Network


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

Hotspot


Dive into the research topics where Jieya Shao is active.

Publication


Featured researches published by Jieya Shao.


Cell Reports | 2013

Endocrine-Therapy-Resistant ESR1 Variants Revealed by Genomic Characterization of Breast-Cancer-Derived Xenografts

Shunqiang Li; Dong Shen; Jieya Shao; Robert Crowder; Wenbin Liu; Aleix Prat; Xiaping He; Shuying Liu; Jeremy Hoog; Charles Lu; Li Ding; Obi L. Griffith; Christopher A. Miller; Dave Larson; Robert S. Fulton; Michelle L. K. Harrison; Tom Mooney; Joshua F. McMichael; Jingqin Luo; Yu Tao; Rodrigo Franco Gonçalves; Christopher Schlosberg; Jeffrey F. Hiken; Laila Saied; César Sánchez; Therese Giuntoli; Caroline Bumb; Crystal Cooper; Robert T. Kitchens; Austin Lin

To characterize patient-derived xenografts (PDXs) for functional studies, we made whole-genome comparisons with originating breast cancers representative of the major intrinsic subtypes. Structural and copy number aberrations were found to be retained with high fidelity. However, at the single-nucleotide level, variable numbers of PDX-specific somatic events were documented, although they were only rarely functionally significant. Variant allele frequencies were often preserved in the PDXs, demonstrating that clonal representation can be transplantable. Estrogen-receptor-positive PDXs were associated with ESR1 ligand-binding-domain mutations, gene amplification, or an ESR1/YAP1 translocation. These events produced different endocrine-therapy-response phenotypes in human, cell line, and PDX endocrine-response studies. Hence, deeply sequenced PDX models are an important resource for the search for genome-forward treatment options and capture endocrine-drug-resistance etiologies that are not observed in standard cell lines. The originating tumor genome provides a benchmark for assessing genetic drift and clonal representation after transplantation.


Neuron | 2003

A rapid cellular FRET assay of polyglutamine aggregation identifies a novel inhibitor

Sonia K Pollitt; Judit Pallos; Jieya Shao; Urvee A. Desai; Aye Aye K. Ma; Leslie M. Thompson; J. Lawrence Marsh; Marc I. Diamond

Many neurodegenerative diseases, including tauopathies, Parkinsons disease, amyotrophic lateral sclerosis, and the polyglutamine diseases, are characterized by intracellular aggregation of pathogenic proteins. It is difficult to study modifiers of this process in intact cells in a high-throughput and quantitative manner, although this could facilitate molecular insights into disease pathogenesis. Here we introduce a high-throughput assay to measure intracellular polyglutamine protein aggregation using fluorescence resonance energy transfer (FRET). We screened over 2800 biologically active small molecules for inhibitory activity and have characterized one lead compound in detail. Y-27632, an inhibitor of the Rho-associated kinase p160ROCK, diminished polyglutamine protein aggregation (EC(50) congruent with 5 microM) and reduced neurodegeneration in a Drosophila model of polyglutamine disease. This establishes a novel high-throughput approach to study protein misfolding and aggregation associated with neurodegenerative diseases and implicates a signaling pathway of previously unrecognized importance in polyglutamine protein processing.


Journal of Biological Chemistry | 2010

Interaction with Polyglutamine Aggregates Reveals a Q/N-rich Domain in TDP-43

Rodrigo A. Fuentealba; Maria Udan; Shaughn Bell; Iga Wegorzewska; Jieya Shao; Marc I. Diamond; Conrad C. Weihl; Robert H. Baloh

The identification of pathologic TDP-43 aggregates in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration, followed by the discovery of dominantly inherited point mutations in TDP-43 in familial ALS, have been critical insights into the mechanism of these untreatable neurodegenerative diseases. However, the biochemical basis of TDP-43 aggregation and the mechanism of how mutations in TDP-43 lead to disease remain enigmatic. In efforts to understand how TDP-43 alters its cellular localization in response to proteotoxic stress, we found that TDP-43 is sequestered into polyglutamine aggregates. Furthermore, we found that binding to polyglutamine aggregates requires a previously uncharacterized glutamine/asparagine (Q/N)-rich region in the C-terminal domain of TDP-43. Sequestration into polyglutamine aggregates causes TDP-43 to be cleared from the nucleus and become detergent-insoluble. Finally, we observed that sequestration into polyglutamine aggregates led to loss of TDP-43-mediated splicing in the nucleus and that polyglutamine toxicity could be partially rescued by increasing expression of TDP-43. These data indicate pathologic sequestration into polyglutamine aggregates, and loss of nuclear TDP-43 function may play an unexpected role in polyglutamine disease pathogenesis. Furthermore, as Q/N domains have a strong tendency to self-aggregate and in some cases can function as prions, the identification of a Q/N domain in TDP-43 has important implications for the mechanism of pathologic aggregation of TDP-43 in ALS and other neurodegenerative diseases.


Molecular and Cellular Biology | 2008

Phosphorylation of Profilin by ROCK1 Regulates Polyglutamine Aggregation

Jieya Shao; William J. Welch; Nicholas A. Diprospero; Marc I. Diamond

ABSTRACT Y-27632, an inhibitor of the Rho-associated kinase ROCK, is a therapeutic lead for Huntington disease (HD). The downstream targets that mediate its inhibitory effects on huntingtin (Htt) aggregation and toxicity are unknown. We have identified profilin, a small actin-binding factor that also interacts with Htt, as being a direct target of the ROCK1 isoform. The overexpression of profilin reduces the aggregation of polyglutamine-expanded Htt and androgen receptor (AR) peptides. This requires profilins G-actin binding activity and its direct interaction with Htt, which are both inhibited by the ROCK1-mediated phosphorylation of profilin at Ser-137. Y-27632 blocks the phosphorylation of profilin in HEK293 cells and primary neurons, which maintains profilin in an active state. The knockdown of profilin blocks the inhibitory effect of Y-27632 on both AR and Htt aggregation. A signaling pathway from ROCK1 to profilin thus controls polyglutamine protein aggregation and is targeted by a promising therapeutic lead for HD.


Journal of Biological Chemistry | 2003

Phosphorylation of serine 13 is required for the proper function of the Hsp90 co-chaperone, Cdc37

Jieya Shao; Thomas Prince; Steven D. Hartson; Robert L. Matts

The Hsp90 co-chaperone Cdc37 provides an essential function for the biogenesis and support of numerous protein kinases. In this report, we demonstrate that mammalian Cdc37 is phosphorylated on Ser13 in situ in rabbit reticulocyte lysate and in cultured K562 cells and that casein kinase II is capable of quantitatively phosphorylating recombinant Cdc37 at this site. Mutation of Ser13 to either Ala or Glu compromises the recruitment of Cdc37 to Hsp90-kinase complexes but has only modest effects on its basal (client-free) binding to Hsp90. Furthermore, Cdc37 containing the complementing Ser to Glu mutation showed altered interactions with Hsp90-kinase complexes consistent with compromised Cdc37 modulation of the Hsp90 ATP-driven reaction cycle. Thus, the data indicate that phosphorylation of Cdc37 on Ser13 is critical for its ability to coordinate Hsp90 nucleotide-mediated conformational switching and kinase binding.


Human Molecular Genetics | 2014

Prion-like nuclear aggregation of TDP-43 during heat shock is regulated by HSP40/70 chaperones

Maria Udan-Johns; Rocio Bengoechea; Shaughn Bell; Jieya Shao; Marc I. Diamond; Heather L. True; Conrad C. Weihl; Robert H. Baloh

TDP-43 aggregation in the cytoplasm or nucleus is a key feature of the pathology of amyotrophic lateral sclerosis and frontotemporal dementia and is observed in numerous other neurodegenerative diseases, including Alzheimers disease. Despite this fact, the inciting events leading to TDP-43 aggregation remain unclear. We observed that endogenous TDP-43 undergoes reversible aggregation in the nucleus after the heat shock and that this behavior is mediated by the C-terminal prion domain. Substitution of the prion domain from TIA-1 or an authentic yeast prion domain from RNQ1 into TDP-43 can completely recapitulate heat shock-induced aggregation. TDP-43 is constitutively bound to members of the Hsp40/Hsp70 family, and we found that heat shock-induced TDP-43 aggregation is mediated by the availability of these chaperones interacting with the inherently disordered C-terminal prion domain. Finally, we observed that the aggregation of TDP-43 during heat shock led to decreased binding to hnRNPA1, and a change in TDP-43 RNA-binding partners suggesting that TDP-43 aggregation alters its function in response to misfolded protein stress. These findings indicate that TDP-43 shares properties with physiologic prions from yeast, in that self-aggregation is mediated by a Q/N-rich disordered domain, is modulated by chaperone proteins and leads to altered function of the protein. Furthermore, they indicate that TDP-43 aggregation is regulated by chaperone availability, explaining the recurrent observation of TDP-43 aggregates in degenerative diseases of both the brain and muscle where protein homeostasis is disrupted.


Clinical Cancer Research | 2015

Efficacy of SERD/SERM Hybrid-CDK4/6 inhibitor combinations in models of endocrine therapy resistant breast cancer

Suzanne E. Wardell; Matthew J. Ellis; Holly M. Alley; Koleen Eisele; Todd VanArsdale; Stephen Dann; Kim Arndt; Tina Primeau; Elizabeth Griffin; Jieya Shao; Robert Crowder; Jin-Ping Lai; John D. Norris; Donald P. McDonnell; Shunqiang Li

Purpose: Endocrine therapy, using tamoxifen or an aromatase inhibitor, remains first-line therapy for the management of estrogen receptor (ESR1)–positive breast cancer. However, ESR1 mutations or other ligand-independent ESR1 activation mechanisms limit the duration of response. The clinical efficacy of fulvestrant, a selective estrogen receptor downregulator (SERD) that competitively inhibits agonist binding to ESR1 and triggers receptor downregulation, has confirmed that ESR1 frequently remains engaged in endocrine therapy–resistant cancers. We evaluated the activity of a new class of selective estrogen receptor modulators (SERM)/SERD hybrids (SSH) that downregulate ESR1 in relevant models of endocrine-resistant breast cancer. Building on the observation that concurrent inhibition of ESR1 and the cyclin-dependent kinases 4 and 6 (CDK4/6) significantly increased progression-free survival in advanced patients, we explored the activity of different SERD– or SSH–CDK4/6 inhibitor combinations in models of endocrine therapy–resistant ESR1+ breast cancer. Experimental Design: SERDs, SSHs, and the CDK4/6 inhibitor palbociclib were evaluated as single agents or in combination in established cellular and animal models of endocrine therapy–resistant ESR1+ breast cancer. Results: The combination of palbociclib with a SERD or an SSH was shown to effectively inhibit the growth of MCF7 cell or ESR1-mutant patient-derived tumor xenografts. In tamoxifen-resistant MCF7 xenografts, the palbociclib/SERD or SSH combination resulted in an increased duration of response as compared with either drug alone. Conclusions: A SERD– or SSH–palbociclib combination has therapeutic potential in breast tumors resistant to endocrine therapies or those expressing ESR1 mutations. Clin Cancer Res; 21(22); 5121–30. ©2015 AACR. See related commentary by DeMichele and Chodosh, p. 4999


Molecular and Cellular Biology | 2000

p50 Cdc37 Can Buffer the Temperature-Sensitive Properties of a Mutant of Hck

Glen M. Scholz; Steven D. Hartson; Kellie Cartledge; Nathan E. Hall; Jieya Shao; Ashley R. Dunn; Robert L. Matts

ABSTRACT Genetic studies have previously revealed that Cdc37p is required for the catalytic competence of v-Src in yeast. We have reasoned that temperature-sensitive mutants of Src family kinases might be more sensitive to the cellular level of p50Cdc37, the mammalian homolog of Cdc37p, than their wild-type counterpart, thus potentially providing a unique opportunity to elucidate the involvement of p50Cdc37 in the folding and stabilization of Src family kinases. A temperature-sensitive mutant of a constitutively active form of Hck (i.e., tsHck499F) was created by mutating two amino acids within the kinase domain of Hck499F. Significantly, overexpression of p50Cdc37 rescues the catalytic activity of tsHck499F at 33°C, while partially buffering it against inactivation at higher temperatures (e.g., 37 and 39°C). Hsp90 function is required for tsHck499F activity and its stabilization by p50Cdc37, but overexpression of Hsp90 is not sufficient to stabilize tsHck499F. Overexpression of p50Cdc37 promotes the association of tsHck499F with Hsp90, suggesting that the cellular level of p50Cdc37might be the rate-limiting step in the association oftsHck499F with Hsp90. A truncation mutant of p50Cdc37 that cannot bind Hsp90 still has a limited capacity to rescue the catalytic activity of tsHck499F and promote its association with Hsp90. This is a particularly important observation, since it argues that rather than solely acting as a passive adapter protein to tether tsHck499F to Hsp90, p50Cdc37 may also act allosterically to enhance the association of tsHck499F with Hsp90. The findings presented here might also have implications for our understanding of the evolution of protein kinases and tumor development.


PLOS ONE | 2010

F-Actin Binding Regions on the Androgen Receptor and Huntingtin Increase Aggregation and Alter Aggregate Characteristics

Suzanne Angeli; Jieya Shao; Marc I. Diamond

Protein aggregation is associated with neurodegeneration. Polyglutamine expansion diseases such as spinobulbar muscular atrophy and Huntington disease feature proteins that are destabilized by an expanded polyglutamine tract in their N-termini. It has previously been reported that intracellular aggregation of these target proteins, the androgen receptor (AR) and huntingtin (Htt), is modulated by actin-regulatory pathways. Sequences that flank the polyglutamine tract of AR and Htt might influence protein aggregation and toxicity through protein-protein interactions, but this has not been studied in detail. Here we have evaluated an N-terminal 127 amino acid fragment of AR and Htt exon 1. The first 50 amino acids of ARN127 and the first 14 amino acids of Htt exon 1 mediate binding to filamentous actin in vitro. Deletion of these actin-binding regions renders the polyglutamine-expanded forms of ARN127 and Htt exon 1 less aggregation-prone, and increases the SDS-solubility of aggregates that do form. These regions thus appear to alter the aggregation frequency and type of polyglutamine-induced aggregation. These findings highlight the importance of flanking sequences in determining the propensity of unstable proteins to misfold.


FEBS Letters | 2008

ROCK and PRK-2 mediate the inhibitory effect of Y-27632 on polyglutamine aggregation

Jieya Shao; William J. Welch; Marc I. Diamond

Polyglutamine expansion in huntingtin (Htt) and the androgen receptor (AR) causes untreatable neurodegenerative diseases. Y‐27632, a therapeutic lead, reduces Htt and AR aggregation in cultured cells, and Htt‐induced neurodegeneration in Drosophila. Y‐27632 inhibits both Rho‐associated kinases ROCK and PRK‐2, making its precise intracellular target uncertain. Over‐expression of either kinase increases Htt and AR aggregation. Three ROCK inhibitors (Y‐27632, HA‐1077, and H‐1152P), and a specific ROCK inhibitory peptide reduce polyglutamine protein aggregation, as does knockdown of ROCK or PRK‐2 by RNAi. RNAi also indicates that each kinase is required for the inhibitory effects of Y‐27632 to manifest fully. These two actin regulatory kinases are thus involved in polyglutamine aggregation, and their simultaneous inhibition may be an important therapeutic goal.

Collaboration


Dive into the Jieya Shao's collaboration.

Top Co-Authors

Avatar

Marc I. Diamond

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robert Crowder

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Shunqiang Li

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Chanpheng Phommaly

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Rogers

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jeremy Hoog

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jonathan T. Lei

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sherri R. Davies

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge