Network


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

Hotspot


Dive into the research topics where Thuy Ai Nguyen is active.

Publication


Featured researches published by Thuy Ai Nguyen.


Current Biology | 2005

Functional conservation of an insect odorant receptor gene across 250 million years of evolution.

Walton D. Jones; Thuy Ai Nguyen; Brian Kloss; Kevin J. Lee; Leslie B. Vosshall

Pest insects have a profound negative impact on agriculture and human health. Significant global losses of crops, stored agricultural products, timber and livestock can be attributed to damage and destruction by insects . Blood-feeding insects such as mosquitoes, flies and ticks transmit many of humanitys most devastating infectious diseases. Insect-borne diseases account for more than one million annual fatalities, and insect-associated illnesses surpass 300 million annual reported cases . The medical and economic impact of these animals can be ascribed in part to the sensitivity and selectivity of their olfactory systems, essential for location of their preferred plant and animal hosts.


Cancer and Metastasis Reviews | 2008

The type 2C phosphatase Wip1: An oncogenic regulator of tumor suppressor and DNA damage response pathways

Xiongbin Lu; Thuy Ai Nguyen; Sung Hwan Moon; Yolanda F. Darlington; Matthias Sommer; Lawrence A. Donehower

The Wild-type p53-induced phosphatase 1, Wip1 (or PPM1D), is unusual in that it is a serine/threonine phosphatase with oncogenic activity. A member of the type 2C phosphatases (PP2Cδ), Wip1 has been shown to be amplified and overexpressed in multiple human cancer types, including breast and ovarian carcinomas. In rodent primary fibroblast transformation assays, Wip1 cooperates with known oncogenes to induce transformed foci. The recent identification of target proteins that are dephosphorylated by Wip1 has provided mechanistic insights into its oncogenic functions. Wip1 acts as a homeostatic regulator of the DNA damage response by dephosphorylating proteins that are substrates of both ATM and ATR, important DNA damage sensor kinases. Wip1 also suppresses the activity of multiple tumor suppressors, including p53, ATM, p16INK4a and ARF. We present evidence that the suppression of p53, p38 MAP kinase, and ATM/ATR signaling pathways by Wip1 are important components of its oncogenicity when it is amplified and overexpressed in human cancers.


Journal of Neuro-oncology | 2008

Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D

Robert C. Castellino; Massimiliano De Bortoli; Xiongbin Lu; Sung Hwan Moon; Thuy Ai Nguyen; Mark A. Shepard; Pulivarthi H. Rao; Lawrence A. Donehower; John Kim

Medulloblastoma is the most common malignant brain tumor of childhood. Despite numerous advances, clinical challenges range from recurrent and progressive disease to long-term toxicities in survivors. The lack of more effective, less toxic therapies results from our limited understanding of medulloblastoma growth. Although TP53 is the most commonly altered gene in cancers, it is rarely mutated in medulloblastoma. Accumulating evidence, however, indicates that TP53 pathways are disrupted in medulloblastoma. Wild-typep53-induced phosphatase 1 (WIP1 or PPM1D) encodes a negative regulator of p53. WIP1 amplification (17q22-q23) and its overexpression have been reported in diverse cancer types. We examined primary medulloblastoma specimens and cell lines, and detected WIP1 copy gain and amplification prevalent among but not exclusively in the tumors with 17q gain and isochromosome 17q (i17q), which are among the most common cytogenetic lesions in medulloblastoma. WIP1 RNA levels were significantly higher in the tumors with 17q gain or i17q. Immunoblots confirmed significant WIP1 protein in primary tumors, generally higher in those with 17q gain or i17q. Under basal growth conditions and in response to the chemotherapeutic agent, etoposide, WIP1 antagonized p53-mediated apoptosis in medulloblastoma cell lines. These results indicate that medulloblastoma express significant levels of WIP1 that modulate genotoxic responsiveness by negatively regulating p53.


Nucleic Acids Research | 2013

Diverse stresses dramatically alter genome-wide p53 binding and transactivation landscape in human cancer cells

Daniel Menendez; Thuy Ai Nguyen; Johannes M. Freudenberg; Viju J. Mathew; Carl W. Anderson; Raja Jothi; Michael A. Resnick

The effects of diverse stresses on promoter selectivity and transcription regulation by the tumor suppressor p53 are poorly understood. We have taken a comprehensive approach to characterizing the human p53 network that includes p53 levels, binding, expression and chromatin changes under diverse stresses. Human osteosarcoma U2OS cells treated with anti-cancer drugs Doxorubicin (DXR) or Nutlin-3 (Nutlin) led to strikingly different p53 gene binding patterns based on chromatin immunoprecipitation with high-throughput sequencing experiments. Although two contiguous RRRCWWGYYY decamers is the consensus binding motif, p53 can bind a single decamer and function in vivo. Although the number of sites bound by p53 was six times greater for Nutlin than DXR, expression changes induced by Nutlin were much less dramatic compared with DXR. Unexpectedly, the solvent dimethylsulphoxide (DMSO) alone induced p53 binding to many sites common to DXR; however, this binding had no effect on target gene expression. Together, these data imply a two-stage mechanism for p53 transactivation where p53 binding only constitutes the first stage. Furthermore, both p53 binding and transactivation were associated with increased active histone modification histone H3 lysine 4 trimethylation. We discovered 149 putative new p53 target genes including several that are relevant to tumor suppression, revealing potential new targets for cancer therapy and expanding our understanding of the p53 regulatory network.


Journal of Biological Chemistry | 2010

Wild-type p53-induced Phosphatase 1 Dephosphorylates Histone Variant γ-H2AX and Suppresses DNA Double Strand Break Repair

Sung Hwan Moon; Lin Lin; Xinna Zhang; Thuy Ai Nguyen; Yolanda F. Darlington; Alan S. Waldman; Xiongbin Lu; Lawrence A. Donehower

In response to DNA double strand breaks, the histone variant H2AX at the break site is phosphorylated at serine 139 by DNA damage sensor kinases such as ataxia telangiectasia-mutated, forming γ-H2AX. This phosphorylation event is critical for sustained recruitment of other proteins to repair the break. After repair, restoration of the cell to a prestress state is associated with γ-H2AX dephosphorylation and dissolution of γ-H2AX-associated damage foci. The phosphatases PP2A and PP4 have previously been shown to dephosphorylate γ-H2AX. Here, we demonstrate that the wild-type p53-induced phosphatase 1 (WIP1) also dephosphorylates γ-H2AX at serine 139 in vitro and in vivo. Overexpression of WIP1 reduces formation of γ-H2AX foci in response to ionizing and ultraviolet radiation and blocks recruitment of MDC1 (mediator of DNA damage checkpoint 1) and 53BP1 (p53 binding protein 1) to DNA damage foci. Finally, these inhibitory effects of WIP1 on γ-H2AX are accompanied by WIP1 suppression of DNA double strand break repair. Thus, WIP1 has a homeostatic role in reversing the effects of ataxia telangiectasia-mutated phosphorylation of H2AX.


Molecular Carcinogenesis | 2006

Augmented cancer resistance and DNA damage response phenotypes in PPM1D null mice

Bonnie Nannenga; Xiongbin Lu; Melissa Dumble; Marc van Maanen; Thuy Ai Nguyen; Richard E. Sutton; T. Rajendra Kumar; Lawrence A. Donehower

The p53‐induced serine/threonine phosphatase, protein phosphatase 1D magnesium‐dependent, delta isoform (PPM1D) (or wild‐type p53‐induced phosphatase 1 (Wip1)), exhibits oncogenic activity in vitro and in vivo. It behaves as an oncogene in rodent fibroblast transformation assays and is amplified and overexpressed in several human tumor types. It may contribute to oncogenesis through functional inactivation of p53. Here, we show that the oncogenic function of PPM1D is associated with its phosphatase activity. While overexpressed PPM1D may be oncogenic, PPM1D null mice are resistant to spontaneous tumors over their entire lifespan. This cancer resistance may be based in part on an augmented stress response following DNA damage. PPM1D null mice treated with ionizing radiation display increased p53 protein levels and increased phosphorylation of p38 MAP kinase, p53, checkpoint kinase 1 (Chk1), and checkpoint kinase 2 (Chk2) in their tissues compared to their wild‐type (WT) counterparts. Male PPM1D null mice show a modest reduction in longevity, reduced serum insulin‐like growth factor 1 (IGF‐1) levels, and reduced body weight compared to WT mice. The PPM1D null mouse phenotypes indicate that PPM1D has a homeostatic role in abrogating the DNA damage response and may regulate aspects of male longevity.


Cell Cycle | 2005

Reversal of the ATM/ATR-mediated DNA damage response by the oncogenic phosphatase PPM1D.

Xiongbin Lu; Thuy Ai Nguyen; Lawrence A. Donehower

The eukaryotic cell has evolved a sophisticated set of cell signaling pathways that respond to DNA damage and efficiently repair that damage, protecting the cell from deleterious mutations, genomic instability, and transformation into a cancerous state. The ATM and ATR serine/threonine kinases are key sensors and transducers of DNA damage signals through phosphorylation of an array of signaling molecules that mediate all aspects of the DNA damage response, including enforcement of cell cycle checkpoints and direct repair of damaged DNA. We have shown that a type 2C serine/threonine phosphatase, PPM1D (or Wip1), can reverse the phosphorylation status of ATM/ATR-phosphorylated proteins p53 and Chk1. This dephosphorylation of p53 and Chk1 by PPM1D may result in reduced functional activities and is accompanied by suppression of DNA damage-induced cell cycle checkpoints and some aspects of DNA repair. Because PPM1D is transcriptionally activated by p53 in response to DNA damage, PPM1D may serve as a critical component of a p53 negative feedback regulatory loop since it now appears that PPM1D can inhibit p53 activity by at least four different molecular mechanisms. This may explain why PPM1D is amplified and overexpressed in a subset of human breast cancers that invariably retain wild type p53 alleles. We hypothesize that PPM1D is a homeostatic regulator of the DNA damage response that returns the cell to a more normal unstressed state following repair of the damage.


Cell Cycle | 2010

Dephosphorylation of γH2AX by WIP1: An important homeostatic regulatory event in DNA repair and cell cycle control

Sung Hwan Moon; Thuy Ai Nguyen; Yolanda F. Darlington; Xiongbin Lu; Lawrence A. Donehower

DNA double strand breaks are a particularly toxic form of DNA damage and the mammalian cell has evolved an intricate set of responses to repair this type of DNA lesion. A key early event in the DNA damage response (DDR) is ATM phosphorylation of the histone variant H2AX at serine 139 at the site of the DNA break. Phosphorylated S139 H2AX, or γ-H2AX, forms a docking site for binding of MDC1, leading to sustained recruitment of other DNA repair factors that mediate the repair of the DNA double strand break. Moreover, recruitment of MDC1 to the break site activates cell cycle checkpoints, protecting the cell from replication of damaged DNA templates. While the molecular events leading to DNA double strand break repair have been well described, the deactivating or homeostatic mechanisms following completion of repair remain largely unexplored. Recent publications by our laboratories and the Medema laboratory shed new light on this issue. Both publications showed that the Wild-type p53-Induced Phosphatase 1 (WIP1) directly dephosphorylates γ-H2AX. WIP1 migrates to the sites of irradiation-induced foci (IRIF), though at a delayed rate relative to MDC1 and mediates γ-H2AX dephosphorylation, presumably after DNA repair is complete. This prevents recruitment of other repair factors such as MDC1 and 53BP1 to the DNA damage sites and promotes the dissolution of IRIF. In addition, overexpression of WIP1 has a suppressive effect on DNA double strand break repair. Taken together, these reports further implicate WIP1 as a critical homeostatic regulator of the DDR.


Cell Cycle | 2008

The Wip1 phosphatase and Mdm2: Cracking the "Wip" on p53 stability

Xiongbin Lu; Thuy Ai Nguyen; Xinna Zhang; Lawrence A. Donehower

The p53 tumor suppressor is essential in maintaining genomic integrity in response to cellular stresses. In response to DNA damage, p53 is activated and stabilized largely through post-translational modifications, including phosphorylation by DNA damage responsive kinases such as ATM and ATR. Activated p53 transactivates a battery of genes that can mediate either cell cycle arrest or apoptosis. In those instances where p53 facilitates cell cycle arrest, a means to return the cell to a pre-stress state with low p53 levels is important. The E3 ubiquitin ligase Mdm2 is one p53 transcriptional target that accumulates after damage and promotes p53 ubiquitination and degradation. Thus, p53 and Mdm2 form a critical negative feedback regulatory loop that helps to maintain appropriate p53 levels in the presence or absence of stress. We propose here that Wip1 (Wildtype p53-Induced Phosphatase 1), also known as PPM1D, plays an important role in the p53-Mdm2 autoregulatory loop. We have recently shown that Wip1, also a p53 target gene, dephosphorylates Mdm2 at Ser395 (an ATM target site), resulting in stabilization of Mdm2, enhanced Mdm2-p53 binding, and enhanced ubiquitination of p53 by Mdm2. Thus, Wip1 facilitates Mdm2-mediated degradation of p53. The p53 inhibitory role of Wip1 implicates it as a potential oncogene and indeed Wip1 is amplified and overexpressed in a number of human cancers. Wip1 may inhibit p53 signaling by multiple mechanisms, but our data suggests that its largest effects are due to dephosphorylation of Mdm2.


DNA Repair | 2010

The oncogenic phosphatase WIP1 negatively regulates nucleotide excision repair.

Thuy Ai Nguyen; Scott D. Slattery; Sung Hwan Moon; Yolanda F. Darlington; Xiongbin Lu; Lawrence A. Donehower

Nucleotide excision repair (NER) is the only mechanism in humans to repair UV-induced DNA lesions such as pyrimidine (6-4) pyrimidone photoproducts and cyclobutane pyrimidine dimers (CPDs). In response to UV damage, the ataxia telangiectasia mutated and Rad3-related (ATR) kinase phosphorylates and activates several downstream effector proteins, such as p53 and XPA, to arrest cell cycle progression, stimulate DNA repair, or initiate apoptosis. However, following the completion of DNA repair, there must be active mechanisms that restore the cell to a prestressed homeostatic state. An important part of this recovery must include a process to reduce p53 and NER activity as well as to remove repair protein complexes from the DNA damage sites. Since activation of the damage response occurs in part through phosphorylation, phosphatases are obvious candidates as homeostatic regulators of the DNA damage and repair responses. Therefore, we investigated whether the serine/threonine wild-type p53-induced phosphatase 1 (WIP1/PPM1D) might regulate NER. WIP1 overexpression inhibits the kinetics of NER and CPD repair, whereas WIP1 depletion enhances NER kinetics and CPD repair. This NER suppression is dependent on WIP1 phosphatase activity, as phosphatase-dead WIP1 mutants failed to inhibit NER. Moreover, WIP1 suppresses the kinetics of UV-induced damage repair largely through effects on NER, as XPD-deficient cells are not further suppressed in repairing UV damage by overexpressed WIP1. Wip1 null mice quickly repair their CPD and undergo less UV-induced apoptosis than their wild-type counterparts. In vitro phosphatase assays identify XPA and XPC as two potential WIP1 targets in the NER pathway. Thus WIP1 may suppress NER kinetics by dephosphorylating and inactivating XPA and XPC and other NER proteins and regulators after UV-induced DNA damage is repaired.

Collaboration


Dive into the Thuy Ai Nguyen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiongbin Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Daniel Menendez

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kevin J. Lee

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Michael A. Resnick

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sung Hwan Moon

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl W. Anderson

Brookhaven National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Alan Herron

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Leping Li

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge