Network


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

Hotspot


Dive into the research topics where N. Sumru Bayin is active.

Publication


Featured researches published by N. Sumru Bayin.


Nature Communications | 2015

Sox2 antagonizes the Hippo pathway to maintain stemness in cancer cells

Upal Basu-Roy; N. Sumru Bayin; Kirk Rattanakorn; Eugenia Han; Dimitris Placantonakis; Alka Mansukhani; Claudio Basilico

The repressive Hippo pathway has a profound tumour suppressive role in cancer by restraining the growth-promoting function of the transcriptional coactivator, YAP. We previously showed that the stem cell transcription factor Sox2 maintains cancer stem cells (CSCs) in osteosarcomas. We now report that in these tumours, Sox2 antagonizes the Hippo pathway by direct repression of two Hippo activators, Nf2 (Merlin) and WWC1 (Kibra), leading to exaggerated YAP function. Repression of Nf2, WWC1 and high YAP expression marks the CSC fraction of the tumor population, while the more differentiated fraction has high Nf2, high WWC1 and reduced YAP expression. YAP depletion sharply reduces CSCs and tumorigenicity of osteosarcomas. Thus, Sox2 interferes with the tumour-suppressive Hippo pathway to maintain CSCs in osteosarcomas. This Sox2-Hippo axis is conserved in other Sox2-dependent cancers such as glioblastomas. Disruption of YAP transcriptional activity could be a therapeutic strategy for Sox2-dependent tumours.


World Journal of Stem Cells | 2014

Brain stem cells as the cell of origin in glioma.

Aram S. Modrek; N. Sumru Bayin; Dimitris Placantonakis

Glioma incidence rates in the United States are near 20000 new cases per year, with a median survival time of 14.6 mo for high-grade gliomas due to limited therapeutic options. The origins of these tumors and their many subtypes remain a matter of investigation. Evidence from mouse models of glioma and human clinical data have provided clues about the cell types and initiating oncogenic mutations that drive gliomagenesis, a topic we review here. There has been mixed evidence as to whether or not the cells of origin are neural stem cells, progenitor cells or differentiated progeny. Many of the existing murine models target cell populations defined by lineage-specific promoters or employ lineage-tracing methods to track the potential cells of origin. Our ability to target specific cell populations will likely increase concurrently with the knowledge gleaned from an understanding of neurogenesis in the adult brain. The cell of origin is one variable in tumorigenesis, as oncogenes or tumor suppressor genes may differentially transform the neuroglial cell types. Knowledge of key driver mutations and susceptible cell types will allow us to understand cancer biology from a developmental standpoint and enable early interventional strategies and biomarker discovery.


PLOS ONE | 2014

Selective Lentiviral Gene Delivery to CD133-Expressing Human Glioblastoma Stem Cells

N. Sumru Bayin; Aram S. Modrek; August Dietrich; Jonathan Lebowitz; Tobias Abel; Hae-Ri Song; Markus Schober; David Zagzag; Christian J. Buchholz; Moses V. Chao; Dimitris Placantonakis

Glioblastoma multiforme (GBM) is a deadly primary brain malignancy. Glioblastoma stem cells (GSC), which have the ability to self-renew and differentiate into tumor lineages, are believed to cause tumor recurrence due to their resistance to current therapies. A subset of GSCs is marked by cell surface expression of CD133, a glycosylated pentaspan transmembrane protein. The study of CD133-expressing GSCs has been limited by the relative paucity of genetic tools that specifically target them. Here, we present CD133-LV, a lentiviral vector presenting a single chain antibody against CD133 on its envelope, as a vehicle for the selective transduction of CD133-expressing GSCs. We show that CD133-LV selectively transduces CD133+ human GSCs in dose-dependent manner and that transduced cells maintain their stem-like properties. The transduction efficiency of CD133-LV is reduced by an antibody that recognizes the same epitope on CD133 as the viral envelope and by shRNA-mediated knockdown of CD133. Conversely, the rate of transduction by CD133-LV is augmented by overexpression of CD133 in primary human GBM cultures. CD133-LV selectively transduces CD133-expressing cells in intracranial human GBM xenografts in NOD.SCID mice, but spares normal mouse brain tissue, neurons derived from human embryonic stem cells and primary human astrocytes. Our findings indicate that CD133-LV represents a novel tool for the selective genetic manipulation of CD133-expressing GSCs, and can be used to answer important questions about how these cells contribute to tumor biology and therapy resistance.


Oncotarget | 2017

Notch signaling regulates metabolic heterogeneity in glioblastoma stem cells

N. Sumru Bayin; Joshua D. Frenster; Rajeev Sen; Sheng Si; Aram S. Modrek; Nataliya Galifianakis; Igor Dolgalev; Valerio Ortenzi; Irineu Illa-Bochaca; Anadjeet Khahera; Jonathan Serrano; Luis Chiriboga; David Zagzag; John G. Golfinos; Werner K. Doyle; Aristotelis Tsirigos; Adriana Heguy; Mitch Chesler; Mary Helen Barcellos-Hoff; Matija Snuderl; Dimitris Placantonakis

Glioblastoma (GBM) stem cells (GSCs) reside in both hypoxic and vascular microenvironments within tumors. The molecular mechanisms that allow GSCs to occupy such contrasting niches are not understood. We used patient-derived GBM cultures to identify GSC subtypes with differential activation of Notch signaling, which co-exist in tumors but occupy distinct niches and match their metabolism accordingly. Multipotent GSCs with Notch pathway activation reside in perivascular niches, and are unable to entrain anaerobic glycolysis during hypoxia. In contrast, most CD133-expressing GSCs do not depend on canonical Notch signaling, populate tumors regardless of local vascularity and selectively utilize anaerobic glycolysis to expand in hypoxia. Ectopic activation of Notch signaling in CD133-expressing GSCs is sufficient to suppress anaerobic glycolysis and resistance to hypoxia. These findings demonstrate a novel role for Notch signaling in regulating GSC metabolism and suggest intratumoral GSC heterogeneity ensures metabolic adaptations to support tumor growth in diverse tumor microenvironments.Glioblastoma (GBM) stem cells (GSCs) reside in both hypoxic and vascular microenvironments within tumors. The molecular mechanisms that allow GSCs to occupy such contrasting niches are not understood. We used patient-derived GBM cultures to identify GSC subtypes with differential activation of Notch signaling, which co-exist in tumors but occupy distinct niches and match their metabolism accordingly. Multipotent GSCs with Notch pathway activation reside in perivascular niches, and are unable to entrain anaerobic glycolysis during hypoxia. In contrast, most CD133-expressing GSCs do not depend on canonical Notch signaling, populate tumors regardless of local vascularity and selectively utilize anaerobic glycolysis to expand in hypoxia. Ectopic activation of Notch signaling in CD133-expressing GSCs is sufficient to suppress anaerobic glycolysis and resistance to hypoxia. These findings demonstrate a novel role for Notch signaling in regulating GSC metabolism and suggest intratumoral GSC heterogeneity ensures metabolic adaptations to support tumor growth in diverse tumor microenvironments.


Neoplasia | 2016

Patient-Specific Screening Using High-Grade Glioma Explants to Determine Potential Radiosensitization by a TGF-β Small Molecule Inhibitor

N. Sumru Bayin; Lin Ma; Cheddhi Thomas; Rabaa Baitalmal; Akhila Sure; Kush Fansiwala; Mark Bustoros; John G. Golfinos; Donato Pacione; Matija Snuderl; David Zagzag; Mary Helen Barcellos-Hoff; Dimitris Placantonakis

High-grade glioma (HGG), a deadly primary brain malignancy, manifests radioresistance mediated by cell-intrinsic and microenvironmental mechanisms. High levels of the cytokine transforming growth factor-β (TGF-β) in HGG promote radioresistance by enforcing an effective DNA damage response and supporting glioma stem cell self-renewal. Our analysis of HGG TCGA data and immunohistochemical staining of phosphorylated Smad2, which is the main transducer of canonical TGF-β signaling, indicated variable levels of TGF-β pathway activation across HGG tumors. These data suggest that evaluating the putative benefit of inhibiting TGF-β during radiotherapy requires personalized screening. Thus, we used explant cultures of seven HGG specimens as a rapid, patient-specific ex vivo platform to test the hypothesis that LY364947, a small molecule inhibitor of the TGF-β type I receptor, acts as a radiosensitizer in HGG. Immunofluorescence detection and image analysis of γ-H2AX foci, a marker of cellular recognition of radiation-induced DNA damage, and Sox2, a stem cell marker that increases post-radiation, indicated that LY364947 blocked these radiation responses in five of seven specimens. Collectively, our findings suggest that TGF-β signaling increases radioresistance in most, but not all, HGGs. We propose that short-term culture of HGG explants provides a flexible and rapid platform for screening context-dependent efficacy of radiosensitizing agents in patient-specific fashion. This time- and cost-effective approach could be used to personalize treatment plans in HGG patients.


Case reports in neurological medicine | 2016

Adult Primary Spinal Epidural Extraosseous Ewing’s Sarcoma: A Case Report and Review of the Literature

Mark Bustoros; Cheddhi Thomas; Joshua D. Frenster; Aram S. Modrek; N. Sumru Bayin; Matija Snuderl; Gerald Rosen; Peter B. Schiff; Dimitris Placantonakis

Background. Extraosseous Ewings sarcoma in the spinal epidural space is a rare malignancy, especially in adults. Case Presentation. A 40-year-old male presented with back pain and urinary hesitancy. MRI revealed a thoracic extradural mass with no osseous involvement. He underwent surgery for gross total resection of the mass, which was diagnosed as Ewings sarcoma. He was subsequently treated with chemoradiotherapy. He remains disease-free 1 year after surgery. Review of the literature indicated only 45 previously reported cases of spinal epidural extraosseous Ewings sarcoma in adults. Conclusions. Extraosseous Ewings sarcoma in the spinal epidural space is a rare clinical entity that should be included in the differential for spinal epidural masses. Its treatment is multidisciplinary but frequently requires surgical intervention due to compressive neurologic symptoms. Gross total resection appears to correlate with improved outcomes.


Archive | 2018

Selective Targeting of CD133-Expressing Glioblastoma Stem Cells Using Lentiviral Vectors

N. Sumru Bayin; Dimitris Placantonakis

Several lines of evidence suggest a cellular hierarchy in glioblastoma (GBM). In this hierarchy, GBM stem-like cells (GSCs) play critical roles in tumor progression and recurrence, by virtue of their robust tumor-propagating potential and resistance to conventional chemoradiotherapy. Therefore, targeting GSCs holds significant therapeutic promise. Expression of CD133 (PROM1), a cell surface glycoprotein, has been associated with the GSC phenotype and used as a GSC marker. Here, we describe a protocol that allows the selective lentiviral transduction of CD133-expressing GBM cells. This selectivity is conferred by pseudotyping the lentiviral envelope with a single-chain antibody against an extracellular epitope on CD133. We previously demonstrated the efficacy and specificity of this lentiviral vector using patient-derived GBM cultures. This chapter outlines the preparation of the vector and the transduction of human GBM cells.


Neurosurgery | 2016

144 GPR133 Promotes Glioblastoma Growth in Hypoxia.

Joshua D. Frenster; Julio Inocencio; Zhongye Xu; Joravar Dhaliwal; Abdulhakeem Alghamdi; David Zagzag; N. Sumru Bayin; Dimitris Placantonakis

INTRODUCTION Microenvironmental diversity in glioblastoma (GBM) is exemplified by normoxic hypervascular areas and hypoxic necrotic regions. GBM stem cells (GSCs) play a central role in tumor growth and therapy resistance. How GSCs adapt to diverse GBM microenvironments remains an important and unanswered question. METHODS Using primary human GBM cultures, we discovered that CD133+ GSCs are metabolically adept at expanding in hypoxic conditions. Transcriptional analysis indicated that CD133+ GSCs have 17.8 ± 8.8-fold enriched expression of GPR133 (n = 3 biospecimens), a member of the adhesion family of G-protein-coupled receptors. We used genetic, biochemical, and computational assays to interrogate the role of GPR133 in GBM. RESULTS Immunostaining of 12 human GBM biospecimens revealed that GPR133 expression is restricted to hypoxic regions of GBM and not present in normal brain. To test whether GPR133 expression is regulated by oxygen tension, we subjected GBM cultures to 1% O2 and found that GPR133 transcript was consistently upregulated (n = 5 cultures) in HIF1a-dependent manner. To elucidate GPR133s role in tumor growth, we used small hairpin RNA-mediated knockdown. GPR133 knockdown depleted CD133+ GSCs and inhibited tumor sphere formation under both normoxic and hypoxic conditions (P < .05). GPR133 knockdown also prevented in vivo tumor formation and increased survival of implanted mice (n = 4/group). CD133+ GSCs have 26.2% ± 12.53% higher cAMP levels than CD133- GBM cells (n = 3). GPR133 knockdown downregulated cAMP levels to 47.25% ± 27.27% of control (n = 3). Forskolin, which activates adenylate cyclase and boosts cAMP production, rescued the knockdown phenotype. These findings suggest that GPR133 canonical signaling is mediated by cAMP. Kaplan-Meier survival analysis of 160 The Cancer Genome Atlas (TCGA) patients indicated that increased GPR133 mRNA in GBM tumors correlated with poor survival (P = .002). CONCLUSION Our results suggest that GPR133 acts promotes GBM growth in hypoxia. We propose that GPR133 represents an attractive novel therapeutic target in GBM.


bioRxiv | 2018

Multiple modes of PRC2 inhibition elicit global chromatin alterations in H3K27M pediatric glioma

James M. Stafford; Chul-Hwan Lee; Philipp Voigt; Nicolas Descostes; Ricardo Saldaña-Meyer; Jia-Ray Yu; Gary LeRoy; Ozgur Oksuz; Jessica R. Chapman; Fernando Suarez; Aram S. Modrek; N. Sumru Bayin; Dimitris Placantonakis; Matthias A. Karajannis; Matija Snuderl; Beatrix Ueberheide; Danny Reinberg

A methionine substitution at lysine 27 on histone H3 variants (H3K27M) characterizes ~80% of diffuse intrinsic pontine gliomas (DIPG) and inhibits PRC2 in a dominant negative fashion. Yet, the mechanisms for this inhibition and abnormal epigenomic landscape have not been resolved. Using quantitative proteomics, we discovered that robust PRC2 inhibition requires levels of H3K27M greatly exceeding those of PRC2, seen in DIPG. While PRC2 inhibition requires interaction with H3K27M, we found this interaction on chromatin is transient with PRC2 largely being released from H3K27M. Unexpectedly, inhibition persisted even after PRC2 dissociated from H3K27M-chromatin suggesting a lasting impact on PRC2. Furthermore, allosterically activated PRC2 is particularly sensitive to K27M leading to a failure to spread H3K27me3 at distinct foci. In turn, levels of Polycomb antagonists such as H3K36me2 are elevated suggesting a more global, downstream effect on the epigenome. Together, these findings reveal the conditions required for H3K27M-mediated PRC2 inhibition and reconcile seemingly paradoxical effects of H3K27M on PRC2 recruitment and activity.


Archive | 2018

Single-Cell RNA Sequencing of Glioblastoma Cells

Rajeev Sen; Igor Dolgalev; N. Sumru Bayin; Adriana Heguy; Aris Tsirigos; Dimitris Placantonakis

Single-cell RNA sequencing (sc-RNASeq) is a recently developed technique used to evaluate the transcriptome of individual cells. As opposed to conventional RNASeq in which entire populations are sequenced in bulk, sc-RNASeq can be beneficial when trying to better understand gene expression patterns in markedly heterogeneous populations of cells or when trying to identify transcriptional signatures of rare cells that may be underrepresented when using conventional bulk RNASeq. In this method, we describe the generation and analysis of cDNA libraries from single patient-derived glioblastoma cells using the C1 Fluidigm system. The protocol details the use of the C1 integrated fluidics circuit (IFC) for capturing, imaging and lysing cells; performing reverse transcription; and generating cDNA libraries that are ready for sequencing and analysis.

Collaboration


Dive into the N. Sumru Bayin'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