012

Polyclonal HER2-specific antibodies induced by vaccination mediate receptor internalization and degradation in tumor cells

Xiu-Rong Ren; Junping Wei; Gangjun Lei; Jiangbo Wang; Jiuyi Lu; Wenle Xia; Neil L. Spector; Larry S. Barak; Timothy M. Clay; Takuya Osada; Erika Paige Hamilton; Kimberly L. Blackwell; Amy Hobeika; Michael A. Morse; H. Kim Lyerly; Wei Chen

IntroductionSustained HER2 signaling at the cell surface is an oncogenic mechanism in a significant proportion of breast cancers. While clinically effective therapies targeting HER2 such as mAbs and tyrosine kinase inhibitors exist, tumors overexpressing HER2 eventually progress despite treatment. Thus, abrogation of persistent HER2 expression at the plasma membrane to synergize with current approaches may represent a novel therapeutic strategy.MethodsWe generated polyclonal anti-HER2 antibodies (HER2-VIA) by vaccinating mice with an adenovirus expressing human HER2, and assessed their signaling effects in vitro and anti-tumor effects in a xenograft model. In addition, we studied the signaling effects of human HER2-specific antibodies induced by vaccinating breast cancer patients with a HER2 protein vaccine.ResultsHER2-VIA bound HER2 at the plasma membrane, initially activating the downstream kinases extracellular signal-regulated protein kinase 1/2 and Akt, but subsequently inducing receptor internalization in clathrin-coated pits in a HER2 kinase-independent manner, followed by ubiquitination and degradation of HER2 into a 130 kDa fragment phosphorylated at tyrosine residues 1,221/1,222 and 1,248. Following vaccination of breast cancer patients with the HER2 protein vaccine, HER2-specific antibodies were detectable and these antibodies bound to cell surface-expressed HER2 and inhibited HER2 signaling through blocking tyrosine 877 phosphorylation of HER2. In contrast to the murine antibodies, human anti-HER2 antibodies induced by protein vaccination did not mediate receptor internalization and degradation.ConclusionThese data provide new insight into HER2 trafficking at the plasma membrane and the changes induced by polyclonal HER2-specific antibodies. The reduction of HER2 membrane expression and HER2 signaling by polyclonal antibodies induced by adenoviral HER2 vaccines supports human clinical trials with this strategy for those breast cancer patients with HER2 therapy-resistant disease.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2013

MARCKS and HSP70 interactions regulate mucin secretion by human airway epithelial cells in vitro

Shijing Fang; Anne L. Crews; Wei Chen; Joungjoa Park; Qi Yin; Xiu-Rong Ren; Kenneth B. Adler

Myristoylated alanine-rich C kinase substrate (MARCKS) protein has been recognized as a key regulatory molecule controlling mucin secretion by airway epithelial cells in vitro and in vivo. We recently showed that two intracellular chaperones, heat shock protein 70 (HSP70) and cysteine string protein (CSP), associate with MARCKS in the secretory mechanism. To elucidate more fully MARCKS-HSP70 interactions in this process, studies were performed in well-differentiated normal human bronchial epithelial (NHBE) cells maintained in air-liquid interface culture utilizing specific pharmacological inhibition of HSP70 with pyrimidinone MAL3-101 and siRNA approaches. The results indicate that HSP70 interaction with MARCKS is enhanced after exposure of the cells to the protein kinase C activator/mucin secretagogue, phorbol 12-myristate 13-acetate (PMA). Pretreatment of NHBEs with MAL3-101 attenuated in a concentration-dependent manner PMA-stimulated mucin secretion and interactions among HSP70, MARCKS, and CSP. In additional studies, trafficking of MARCKS in living NHBE cells was investigated after transfecting cells with fluorescently tagged DNA constructs: MARCKS-yellow fluorescent protein, and/or HSP70-cyan fluorescent protein. Cells were treated with PMA 48 h posttransfection, and trafficking of the constructs was examined by confocal microscopy. MARCKS translocated rapidly from plasma membrane to cytoplasm, whereas HSP70 was observed in the cytoplasm and appeared to associate with MARCKS after PMA exposure. Pretreatment of cells with either MAL3-101 or HSP70 siRNA inhibited translocation of MARCKS. These results provide evidence of a role for HSP70 in mediating mucin secretion via interactions with MARCKS and that these interactions are critical for the cytoplasmic translocation of MARCKS upon its phosphorylation.


Breast Cancer Research | 2015

Perhexiline promotes HER3 ablation through receptor internalization and inhibits tumor growth

Xiu-Rong Ren; Jiangbo Wang; Takuya Osada; Robert A. Mook; Michael A. Morse; Larry S. Barak; Herbert Kim Lyerly; Wei Chen

IntroductionHuman epidermal growth factor receptor HER3 has been implicated in promoting the aggressiveness and metastatic potential of breast cancer. Upregulation of HER3 has been found to be a major mechanism underlying drug resistance to EGFR and HER2 tyrosine kinase inhibitors and to endocrine therapy in the treatment of breast cancer. Thus, agents that reduce HER3 expression at the plasma membrane may synergize with current therapies and offer a novel therapeutic strategy to improve treatment.MethodsWe devised an image-based screening platform using membrane localized HER3-YFP to identify small molecules that promote HER3 internalization and degradation. In vitro and in vivo tumor models were used to characterize the signaling effects of perhexiline, an anti-anginal drug, identified by the screening platform.ResultsWe found perhexiline, an anti-anginal drug, selectively internalized HER3, decreased HER3 expression, and subsequently inhibited signaling downstream of HER3. Consistent with these results, perhexiline inhibited breast cancer cell proliferation in vitro and tumor growth in vivo.ConclusionsThis is the first demonstration that HER3 can be targeted with small molecules by eliminating it from the cell membrane. The novel approach used here led to the discovery that perhexiline ablates HER3 expression, and offers an opportunity to identify HER3 ablation modulators as innovative therapeutics to improve survival in breast cancer patients.


PLOS ONE | 2013

Regulation of Hedgehog Signaling by Myc-Interacting Zinc Finger Protein 1, Miz1

Jiuyi Lu; Minyong Chen; Xiu-Rong Ren; Jiangbo Wang; H. Kim Lyerly; Larry S. Barak; Wei Chen

Smoothened (Smo) mediated Hedgehog (Hh) signaling plays an essential role in regulating embryonic development and postnatal tissue homeostasis. Aberrant activation of the Hh pathway contributes to the formation and progression of various cancers. In vertebrates, however, key regulatory mechanisms responsible for transducing signals from Smo to the nucleus remain to be delineated. Here, we report the identification of Myc-interacting Zinc finger protein 1 (Miz1) as a Smo and Gli2 binding protein that positively regulates Hh signaling. Overexpression of Miz1 increases Gli luciferase reporter activity, whereas knockdown of endogenous Miz1 has the opposite effect. Activation of Smo induces translocation of Miz1 to the primary cilia together with Smo and Gli2. Furthermore, Miz1 is localized to the nucleus upon Hh activation in a Smo-dependent manner, and loss of Miz1 prevents the nuclear translocation of Gli2. More importantly, silencing Miz1 expression inhibits cell proliferation in vitro and the growth of Hh-driven medulloblastoma tumors allografted in SCID mice. Taken together, these results identify Miz1 as a novel regulator in the Hh pathway that plays an important role in mediating Smo-dependent oncogenic signaling.


Bioorganic & Medicinal Chemistry | 2017

Benzimidazole inhibitors from the Niclosamide chemotype inhibit Wnt/β-catenin signaling with selectivity over effects on ATP homeostasis

Robert A. Mook; Xiu-Rong Ren; Jiangbo Wang; Hailan Piao; Larry S. Barak; H. Kim Lyerly; Wei Chen

The Wnt signaling pathway plays a key role in organ and tissue homeostasis, and when dysregulated, can become a major underlying mechanism of disease, particularly cancer. We reported previously that the anthelmintic drug Niclosamide inhibits Wnt/β-catenin signaling and suppresses colon cancer cell growth in vitro and in vivo. To define Niclosamides mechanism of Wnt/β-catenin inhibition, and to improve its selectivity and pharmacokinetic properties as an anticancer treatment, we designed a novel class of benzimidazole inhibitors of Wnt/β-catenin signaling based on SAR studies of the Niclosamide salicylanilide chemotype. Niclosamide has multiple biological activities. To address selectivity in our design, we interrogated a protonophore SAR model and used the principle of conformational restriction to identify novel Wnt/β-catenin inhibitors with less effect on ATP cellular homeostasis. These studies led to the identification of 4-chloro-2-(5-(trifluoromethyl)-1H-benzo[d]imidazol-2-yl) phenol (4) and related derivatives with greater selectivity for Wnt/β-catenin signaling inhibition vs. differential effects on cellular ATP homeostasis. This is the first report that the Wnt signaling inhibitory activity of Niclosamide can be translated into a new chemical class and to show that its effects on ATP homeostasis can be separated from its inhibitory effects on Wnt signaling. These compounds could be useful tools to elucidate the mechanism of Niclosamides inhibition of Wnt signaling, and aid the discovery of inhibitors with improved pharmacologic properties to treat cancer and diseases in which Niclosamide has important biological activity.


Bioorganic & Medicinal Chemistry | 2018

Identification of DK419, a potent inhibitor of Wnt/β-catenin signaling and colorectal cancer growth

Jiangbo Wang; Robert A. Mook; Xiu-Rong Ren; Qingfu Zhang; Genevieve Jing; Min Lu; Ivan Spasojevic; H. Kim Lyerly; David S. Hsu; Wei Chen

The Wnt signaling pathway is critical for normal tissue development and is an underlying mechanism of disease when dysregulated. Previously, we reported that the drug Niclosamide inhibits Wnt/β-catenin signaling by decreasing the cytosolic levels of Dishevelled and β-catenin, and inhibits the growth of colon cancers both in vitro and in vivo. Since the discovery of Niclosamides anthelmintic activity, a growing body of literature indicates that Niclosamide is a multifunctional drug. In an effort to identify derivatives of Niclosamide with improved pharmacokinetic properties that maintain the multifunctional drug activity of Niclosamide for clinical evaluation, we designed DK419, a derivative containing a 1H-benzo[d]imidazole-4-carboxamide substructure, using the structure-activity relationships (SAR) of the Niclosamide salicylanilide chemotype. Similar to Niclosamide, we found DK419 inhibited Wnt/β-catenin signaling, altered cellular oxygen consumption rate and induced production of pAMPK. Moreover, we found DK419 inhibited the growth of CRC tumor cells in vitro, had good plasma exposure when dosed orally, and inhibited the growth of patient derived CRC240 tumor explants in mice dosed orally. DK419, a derivative of Niclosamide with multifunctional activity and improved pharmacokinetic properties, is a promising agent to treat colorectal cancer, Wnt-related diseases and other diseases in which Niclosamide has demonstrated functional activity.


Journal of Translational Medicine | 2013

Correction: phase 1 clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibtion

Erika Paige Hamilton; Kimberly L. Blackwell; Amy Hobeika; Timothy M. Clay; Gloria Broadwater; Xiu-Rong Ren; Wei Chen; Henry Castro; Frederic Lehmann; Neil L. Spector; Junping Wei; Takuya Osada; H. Kim Lyerly; Michael A. Morse

Corrections In our original manuscript [1], the corresponding author, Michael A Morse, was missed from the authors’ list. Therefore the correct author list should be: Erika Hamilton, Kimberly Blackwell, Amy C Hobeika, Timothy M Clay, Gloria Broadwater, Xiu-Rong Ren, Wei Chen, Henry Castro, Frederic Lehmann, Neil Spector, Junping Wei, Takuya Osada, H Kim Lyerly and Michael A Morse. Also the title of our original manuscript ‘Phase 1 clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibtion’ is also incorrect. The title should read:‘Phase 1 clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibition’. Journal of Translational Medicine regret any inconvenience that this inaccuracy might have caused.

Collaboration


Dive into the Xiu-Rong Ren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erika Paige Hamilton

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge