Network


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

Hotspot


Dive into the research topics where Xiaoling Puyang is active.

Publication


Featured researches published by Xiaoling Puyang.


Nature Communications | 2017

Splicing modulators act at the branch point adenosine binding pocket defined by the PHF5A–SF3b complex

Teng Teng; Jennifer Tsai; Xiaoling Puyang; Michael Seiler; Shouyong Peng; Sudeep Prajapati; Daniel Aird; Silvia Buonamici; Benjamin Caleb; Betty Chan; Laura Corson; Jacob Feala; Peter Fekkes; Baudouin Gerard; Craig Karr; Manav Korpal; Xiang Liu; Jason T. Lowe; Yoshiharu Mizui; James Palacino; Eunice Park; P.G.R. Smith; V. Subramanian; Zhenhua Jeremy Wu; Jian Zou; Lihua Yu; Agustin Chicas; Markus Warmuth; Nicholas A. Larsen; Ping Zhu

Pladienolide, herboxidiene and spliceostatin have been identified as splicing modulators that target SF3B1 in the SF3b subcomplex. Here we report that PHF5A, another component of this subcomplex, is also targeted by these compounds. Mutations in PHF5A-Y36, SF3B1-K1071, SF3B1-R1074 and SF3B1-V1078 confer resistance to these modulators, suggesting a common interaction site. RNA-seq analysis reveals that PHF5A-Y36C has minimal effect on basal splicing but inhibits the global action of splicing modulators. Moreover, PHF5A-Y36C alters splicing modulator-induced intron-retention/exon-skipping profile, which correlates with the differential GC content between adjacent introns and exons. We determine the crystal structure of human PHF5A demonstrating that Y36 is located on a highly conserved surface. Analysis of the cryo-EM spliceosome Bact complex shows that the resistance mutations cluster in a pocket surrounding the branch point adenosine, suggesting a competitive mode of action. Collectively, we propose that PHF5A–SF3B1 forms a central node for binding to these splicing modulators.


Nature Communications | 2017

Evasion of immunosurveillance by genomic alterations of PPARγ/RXRα in bladder cancer

Manav Korpal; Xiaoling Puyang; Zhenhua Jeremy Wu; Roland Seiler; Craig Furman; Htoo Zarni Oo; Michael Seiler; Sean Irwin; V. Subramanian; Jaya Julie Joshi; Chris K. Wang; Victoria Rimkunas; Davide Tortora; Hua Yang; Namita Kumar; Galina Kuznetsov; Mark Matijevic; Jesse Chow; P.V. Kumar; Jian Zou; Jacob Feala; Laura Corson; Ryan Henry; Anand Selvaraj; Allison Davis; Kristjan Bloudoff; James Douglas; Bernhard Kiss; Morgan E. Roberts; Ladan Fazli

Muscle-invasive bladder cancer (MIBC) is an aggressive disease with limited therapeutic options. Although immunotherapies are approved for MIBC, the majority of patients fail to respond, suggesting existence of complementary immune evasion mechanisms. Here, we report that the PPARγ/RXRα pathway constitutes a tumor-intrinsic mechanism underlying immune evasion in MIBC. Recurrent mutations in RXRα at serine 427 (S427F/Y), through conformational activation of the PPARγ/RXRα heterodimer, and focal amplification/overexpression of PPARγ converge to modulate PPARγ/RXRα-dependent transcription programs. Immune cell-infiltration is controlled by activated PPARγ/RXRα that inhibits expression/secretion of inflammatory cytokines. Clinical data sets and an in vivo tumor model indicate that PPARγHigh/RXRαS427F/Y impairs CD8+ T-cell infiltration and confers partial resistance to immunotherapies. Knockdown of PPARγ or RXRα and pharmacological inhibition of PPARγ significantly increase cytokine expression suggesting therapeutic approaches to reviving immunosurveillance and sensitivity to immunotherapies. Our study reveals a class of tumor cell-intrinsic “immuno-oncogenes” that modulate the immune microenvironment of cancer.Muscle-invasive bladder cancer (MIBC) is a potentially lethal disease. Here the authors characterize diverse genetic alterations in MIBC that convergently lead to constitutive activation of PPARgamma/RXRalpha and result in immunosurveillance escape by inhibiting CD8+ T-cell recruitment.


Cancer Research | 2017

Abstract DDT01-04: Discovery and development of H3B-6545: A novel, oral, selective estrogen receptor covalent antagonist (SERCA) for the treatment of breast cancer

P.G.R. Smith; Xiaoling Puyang; Craig Furman; Guo Zhu Zheng; Deepti Banka; Mike Thomas; V. Subramanian; Sean Irwin; Nicholas A. Larsen; Benjamin Caleb; Craig Karr; Jeremy Wu; Morgan O’Shea; Joyce Yang; Allison M. Davis; Amy Kim; Nathalie Rioux; Victoria Rimkunas; Huilan Yao; Crystal MacKenzie; P.V. Kumar; Sherri Smith; Sean Eckley; Andrew Hart; George Lai; Christopher Rowbottom; Peter Fekkes; Silvia Buonamici; Dominic Reynolds; Lihua Yu

Mutations in the estrogen receptor (ER) are detected in up to 30% of patients that initially respond but subsequently relaps to anti-endocrine therapies. ERα mutations, likely through constitutively activating ERα, can functionally confer resistance to existing classes of endocrine therapies. Current endocrine therapies are only partially effective in the ERα mutant setting and a significant proportion of endocrine-therapy resistant breast cancer metastases continue to remain dependent on ERα signaling for growth/survival indicating a critical need to develop the next generation of ERα antagonists that can overcome ERα wild-type and mutant activity. Here we describe a novel series of compounds with a unique mode of inhibition that potently target both wild-type and mutant ERα. These compounds are Selective Estrogen Receptor Covalent Antagonists (SERCAs) that inactivate the estrogen receptor by targeting a cysteine that is not present in other nuclear hormone receptors, leading to a unique biological and activity profile differentiated from Selective Estrogen Receptor Modulators (SERMs) and Selective Estrogen Receptor Degraders (SERDs). Using structure-based drug design approaches we have identified a first-in-class clinical candidate, H3B-6545. H3B-6545 is a highly selective small molecule that potently antagonizes wild-type and mutant ERα in biochemical and cell based assays. In vitro comparisons with standard of care and other experimental agents confirm increased cell potency of H3B-6545 under continuous as well as washout treatment conditions. In vivo, once daily oral dosing of H3B-6545 shows potent activity and superior efficacy to fulvestrant in the MCF-7 xenograft model with maximal antitumor activity at doses >10x below the maximum tolerated dose in mice. In addition, H3B-6545 shows superior antitumor activity to tamoxifen and fulvestrant in patient derived xenograft models of estrogen receptor positive breast cancer including models carrying ERα mutations In non-clinical safety studies in rat and monkeys, H3B-6545 is well tolerated across a broad dose range and at exposures that significantly exceed those required for efficacy in mouse xenograft models. In summary, H3B-6545 is a first-in-class, orally available and potent selective estrogen receptor covalent antagonist with a compelling preclinical efficacy and safety profile that is being developed for the treatment of breast cancer. Citation Format: Peter G. Smith, Xiaoling Puyang, Craig Furman, Guo Zhu Zheng, Deepti Banka, Michael Thomas, Vanitha Subramanian, Sean Irwin, Nicholas Larsen, Benjamin Caleb, Craig Karr, Jeremy Wu, Morgan O’Shea, Joyce Yang, Allison Davis, Amy Kim, Nathalie Rioux, Victoria Rimkunas, Huilan Yao, Crystal MacKenzie, Pavan Kumar, Sherri Smith, Sean Eckley, Andrew Hart, George Lai, Christopher Rowbottom, Peter Fekkes, Silvia Buonamici, Dominic Reynolds, Lihua Yu, Tarek Sahmoud, Markus Warmuth, Lorna Mitchell, Ping Zhu, Manav Korpal. Discovery and development of H3B-6545: A novel, oral, selective estrogen receptor covalent antagonist (SERCA) for the treatment of 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 DDT01-04. doi:10.1158/1538-7445.AM2017-DDT01-04


Cancer Research | 2016

Abstract 3013: Identification of PHF5A as a common cellular target of splicing-modulating chemical probes

Teng Teng; Xiaoling Puyang; Shouyong Peng; Jacob Feala; Betty Chan; Jennifer Tsai; Benjamin Caleb; Craig Karr; Eunice Park; Laura Corson; Yoshiharu Mizui; P.G.R. Smith; Nicholas A. Larsen; Lihua Yu; Markus Warmuth; Ping Zhu; Agustin Chicas

Recent discoveries that splicing factors such SF3B1, U2AF1, SRSF2 are frequently mutated in multiple hematological malignancies including chronic lymphocytic leukaemia and myelodysplastic syndromes have generated interest in therapeutic approaches to target the splicesome dependency in cancer cells bearing mutations in splicing factors. Previously, several structurally unrelated natural compounds including pladienolide, herboxidiene, and FR901464 have been shown to exert potent anti-proliferative effects in cancer cells grown in vitro. Further optimization has led to the discovery of natural product analogs (e.g. E7107) with anti-tumor efficacy in vivo in xenograft models. Target identification has revealed the SF3B complex of the splicesome as the common action site for these compounds. Recent work has demonstrated biological and genetic evidence that single amino acid substitution (R1074H) in SF3B1 completely abolished the anti-proliferative effect of pladienolide derivative E7107, suggesting that SF3B1 is the direct binding partner for pladienolides. However, the same SF3B1 R1074H mutation does not provide equal level of protections for cells treated with herboxidiene derivatives, indicating differential mechanism of action for these two classes of splicing modulators. To identify targets for herboxidiene-like compounds, we have generated resistant HCT116 clones upon continuous administration of herboxidiene derivative H3B-37045 in vitro. Whole exome sequencing from 6 resistant clones revealed a common Y36C mutation in SF3B subunit component PHF5A (SF3B14b). Over-expression of PHF5A Y36C but not the wild-type form in parental HCT116 cells confirmed the protective effect of this mutation to H3B-37045. Surprisingly, PHF5A Y36C expression also conferred resistance to the pladienolide derivative E7107, which indicates that, unlike the SF3B1 R1074H mutation, PHF5A resides within a common node of action site among different splicing modulators. RNA-seq, biochemical and structure homology-modeling analysis suggested that PHF5A Y36C mutation disrupted the action of splicing modulators through interfering with the compounds’ interaction with the SF3B complex. Detailed analysis of the function of the Y36C mutant and wild-type PHF5A in the SF3B complex is currently ongoing. Understanding the function of PHF5A in splicing and the molecular mechanism of Y36C mutation shall provide new insights of the biological role of splicesome, and guide the development of next generation splicesome inhibitors. Citation Format: Teng Teng, Xiaoling Puyang, Shouyong Peng, Jacob Feala, Betty Chan, Jennifer Tsai, Benjamin Caleb, Craig Karr, Eunice Park, Laura Corson, Yoshiharu Mizui, Peter Smith, Nicholas Larsen, Lihua Yu, Markus Warmuth, Ping Zhu, Agustin Chicas. Identification of PHF5A as a common cellular target of splicing-modulating chemical probes. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3013.


Journal of Visualized Experiments | 2015

Implementation of In Vitro Drug Resistance Assays: Maximizing the Potential for Uncovering Clinically Relevant Resistance Mechanisms

Manav Korpal; Jacob Feala; Xiaoling Puyang; Jian Zou; Alex H. Ramos; Jeremy Wu; Timm Baumeister; Lihua Yu; Markus Warmuth; Ping Zhu

Although targeted therapies are initially effective, resistance inevitably emerges. Several methods, such as genetic analysis of resistant clinical specimens, have been applied to uncover these resistance mechanisms to facilitate follow-up care. Although these approaches have led to clinically relevant discoveries, difficulties in attaining the relevant patient material or in deconvoluting the genomic data collected from these specimens have severely hampered the path towards a cure. To this end, we here describe a tool for expeditious discovery that may guide improvement in first-line therapies and alternative clinical management strategies. By coupling preclinical in vitro or in vivo drug selection with next-generation sequencing, it is possible to identify genomic structural variations and/or gene expression alterations that may serve as functional drivers of resistance. This approach facilitates the spontaneous emergence of alterations, enhancing the probability that these mechanisms may be observed in the patients. In this protocol we provide guidelines to maximize the potential for uncovering single nucleotide variants that drive resistance using adherent lines.


Cancer Research | 2014

Abstract 2932: SF3B1 mutations induce aberrant mRNA splicing in cancer and confer sensitivity to spliceosome inhibition

Silvia Buonamici; Kian-Huat Lim; Jacob Feala; Eunice Park; Laura Corson; Michelle Aicher; Daniel Aird; Betty Chan; Erik Corcoran; Rachel Darman; Peter Fekkes; Gregg F. Keaney; Pavan Kumar; Kaiko Kunii; Linda Lee; Xiaoling Puyang; Jose Rodrigues; Anand Selvaraj; Mike Thomas; John Q. Wang; Markus Warmuth; Lihua Yu; Ping Zhu; P.G.R. Smith; Yoshiharu Mizui

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Recurrent heterozygous mutations of the spliceosome protein SF3B1 have been identified in myelodysplastic syndromes, chronic lymphocytic leukemia (CLL), breast, pancreatic and skin cancers. SF3B1 is a component of the U2 snRNP complex which binds to the pre-mRNA branch point site and is involved in recognition and stabilization of the spliceosome at the 3′ splice site. To understand the impact of SF3B1 mutations, we compared RNAseq profiles from tumor samples with SF3B1 hotspot mutations (SF3B1-MUT) or wild-type SF3B1 (SF3B1-WT) in breast cancer, melanoma and CLL. This analysis revealed significant increases in the usage of novel alternative splice junctions in SF3B1-MUT samples including selection of alternative 3′ splice sites and less frequently exon skipping. These events induce expression of alternative mRNAs that are translated into novel proteins or aberrant mRNAs that are decayed by cells. A common alternative splicing profile was shared across different hotspot mutations and lineages (e.g. ZDHHC16 and COASY); however, unique alternative splicing profiles were also observed suggesting lineage specific effects. RNAseq analysis of several cell lines with endogenous SF3B1 hotspot mutations confirmed the presence of the same spliced isoforms as observed in tumor samples. To prove that SF3B1-MUT were inducing alternative splicing, transient transfection of several SF3B1 hotspot mutations in 293FT cells induced the expression of the common alternatively spliced genes suggesting functional similarity. Selective shRNA depletion of mutant SF3B1 allele in SF3B1-MUT cells resulted in downregulation of the same splice isoforms. Furthermore, isogenic B-cell lines (NALM-6) expressing the most frequent SF3B1 mutation (K700E) were generated and profiled by RNAseq. As expected, similar alternatively spliced genes were observed in NALM-6 SF3B1-K700E cells exclusively. To investigate the role of nonsense-mediated mRNA decay (NMD) in eliminating aberrant mRNAs induced by SF3B1-MUT, we treated NALM-6 SF3B1-K700E cells with cycloheximide, a translation inhibitor known to inhibit NMD. In the treated samples, expression of several aberrant mRNAs was revealed and some of these transcripts were shown to be downregulated in patient samples. Taken together, these results confirm the association between different SF3B1 hotspot mutations and the presence of novel splice isoforms. We demonstrated that E7107, a potent and selective inhibitor of wild-type SF3B1, also binds and inhibits SF3B1-MUT protein. In addition, E7107 represses the expression of several common aberrant splice mRNA products in SF3B1-MUT cells in vitro and in vivo. When tested in a NALM-6 mouse model, E7107 induced tumor regression and increased the overall survival of animals implanted with NALM-6 SF3B1-K700E cells. These data suggest splicing inhibitors as a promising therapeutic approach for cancer patients carrying SF3B1 mutations. Citation Format: Silvia Buonamici, Kian Huat Lim, Jacob Feala, Eunice Park, Laura Corson, Michelle Aicher, Daniel Aird, Betty Chan, Erik Corcoran, Rachel Darman, Peter Fekkes, Gregg Keaney, Pavan Kumar, Kaiko Kunii, Linda Lee, Xiaoling Puyang, Jose Rodrigues, Anand Selvaraj, Michael Thomas, John Wang, Markus Warmuth, Lihua Yu, Ping Zhu, Peter Smith, Yoshiharu Mizui. SF3B1 mutations induce aberrant mRNA splicing in cancer and confer sensitivity to spliceosome inhibition. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2932. doi:10.1158/1538-7445.AM2014-2932


Cell Reports | 2015

Cancer-Associated SF3B1 Hotspot Mutations Induce Cryptic 3′ Splice Site Selection through Use of a Different Branch Point

Rachel Darman; Michael Seiler; Anant A. Agrawal; Kian Huat Lim; Shouyong Peng; Daniel Aird; Suzanna Bailey; Erica B. Bhavsar; Betty Chan; Simona Colla; Laura Corson; Jacob Feala; Peter Fekkes; Kana Ichikawa; Gregg F. Keaney; Linda Lee; Pavan Kumar; Kaiko Kunii; Crystal MacKenzie; Mark Matijevic; Yoshiharu Mizui; Khin Than Myint; Eun Sun Park; Xiaoling Puyang; Anand Selvaraj; Michael Thomas; Jennifer J. Tsai; John Wang; Markus Warmuth; Hui Yang


Archive | 2017

DERIVADOS DE INDAZOL Y SU USO EN EL TRATAMIENTO DEL CÁNCER DE MAMA ER

Ping Zhu; Guo Zhu Zheng; John Wang; Peter G. Smith; Susanta Samajdar; Xiaoling Puyang; Vijay Kumar Nyavanandi; Manav Korpal; Ming Hao; Mark Hong Bock


Cancer Research | 2017

Abstract 126: A chemogenomic approach reveals the action of splicing modulators at the branch point adenosine binding pocket defined by the PHF5A/SF3b complex

Teng Teng; Jennifer Tsai; Xiaoling Puyang; Michael Seiler; Shouyong Peng; Daniel Aird; Silvia Buonamici; Benjamin Caleb; Betty Chan; Laura Corson; Jacob Feala; Peter Fekkes; Craig Karr; Manav Korpal; Yoshiharu Mizui; Eunice Park; James Palacino; Peter Smith; V. Subramanian; Jeremy Wu; Lihua Yu; Agustin Chicas; Markus Warmuth; Nicholas A. Larsen; Ping Zhu


Archive | 2016

TETRASUBSTITUTED ALKENE COMPOUNDS AND THEIR USE

Mark Hong Bock; Ming-Hong Hao; Manav Korpal; Vijay Kumar Nyavanandi; Xiaoling Puyang; Susanta Samajdar; Peter Gerard Smith; John Wang; Guo Zhu Zheng; Ping Zhu

Collaboration


Dive into the Xiaoling Puyang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

P.G.R. Smith

University of Southampton

View shared research outputs
Top Co-Authors

Avatar

V. Subramanian

Central Leather Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge