Network


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

Hotspot


Dive into the research topics where Kadir C. Akdemir is active.

Publication


Featured researches published by Kadir C. Akdemir.


Nature | 2010

TRIM24 links a non-canonical histone signature to breast cancer

Wen Wei Tsai; Zhanxin Wang; Teresa T. Yiu; Kadir C. Akdemir; Weiya Xia; Stefan Winter; Cheng Yu Tsai; Xiaobing Shi; Dirk Schwarzer; William Plunkett; Bruce J. Aronow; Or Gozani; Wolfgang Fischle; Mien Chie Hung; Dinshaw J. Patel; Michelle Craig Barton

Recognition of modified histone species by distinct structural domains within ‘reader’ proteins plays a critical role in the regulation of gene expression. Readers that simultaneously recognize histones with multiple marks allow transduction of complex chromatin modification patterns into specific biological outcomes. Here we report that chromatin regulator tripartite motif-containing 24 (TRIM24) functions in humans as a reader of dual histone marks by means of tandem plant homeodomain (PHD) and bromodomain (Bromo) regions. The three-dimensional structure of the PHD-Bromo region of TRIM24 revealed a single functional unit for combinatorial recognition of unmodified H3K4 (that is, histone H3 unmodified at lysine 4, H3K4me0) and acetylated H3K23 (histone H3 acetylated at lysine 23, H3K23ac) within the same histone tail. TRIM24 binds chromatin and oestrogen receptor to activate oestrogen-dependent genes associated with cellular proliferation and tumour development. Aberrant expression of TRIM24 negatively correlates with survival of breast cancer patients. The PHD-Bromo of TRIM24 provides a structural rationale for chromatin activation through a non-canonical histone signature, establishing a new route by which chromatin readers may influence cancer pathogenesis.


Nature Genetics | 2017

Systematic analysis of telomere length and somatic alterations in 31 cancer types

Floris P. Barthel; Wei Wei; Ming Tang; Emmanuel Martinez-Ledesma; Xin Hu; Samirkumar Amin; Kadir C. Akdemir; Sahil Seth; Xingzhi Song; Qianghu Wang; Tara M. Lichtenberg; Jian Hu; Jianhua Zhang; Siyuan Zheng; Roel G.W. Verhaak

Cancer cells survive cellular crisis through telomere maintenance mechanisms. We report telomere lengths in 18,430 samples, including tumors and non-neoplastic samples, across 31 cancer types. Telomeres were shorter in tumors than in normal tissues and longer in sarcomas and gliomas than in other cancers. Among 6,835 cancers, 73% expressed telomerase reverse transcriptase (TERT), which was associated with TERT point mutations, rearrangements, DNA amplifications and transcript fusions and predictive of telomerase activity. TERT promoter methylation provided an additional deregulatory TERT expression mechanism. Five percent of cases, characterized by undetectable TERT expression and alterations in ATRX or DAXX, demonstrated elongated telomeres and increased telomeric repeat–containing RNA (TERRA). The remaining 22% of tumors neither expressed TERT nor harbored alterations in ATRX or DAXX. In this group, telomere length positively correlated with TP53 and RB1 mutations. Our analysis integrates TERT abnormalities, telomerase activity and genomic alterations with telomere length in cancer.


Nucleic Acids Research | 2014

Genome-wide profiling reveals stimulus-specific functions of p53 during differentiation and DNA damage of human embryonic stem cells

Kadir C. Akdemir; Abhinav K. Jain; Kendra Allton; Bruce J. Aronow; Xueping Xu; Austin J. Cooney; Wei Li; Michelle Craig Barton

How tumor suppressor p53 selectively responds to specific signals, especially in normal cells, is poorly understood. We performed genome-wide profiling of p53 chromatin interactions and target gene expression in human embryonic stem cells (hESCs) in response to early differentiation, induced by retinoic acid, versus DNA damage, caused by adriamycin. Most p53-binding sites are unique to each state and define stimulus-specific p53 responses in hESCs. Differentiation-activated p53 targets include many developmental transcription factors and, in pluripotent hESCs, are bound by OCT4 and NANOG at chromatin enriched in both H3K27me3 and H3K4me3. Activation of these genes occurs with recruitment of p53 and H3K27me3-specific demethylases, UTX and JMJD3, to chromatin. In contrast, genes associated with cell migration and motility are bound by p53 specifically after DNA damage. Surveillance functions of p53 in cell death and cell cycle regulation are conserved in both DNA damage and differentiation. Comparative genomic analysis of p53-targets in mouse and human ESCs supports an inter-species divergence in p53 regulatory functions during evolution. Our findings expand the registry of p53-regulated genes to define p53-regulated opposition to pluripotency during early differentiation, a process highly distinct from stress-induced p53 response in hESCs.


Genome Biology | 2015

HiCPlotter integrates genomic data with interaction matrices

Kadir C. Akdemir; Lynda Chin

AbstractMetazoan genomic material is folded into stable non-randomly arranged chromosomal structures that are tightly associated with transcriptional regulation and DNA replication. Various factors including regulators of pluripotency, long non-coding RNAs, or the presence of architectural proteins have been implicated in regulation and assembly of the chromatin architecture. Therefore, comprehensive visualization of this multi-faceted structure is important to unravel the connections between nuclear architecture and transcriptional regulation. Here, we present an easy-to-use open-source visualization tool, HiCPlotter, to facilitate juxtaposition of Hi-C matrices with diverse genomic assay outputs, as well as to compare interaction matrices between various conditions. https://github.com/kcakdemir/HiCPlotter


Molecular and Cellular Biology | 2011

Ubp8 and SAGA Regulate Snf1 AMP Kinase Activity

Marenda A. Wilson; Evangelia Koutelou; Calley Hirsch; Kadir C. Akdemir; Andria Schibler; Michelle Craig Barton; Sharon Y.R. Dent

ABSTRACT Posttranslational modifications of histone proteins play important roles in the modulation of gene expression. The Saccharomyces cerevisiae (yeast) 2-MDa SAGA (Spt-Ada-Gcn5) complex, a well-studied multisubunit histone modifier, regulates gene expression through Gcn5-mediated histone acetylation and Ubp8-mediated histone deubiquitination. Using a proteomics approach, we determined that the SAGA complex also deubiquitinates nonhistone proteins, including Snf1, an AMP-activated kinase. Ubp8-mediated deubiquitination of Snf1 affects the stability and phosphorylation state of Snf1, thereby affecting Snf1 kinase activity. Others have reported that Gal83 is phosphorylated by Snf1, and we found that deletion of UBP8 causes decreased phosphorylation of Gal83, which is consistent with the effects of Ubp8 loss on Snf1 kinase functions. Overall, our data indicate that SAGA modulates the posttranslational modifications of Snf1 in order to fine-tune gene expression levels.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Truncating PREX2 mutations activate its GEF activity and alter gene expression regulation in NRAS-mutant melanoma

Yonathan Lissanu Deribe; Yanxia Shi; Kunal Rai; Luigi Nezi; Samir B. Amin; Chia Chin Wu; Kadir C. Akdemir; Mozhdeh Mahdavi; Qian Peng; Qing Edward Chang; Kirsti Hornigold; Stefan T. Arold; Heidi C. E. Welch; Levi A. Garraway; Lynda Chin

Significance Mutations in the PI3K/PTEN/Akt signaling pathway occur frequently across multiple tumor types. These mutations primarily serve to activate PI-3 and Akt kinases. PREX2 is a guanine nucleotide exchanger for Rac1 that is significantly mutated in melanoma and pancreatic ductal adenocarcinoma. Here we report that a mouse model of a truncating PREX2 mutation shows accelerated melanoma development in the context of mutant NRAS. Truncating PREX2 mutations have increased Rac1 guanine nucleotide exchange factor activity, and tumors harboring these mutations have elevated PI3K/Akt pathway activation and reduced expression of critical negative cell cycle regulators leading to increased cell proliferation. This work provides evidence for a previously unidentified mechanism of activating Rac1, the PI3K pathway, and regulation of cell cycle progression in melanoma. PREX2 (phosphatidylinositol-3,4,5-triphosphate-dependent Rac-exchange factor 2) is a PTEN (phosphatase and tensin homolog deleted on chromosome 10) binding protein that is significantly mutated in cutaneous melanoma and pancreatic ductal adenocarcinoma. Here, genetic and biochemical analyses were conducted to elucidate the nature and mechanistic basis of PREX2 mutation in melanoma development. By generating an inducible transgenic mouse model we showed an oncogenic role for a truncating PREX2 mutation (PREX2E824*) in vivo in the context of mutant NRAS. Using integrative cross-species gene expression analysis, we identified deregulated cell cycle and cytoskeleton organization as significantly perturbed biological pathways in PREX2 mutant tumors. Mechanistically, truncation of PREX2 activated its Rac1 guanine nucleotide exchange factor activity, abolished binding to PTEN and activated the PI3K (phosphatidyl inositol 3 kinase)/Akt signaling pathway. We further showed that PREX2 truncating mutations or PTEN deletion induces down-regulation of the tumor suppressor and cell cycle regulator CDKN1C (also known as p57KIP2). This down-regulation occurs, at least partially, through DNA hypomethylation of a differentially methylated region in chromosome 11 that is a known regulatory region for expression of the CDKN1C gene. Together, these findings identify PREX2 as a mediator of NRAS-mutant melanoma development that acts through the PI3K/PTEN/Akt pathway to regulate gene expression of a cell cycle regulator.


Hepatology | 2010

Direct activation of forkhead box O3 by tumor suppressors p53 and p73 is disrupted during liver regeneration in mice.

Svitlana Kurinna; Sabrina A. Stratton; Wen Wei Tsai; Kadir C. Akdemir; Weisong Gu; Pallavi Singh; Triona Goode; Gretchen J. Darlington; Michelle C. Barton

The p53 family of proteins regulates the expression of target genes that promote cell cycle arrest and apoptosis, which may be linked to cellular growth control as well as tumor suppression. Within the p53 family, p53 and the transactivating p73 isoform (TA‐p73) have hepatic‐specific functions in development and tumor suppression. Here, we determined TA‐p73 interactions with chromatin in the adult mouse liver and found forkhead box O3 (Foxo3) to be one of 158 gene targets. Global profiling of hepatic gene expression in the regenerating liver versus the quiescent liver revealed specific, functional categories of genes regulated over the time of regeneration. Foxo3 is the most responsive gene among transcription factors with altered expression during regenerative cellular proliferation. p53 and TA‐p73 bind a Foxo3 p53 response element (p53RE) and maintain active expression in the quiescent liver. During regeneration of the liver, the binding of p53 and TA‐p73, the recruitment of acetyltransferase p300, and the active chromatin structure of Foxo3 are disrupted along with a loss of Foxo3 expression. In agreement with the loss of Foxo3 transcriptional activation, a decrease in histone activation marks (dimethylated histone H3 at lysine 4, acetylated histone H3 at lysine 14, and acetylated H4) at the Foxo3 p53RE was detected after partial hepatectomy in mice. These parameters of Foxo3 regulation are reestablished with the completion of liver growth and regeneration and support a temporary suspension of p53 and TA‐p73 regulatory functions in normal cells during tissue regeneration. p53‐dependent and TA‐p73–dependent activation of Foxo3 was also observed in mouse embryonic fibroblasts and in mouse hepatoma cells overexpressing p53, TA‐p73α, and TA‐p73β isoforms. Conclusion: p53 and p73 directly bind and activate the expression of the Foxo3 gene in the adult mouse liver and murine cell lines. p53, TA‐p73, and p300 binding and Foxo3 expression decrease during liver regeneration, and this suggests a critical growth control mechanism mediated by these transcription factors in vivo. (HEPATOLOGY 2010;)


Cell Reports | 2017

Systematic Epigenomic Analysis Reveals Chromatin States Associated with Melanoma Progression

Petko Fiziev; Kadir C. Akdemir; John P. Miller; Emily Z. Keung; Neha S. Samant; Sneha Sharma; Christopher A. Natale; Christopher Terranova; Mayinuer Maitituoheti; Samirkumar Amin; Emmanuel Martinez-Ledesma; Mayura Dhamdhere; Jacob B. Axelrad; Amiksha Shah; Christine S. Cheng; Harshad S. Mahadeshwar; Sahil Seth; Michelle Craig Barton; Alexei Protopopov; Kenneth Y. Tsai; Michael A. Davies; Benjamin A. Garcia; Ido Amit; Lynda Chin; Jason Ernst; Kunal Rai

The extent and nature of epigenomic changes associated with melanoma progression is poorly understood. Through systematic epigenomic profiling of 35 epigenetic modifications and transcriptomic analysis, we define chromatin state changes associated with melanomagenesis by using a cell phenotypic model of non-tumorigenic and tumorigenic states. Computation of specific chromatin state transitions showed loss of histone acetylations and H3K4me2/3 on regulatory regions proximal to specific cancer-regulatory genes in important melanoma-driving cell signaling pathways. Importantly, such acetylation changes were also observed between benign nevi and malignant melanoma human tissues. Intriguingly, only a small fraction of chromatin state transitions correlated with expected changes in gene expression patterns. Restoration of acetylation levels on deacetylated loci by histone deacetylase (HDAC) inhibitors selectively blocked excessive proliferation in tumorigenic cells and human melanoma cells, suggesting functional roles of observed chromatin state transitions in driving hyperproliferative phenotype. Through these results, we define functionally relevant chromatin states associated with melanoma progression.


Nature | 2017

Synthetic vulnerabilities of mesenchymal subpopulations in pancreatic cancer

Giannicola Genovese; Alessandro Carugo; James Tepper; Frederick Robinson; Liren Li; Maria Svelto; Luigi Nezi; Denise Corti; Rosalba Minelli; Piergiorgio Pettazzoni; Tony Gutschner; Chia Chin Wu; Sahil Seth; Kadir C. Akdemir; Elisabetta Leo; Samirkumar Amin; Marco Dal Molin; Haoqiang Ying; Lawrence N. Kwong; Simona Colla; Koichi Takahashi; Papia Ghosh; Virginia Giuliani; Florian Muller; Prasenjit Dey; Shan Jiang; Jill Garvey; Chang Gong Liu; Jianhua Zhang; Timothy P. Heffernan

Malignant neoplasms evolve in response to changes in oncogenic signalling. Cancer cell plasticity in response to evolutionary pressures is fundamental to tumour progression and the development of therapeutic resistance. Here we determine the molecular and cellular mechanisms of cancer cell plasticity in a conditional oncogenic Kras mouse model of pancreatic ductal adenocarcinoma (PDAC), a malignancy that displays considerable phenotypic diversity and morphological heterogeneity. In this model, stochastic extinction of oncogenic Kras signalling and emergence of Kras-independent escaper populations (cells that acquire oncogenic properties) are associated with de-differentiation and aggressive biological behaviour. Transcriptomic and functional analyses of Kras-independent escapers reveal the presence of Smarcb1–Myc-network-driven mesenchymal reprogramming and independence from MAPK signalling. A somatic mosaic model of PDAC, which allows time-restricted perturbation of cell fate, shows that depletion of Smarcb1 activates the Myc network, driving an anabolic switch that increases protein metabolism and adaptive activation of endoplasmic-reticulum-stress-induced survival pathways. Increased protein turnover renders mesenchymal sub-populations highly susceptible to pharmacological and genetic perturbation of the cellular proteostatic machinery and the IRE1-α–MKK4 arm of the endoplasmic-reticulum-stress-response pathway. Specifically, combination regimens that impair the unfolded protein responses block the emergence of aggressive mesenchymal subpopulations in mouse and patient-derived PDAC models. These molecular and biological insights inform a potential therapeutic strategy for targeting aggressive mesenchymal features of PDAC.


Human Genetics | 2016

Pathogenetics of alveolar capillary dysplasia with misalignment of pulmonary veins

Przemyslaw Szafranski; Tomasz Gambin; Avinash V. Dharmadhikari; Kadir C. Akdemir; Shalini N. Jhangiani; Jennifer Schuette; Nihal Godiwala; Svetlana A. Yatsenko; Jessica Sebastian; Suneeta Madan-Khetarpal; Urvashi Surti; Rosanna G. Abellar; David A. Bateman; Ashley Wilson; Melinda Markham; Jill Slamon; Fernando Santos-Simarro; María Palomares; Julián Nevado; Pablo Lapunzina; Brian Hon-Yin Chung; Wai Lap Wong; Yoyo W. Y. Chu; Gary Tsz Kin Mok; Eitan Kerem; Joel Reiter; Namasivayam Ambalavanan; Scott A. Anderson; David R. Kelly; Joseph T.C. Shieh

Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal lung developmental disorder caused by heterozygous point mutations or genomic deletion copy-number variants (CNVs) of FOXF1 or its upstream enhancer involving fetal lung-expressed long noncoding RNA genes LINC01081 and LINC01082. Using custom-designed array comparative genomic hybridization, Sanger sequencing, whole exome sequencing (WES), and bioinformatic analyses, we studied 22 new unrelated families (20 postnatal and two prenatal) with clinically diagnosed ACDMPV. We describe novel deletion CNVs at the FOXF1 locus in 13 unrelated ACDMPV patients. Together with the previously reported cases, all 31 genomic deletions in 16q24.1, pathogenic for ACDMPV, for which parental origin was determined, arose de novo with 30 of them occurring on the maternally inherited chromosome 16, strongly implicating genomic imprinting of the FOXF1 locus in human lungs. Surprisingly, we have also identified four ACDMPV families with the pathogenic variants in the FOXF1 locus that arose on paternal chromosome 16. Interestingly, a combination of the severe cardiac defects, including hypoplastic left heart, and single umbilical artery were observed only in children with deletion CNVs involving FOXF1 and its upstream enhancer. Our data demonstrate that genomic imprinting at 16q24.1 plays an important role in variable ACDMPV manifestation likely through long-range regulation of FOXF1 expression, and may be also responsible for key phenotypic features of maternal uniparental disomy 16. Moreover, in one family, WES revealed a de novo missense variant in ESRP1, potentially implicating FGF signaling in the etiology of ACDMPV.

Collaboration


Dive into the Kadir C. Akdemir's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michelle Craig Barton

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kunal Rai

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bruce J. Aronow

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Emily Z. Keung

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sahil Seth

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Abhinav K. Jain

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kendra Allton

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sneha Sharma

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Wei Li

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge