Network


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

Hotspot


Dive into the research topics where Safak Yalcin is active.

Publication


Featured researches published by Safak Yalcin.


Journal of Clinical Investigation | 2007

Foxo3 is required for the regulation of oxidative stress in erythropoiesis

Dragan Marinkovic; Xin Zhang; Safak Yalcin; Julia P. Luciano; Carlo Brugnara; Tara Huber; Saghi Ghaffari

Erythroid cells accumulate hemoglobin as they mature and as a result are highly prone to oxidative damage. However, mechanisms of transcriptional control of antioxidant defense in erythroid cells have thus far been poorly characterized. We observed that animals deficient in the forkhead box O3 (Foxo3) transcription factor died rapidly when exposed to erythroid oxidative stress-induced conditions, while wild-type mice showed no decreased viability. In view of this striking finding, we investigated the potential role of Foxo3 in the regulation of ROS in erythropoiesis. Foxo3 expression, nuclear localization, and transcriptional activity were all enhanced during normal erythroid cell maturation. Foxo3-deficient erythrocytes exhibited decreased expression of ROS scavenging enzymes and had a ROS-mediated shortened lifespan and evidence of oxidative damage. Furthermore, loss of Foxo3 induced mitotic arrest in erythroid precursor cells, leading to a significant decrease in the rate of in vivo erythroid maturation. We identified ROS-mediated upregulation of p21(CIP1/WAF1/Sdi1) (also known as Cdkn1a) as a major contributor to the interference with cell cycle progression in Foxo3-deficient erythroid precursor cells. These findings establish an essential nonredundant function for Foxo3 in the regulation of oxidative stress, cell cycle, maturation, and lifespan of erythroid cells. These results may have an impact on the understanding of human disorders in which ROS play a role.


Journal of Biological Chemistry | 2008

Foxo3 Is Essential for the Regulation of Ataxia Telangiectasia Mutated and Oxidative Stress-mediated Homeostasis of Hematopoietic Stem Cells

Safak Yalcin; Xin Zhang; Julia P. Luciano; Sathish Kumar Mungamuri; Dragan Marinkovic; Cécile Vercherat; Abby Sarkar; Marcos Grisotto; Reshma Taneja; Saghi Ghaffari

Unchecked accumulation of reactive oxygen species (ROS) compromises maintenance of hematopoietic stem cells. Regulation of ROS by the tumor suppressor protein ataxia telangiectasia mutated (ATM) is critical for preserving the hematopoietic stem cell pool. In this study we demonstrate that the Foxo3 member of the Forkhead Box O (FoxO) family of transcription factors is essential for normal ATM expression. In addition, we show that loss of Foxo3 leads to defects in hematopoietic stem cells, and these defects result from an overaccumulation of ROS. Foxo3 suppression of ROS in hematopoietic stem cells is mediated partly by regulation of ATM expression. We identify ROS-independent modulations of ATM and p16INK4a and ROS-mediated activation of p53/p21CIP1/WAF1/Sdi1 tumor suppressor pathways as major contributors to Foxo3-null hematopoietic stem cells defects. Our studies demonstrate that Foxo3 represses ROS in part via regulation of ATM and that this repression is required for maintenance of the hematopoietic stem cell pool.


The EMBO Journal | 2010

ROS‐mediated amplification of AKT/mTOR signalling pathway leads to myeloproliferative syndrome in Foxo3 −/− mice

Safak Yalcin; Dragan Marinkovic; Sathish Kumar Mungamuri; Xin Zhang; Wei Tong; Rani S. Sellers; Saghi Ghaffari

Reactive oxygen species (ROS) participate in normal intracellular signalling and in many diseases including cancer and aging, although the associated mechanisms are not fully understood. Forkhead Box O (FoxO) 3 transcription factor regulates levels of ROS concentrations, and is essential for maintenance of hematopoietic stem cells. Here, we show that loss of Foxo3 causes a myeloproliferative syndrome with splenomegaly and increased hematopoietic progenitors (HPs) that are hypersensitive to cytokines. These mutant HPs contain increased ROS, overactive intracellular signalling through the AKT/mammalian target of rapamycin signalling pathway and relative deficiency of Lnk, a negative regulator of cytokine receptor signalling. In vivo treatment with ROS scavenger N‐acetyl‐cysteine corrects these biochemical abnormalities and relieves the myeloproliferation. Moreover, enforced expression of Lnk by retroviral transfer corrects the abnormal expansion of Foxo3−/− HPs in vivo. Our combined results show that loss of Foxo3 causes increased ROS accumulation in HPs. In turn, this inhibits Lnk expression that contributes to exaggerated cytokine responses that lead to myeloproliferation. Our findings could explain the mechanisms by which mutations that alter Foxo3 function induce malignancy. More generally, the work illustrates how deregulated ROS may contribute to malignant progression.


American Journal of Hematology | 2014

FOXO3‐mTOR metabolic cooperation in the regulation of erythroid cell maturation and homeostasis

Xin Zhang; Genís Campreciós; Pauline Rimmele; Raymond Liang; Safak Yalcin; Sathish Kumar Mungamuri; Jeffrey Barminko; Valentina d'Escamard; Margaret H. Baron; Carlo Brugnara; Dmitri Papatsenko; Stefano Rivella; Saghi Ghaffari

Ineffective erythropoiesis is observed in many erythroid disorders including β‐thalassemia and anemia of chronic disease in which increased production of erythroblasts that fail to mature exacerbate the underlying anemias. As loss of the transcription factor FOXO3 results in erythroblast abnormalities similar to the ones observed in ineffective erythropoiesis, we investigated the underlying mechanisms of the defective Foxo3−/− erythroblast cell cycle and maturation. Here we show that loss of Foxo3 results in overactivation of the JAK2/AKT/mTOR signaling pathway in primary bone marrow erythroblasts partly mediated by redox modulation. We further show that hyperactivation of mTOR signaling interferes with cell cycle progression in Foxo3 mutant erythroblasts. Importantly, inhibition of mTOR signaling, in vivo or in vitro enhances significantly Foxo3 mutant erythroid cell maturation. Similarly, in vivo inhibition of mTOR remarkably improves erythroid cell maturation and anemia in a model of β‐thalassemia. Finally we show that FOXO3 and mTOR are likely part of a larger metabolic network in erythroblasts as together they control the expression of an array of metabolic genes some of which are implicated in erythroid disorders. These combined findings indicate that a metabolism‐mediated regulatory network centered by FOXO3 and mTOR control the balanced production and maturation of erythroid cells. They also highlight physiological interactions between these proteins in regulating erythroblast energy. Our results indicate that alteration in the function of this network might be implicated in the pathogenesis of ineffective erythropoiesis. Am. J. Hematol. 89:954–963, 2014.


Current Drug Targets | 2011

Regulation and Function of FoxO Transcription Factors in Normal and Cancer Stem Cells: What Have We Learned?

Xin Zhang; Maite Rielland; Safak Yalcin; Saghi Ghaffari

Forkhead FoxO transcription factors exert critical biological functions in response to genotoxic stress. In mammals four FoxOs proteins are known. FoxOs induce cell cycle arrest, repair damaged DNA, or initiate apoptosis by modulating genes that control these processes. In particular, FoxO proteins are critical regulators of oxidative stress by modulating the expression of several anti-oxidant enzyme genes. This function of FoxO is essential for the regulation of stem and progenitor cell pool in the hematopoietic system and possibly other stem cells. Overall functions of FoxOs are consistent with their role as tumor suppressors as has been shown in animal models. As such, FoxOs are suppressed in various cancer cells. However, recent reports strongly suggest that FoxOs are critical for the maintenance of leukemic stem cells. The diverse functions of FoxOs are orchestrated by tight regulations of expression and activity of its family members. Here we discuss the recent progress in understanding the function of FoxOs specifically in normal and cancer stem cells and what is known about the regulation of these proteins in various cell types and tissues including in the physiological setting of primary cells in vivo. These studies underscore the importance of regulation of FoxO proteins and whether these factors play distinct or redundant functions. Understanding how FoxOs are modulated is critical for devising novel therapies based on targeted restoration/or inhibition of FoxO function in cancer and in other diseased cells in which FoxOs have a key function.


Human Molecular Genetics | 2009

Stra13 regulates oxidative stress mediated skeletal muscle degeneration

Cécile Vercherat; Teng-Kai Chung; Safak Yalcin; Neriman Tuba Gulbagci; Suma Gopinadhan; Saghi Ghaffari; Reshma Taneja

Duchenne Muscular Dystrophy (DMD), caused by loss of dystrophin is characterized by progressive muscle cell necrosis. However, the mechanisms leading to muscle degeneration in DMD are poorly understood. Here, we demonstrate that Stra13 protects muscle cells from oxidative damage, and its absence leads to muscle necrosis in response to injury in Stra13-deficient mice. Interestingly, Stra13-/- mutants express elevated levels of TNFalpha, reduced levels of heme-oxygenase-1, and display apparent signs of oxidative stress prior to muscle death. Moreover, Stra13-/- muscle cells exhibit an increased sensitivity to pro-oxidants, and conversely, Stra13 overexpression provides resistance to oxidative damage. Consistently, treatment with anti-oxidant N-acetylcysteine ameliorates muscle necrosis in Stra13-/- mice. We also demonstrate that Stra13 expression is elevated in muscles from dystrophin-deficient (mdx) mice, and mdx/Stra13-/- double mutants exhibit an early onset of muscle degeneration. Our studies underscore the importance of oxidative stress-mediated muscle degeneration in muscular dystrophy, and reveal the contribution of Stra13 in maintenance of muscle integrity.


Cell Reports | 2015

Myeloid Dysregulation in a Human Induced Pluripotent Stem Cell Model of PTPN11-Associated Juvenile Myelomonocytic Leukemia

Sonia Mulero-Navarro; Ana Sevilla; Ángel C. Román; Dung Fang Lee; Sunita L. D’Souza; Sherly Pardo; Ilan Riess; Jie Su; Ninette Cohen; Christoph Schaniel; Nelson A. Rodriguez; Alessia Baccarini; Brian D. Brown; Hélène Cavé; Aurélie Caye; Marion Strullu; Safak Yalcin; Christopher Y. Park; Perundurai S. Dhandapany; Ge Yongchao; Lisa Edelmann; Sawsan Bahieg; Patrick Raynal; Elisabetta Flex; Marco Tartaglia; Kateri Moore; Ihor R. Lemischka; Bruce D. Gelb

Somatic PTPN11 mutations cause juvenile myelomonocytic leukemia (JMML). Germline PTPN11 defects cause Noonan syndrome (NS), and specific inherited mutations cause NS/JMML. Here, we report that hematopoietic cells differentiated from human induced pluripotent stem cells (hiPSCs) harboring NS/JMML-causing PTPN11 mutations recapitulated JMML features. hiPSC-derived NS/JMML myeloid cells exhibited increased signaling through STAT5 and upregulation of miR-223 and miR-15a. Similarly, miR-223 and miR-15a were upregulated in 11/19 JMML bone marrow mononuclear cells harboring PTPN11 mutations, but not those without PTPN11 defects. Reducing miR-223s function in NS/JMML hiPSCs normalized myelogenesis. MicroRNA target gene expression levels were reduced in hiPSC-derived myeloid cells as well as in JMML cells with PTPN11 mutations. Thus, studying an inherited human cancer syndrome with hiPSCs illuminated early oncogenesis prior to the accumulation of secondary genomic alterations, enabling us to discover microRNA dysregulation, establishing a genotype-phenotype association for JMML and providing therapeutic targets.


Archive | 2014

The Role of MicroRNAs in Hematopoietic Stem Cells and Leukemia Development

Safak Yalcin; Stephen S. Chung; Christopher Y. Park

Hematopoietic stem cells (HSCs) are multipotent cells capable of self-renewal as well as differentiation into all mature blood cell types to sustain lifelong hematopoiesis. Leukemias are thought to be initiated and maintained by leukemic stem cells (LSCs), which also have the capacity for self-renewal, but they are also characterized by varying levels of impaired differentiation and increased proliferation which give rise to disease phenotypes. LSCs and HSCs share the common ability to self-renew, and they may rely on similar pathways for this unique function. Thus, it is critical to understand the molecular mechanisms shared between these two cell populations, as these are likely to represent key features that drive leukemic transformation of HSCs and/or their downstream progeny. Recently, microRNAs (miRNAs) have been implicated as important regulators of self-renewal and differentiation in the hematopoietic system. Profiling of normal and malignant hematopoietic cells, corroborated with a limited but growing number of functional studies, has demonstrated that miRNAs are critical regulators of HSC function, are dysregulated in leukemias, and likely play an important role in leukemogenesis. Herein, we will review these studies and discuss their contributions toward our understanding of the importance of miRNAs in normal and malignant stem cell function in the hematopoietic system.


Nature Cell Biology | 2011

FOXO1 is an essential regulator of pluripotency in human embryonic stem cells

Xin Zhang; Safak Yalcin; Dung Fang Lee; Tsung Yin J Yeh; Seung Min Lee; Jie Su; Sathish Kumar Mungamuri; Pauline Rimmele; Marion Kennedy; Rani S. Sellers; Markus Landthaler; Thomas Tuschl; Nai Wen Chi; Ihor R. Lemischka; Gordon Keller; Saghi Ghaffari


Cell Cycle | 2016

Evidence for AKT-independent regulation of FOXO1 and FOXO3 in haematopoietic stem and progenitor cells.

Raymond Liang; Pauline Rimmele; Carolina L. Bigarella; Safak Yalcin; Saghi Ghaffari

Collaboration


Dive into the Safak Yalcin's collaboration.

Top Co-Authors

Avatar

Saghi Ghaffari

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Xin Zhang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Christopher Y. Park

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sathish Kumar Mungamuri

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Julia P. Luciano

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Reshma Taneja

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Cécile Vercherat

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Joseph Yusup Shin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pauline Rimmele

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge