Network


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

Hotspot


Dive into the research topics where Fatih Kocabas is active.

Publication


Featured researches published by Fatih Kocabas.


Cell Stem Cell | 2010

The Distinct Metabolic Profile of Hematopoietic Stem Cells Reflects Their Location in a Hypoxic Niche

Tugba Simsek; Fatih Kocabas; Junke Zheng; Ralph J. DeBerardinis; Ahmed I. Mahmoud; Eric N. Olson; Jay W. Schneider; Cheng Cheng Zhang; Hesham A. Sadek

Bone marrow transplantation is the primary therapy for numerous hematopoietic disorders. The efficiency of bone marrow transplantation depends on the function of long-term hematopoietic stem cells (LT-HSCs), which is markedly influenced by their hypoxic niche. Survival in this low-oxygen microenvironment requires significant metabolic adaptation. Here, we show that LT-HSCs utilize glycolysis instead of mitochondrial oxidative phosphorylation to meet their energy demands. We used flow cytometry to identify a unique low mitochondrial activity/glycolysis-dependent subpopulation that houses the majority of hematopoietic progenitors and LT-HSCs. Finally, we demonstrate that Meis1 and Hif-1alpha are markedly enriched in LT-HSCs and that Meis1 regulates HSC metabolism through transcriptional activation of Hif-1alpha. These findings reveal an important transcriptional network that regulates HSC metabolism.


Nature | 2013

Meis1 regulates postnatal cardiomyocyte cell cycle arrest

Ahmed I. Mahmoud; Fatih Kocabas; Shalini Muralidhar; Wataru Kimura; Ahmed S. Koura; Suwannee Thet; Enzo R. Porrello; Hesham A. Sadek

The neonatal mammalian heart is capable of substantial regeneration following injury through cardiomyocyte proliferation. However, this regenerative capacity is lost by postnatal day 7 and the mechanisms of cardiomyocyte cell cycle arrest remain unclear. The homeodomain transcription factor Meis1 is required for normal cardiac development but its role in cardiomyocytes is unknown. Here we identify Meis1 as a critical regulator of the cardiomyocyte cell cycle. Meis1 deletion in mouse cardiomyocytes was sufficient for extension of the postnatal proliferative window of cardiomyocytes, and for re-activation of cardiomyocyte mitosis in the adult heart with no deleterious effect on cardiac function. In contrast, overexpression of Meis1 in cardiomyocytes decreased neonatal myocyte proliferation and inhibited neonatal heart regeneration. Finally, we show that Meis1 is required for transcriptional activation of the synergistic CDK inhibitors p15, p16 and p21. These results identify Meis1 as a critical transcriptional regulator of cardiomyocyte proliferation and a potential therapeutic target for heart regeneration.


Blood | 2012

Meis1 regulates the metabolic phenotype and oxidant defense of hematopoietic stem cells

Fatih Kocabas; Junke Zheng; Suwannee Thet; Neal G. Copeland; Nancy A. Jenkins; Ralph J. DeBerardinis; Cheng Cheng Zhang; Hesham A. Sadek

The role of Meis1 in leukemia is well established, but its role in hematopoietic stem cells (HSCs) remains poorly understood. Previously, we showed that HSCs use glycolytic metabolism to meet their energy demands. However, the mechanism of regulation of HSC metabolism, and the importance of maintaining this distinct metabolic phenotype on HSC function has not been determined. More importantly, the primary function of Meis1 in HSCs remains unknown. Here, we examined the effect of loss of Meis1 on HSC function and metabolism. Inducible Meis1 deletion in adult mouse HSCs resulted in loss of HSC quiescence, and failure of bone marrow repopulation after transplantation. While we previously showed that Meis1 regulates Hif-1α transcription in vitro, we demonstrate here that loss of Meis1 results in down-regulation of both Hif-1α and Hif-2α in HSCs. This resulted in a shift to mitochondrial metabolism, increased reactive oxygen species production, and apoptosis of HSCs. Finally, we demonstrate that the effect of Meis1 knockout on HSCs is entirely mediated through reactive oxygen species where treatment of the Meis1 knockout mice with the scavenger N-acetylcystein restored HSC quiescence and rescued HSC function. These results uncover an important transcriptional network that regulates metabolism, oxidant defense, and maintenance of HSCs.


Molecular Pharmaceutics | 2013

Synergistic Interaction of Paclitaxel and Curcumin with Cyclodextrin Polymer Complexation in Human Cancer Cells

Ali O Boztas; Ozgur Karakuzu; Gabriela Galante; Zafer Ugur; Fatih Kocabas; Cengiz Z. Altuntas; A. Ozgur Yazaydin

The use of cytotoxic chemotherapic agents is the most common method for the treatment of metastatic cancers. Poor water solubility and low efficiency of chemotherapic agents are among the major hurdles of effective chemotherapy treatments. Curcumin and paclitaxel are well-known chemotherapic agents with poor water solubility and undesired side effects. In this study, a novel drug nanocarrier system was formulated by encapsulating curcumin and paclitaxel in poly(β-cyclodextrin triazine) (PCDT) for the therapy of four cancer models; ovarian, lung, prostate, and breast cancer. Cell viability and colony formation assays revealed enhanced curcumin cytotoxicity upon complexation. Annexin V apoptotic studies showed that the PCDT complexation improved curcumin induced apoptosis in human ovarian cancer cell lines A2780 and SKOV-3, human nonsmall cell lung carcinoma cell line H1299, and human prostate cancer line DU-145, while no significant effect was observed with paclitaxel/PCDT complexation. The bioactivity of combining curcumin and paclitaxel was also investigated. A synergism was found between curcumin and paclitaxel, particularly when complexed with PCDT on A2780, SKOV-3, and H1299 cancer cell lines.


eLife | 2013

Intraflagellar transport drives flagellar surface motility

Sheng Min Shih; Benjamin D. Engel; Fatih Kocabas; Thomas Bilyard; Arne Gennerich; Wallace F. Marshall; Ahmet Yildiz

The assembly and maintenance of all cilia and flagella require intraflagellar transport (IFT) along the axoneme. IFT has been implicated in sensory and motile ciliary functions, but the mechanisms of this relationship remain unclear. Here, we used Chlamydomonas flagellar surface motility (FSM) as a model to test whether IFT provides force for gliding of cells across solid surfaces. We show that IFT trains are coupled to flagellar membrane glycoproteins (FMGs) in a Ca2+-dependent manner. IFT trains transiently pause through surface adhesion of their FMG cargos, and dynein-1b motors pull the cell towards the distal tip of the axoneme. Each train is transported by at least four motors, with only one type of motor active at a time. Our results demonstrate the mechanism of Chlamydomonas gliding motility and suggest that IFT plays a major role in adhesion-induced ciliary signaling pathways. DOI: http://dx.doi.org/10.7554/eLife.00744.001


EMBO Reports | 2015

Mitochondrial metabolism in hematopoietic stem cells requires functional FOXO3

Pauline Rimmele; Raymond Liang; Carolina L. Bigarella; Fatih Kocabas; Jingjing Xie; Madhavika N. Serasinghe; Jerry E. Chipuk; Hesham A. Sadek; Cheng Cheng Zhang; Saghi Ghaffari

Hematopoietic stem cells (HSC) are primarily dormant but have the potential to become highly active on demand to reconstitute blood. This requires a swift metabolic switch from glycolysis to mitochondrial oxidative phosphorylation. Maintenance of low levels of reactive oxygen species (ROS), a by‐product of mitochondrial metabolism, is also necessary for sustaining HSC dormancy. Little is known about mechanisms that integrate energy metabolism with hematopoietic stem cell homeostasis. Here, we identify the transcription factor FOXO3 as a new regulator of metabolic adaptation of HSC. ROS are elevated in Foxo3−/− HSC that are defective in their activity. We show that Foxo3−/− HSC are impaired in mitochondrial metabolism independent of ROS levels. These defects are associated with altered expression of mitochondrial/metabolic genes in Foxo3−/− hematopoietic stem and progenitor cells (HSPC). We further show that defects of Foxo3−/− HSC long‐term repopulation activity are independent of ROS or mTOR signaling. Our results point to FOXO3 as a potential node that couples mitochondrial metabolism with HSC homeostasis. These findings have critical implications for mechanisms that promote malignant transformation and aging of blood stem and progenitor cells.


Journal of Cardiovascular Translational Research | 2012

The Hypoxic Epicardial and Subepicardial Microenvironment

Fatih Kocabas; Ahmed I. Mahmoud; Drazen Sosic; Enzo R. Porrello; Rui Chen; Joseph A. Garcia; Ralph J. DeBerardinis; Hesham A. Sadek

Recent reports indicate that the adult mammalian heart is capable of limited, but measurable, cardiomyocyte turnover. While the lineage origin of the newly formed cardiomyocytes is not entirely understood, mounting evidence suggest that the epicardium and subepicardium may represent an important source of cardiac stem or progenitor cells. Stem cell niches are characterized by low oxygen tension, where stem cells preferentially utilize cytoplasmic glycolysis to meet their energy demands. However, it is unclear if the heart harbors similar hypoxic regions, or whether these regions house metabolically distinct cardiac progenitor populations. Here we identify the epicardium and subepicardium as the cardiac hypoxic niche-based capillary density quantification, and localization of Hif-1α in the uninjured heart. We further demonstrate that this hypoxic microenvironment houses a metabolically distinct population of glycolytic progenitor cells. Finally, we show that Hif-1α regulates the glycolytic phenotype and progenitor properties of these cells. These findings highlight important anatomical and functional properties of the epicardial and subepicardial microenvironment, and the potential role of hypoxia signaling in regulation of cardiac progenitors.


Blood | 2014

Profilin 1 is essential for retention and metabolism of mouse hematopoietic stem cells in bone marrow

Junke Zheng; Zhigang Lu; Fatih Kocabas; Ralph T. Böttcher; Mercedes Costell; Xunlei Kang; Xiaoye Liu; Ralph J. DeBerardinis; Qianming Wang; Guo-Qiang Chen; Hesham A. Sadek; Cheng Cheng Zhang

How stem cells interact with the microenvironment to regulate their cell fates and metabolism is largely unknown. Here we demonstrated that the deletion of the cytoskeleton-modulating protein profilin 1 (pfn1) in hematopoietic stem cell (HSCs) led to bone marrow failure, loss of quiescence, and mobilization and apoptosis of HSCs in vivo. A switch from glycolysis to mitochondrial respiration with increased reactive oxygen species (ROS) level was also observed in HSCs on pfn1 deletion. Importantly, treatment of pfn1-deficient mice with the antioxidant N-acetyl-l-cysteine reversed the ROS level and loss of quiescence of HSCs, suggesting that the metabolism is mechanistically linked to the cell cycle quiescence of stem cells. The actin-binding and proline-binding activities of pfn1 are required for its function in HSCs. Our study provided evidence that pfn1 at least partially acts through the axis of pfn1/Gα13/EGR1 to regulate stem cell retention and metabolism in the bone marrow.


Cell & Bioscience | 2015

Hypoxic metabolism in human hematopoietic stem cells

Fatih Kocabas; Li Xie; Jingjing Xie; Zhuo Yu; Ralph J. DeBerardinis; Wataru Kimura; Suwannee Thet; Ahmed F. Elshamy; Hesham Abouellail; Shalini Muralidhar; Xiaoye Liu; Chiqi Chen; Hesham A. Sadek; Cheng Cheng Zhang; Junke Zheng

BackgroundAdult hematopoietic stem cells (HSCs) are maintained in a microenvironment, known as niche in the endosteal regions of the bone marrow. This stem cell niche with low oxygen tension requires HSCs to adopt a unique metabolic profile. We have recently demonstrated that mouse long-term hematopoietic stem cells (LT-HSCs) utilize glycolysis instead of mitochondrial oxidative phosphorylation as their main energy source. However, the metabolic phenotype of human hematopoietic progenitor and stem cells (HPSCs) remains unknown.ResultsWe show that HPSCs have a similar metabolic phenotype, as shown by high rates of glycolysis, and low rates of oxygen consumption. Fractionation of human mobilized peripheral blood cells based on their metabolic footprint shows that cells with a low mitochondrial potential are highly enriched for HPSCs. Remarkably, low MP cells had much better repopulation ability as compared to high MP cells. Moreover, similar to their murine counterparts, we show that Hif-1α is upregulated in human HPSCs, where it is transcriptionally regulated by Meis1. Finally, we show that Meis1 and its cofactors Pbx1 and HoxA9 play an important role in transcriptional activation of Hif-1α in a cooperative manner.ConclusionsThese findings highlight the unique metabolic properties of human HPSCs and the transcriptional network that regulates their metabolic phenotype.


Anatolian Journal of Cardiology | 2016

Evolving approaches to heart regeneration by therapeutic stimulation of resident cardiomyocyte cell cycle

Raife Dilek Turan; Galip Servet Aslan; Dogacan Yucel; Remziye Döğer; Fatih Kocabas

Heart has long been considered a terminally differentiated organ. Recent studies, however, have suggested that there is a modest degree of cardiomyocyte (CM) turnover in adult mammalian heart, albeit not sufficient for replacement of lost CMs following cardiac injuries. Cardiac regeneration studies in various model organisms including zebrafish, newt, and more recently in neonatal mouse, have demonstrated that CM dedifferentiation and concomitant proliferation play important roles in replacement of lost CMs and restoration of cardiac contractility. Further studies with neonatal cardiac regeneration mouse model suggested that major source of new CMs is existing CMs, with the possibility of involvement of cardiac stem cells. Numerous studies have now been conducted on induction of cardiac regeneration and have identified various cardiogenic factors, cardiogenic micro ribonucleic acid and cardiogenic small molecules. This report is a review of studies regarding generation of CM and prospects for application.

Collaboration


Dive into the Fatih Kocabas's collaboration.

Top Co-Authors

Avatar

Hesham A. Sadek

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cheng Cheng Zhang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ralph J. DeBerardinis

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ahmed I. Mahmoud

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Junke Zheng

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmet Yildiz

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge