Network


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

Hotspot


Dive into the research topics where Kaya Zhu is active.

Publication


Featured researches published by Kaya Zhu.


Stem Cells | 2010

Hematopoietic Stem Cell Defects in Mice with Deficiency of Fancd2 or Usp1

Kalindi Parmar; Jungmin Kim; Stephen M. Sykes; Akiko Shimamura; Patricia Stuckert; Kaya Zhu; Abigail Hamilton; Mary Kathryn Deloach; Jeffery L. Kutok; Koichi Akashi; D. Gary Gilliland; Alan D. D'Andrea

Fanconi anemia (FA) is a human genetic disease characterized by a DNA repair defect and progressive bone marrow failure. Central events in the FA pathway are the monoubiquitination of the Fancd2 protein and the removal of ubiquitin by the deubiquitinating enzyme, Usp1. Here, we have investigated the role of Fancd2 and Usp1 in the maintenance and function of murine hematopoietic stem cells (HSCs). Bone marrow from Fancd2−/− mice and Usp1−/− mice exhibited marked hematopoietic defects. A decreased frequency of the HSC populations including Lin‐Sca‐1+Kit+ cells and cells enriched for dormant HSCs expressing signaling lymphocyte activation molecule (SLAM) markers, was observed in the bone marrow of Fancd2‐deficient mice. In addition, bone marrow from Fancd2−/− mice contained significantly reduced frequencies of late‐developing cobblestone area‐forming cell activity in vitro compared to the bone marrow from wild‐type mice. Furthermore, Fancd2‐deficient and Usp1‐deficient bone marrow had defective long‐term in vivo repopulating ability. Collectively, our data reveal novel functions of Fancd2 and Usp1 in maintaining the bone marrow HSC compartment and suggest that FA pathway disruption may account for bone marrow failure in FA patients. STEM CELLS 2010;28:1186–1195


Surgery | 2011

Necroptosis is a novel mechanism of radiation-induced cell death in anaplastic thyroid and adrenocortical cancers

Matthew A. Nehs; Chi-Iou Lin; David Kozono; Edward E. Whang; Nancy L. Cho; Kaya Zhu; Jacob Moalem; Francis D. Moore; Daniel T. Ruan

BACKGROUND Necroptosis is a recently described mechanism of programmed cellular death. We hypothesize that necroptosis plays an important role in radiation-induced cell death in endocrine cancers. METHODS Thyroid and adrenocortical carcinoma cell lines were exposed to increasing doses of radiation in the presence of necroptosis inhibitor Nec-1 and/or apoptosis-inhibitor zVAD. H295R cells deficient in receptor interacting protein 1 (RIP1), an essential kinase for necroptosis, were used as controls. Survival curves were generated at increasing doses of radiation. RESULTS Nec-1 and zVAD increased cellular survival with increasing doses of radiotherapy in 8505c, TPC-1, and SW13. Both inhibitors used together had an additive effect. At 6 Gy, 8505c, TPC-1, and SW13 cell survival was significantly increased compared to controls by 40%, 33%, and 31% with Nec-1 treatment, by 53%, 47%, and 44% with zVAD treatment, and by 80%, 70%, and 65% with both compounds, respectively (P < .05). H295R showed no change in survival with Nec-1 treatment. The radiobiologic parameter quasithreshold dose was significantly increased in 8505c, TPC-1, and SW13 cells when both Nec-1 and zVAD were used in combination to inhibit necroptosis and apoptosis together, revealing resistance to standard doses of fractionated therapeutic radiation. CONCLUSION Necroptosis contributes to radiation-induced cell death. Future studies should investigate ways to promote the activation of necroptosis to improve radiosensitivity.


PLOS ONE | 2013

Proteasome Inhibitors Block DNA Repair and Radiosensitize Non-Small Cell Lung Cancer

Kyle R. Cron; Kaya Zhu; Deepa Kushwaha; Grace Hsieh; Dmitry Merzon; Jonathan Rameseder; Clark C. Chen; Alan D. D’Andrea; David Kozono

Despite optimal radiation therapy (RT), chemotherapy and/or surgery, a majority of patients with locally advanced non-small cell lung cancer (NSCLC) fail treatment. To identify novel gene targets for improved tumor control, we performed whole genome RNAi screens to identify knockdowns that most reproducibly increase NSCLC cytotoxicity. These screens identified several proteasome subunits among top hits, including the topmost hit PSMA1, a component of the core 20 S proteasome. Radiation and proteasome inhibition showed synergistic effects. Proteasome inhibition resulted in an 80–90% decrease in homologous recombination (HR), a 50% decrease in expression of NF-κB-inducible HR genes BRCA1 and FANCD2, and a reduction of BRCA1, FANCD2 and RAD51 ionizing radiation-induced foci. IκBα RNAi knockdown rescued NSCLC radioresistance. Irradiation of mice with NCI-H460 xenografts after inducible PSMA1 shRNA knockdown markedly increased murine survival compared to either treatment alone. Proteasome inhibition is a promising strategy for NSCLC radiosensitization via inhibition of NF-κB-mediated expression of Fanconi Anemia/HR DNA repair genes.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Dynamic epigenetic regulation of glioblastoma tumorigenicity through LSD1 modulation of MYC expression

David Kozono; Jie Li; Masayuki Nitta; Oltea Sampetrean; David D. Gonda; Deepa Kushwaha; Dmitry Merzon; Valya Ramakrishnan; Shan Zhu; Kaya Zhu; Hiroko Matsui; Olivier Harismendy; Wei Hua; Ying Mao; Chang-Hyuk Kwon; Hideyuki Saya; Ichiro Nakano; Donald P. Pizzo; Scott R. VandenBerg; Clark C. Chen

Significance Glioblastoma is the most common type of adult brain cancer, with near-uniform fatality within 2 y of diagnosis. Therapeutic failure is thought to be related to small subpopulations of cells that exhibit tumorigenicity, the cellular capacity to reconstitute the entire tumor mass. One fundamental issue is whether tumorigenicity exists within a static subpopulation of cells or whether the capacity is stochastically acquired. We provide evidence that tumorigenicity is a cellular property that is durable yet undergoes low-frequency stochastic changes. We showed that these changes are driven by lysine-specific demethylase 1 (LSD1)-mediated epigenetic (heritable non-DNA sequence-altering) modifications that impact expression of key transcription factors, which in turn govern transitions between tumorigenic states. These findings harbor implications for glioblastoma therapeutic development. The available evidence suggests that the lethality of glioblastoma is driven by small subpopulations of cells that self-renew and exhibit tumorigenicity. It remains unclear whether tumorigenicity exists as a static property of a few cells or as a dynamically acquired property. We used tumor-sphere and xenograft formation as assays for tumorigenicity and examined subclones isolated from established and primary glioblastoma lines. Our results indicate that glioblastoma tumorigenicity is largely deterministic, yet the property can be acquired spontaneously at low frequencies. Further, these dynamic transitions are governed by epigenetic reprogramming through the lysine-specific demethylase 1 (LSD1). LSD depletion increases trimethylation of histone 3 lysine 4 at the avian myelocytomatosis viral oncogene homolog (MYC) locus, which elevates MYC expression. MYC, in turn, regulates oligodendrocyte lineage transcription factor 2 (OLIG2), SRY (sex determining region Y)-box 2 (SOX2), and POU class 3 homeobox 2 (POU3F2), a core set of transcription factors required for reprogramming glioblastoma cells into stem-like states. Our model suggests epigenetic regulation of key transcription factors governs transitions between tumorigenic states and provides a framework for glioblastoma therapeutic development.


PLOS ONE | 2011

Activation of Hif1α by the prolylhydroxylase inhibitor dimethyoxalyglycine decreases radiosensitivity.

Marina K. Ayrapetov; Chang Xu; Yingli Sun; Kaya Zhu; Kalindi Parmar; Alan D. D'Andrea; Brendan D. Price

Hypoxia inducible factor 1α (Hif1α) is a stress responsive transcription factor, which regulates the expression of genes required for adaption to hypoxia. Hif1α is normally hydroxylated by an oxygen-dependent prolylhydroxylase, leading to degradation and clearance of Hif1α from the cell. Under hypoxic conditions, the activity of the prolylhydroxylase is reduced and Hif1α accumulates. Hif1α is also constitutively expressed in tumor cells, where it is associated with resistance to ionizing radiation. Activation of the Hif1α transcriptional regulatory pathway may therefore function to protect normal cells from DNA damage caused by ionizing radiation. Here, we utilized the prolylhydroxylase inhibitor dimethyloxalylglycine (DMOG) to elevate Hif1α levels in mouse embryonic fibroblasts (MEFs) to determine if DMOG could function as a radioprotector. The results demonstrate that DMOG increased Hif1α protein levels and decreased the sensitivity of MEFs to ionizing radiation. Further, the ability of DMOG to function as a radioprotector required Hif1α, indicating a key role for Hif1αs transcriptional activity. DMOG also induced the Hif1α -dependent accumulation of several DNA damage response proteins, including CHD4 and MTA3 (sub-units of the NuRD deacetylase complex) and the Suv39h1 histone H3 methyltransferase. Depletion of Suv39h1, but not CHD4 or MTA3, reduced the ability of DMOG to protect cells from radiation damage, implicating increased histone H3 methylation in the radioprotection of cells. Finally, treatment of mice with DMOG prior to total body irradiation resulted in significant radioprotection of the mice, demonstrating the utility of DMOG and related prolylhydroxylase inhibitors to protect whole organisms from ionizing radiation. Activation of Hif1α through prolylhydroxylase inhibition therefore identifies a new pathway for the development of novel radiation protectors.


Cancer Biology & Therapy | 2015

USP9X inhibition promotes radiation-induced apoptosis in non-small cell lung cancer cells expressing mid-to-high MCL1

Deepa Kushwaha; Colin O’Leary; Kyle R. Cron; Peter V. Deraska; Kaya Zhu; Alan D. D’Andrea; David Kozono

Background and Purpose: Radiotherapy (RT) is vital for the treatment of locally advanced non-small cell lung cancer (NSCLC), yet its delivery is limited by tolerances of adjacent organs. We sought therefore to identify and characterize gene targets whose inhibition may improve RT. Materials and Methods: Whole genome pooled shRNA cytotoxicity screens were performed in A549 and NCI-H460 using a retroviral library of 74,705 sequences. Cells were propagated with or without daily radiation Monday–Friday. Radiosensitization by top differential dropout hits was assessed by clonogenic assays. Apoptosis was assessed using a caspase 3/7 cell-based activity assay and by annexin V-FITC and PI staining. MCL1 expression was assessed by qPCR and Western blotting. Results: USP9X, a deubiquitinase, was a top hit among druggable gene products. WP1130, a small molecule USP9X inhibitor, showed synergistic cytotoxicity with IR. MCL1, an anti-apoptotic protein deubiquitinated by USP9X, decreased with USP9X inhibition and IR. This was accompanied by increases in caspase 3/7 activity and apoptosis. In a panel of NSCLC lines, MCL1 and USP9X protein and gene expression levels were highly correlated. Lines showing high levels of MCL1 expression were the most sensitive to USP9X inhibition. Conclusions: These data support the use of MCL1 expression as a predictive biomarker for USP9X inhibitors in NSCLC therapy.


Cancer Research | 2015

Abstract 979: Dynamic epigenetic regulation of glioblastoma tumorigenicity through LSD1 modulation of MYC expression

Jie Li; David Kozono; Masayuki Nitta; Oltea Sampetrean; David D. Gonda; Deepa Kushwaha; Dmitry Merzon; Valya Ramakrishnan; Shan Zhu; Kaya Zhu; Hiroko Matsui; Olivier Harismendy; Wei Hua; Ying Mao; Chang-Hyuk Kwon; Hideyuki Saya; Bob S. Carter; Donald P. Pizzo; Scott R. VandenBerg; Clark C. Chen

Glioblastoma is the most common form of primary brain cancer and remains one of the deadliest of human cancers with near-uniform fatality. Increasing evidence suggests that the lethality of glioblastoma is driven by small subpopulations of cells with self-renew ability and tumorigenicity, termed as tumor initiating cells. The mechanism how the tumor initiating cells maintain and gain tumorigenicity in glioblastoma still remains unclear. Here, we used sphere formation and tumor propagating potential to measure the tumorigenicity in established cell line and primary glioblastoma cells. The results indicated that glioblastoma tumorigenicity appears largely deterministic, though spontaneous gain and loss of this property occur at low frequency. Mechanically, this dynamic transition in tumorigenicity was governed by MYC level which was modulated epigenetically by the lysine-specific demethylase 1 (LSD1). Elevated MYC expression, in turn, regulates OLIG2, SOX2 and POU3F2, a core set of transcription factors required for reprogramming glioblastoma cells into stem-like states. Our model suggests epigenetic regulation of key transcription factors facilitates transitions between tumorigenic states and provides a framework for glioblastoma therapeutic development. Importantly, the effect of LSD1 on tumorigenity is “Janus”-like; partial depletion of LSD1 caused increased MYC expression and a pro-tumorigenic state. In contrast, complete suppression of LSD1 induced cell death. As such, therapeutic strategies targeting LSD1 and other targets manifesting similar “Janus” effect should be designed to prevent unintended induction of tumorigenesis during treatment. Citation Format: Jie Li, David Kozono, Masayuki Nitta, Oltea Sampetrean, David Gonda, Deepa S. Kushwaha, Dmitry Merzon, Valya Ramakrishnan, Shan Zhu, Kaya Zhu, Hiroko Matsui, Olivier Harismendy, Wei Hua, Ying Mao, Chang-Hyuk Kwon, Hideyuki Saya, Bob S. Carter, Donald P. Pizzo, Scott R. VandenBerg, Clark C. Chen. Dynamic epigenetic regulation of glioblastoma tumorigenicity through LSD1 modulation of MYC expression. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 979. doi:10.1158/1538-7445.AM2015-979


Molecular Cancer Research | 2015

Abstract PR02: Dynamic epigenetic regulation of glioblastoma tumorigenicity through a LSD1-MYC-OLIG2 axis

Clark Chen; David Kozono; Jie Li; Masayuki Nitta; Oltea Sampetrean; Kimberly Ng; David D. Gonda; Deepa Kushwaha; Dmitry Merzon; Valya Ramakrishnan; Shan Zhu; Kaya Zhu; Hiroko Matsui; Olivier Harismendy; Wei Hua; Ying Mao; Chang-Hyuk Kwon; Keith L. Ligon; Hideyuki Saya; Bob S. Carter; Donald P. Pizzo; Scott R. VandenBerg; Frank Furnari; Webster K. Cavenee

Glioblastoma is one of the most devastating of human cancers, with near-uniform fatality within two years of diagnosis. Therapeutic failure is thought to be related to small subpopulation of cells that exhibit the properties of self-renewal and tumorigenicity. Understanding how such subpopulations attain and retain these properties remains a central question in oncology. One fundamental issue is whether tumorigenicity exists within a static population of elite cells or whether the capacity is stochastically acquired. To test these models, we assayed the tumorigenicity of single-cell subclones derived from long-terms passaged and primary patient-derived xenograft (PDX) glioblastoma lines. Our findings were best described by a hybrid model that is largely deterministic (elite) but with opportunities for dynamic (stochastic) interchange between non-tumorigenic and tumorigenic states. To identify molecular determinants of tumorigenicity, we performed gene expression profiling of the subclones. Analysis of the data suggested that tumorigenicity in glioblastoma is a dynamic property driven by variation in MYC expression, which in turn regulates Olig2 expression, a neural stem cell marker. Ectopic expression of MYC conferred tumorigenicty and MYC silencing abolished tumorigenicity in vitro and in vivo for multiple PDX and GEMM models. Transition between tumorigenic and non-tumorigenic cell states was associated with changes in histone modification at the MYC locus mediated by expression of lysine-specific demethylase 1 (LSD1). The model suggests a critical LSD1-MYC-OLIG2 axis that regulates the dynamic transition between glioblastoma cell states of differing tumorigenicity and unveils a novel framework for glioblastoma therapeutic development. Citation Format: Clark Chen, David Kozono, Jie Li, Masayuki Nitta, Oltea Sampetrean, Kimberly Ng, David Gonda, Deepa S. Kushwaha, Dmitry Merzon, Valya Ramakrishnan, Shan Zhu, Kaya Zhu, Hiroko Matsui, Olivier Harismendy, Wei Hua, Ying Mao, Chang-Hyuk Kwon, Keith L. Ligon, Hideyuki Saya, Bob S. Carter, Donald P. Pizzo, Scott R. VandenBerg, Frank Furnari, Webster Cavenee. Dynamic epigenetic regulation of glioblastoma tumorigenicity through a LSD1-MYC-OLIG2 axis. [abstract]. In: Proceedings of the AACR Special Conference on Myc: From Biology to Therapy; Jan 7-10, 2015; La Jolla, CA. Philadelphia (PA): AACR; Mol Cancer Res 2015;13(10 Suppl):Abstract nr PR02.


Cancer Research | 2013

Abstract 1583: Proteasome inhibition as a strategy for non-small cell lung carcinoma via inhibition of DNA double strand break repair.

Kyle R. Cron; Kaya Zhu; Deepa Kushwaha; Grace Hsieh; Dmitry Merzon; Jack Monahan; Clark C. Chen; Alan D. D'Andrea; David Kozono

Background: Each year, ∼60,000 US patients are diagnosed with locally advanced non-small cell lung carcinoma (NSCLC). Despite optimal radiation therapy (RT), chemotherapy +/- surgery, over 30% of patients fails locoregionally. The addition of concurrent chemotherapy to RT adds only ∼5% absolute survival benefit. Novel radiosensitizers may improve the therapeutic index. To identify the best potential targets, we performed whole genome RNAi screens that identified several proteasome subunits among top genes whose knockdown increased NSCLC cytotoxicity. Materials and Methods: Cytotoxicity was assessed by luminescent cell viability and clonogenic assays. Homologous recombination (HR) and non-homologous end-joining (NHEJ) were assessed using NSCLC cell lines transduced with reporter constructs that express GFP upon repair of I-SceI-induced DNA double strand breaks (DSBs). DNA damage-induced focus formation was assessed by immunofluorescence, scoring % of cells showing ≥5 foci. In vivo radiosensitization by proteasome gene knockdown was assessed using NCr nude mice injected with 1x106 NCI-H460 cells stably transfected with inducible PSMA1 shRNA. Once tumors reached ≥3 mm, knockdown was induced with doxycycline, and then one week later, RT to a dose of 20 Gy in 5 fractions was initiated. Results: Radiation and proteasome inhibition showed synergistic cytotoxicity. Irradiation of A549 cells with 1 Gy x 3 decreased clonogenic survival by 58% compared to control, while ionizing radiation (IR) plus 10 nM bortezomib decreased survival by 74% compared to bortezomib alone. Similar results were seen in NCI-H460. Proteasome inhibition via bortezomib or PSMA1 siRNA knockdown resulted in 80-90% decreased HR and NHEJ. Additionally, bortezomib or PSMA1 shRNA knockdown resulted in ≥50% decreases in BRCA1, FANCD2 or RAD51 IR-induced focus formation. Treatment of NCI-H460 xenografts with RT in the setting of PSMA1 knockdown showed marked improvements in survival; at 100 days post treatment initiation, only 20% of RT-only treated mice and 30% of doxycycline-treated mice (for PSMA1 shRNA knockdown in tumors) survived, compared to 100% following dual treatment (n = 10 per arm). There was a statistically significant difference in survival between mice with tumors treated with RT alone vs. RT + PSMA1 knockdown, with median survivals of 43 days vs. not reached, P = .0003 by the log-rank test. Conclusions: Proteasome inhibition has emerged as a promising target in NSCLC radiosensitization with evidence indicating the mechanism is through inhibition of DNA DSB repair pathways HR and NHEJ. Citation Format: Kyle R. Cron, Kaya Zhu, Deepa Kushwaha, Grace Hsieh, Dmitry Merzon, Jack Monahan, Clark C. Chen, Alan D9Andrea, David Kozono. Proteasome inhibition as a strategy for non-small cell lung carcinoma via inhibition of DNA double strand break repair. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1583. doi:10.1158/1538-7445.AM2013-1583


Cancer Research | 2012

Abstract 5218: A c-Myc driven threshold model of tumor initiating in glioblastoma

David Kozono; Masayuki Nitta; Oltea Sampetrean; Kimberly Ng; Dmitry Merzon; Deepa Kushwaha; Kaya Zhu; Keith L. Ligon; Shan Zhu; Hediyuki Saya; Clark C. Chen

Recent evidence suggests that glioblastoma is driven by a subset of Tumor Initiating (TI) cells characterized by their capacity to form tumors in xenograft models and self-renew in vitro. These TI cells share many properties of neural stem/progenitor cells, including the expression of certain cell surface markers. With serial passage, many cells lose their capacity to TI. The transition between TI proficient and deficient states remains poorly understood. There are two theoretic models for the maintenance of TI states. In the “elite” model, TI activity is restricted to a predetermined subpopulation of cells. The alternative “stochastic” model suggests that any tumor cell has a finite chance of acquiring TI capacity through random fluctuations in cell physiology. To distinguish between these two models, we established culturing conditions capable of sustaining a single cell line in both low and high TI states. For both primary and long-term passaged glioblastoma lines, culturing under sphere forming conditions lead to increased in vivo tumorigenicity relative to serum based conditions. Further, cells cultured under sphere forming conditions exhibited increased expression of neural stem cell markers, including Nestin, Musashi, Olig2, and Sox2. Moreover, the ability of individual subclones to form xenografts closely correlated with their ability to grow under sphere forming conditions. In this context, the “elite” model predicts that only a subset of subclones (derived from a single line) cultured under serum conditions would possess high TI capacity and grow under sphere forming conditions. Moreover, individual cells derived from the same subclone would exhibit similar capacity for growth under sphere forming conditions. Surprisingly, single cells derived from a single glioblastoma subclone exhibited a wide range of growth capacity under sphere forming conditions, suggesting the TI state is in part driven by a stochastic process. On the other hand, only a subset of subclones from a single glioblastoma line displayed capacity for growth under sphere forming conditions, as predicted by the elite model. Transcriptome profiling of the different subclones revealed a gene signature associated with TI capacity. Analysis of this signature showed enrichment for genes regulated by c-Myc. Indeed, clones with increased TI capacity tend to harbor increased c-Myc expression. Additionally, over-expression of c-myc increased the TI capacity of glioblastoma cells in xenograft models and led to the formation of malignant brain tumor in an Ink4a/ARF null transgenic murine model. Finally, analysis of The Cancer Genome Atlas Project (TCGA) glioblastoma database revealed c-Myc over-expression in glioblastoma cells, particularly in the pro-neural and mesenchymal subtypes. Our results are most consistent with a threshold model in which TI states in glioblastomas are driven by expression levels of critical factors such as c-Myc. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5218. doi:1538-7445.AM2012-5218

Collaboration


Dive into the Kaya Zhu'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge