Network


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

Hotspot


Dive into the research topics where Süleyman Coşkun is active.

Publication


Featured researches published by Süleyman Coşkun.


Cell Stem Cell | 2010

Mdm2 Is Required for Survival of Hematopoietic Stem Cells/Progenitors via Dampening of ROS-Induced p53 Activity

Hussein A. Abbas; Daniela R. Maccio; Süleyman Coşkun; James G. Jackson; Amy L. Hazen; Tiffany M. Sills; M. James You; Karen K. Hirschi; Guillermina Lozano

Mdm2 is an E3 ubiquitin ligase that targets p53 for degradation. p53(515C) (encoding p53R172P) is a hypomorphic allele of p53 that rescues the embryonic lethality of Mdm2(-/-) mice. Mdm2(-/-) p53(515C/515C) mice, however, die by postnatal day 13 resulting from hematopoietic failure. Hematopoietic stem cells and progenitors of Mdm2(-/-) p53(515C/515C) mice were normal in fetal livers but were depleted in postnatal bone marrows. After birth, these mice had elevated reactive oxygen species (ROS) thus activating p53R172P. In the absence of Mdm2, stable p53R172P induced ROS and cell cycle arrest, senescence, and cell death in the hematopoietic compartment. This phenotype was partially rescued with antioxidant treatment and upon culturing of hematopoietic cells in methycellulose at 3% oxygen. p16 was also stabilized because of ROS, and its loss increased cell cycling and partially rescued hematopoiesis and survival. Thus, Mdm2 is required to control ROS-induced p53 levels for sustainable hematopoiesis.


Nature Genetics | 2016

Integrated genomic characterization of IDH1-mutant glioma malignant progression

Hanwen Bai; Akdes Serin Harmancı; E. Zeynep Erson-Omay; Jie Li; Süleyman Coşkun; Matthias Simon; Boris Krischek; Koray Özduman; S. Bulent Omay; Eric A. Sorensen; Şevin Turcan; Mehmet Bakırcığlu; Geneive Carrión-Grant; Phillip B. Murray; Victoria E. Clark; A. Gulhan Ercan-Sencicek; James Knight; Leman Sencar; Selin Altınok; Leon D. Kaulen; Burcu Gülez; Marco Timmer; Johannes Schramm; Octavian Henegariu; Jennifer Moliterno; Angeliki Louvi; Timothy A. Chan; Stacey L. Tannheimer; M. Necmettin Pamir; Alexander O. Vortmeyer

Gliomas represent approximately 30% of all central nervous system tumors and 80% of malignant brain tumors. To understand the molecular mechanisms underlying the malignant progression of low-grade gliomas with mutations in IDH1 (encoding isocitrate dehydrogenase 1), we studied paired tumor samples from 41 patients, comparing higher-grade, progressed samples to their lower-grade counterparts. Integrated genomic analyses, including whole-exome sequencing and copy number, gene expression and DNA methylation profiling, demonstrated nonlinear clonal expansion of the original tumors and identified oncogenic pathways driving progression. These include activation of the MYC and RTK-RAS-PI3K pathways and upregulation of the FOXM1- and E2F2-mediated cell cycle transitions, as well as epigenetic silencing of developmental transcription factor genes bound by Polycomb repressive complex 2 in human embryonic stem cells. Our results not only provide mechanistic insight into the genetic and epigenetic mechanisms driving glioma progression but also identify inhibition of the bromodomain and extraterminal (BET) family as a potential therapeutic approach.


Developmental Cell | 2013

Hemogenic Endothelial Cell Specification Requires c-Kit, Notch Signaling, and p27-Mediated Cell-Cycle Control

Kathrina L. Marcelo; Tiffany M. Sills; Süleyman Coşkun; Hema Vasavada; Supriya Sanglikar; Lauren C. Goldie; Karen K. Hirschi

Delineating the mechanism or mechanisms that regulate the specification of hemogenic endothelial cells from primordial endothelium is critical for optimizing their derivation from human stem cells for clinical therapies. We previously determined that retinoic acid (RA) is required for hemogenic specification, as well as cell-cycle control, of endothelium during embryogenesis. Herein, we define the molecular signals downstream of RA that regulate hemogenic endothelial cell development and demonstrate that cell-cycle control is required for this process. We found that re-expression of c-Kit in RA-deficient (Raldh2(-/-)) primordial endothelium induced Notch signaling and p27 expression, which restored cell-cycle control and rescued hemogenic endothelial cell specification and function. Re-expression of p27 in RA-deficient and Notch-inactivated primordial endothelial cells was sufficient to correct their defects in cell-cycle regulation and hemogenic endothelial cell development. Thus, RA regulation of hemogenic endothelial cell specification requires c-Kit, notch signaling, and p27-mediated cell-cycle control.


Nature Communications | 2017

Integrated genomic analyses of de novo pathways underlying atypical meningiomas

Akdes Serin Harmancı; Mark W. Youngblood; Victoria E. Clark; Süleyman Coşkun; Octavian Henegariu; Daniel Duran; E. Zeynep Erson-Omay; Leon D. Kaulen; Tong Ihn Lee; Brian J. Abraham; Matthias Simon; Boris Krischek; Marco Timmer; Roland Goldbrunner; S. Bulent Omay; Jacob F. Baranoski; Burçin Baran; Geneive Carrión-Grant; Hanwen Bai; Johannes Schramm; Jennifer Moliterno; Alexander O. Vortmeyer; Kaya Bilguvar; Katsuhito Yasuno; Richard A. Young; Murat Gunel

Meningiomas are mostly benign brain tumours, with a potential for becoming atypical or malignant. On the basis of comprehensive genomic, transcriptomic and epigenomic analyses, we compared benign meningiomas to atypical ones. Here, we show that the majority of primary (de novo) atypical meningiomas display loss of NF2, which co-occurs either with genomic instability or recurrent SMARCB1 mutations. These tumours harbour increased H3K27me3 signal and a hypermethylated phenotype, mainly occupying the polycomb repressive complex 2 (PRC2) binding sites in human embryonic stem cells, thereby phenocopying a more primitive cellular state. Consistent with this observation, atypical meningiomas exhibit upregulation of EZH2, the catalytic subunit of the PRC2 complex, as well as the E2F2 and FOXM1 transcriptional networks. Importantly, these primary atypical meningiomas do not harbour TERT promoter mutations, which have been reported in atypical tumours that progressed from benign ones. Our results establish the genomic landscape of primary atypical meningiomas and potential therapeutic targets.


Biotechnology and Bioengineering | 2016

Bioactive poly(ethylene glycol) hydrogels to recapitulate the HSC niche and facilitate HSC expansion in culture.

Maude L. Cuchiara; Süleyman Coşkun; Omar A. Banda; Kelsey L. Horter; Karen K. Hirschi; Jennifer L. West

Hematopoietic stem cells (HSCs) have been used therapeutically for decades, yet their widespread clinical use is hampered by the inability to expand HSCs successfully in vitro. In culture, HSCs rapidly differentiate and lose their ability to self‐renew. We hypothesize that by mimicking aspects of the bone marrow microenvironment in vitro we can better control the expansion and differentiation of these cells. In this work, derivatives of poly(ethylene glycol) diacrylate hydrogels were used as a culture substrate for hematopoietic stem and progenitor cell (HSPC) populations. Key HSC cytokines, stem cell factor (SCF) and interferon‐γ (IFNγ), as well as the cell adhesion ligands RGDS and connecting segment 1 were covalently immobilized onto the surface of the hydrogels. With the use of SCF and IFNγ, we observed significant expansion of HSPCs, ∼97 and ∼104 fold respectively, while maintaining c‐kit+lin− and c‐kit+Sca1+lin− (KSL) populations and the ability to form multilineage colonies after 14 days. HSPCs were also encapsulated within degradable poly(ethylene glycol) hydrogels for three‐dimensional culture. After expansion in hydrogels, ∼60% of cells were c‐kit+, demonstrating no loss in the proportion of these cells over the 14 day culture period, and ∼50% of colonies formed were multilineage, indicating that the cells retained their differentiation potential. The ability to tailor and use this system to support HSC growth could have implications on the future use of HSCs and other blood cell types in a clinical setting. Biotechnol. Bioeng. 2016;113: 870–881.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2017

Rac2 Modulates Atherosclerotic Calcification by Regulating Macrophage Interleukin-1β Production

Nicolle Ceneri; Lina Zhao; Bryan D. Young; Abigail L Healy; Süleyman Coşkun; Hema Vasavada; Timur O. Yarovinsky; Kenneth Ike; Ruggero Pardi; Lingfen Qin; Li Qin; George Tellides; Karen K. Hirschi; Judith Meadows; Robert Soufer; Hyung J. Chun; Mehran M. Sadeghi; Jeffrey R. Bender; Alan R. Morrison

Objective— The calcium composition of atherosclerotic plaque is thought to be associated with increased risk for cardiovascular events, but whether plaque calcium itself is predictive of worsening clinical outcomes remains highly controversial. Inflammation is likely a key mediator of vascular calcification, but immune signaling mechanisms that promote this process are minimally understood. Approach and Results— Here, we identify Rac2 as a major inflammatory regulator of signaling that directs plaque osteogenesis. In experimental atherogenesis, Rac2 prevented progressive calcification through its suppression of Rac1-dependent macrophage interleukin-1&bgr; (IL-1&bgr;) expression, which in turn is a key driver of vascular smooth muscle cell calcium deposition by its ability to promote osteogenic transcriptional programs. Calcified coronary arteries from patients revealed decreased Rac2 expression but increased IL-1&bgr; expression, and high coronary calcium burden in patients with coronary artery disease was associated with significantly increased serum IL-1&bgr; levels. Moreover, we found that elevated IL-1&bgr; was an independent predictor of cardiovascular death in those subjects with high coronary calcium burden. Conclusions— Overall, these studies identify a novel Rac2-mediated regulation of macrophage IL-1&bgr; expression, which has the potential to serve as a powerful biomarker and therapeutic target for atherosclerosis.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

The Transcription Factor E74-Like Factor Controls Quiescence of Endothelial Cells and Their Resistance to Myeloablative Treatments in Bone Marrow

Mariela Sivina; Takeshi Yamada; Chun Shik Park; Monica Puppi; Süleyman Coşkun; Karen K. Hirschi; H. Daniel Lacorazza

Objective—The regeneration of the hematopoietic system in bone marrow after chemotherapy depends on a balance between the quiescence and proliferation of lineage-specific progenitor cells. Even though the vascular network in bone is damaged by cytoablation, the transcriptional control of quiescence in endothelial cells is not well known. In this study, we investigated the role of the transcription factor E74-like factor (ELF4) in the proliferation of endothelial cells in bone marrow. Methods and Results—Loss-of-function models were used to study the role of ELF4 in human and murine endothelial cells. ELF4 promotes cell cycle entry by activating cyclin-dependent kinase-4 in human umbilical vein endothelial cells. Elf4-null mice exhibited enhanced recovery of bone marrow CD45− CD31+ endothelial cells and sinusoidal blood vessels following administration of 5-fluorouracil. Conclusion—Loss of ELF4 leads to increased quiescence in bone marrow endothelial cells by the deregulation of cyclin-dependent kinase-4 expression and to enhanced regeneration of sinusoidal blood vessels.


Journal of Human Genetics | 2016

A patient with a novel homozygous missense mutation in FTO and concomitant nonsense mutation in CETP

Ahmet Okay Caglayan; Beyhan Tüysüz; Süleyman Coşkun; Jennifer L. Quon; Akdes Serin Harmancı; Jacob F. Baranoski; Burçin Baran; E. Zeynep Erson-Omay; Octavian Henegariu; Shrikant Mane; Kaya Bilguvar; Katsuhito Yasuno; Murat Gunel

The fat mass and obesity associated (FTO) gene has previously been associated with a variety of diseases and conditions, notably obesity, acute coronary syndrome and metabolic syndrome. Reports describing mutations in FTO as well as in FTO animal models have further demonstrated a role for FTO in the development of the brain and other organs. Here, we describe a patient born of consanguineous union who presented with microcephaly, developmental delay, behavioral abnormalities, dysmorphic facial features, hypotonia and other various phenotypic abnormalities. Whole-exome sequencing revealed a novel homozygous missense mutation in FTO and a nonsense mutation in the cholesteryl ester transfer protein (CETP). Exome copy number variation analysis revealed no disease-causing large duplications or deletions within coding regions. Patient’s, her parents’ and non-related control’ fibroblasts were analyzed for morphologic defects, abnormal proliferation, apoptosis and transcriptome profile. We have shown that FTO is located in the nucleus of cells from each tested sample. Western blot analysis demonstrated no changes in patient FTO. Quantitative (qPCR) analysis revealed slightly decreased levels of FTO expression in patient cells compared with controls. No morphological or proliferation differences between the patient and control fibroblasts were observed. There is still much to be learned about the molecular mechanisms by which mutations in FTO contribute to such severe phenotypes.


Nature Communications | 2018

Author Correction: Integrated genomic analyses of de novo pathways underlying atypical meningiomas

Akdes Serin Harmancı; Mark W. Youngblood; Victoria E. Clark; Süleyman Coşkun; Octavian Henegariu; Daniel Duran; E. Zeynep Erson-Omay; Leon D. Kaulen; Tong Ihn Lee; Brian J. Abraham; Matthias Simon; Boris Krischek; Marco Timmer; Roland Goldbrunner; S. Bulent Omay; Jacob F. Baranoski; Burçin Baran; Geneive Carrión-Grant; Hanwen Bai; Johannes Schramm; Jennifer Moliterno; Alexander O. Vortmeyer; Kaya Bilguvar; Katsuhito Yasuno; Richard A. Young; Murat Gunel

This corrects the article DOI: 10.1038/ncomms14433.


Nature Communications | 2018

In utero nanoparticle delivery for site-specific genome editing

Adele S. Ricciardi; Raman Bahal; James S. Farrelly; Elias Quijano; Anthony H. Bianchi; Valerie L. Luks; Rachael Putman; Francesc López-Giráldez; Süleyman Coşkun; Eric Song; Yanfeng Liu; Wei-Che Hsieh; Danith H. Ly; David H. Stitelman; Peter M. Glazer; W. Mark Saltzman

Genetic diseases can be diagnosed early during pregnancy, but many monogenic disorders continue to cause considerable neonatal and pediatric morbidity and mortality. Early intervention through intrauterine gene editing, however, could correct the genetic defect, potentially allowing for normal organ development, functional disease improvement, or cure. Here we demonstrate safe intravenous and intra-amniotic administration of polymeric nanoparticles to fetal mouse tissues at selected gestational ages with no effect on survival or postnatal growth. In utero introduction of nanoparticles containing peptide nucleic acids (PNAs) and donor DNAs corrects a disease-causing mutation in the β-globin gene in a mouse model of human β-thalassemia, yielding sustained postnatal elevation of blood hemoglobin levels into the normal range, reduced reticulocyte counts, reversal of splenomegaly, and improved survival, with no detected off-target mutations in partially homologous loci. This work may provide the basis for a safe and versatile method of fetal gene editing for human monogenic disorders.The correction of genetic defects in utero could allow for improved outcomes of gene therapy. Here, the authors demonstrate safe delivery of nanoparticles to fetal mouse tissues, and show that nanoparticles containing peptide nucleic acids to edit the beta-globin gene are effective in a mouse model of beta-thalassemia.

Collaboration


Dive into the Süleyman Coşkun'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