Network


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

Hotspot


Dive into the research topics where Katriana Nugent is active.

Publication


Featured researches published by Katriana Nugent.


Neoplasia | 2015

Structure-Function Studies of the bHLH Phosphorylation Domain of TWIST1 in Prostate Cancer Cells

Rajendra P. Gajula; Sivarajan T. Chettiar; Russell Williams; Katriana Nugent; Yoshinori Kato; Hailun Wang; Reem Malek; Kekoa Taparra; Jessica Cades; Anvesh Annadanam; A-Rum Yoon; Elana Fertig; Beth A. Firulli; Lucia Mazzacurati; Timothy F. Burns; Anthony B. Firulli; Steven S. An; Phuoc T. Tran

The TWIST1 gene has diverse roles in development and pathologic diseases such as cancer. TWIST1 is a dimeric basic helix-loop-helix (bHLH) transcription factor existing as TWIST1-TWIST1 or TWIST1-E12/47. TWIST1 partner choice and DNA binding can be influenced during development by phosphorylation of Thr125 and Ser127 of the Thr-Gln-Ser (TQS) motif within the bHLH of TWIST1. The significance of these TWIST1 phosphorylation sites for metastasis is unknown. We created stable isogenic prostate cancer cell lines overexpressing TWIST1 wild-type, phospho-mutants, and tethered versions. We assessed these isogenic lines using assays that mimic stages of cancer metastasis. In vitro assays suggested the phospho-mimetic Twist1-DQD mutation could confer cellular properties associated with pro-metastatic behavior. The hypo-phosphorylation mimic Twist1-AQA mutation displayed reduced pro-metastatic activity compared to wild-type TWIST1 in vitro, suggesting that phosphorylation of the TWIST1 TQS motif was necessary for pro-metastatic functions. In vivo analysis demonstrates that the Twist1-AQA mutation exhibits reduced capacity to contribute to metastasis, whereas the expression of the Twist1-DQD mutation exhibits proficient metastatic potential. Tethered TWIST1-E12 heterodimers phenocopied the Twist1-DQD mutation for many in vitro assays, suggesting that TWIST1 phosphorylation may result in heterodimerization in prostate cancer cells. Lastly, the dual phosphatidylinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibitor BEZ235 strongly attenuated TWIST1-induced migration that was dependent on the TQS motif. TWIST1 TQS phosphorylation state determines the intensity of TWIST1-induced pro-metastatic ability in prostate cancer cells, which may be partly explained mechanistically by TWIST1 dimeric partner choice.


Cancer Research | 2017

TWIST1-WDR5-Hottip regulates Hoxa9 chromatin to facilitate prostate cancer metastasis.

Reem Malek; Rajendra P. Gajula; Russell Williams; Belinda Nghiem; Brian W. Simons; Katriana Nugent; Hailun Wang; Kekoa Taparra; Ghali Lemtiri-Chlieh; A-Rum Yoon; Lawrence D. True; Steven S. An; Theodore L. DeWeese; Ashley E. Ross; Edward M. Schaeffer; Kenneth J. Pienta; Paula J. Hurley; Colm Morrissey; Phuoc T. Tran

TWIST1 is a transcription factor critical for development that can promote prostate cancer metastasis. During embryonic development, TWIST1 and HOXA9 are coexpressed in mouse prostate and then silenced postnatally. Here we report that TWIST1 and HOXA9 coexpression are reactivated in mouse and human primary prostate tumors and are further enriched in human metastases, correlating with survival. TWIST1 formed a complex with WDR5 and the lncRNA Hottip/HOTTIP, members of the MLL/COMPASS-like H3K4 methylases, which regulate chromatin in the Hox/HOX cluster during development. TWIST1 overexpression led to coenrichment of TWIST1 and WDR5 as well as increased H3K4me3 chromatin at the Hoxa9/HOXA9 promoter, which was dependent on WDR5. Expression of WDR5 and Hottip/HOTTIP was also required for TWIST1-induced upregulation of HOXA9 and aggressive cellular phenotypes such as invasion and migration. Pharmacologic inhibition of HOXA9 prevented TWIST1-induced aggressive prostate cancer cellular phenotypes in vitro and metastasis in vivo This study demonstrates a novel mechanism by which TWIST1 regulates chromatin and gene expression by cooperating with the COMPASS-like complex to increase H3K4 trimethylation at target gene promoters. Our findings highlight a TWIST1-HOXA9 embryonic prostate developmental program that is reactivated during prostate cancer metastasis and is therapeutically targetable. Cancer Res; 77(12); 3181-93. ©2017 AACR.


Cancer Research | 2016

RK-33 Radiosensitizes Prostate Cancer Cells by Blocking the RNA Helicase DDX3

Min Xie; Farhad Vesuna; Saritha Tantravedi; Guus M. Bol; Marise R. Heerma van Voss; Katriana Nugent; Reem Malek; Kathleen L. Gabrielson; Paul J. van Diest; Phuoc T. Tran; Venu Raman

Despite advances in diagnosis and treatment, prostate cancer is the most prevalent cancer in males and the second highest cause of cancer-related mortality. We identified an RNA helicase gene, DDX3 (DDX3X), which is overexpressed in prostate cancers, and whose expression is directly correlated with high Gleason scores. Knockdown of DDX3 in the aggressive prostate cancer cell lines DU145 and 22Rv1 resulted in significantly reduced clonogenicity. To target DDX3, we rationally designed a small molecule, RK-33, which docks into the ATP-binding domain of DDX3. Functional studies indicated that RK-33 preferentially bound to DDX3 and perturbed its activity. RK-33 treatment of prostate cancer cell lines DU145, 22Rv1, and LNCaP (which have high DDX3 levels) decreased proliferation and induced a G1 phase cell-cycle arrest. Conversely, the low DDX3-expressing cell line, PC3, exhibited few changes following RK-33 treatment. Importantly, combination studies using RK-33 and radiation exhibited synergistic effects both in vitro and in a xenograft model of prostate cancer demonstrating the role of RK-33 as a radiosensitizer. Taken together, these results indicate that blocking DDX3 by RK-33 in combination with radiation treatment is a viable option for treating locally advanced prostate cancer. Cancer Res; 76(21); 6340-50. ©2016 AACR.


Molecular Cancer Research | 2017

A First-in-Class TWIST1 Inhibitor with Activity in Oncogene-driven Lung Cancer

Zachary A. Yochum; Jessica Cades; Lucia Mazzacurati; Neil M. Neumann; Susheel Khetarpal; Suman Chatterjee; Hailun Wang; Myriam Attar; Eric H.-B. Huang; Sarah N. Chatley; Katriana Nugent; Ashwin Somasundaram; Johnathan A. Engh; Andrew J. Ewald; Yoon Jae Cho; Charles M. Rudin; Phuoc T. Tran; Timothy F. Burns

TWIST1, an epithelial–mesenchymal transition (EMT) transcription factor, is critical for oncogene-driven non–small cell lung cancer (NSCLC) tumorigenesis. Given the potential of TWIST1 as a therapeutic target, a chemical–bioinformatic approach using connectivity mapping (CMAP) analysis was used to identify TWIST1 inhibitors. Characterization of the top ranked candidates from the unbiased screen revealed that harmine, a harmala alkaloid, inhibited multiple TWIST1 functions, including single-cell dissemination, suppression of normal branching in 3D epithelial culture, and proliferation of oncogene driver-defined NSCLC cells. Harmine treatment phenocopied genetic loss of TWIST1 by inducing oncogene-induced senescence or apoptosis. Mechanistic investigation revealed that harmine targeted the TWIST1 pathway through its promotion of TWIST1 protein degradation. As dimerization is critical for TWIST1 function and stability, the effect of harmine on specific TWIST1 dimers was examined. TWIST1 and its dimer partners, the E2A proteins, which were found to be required for TWIST1-mediated functions, regulated the stability of the other heterodimeric partner posttranslationally. Harmine preferentially promoted degradation of the TWIST1-E2A heterodimer compared with the TWIST-TWIST1 homodimer, and targeting the TWIST1-E2A heterodimer was required for harmine cytotoxicity. Finally, harmine had activity in both transgenic and patient-derived xenograft mouse models of KRAS-mutant NSCLC. These studies identified harmine as a first-in-class TWIST1 inhibitor with marked anti-tumor activity in oncogene-driven NSCLC including EGFR mutant, KRAS mutant and MET altered NSCLC. Implications: TWIST1 is required for oncogene-driven NSCLC tumorigenesis and EMT; thus, harmine and its analogues/derivatives represent a novel therapeutic strategy to treat oncogene-driven NSCLC as well as other solid tumor malignancies. Mol Cancer Res; 15(12); 1764–76. ©2017 AACR.


Prostate Cancer and Prostatic Diseases | 2018

Combining immune check-point blockade and cryoablation in an immunocompetent hormone sensitive murine model of prostate cancer

Benjamin Benzon; Stephanie Glavaris; Brian W. Simons; Robert M. Hughes; Kamyar Ghabili; Patrick Mullane; Rebecca Miller; Katriana Nugent; Brian Shinder; Jeffrey J. Tosoian; Ephraim J. Fuchs; Phuoc T. Tran; Paula J. Hurley; Milena Vuica-Ross; Edward M. Schaeffer; Charles G. Drake; Ashley E. Ross

BackgroundProstate cancer remains the second leading cause of cancer related death in men. Immune check point blocking antibodies have revolutionized treatment of multiple solid tumors, but results in prostate cancer remain marginal. Previous reports have suggested that local therapies, in particular cryoablation might increase tumor immunogenicity. In this work, we examine potential synergism between tumor cryoabalation and check point blocking antibodies.MethodsFVB/NJ mice were injected subcutaneously into each flank with either 1u2009×u2009106 or 0.2u2009×u2009106 isogenic hormone sensitive Myc-Cap cells to establish synchronous grafts. Mice were treated with four intraperitoneal injections of anti-PD-1 (10u2009mg/kg), anti-CTLA-4 (1u2009mg/kg), or isotype control antibody with or without adjuvant cryoablation of the larger tumor graft and with or without neo-adjuvant androgen deprivation with degarelix (ADT). Mouse survival and growth rates of tumor grafts were measured. The immune dependency of observed oncological effects was evaluated by T cell depletion experiments.ResultsTreatment with anti-CTLA-4 antibody and cryoablation delayed the growth of the distant tumor by 14.8 days (pu2009=u20090.0006) and decreased the mortality rate by factor of 4 (pu2009=u20090.0003) when compared to cryoablation alone. This synergy was found to be dependent on CD3+u2009and CD8+u2009cells. Combining PD-1 blockade with cryoablation did not show a benefit over use of either treatment alone. Addition of ADT to anti-PD1 therapy and cryoablation doubled the time to accelerated growth in the untreated tumors (pu2009=u20090.0021) and extended survival when compared to cryoablation combined with ADT in 25% of the mice. Effects of combining anti-PD1 with ADT and cryoablation on mouse survival were obviated by T cell depletion.ConclusionTrimodal therapy consisting of androgen deprivation, cryoablation and PD-1 blockade, as well as the combination of cryoablation and low dose anti-CTLA-4 blockade showed that local therapies with cryoablation could be considered to augment the effects of checkpoint blockade in prostate cancer.


Radiation Research | 2016

Evaluation of On- and Off-Line Bioluminescence Tomography System for Focal Irradiation Guidance

Bin Zhang; John Wong; Iulian Iordachita; Juvenal Reyes; Katriana Nugent; Phuoc T. Tran; Stephen W. Tuttle; Constantinos Koumenis; K Wang

In response to the limitations of computed tomography (CT) and cone-beam CT (CBCT) in irradiation guidance, especially for soft-tissue targets without the use of contrast agents, our group developed a solution that implemented bioluminescence tomography (BLT) as the image-guidance modality for preclinical radiation research. However, adding such a system to existing small animal irradiators is no small task. A potential solution is to utilize an off-line BLT system in close proximity to the irradiator, with stable and effective animal transport between the two systems. In this study, we investigated the localization accuracy of an off-line BLT system when used for the small animal radiation research platform (SARRP) and compared the results with those of an on-line system. The CBCT was equipped on both the off-line BLT system and the SARRP, with a distance of 5 m between them. To evaluate the setup error during animal transport between the two systems, the mice underwent CBCT imaging on the SARRP and were then transported to the off-line system for a second CBCT imaging session. The normalized intensity difference of the two images and the corresponding histogram and correlation were computed to evaluate if the transport process perturbed animal positioning. Strong correlation (correlation coefficients >0.95) between the SARRP and the off-line mouse CBCT was observed. The offset of the implanted light source center can be maintained within 0.2 mm during transport. To compare the target localization accuracy using the on-line SARRP BLT and the off-line system, a self-illuminated bioluminescent source was implanted in the abdomen of anesthetized mice. In addition to the application for dose calculation, CBCT imaging was also employed to generate the mesh grid of the imaged mouse for BLT reconstruction. Two scenarios were devised and compared, which involved localization of the luminescence source based on either: 1. on-line SARRP bioluminescence image and CBCT; or 2. off-line bioluminescence image and SARRP CBCT. The first scenario is assumed to have the least setup error, because no animal transport was involved. The second scenario examines if an off-line BLT system, with the mesh generated from the SARRP CBCT, can be used to guide SARRP irradiation when there is minimal target contrast in CBCT. Stability during animal transport between the two systems was maintained. The center of mass (CoM) of the light source reconstructed by the off-line BLT had an offset of 1.0 ± 0.4 mm from the true CoM derived from the SARRP CBCT. These results are comparable to the offset of 1.0 ± 0.2 mm using on-line BLT. With CBCT information provided by the SARRP and effective animal immobilization during transport, these findings support the utilization of an off-line BLT-guided system, in close proximity to the SARRP, for accurate soft-tissue target localization. In addition, a dedicated standalone BLT system for our partner site at the University of Pennsylvania was introduced in this study.


Journal of Clinical Investigation | 2018

O-GlcNAcylation is required for mutant KRAS -induced lung tumorigenesis

Kekoa Taparra; Hailun Wang; Reem Malek; Audrey Lafargue; Mustafa A. Barbhuiya; Xing Wang; Brian W. Simons; Matthew Ballew; Katriana Nugent; Jennifer Groves; Russell Williams; Takumi Shiraishi; James E. Verdone; Gokben Yildirir; Roger Henry; Bin Zhang; John Wong; Ken Kang-Hsin Wang; Barry D. Nelkin; Kenneth J. Pienta; Dean W. Felsher; Natasha E. Zachara; Phuoc T. Tran

Mutant KRAS drives glycolytic flux in lung cancer, potentially impacting aberrant protein glycosylation. Recent evidence suggests aberrant KRAS drives flux of glucose into the hexosamine biosynthetic pathway (HBP). HBP is required for various glycosylation processes, such as protein N- or O-glycosylation and glycolipid synthesis. However, its function during tumorigenesis is poorly understood. One contributor and proposed target of KRAS-driven cancers is a developmentally conserved epithelial plasticity program called epithelial-mesenchymal transition (EMT). Here we showed in novel autochthonous mouse models that EMT accelerated KrasG12D lung tumorigenesis by upregulating expression of key enzymes of the HBP pathway. We demonstrated that HBP was required for suppressing KrasG12D-induced senescence, and targeting HBP significantly delayed KrasG12D lung tumorigenesis. To explore the mechanism, we investigated protein glycosylation downstream of HBP and found elevated levels of O-linked &bgr;-N-acetylglucosamine (O-GlcNAcylation) posttranslational modification on intracellular proteins. O-GlcNAcylation suppressed KrasG12D oncogene-induced senescence (OIS) and accelerated lung tumorigenesis. Conversely, loss of O-GlcNAcylation delayed lung tumorigenesis. O-GlcNAcylation of proteins SNAI1 and c-MYC correlated with the EMT-HBP axis and accelerated lung tumorigenesis. Our results demonstrated that O-GlcNAcylation was sufficient and required to accelerate KrasG12D lung tumorigenesis in vivo, which was reinforced by epithelial plasticity programs.


Journal of Clinical Investigation | 2018

ATR kinase inhibitor AZD6738 potentiates CD8+ T cell-dependent antitumor activity following radiation

Frank P. Vendetti; Pooja Karukonda; David A. Clump; Troy Teo; R Lalonde; Katriana Nugent; Matthew Ballew; Brian F. Kiesel; Jan H. Beumer; Saumendra N. Sarkar; Thomas P. Conrads; Mark J. O'Connor; Robert L. Ferris; Phuoc T. Tran; Greg M. Delgoffe; Christopher J. Bakkenist

DNA-damaging chemotherapy and radiation therapy are integrated into the treatment paradigm of the majority of cancer patients. Recently, immunotherapy that targets the immunosuppressive interaction between programmed death 1 (PD-1) and its ligand PD-L1 has been approved for malignancies including non–small cell lung cancer, melanoma, and head and neck squamous cell carcinoma. ATR is a DNA damage–signaling kinase activated at damaged replication forks, and ATR kinase inhibitors potentiate the cytotoxicity of DNA-damaging chemotherapies. We show here that the ATR kinase inhibitor AZD6738 combines with conformal radiation therapy to attenuate radiation-induced CD8+ T cell exhaustion and potentiate CD8+ T cell activity in mouse models of Kras-mutant cancer. Mechanistically, AZD6738 blocks radiation-induced PD-L1 upregulation on tumor cells and dramatically decreases the number of tumor-infiltrating Tregs. Remarkably, AZD6738 combines with conformal radiation therapy to generate immunologic memory in complete responder mice. Our work raises the possibility that a single pharmacologic agent may enhance the cytotoxic effects of radiation while concurrently potentiating radiation-induced antitumor immune responses.


Clinical Cancer Research | 2018

Abstract B12: Centrosome clustering inhibition as a novel strategy to sensitize non-small cell lung cancer to radiation treatment and immunotherapy

Hailun Wang; Katriana Nugent; Matthew Ballew; Ghali Lemtiri-Chlieh; Natasha Raman; Michelle S. Levine; Andrew J. Holland; Phuoc T. Tran

The centrosome is a microtubule-organizing center that plays an important role during M-phase and cell division where it organizes the two poles of the bipolar microtubule spindle apparatus from which chromosomes are segregated. In contrast to most normal cells, the vast majority of lung cancers contain extra copies of centrosomes, and a large body of circumstantial evidence links extra centrosomes to the development of cancer. Cells with supernumerary centrosomes form multipolar mitotic spindles, which, if not corrected, lead to lethal multipolar divisions or “mitotic catastrophe.” To overcome this, lung cancer cells cluster their centrosomes into two spindle poles, enabling tumor cells to survive. Thus, inhibition of centrosome clustering (CCi) can force cancer cells into multipolar divisions, lead to chromosomal instability, make cancer cells more sensitive to radiation treatment, and eventually result in the selective killing of cancer cells. At the same time, this approach should spare healthy tissues with normal centrosome numbers, resulting in a favorable therapeutic index. To examine the relationship between CCi and radiation-treatment sensitivity, we first treated three NSCLC cell lines (A549, H460 and H358) and one immortalized primary human bronchial epithelial cell (HBEC) with griseofulvin—a centrosomal clustering inhibitor. Immunofluorescent staining for pericentrin and alpha-tubulin, we observed a significant increase in the number of cells with multipolar spindles in NSCLC cells compared to HBECs (40-80% vs. 15%). Next, we examined the ability of griseofulvin to radiosensitize these cells to radiation treatment. By using clonogenic assays, we saw significantly less clonogenic potential of NSCLC cells following radiation treatment after exposure to 30 uM of griseofulvin, but griseofulvin did not radiosensitize HBECs. To further confirm our results, we genetically targeted KIFC1, a member of kinesin-14 family of motor proteins that have been shown to be essential for clustering centrosomes in cancer cells, but are not required for division in normal cells. In our experiments, knockdown of KIFC1 expression with siRNA also significantly sensitized NSCLC cells to radiation treatment. Finally, when we treated the NSCLC cells with griseofulvin, we saw a significant increase in the formation of micronuclei in cancer cells. Recently, it has been shown that micronuclei are important sources of immunostimulatory DNA via activation of the cGAS-STING pathway. Therefore, inhibition of CC could potentially augment immunogenicity and increase efficiency of immunotherapy (IMT). In conclusion, inhibition of CC resulted in the formation of multipolar spindle in NSCLCs and further sensitized NSCLCs to radiation treatment. CC is not typically required for normal cells, and thus CCi may be a more specific and perhaps less toxic therapy for NSCLCs. The potential of CCi to enhance IMT is under investigation. Citation Format: Hailun Wang, Katriana Nugent, Matthew Ballew, Ghali Lemtiri-Chlieh, Natasha Raman, Michelle Levine, Andrew Holland, Phuoc Tran. Centrosome clustering inhibition as a novel strategy to sensitize non-small cell lung cancer to radiation treatment and immunotherapy [abstract]. In: Proceedings of the Fifth AACR-IASLC International Joint Conference: Lung Cancer Translational Science from the Bench to the Clinic; Jan 8-11, 2018; San Diego, CA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(17_Suppl):Abstract nr B12.


Cancer Research | 2018

Abstract B47: Modeling epithelial plasticity-induced erlotinib resistance in non-small cell lung cancer

Hailun Wang; Jessica Cades; Zachary A. Yochum; Katriana Nugent; Charles M. Rudin; Timothy F. Burns; Phuoc T. Tran

Background: Advanced non-small lung cancer (NSCLC) patients with EGFR mutations initially respond to treatment with the EGFR-targeted tyrosine kinase inhibitors (TKIs) such as erlotinib, but will invariably acquire resistance with progression of disease within 10–16 months. Mechanisms of EGFR TKIs resistance include second-site EGFR mutations (>50%), MET amplfication (5–10%), and mutations in PIK3CA ( Methods: To examine the relationship between epithelial plasticity and erlotinib resistance in EGFR mutant lung cancers, we created an inducible CCSP-rtTA/tetO-EgfrL858R/Twist1 (CET) transgenic mice (Twist1 is a key regulator of EMT). We utilized the tetracycline-inducible gene expression system to control EgfrL858R/Twist1 gene expression in the lung by providing or withdrawing doxycycline to the mice. The mice were treated for 3 weeks with erlotinib and scanned by CT each week, and survival of the mice was also recorded. The tumor tissues were collected 1 week and 3 weeks after the start of treatment and used for immunohistochemical staining for HE 2017 Sep 24-27; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(10 Suppl):Abstract nr B47.

Collaboration


Dive into the Katriana Nugent's collaboration.

Top Co-Authors

Avatar

Phuoc T. Tran

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hailun Wang

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Reem Malek

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Kekoa Taparra

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Jessica Cades

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian W. Simons

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph M. Herman

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge