Network


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

Hotspot


Dive into the research topics where Ilgen Mender is active.

Publication


Featured researches published by Ilgen Mender.


PLOS Biology | 2016

Regulation of the Human Telomerase Gene TERT by Telomere Position Effect—Over Long Distances (TPE-OLD): Implications for Aging and Cancer

Wanil Kim; Andrew T. Ludlow; Jaewon Min; Jerome D. Robin; Guido Stadler; Ilgen Mender; Tsung Po Lai; Ning Zhang; Woodring E. Wright; Jerry W. Shay

Telomerase is expressed in early human development and then becomes silenced in most normal tissues. Because ~90% of primary human tumors express telomerase and generally maintain very short telomeres, telomerase is carefully regulated, particularly in large, long-lived mammals. In the current report, we provide substantial evidence for a new regulatory control mechanism of the rate limiting catalytic protein component of telomerase (hTERT) that is determined by the length of telomeres. We document that normal, young human cells with long telomeres have a repressed hTERT epigenetic status (chromatin and DNA methylation), but the epigenetic status is altered when telomeres become short. The change in epigenetic status correlates with altered expression of TERT and genes near to TERT, indicating a change in chromatin. Furthermore, we identified a chromosome 5p telomere loop to a region near TERT in human cells with long telomeres that is disengaged with increased cell divisions as telomeres progressively shorten. Finally, we provide support for a role of the TRF2 protein, and possibly TERRA, in the telomere looping maintenance mechanism through interactions with interstitial TTAGGG repeats. This provides new insights into how the changes in genome structure during replicative aging result in an increased susceptibility to age-related diseases and cancer prior to the initiation of a DNA damage signal.


International Journal of Oncology | 2013

Imetelstat (a telomerase antagonist) exerts off‑target effects on the cytoskeleton.

Ilgen Mender; Serif Senturk; Nuriman Ozgunes; K. Can Akcali; Dimitris Kletsas; Sergei M. Gryaznov; Alp Can; Jerry W. Shay; Z. Gunnur Dikmen

Telomerase is a cellular ribonucleoprotein reverse transcriptase that plays a crucial role in telomere maintenance. This enzyme is expressed in approximately 90% of human tumors, but not in the majority of normal somatic cells. Imetelstat sodium (GRN163L), is a 13-mer oligonucleotide N3′→P5′ thio-phosphoramidate lipid conjugate, which represents the latest generation of telomerase inhibitors targeting the template region of the human functional telomerase RNA (hTR) subunit. In preclinical trials, this compound has been found to inhibit telomerase activity in multiple cancer cell lines, as well as in vivo xenograft mouse models. Currently, GRN163L is being investigated in several clinical trials, including a phase II human non-small cell lung cancer clinical trial, in a maintenance setting following standard doublet chemotherapy. In addition to the inhibition of telomerase activity in cancer cell lines, GRN163L causes morphological cell rounding changes, independent of hTR expression or telomere length. This leads to the loss of cell adhesion properties; however, the mechanism underlying this effect is not yet fully understood. In the present study, we observed that GRN163L treatment leads to the loss of adhesion in A549 lung cancer cells, due to decreased E-cadherin expression, leading to the disruption of the cytoskeleton through the alteration of actin, tubulin and intermediate filament organization. Consequently, the less adherent cancer cells initially cease to proliferate and are arrested in the G1 phase of the cell cycle, accompanied by decreased matrix metalloproteinase-2 (MMP-2) expression. These effects of GRN163L are independent of its telomerase catalytic activity and may increase the therapeutic efficacy of GRN163L by decreasing the adhesion, proliferation and metastatic potential of cancer cells in vivo.


Journal of Cutaneous Pathology | 2015

A primary melanoma and its asynchronous metastasis highlight the role of BRAF, CDKN2A, and TERT

Gregory A. Hosler; Teresa Davoli; Ilgen Mender; Brandon R. Litzner; Jaehyuk Choi; Payal Kapur; Jerry W. Shay; Richard C. Wang

Alterations in pathways including BRAF, CDKN2A, and TERT contribute to the development of melanoma, but the sequence in which the genetic alterations occur and their prognostic significance remains unclear. To clarify the role of these pathways, we analyzed a primary melanoma and its metastasis.


Molecular Cancer Therapeutics | 2018

Induced Telomere Damage to Treat Telomerase Expressing Therapy-Resistant Pediatric Brain Tumors

Satarupa Sengupta; Matthew Sobo; Kyungwoo Lee; Shiva Senthil Kumar; Angela R. White; Ilgen Mender; Christine Fuller; Lionel M.L. Chow; Maryam Fouladi; Jerry W. Shay

Brain tumors remain the leading cause of cancer-related deaths in children and often are associated with long-term sequelae among survivors of current therapies. Hence, there is an urgent need to identify actionable targets and to develop more effective therapies. Telomerase and telomeres play important roles in cancer, representing attractive therapeutic targets to treat children with poor-prognosis brain tumors such as diffuse intrinsic pontine glioma (DIPG), high-grade glioma (HGG), and high-risk medulloblastoma. We have previously shown that DIPG, HGG, and medulloblastoma frequently express telomerase activity. Here, we show that the telomerase-dependent incorporation of 6-thio-2′deoxyguanosine (6-thio-dG), a telomerase substrate precursor analogue, into telomeres leads to telomere dysfunction–induced foci (TIF) along with extensive genomic DNA damage, cell growth inhibition, and cell death of primary stem-like cells derived from patients with DIPG, HGG, and medulloblastoma. Importantly, the effect of 6-thio-dG is persistent even after drug withdrawal. Treatment with 6-thio-dG elicits a sequential activation of ATR and ATM pathways and induces G2–M arrest. In vivo treatment of mice bearing medulloblastoma xenografts with 6-thio-dG delays tumor growth and increases in-tumor TIFs and apoptosis. Furthermore, 6-thio-dG crosses the blood–brain barrier and specifically targets tumor cells in an orthotopic mouse model of DIPG. Together, our findings suggest that 6-thio-dG is a promising novel approach to treat therapy-resistant telomerase-positive pediatric brain tumors. Mol Cancer Ther; 17(7); 1504–14. ©2018 AACR.


Clinical Cancer Research | 2018

Induction of Telomere Dysfunction Prolongs Disease Control of Therapy-Resistant Melanoma

Gao Zhang; Lawrence W. Wu; Ilgen Mender; Michal Barzily-Rokni; Marc R. Hammond; Omotayo Ope; Chaoran Cheng; Themistoklis Vasilopoulos; Sergio Randell; Norah Sadek; Aurelie Beroard; Min Xiao; Tian Tian; Jiufeng Tan; Umar Saeed; Eric Sugarman; Clemens Krepler; Patricia Brafford; Katrin Sproesser; Sengottuvelan Murugan; Rajasekharan Somasundaram; Bradley Garman; Bradley Wubbenhorst; Jonathan Woo; Xiangfan Yin; Qin Liu; Dennie T. Frederick; Benchun Miao; Wei Xu; Giorgos C. Karakousis

Purpose: Telomerase promoter mutations are highly prevalent in human tumors including melanoma. A subset of patients with metastatic melanoma often fail multiple therapies, and there is an unmet and urgent need to prolong disease control for those patients. Experimental Design: Numerous preclinical therapy-resistant models of human and mouse melanoma were used to test the efficacy of a telomerase-directed nucleoside, 6-thio-2′-deoxyguanosine (6-thio-dG). Integrated transcriptomics and proteomics approaches were used to identify genes and proteins that were significantly downregulated by 6-thio-dG. Results: We demonstrated the superior efficacy of 6-thio-dG both in vitro and in vivo that results in telomere dysfunction, leading to apoptosis and cell death in various preclinical models of therapy-resistant melanoma cells. 6-thio-dG concomitantly induces telomere dysfunction and inhibits the expression level of AXL. Conclusions: In summary, this study shows that indirectly targeting aberrant telomerase in melanoma cells with 6-thio-dG is a viable therapeutic approach in prolonging disease control and overcoming therapy resistance. Clin Cancer Res; 24(19); 4771–84. ©2018 AACR. See related commentary by Teh and Aplin, p. 4629


Oncogene | 2018

Exploiting TERT dependency as a therapeutic strategy for NRAS-mutant melanoma

Patricia Reyes-Uribe; Maria Paz Adrianzen-Ruesta; Zhong Deng; Ileabett M. Echevarría-Vargas; Ilgen Mender; Steven Saheb; Qin Liu; Dario C. Altieri; Maureen E. Murphy; Jerry W. Shay; Paul M. Lieberman; Jessie Villanueva

Targeting RAS is one of the greatest challenges in cancer therapy. Oncogenic mutations in NRAS are present in over 25% of melanomas and patients whose tumors harbor NRAS mutations have limited therapeutic options and poor prognosis. Thus far, there are no clinical agents available to effectively target NRAS or any other RAS oncogene. An alternative approach is to identify and target critical tumor vulnerabilities or non-oncogene addictions that are essential for tumor survival. We investigated the consequences of NRAS blockade in NRAS-mutant melanoma and show that decreased expression of the telomerase catalytic subunit, TERT, is a major consequence. TERT silencing or treatment of NRAS-mutant melanoma with the telomerase-dependent telomere uncapping agent, 6-thio-2′-deoxyguanosine (6-thio-dG), led to rapid cell death, along with evidence of both telomeric and non-telomeric DNA damage, increased ROS levels, and upregulation of a mitochondrial antioxidant adaptive response. Combining 6-thio-dG with the mitochondrial inhibitor Gamitrinib attenuated this adaptive response and more effectively suppressed NRAS-mutant melanoma. Our study uncovers a robust dependency of NRAS-mutant melanoma on TERT, and provides proof-of-principle for a new combination strategy to combat this class of tumors, which could be expanded to other tumor types.


Neoplasia | 2018

Telomerase-Mediated Strategy for Overcoming Non–Small Cell Lung Cancer Targeted Therapy and Chemotherapy Resistance

Ilgen Mender; Ryan LaRanger; Krishna Luitel; Michael Peyton; Luc Girard; Tsung Po Lai; Kimberly Batten; Crystal Cornelius; Maithili P. Dalvi; Michael Ramirez; Wenting Du; Lani F. Wu; Steven J. Altschuler; Rolf A. Brekken; Elisabeth D. Martinez; John D. Minna; Woodring E. Wright; Jerry W. Shay

Standard and targeted cancer therapies for late-stage cancer patients almost universally fail due to tumor heterogeneity/plasticity and intrinsic or acquired drug resistance. We used the telomerase substrate nucleoside precursor, 6-thio-2′-deoxyguanosine (6-thio-dG), to target telomerase-expressing non–small cell lung cancer cells resistant to EGFR-inhibitors and commonly used chemotherapy combinations. Colony formation assays, human xenografts as well as syngeneic and genetically engineered immune competent mouse models of lung cancer were used to test the effect of 6-thio-dG on targeted therapy– and chemotherapy-resistant lung cancer human cells and mouse models. We observed that erlotinib-, paclitaxel/carboplatin-, and gemcitabine/cisplatin-resistant cells were highly sensitive to 6-thio-dG in cell culture and in mouse models. 6-thio-dG, with a known mechanism of action, is a potential novel therapeutic approach to prolong disease control of therapy-resistant lung cancer patients with minimal toxicities.


Nature Communications | 2018

NOVA1 regulates hTERT splicing and cell growth in non-small cell lung cancer

Andrew T. Ludlow; Mandy S. Wong; Jerome D. Robin; Kimberly Batten; Laura Yuan; Tsung Po Lai; Nicole Dahlson; Lu Zhang; Ilgen Mender; Enzo Tedone; Mohammed E. Sayed; Woodring E. Wright; Jerry W. Shay

Alternative splicing is dysregulated in cancer and the reactivation of telomerase involves the splicing of TERT transcripts to produce full-length (FL) TERT. Knowledge about the splicing factors that enhance or silence FL hTERT is lacking. We identified splicing factors that reduced telomerase activity and shortened telomeres using a siRNA minigene reporter screen and a lung cancer cell bioinformatics approach. A lead candidate, NOVA1, when knocked down resulted in a shift in hTERT splicing to non-catalytic isoforms, reduced telomerase activity, and progressive telomere shortening. NOVA1 knockdown also significantly altered cancer cell growth in vitro and in xenografts. Genome engineering experiments reveal that NOVA1 promotes the inclusion of exons in the reverse transcriptase domain of hTERT resulting in the production of FL hTERT transcripts. Utilizing hTERT splicing as a model splicing event in cancer may provide new insights into potentially targetable dysregulated splicing factors in cancer.Splicing of the telomerase transcript, hTERT, can be altered in cancer cells. Here the authors report NOVA1, as a splicing regulator that directly interacts with hTERT pre-mRNA enhancing the number of full-length transcripts and thus telomerase activity in lung cancer cells.


Turkish Journal of Biochemistry-turk Biyokimya Dergisi | 2017

How can I protect my telomeres and slow aging

Z. Gunnur Dikmen; Ilgen Mender

Abstract Recently, short telomeres have become a widely accepted cellular hallmark of aging. Telomere lengths in a single cell are heterogeneous and it is believed that the shortest telomere in a cell drives the induction of senescence. Hence, measuring the shortest telomere lengths (not just average) can provide more information about aging, cancer progression and telomere related diseases.Chronic exposure to DNA damaging agents, oxidative stress, inflammation, smoking, alcohol, exposure to acute and chronic stress promote telomere shortening and earlier onset of cell aging. Healthy life style including Mediterranean diet, moderate exercise, managing stress (breathing, meditation, yoga), spending time with loved ones and lots of laughter will help us to keep our telomeres long and safe.


Cancer Research | 2013

Abstract LB-125: A novel telomerase inhibitor.

Ilgen Mender; Sergei M. Gryaznov; Zeliha Gunnur Dikmen; Woodring E. Wright; Jerry W. Shay

Telomeres are highly specialized structures that are found at the very ends of linear chromosomes. Normal human somatic cells progressively lose their telomeres with each cell division due to the end replication problem. Unlike most normal somatic cells, the vast majority of advanced cancers have telomerase activity, which overcomes the end replication problem by adding multiple TTAGGG sequences to the end of linear chromosomes. Because of the selective expression of this enzyme in cancer cells, it is an almost universal target for antitumor drug development. However, there are surprisingly few telomerase inhibitors in clinical trials. We have modified a chemotherapeutic antimetabolite agent, 6-thioguanine (6-thio-G) so that this newly synthesized compound targets telomeres while retaining its antimetabolite activity. 6-thio-dG is a nucleoside analogue that is recognized by telomerase and is incorporated into telomeres. Telomeric incorporation results in structurally modified telomeres, functionally uncapping the shelterin protective complex, resulting in telomere associated DNA damage foci. Here, we report that 6-thio-dG causes progressive telomere shortening independent from inhibition of telomerase activity and induces telomere dysfunction in HCT116 human colon cancer cell line. Combination of the telomerase inhibitor GRN163L, which is in advanced clinical trials, and 6-thio-dG have additive effects on telomere shortening in HCT116 cells. 6-thio-dG treatment altered HCT116 cellular morphology and killed most of these cells within 1 week. Normal BJ fibroblast cells, which are telomerase silent, were not affected by 6-thio-dG in either cell morphology after 1 week or in enhanced telomere shortening after long-term treatment. These results show that 6-thio-dG is a novel anti-cancer inhibitor and a potential therapeutic agent targeting both telomeres and general DNA damage. Citation Format: Ilgen Mender, Sergei Gryaznov, Zeliha Gunnur Dikmen, Woodring Wright, Jerry Shay. A novel telomerase inhibitor. [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 LB-125. doi:10.1158/1538-7445.AM2013-LB-125

Collaboration


Dive into the Ilgen Mender's collaboration.

Top Co-Authors

Avatar

Jerry W. Shay

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Woodring E. Wright

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tsung Po Lai

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Enzo Tedone

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jerome D. Robin

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kimberly Batten

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew Sobo

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ning Zhang

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge