Network


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

Hotspot


Dive into the research topics where Mesut Eren is active.

Publication


Featured researches published by Mesut Eren.


Circulation | 2002

Age-Dependent Spontaneous Coronary Arterial Thrombosis in Transgenic Mice That Express a Stable Form of Human Plasminogen Activator Inhibitor-1

Mesut Eren; Corrie A. Painter; James B. Atkinson; Paul Declerck; Douglas E. Vaughan

Background—Plasminogen activator inhibitor-1 (PAI-1) regulates fibrinolysis and has been reported to be an independent risk factor for ischemic cardiovascular events. This study describes the age-dependent development of spontaneous coronary arterial thrombi that are associated with evidence of subendocardial myocardial infarction in mice transgenic for human PAI-1. Methods and Results—We generated two independent transgenic mice founder lines that express a stable variant of active human PAI-1 under control of the murine preproendothelin-1 (mPPET-1) promoter. Backcrossed homozygous transgenic animals from founder line I had plasma PAI-1 levels of 23±12 ng/mL. PAI-1 transgenic animals younger than 4 months do not exhibit any evidence of arterial or venous thrombosis. Ninety percent of transgenic animals (n=10) older than 6 months developed spontaneous occlusions of typically multiple, penetrating coronary arteries, with histological evidence of subendocardial infarction identified in 50% of animals. Conclusions—This study shows that chronically elevated levels of PAI-1 are associated with age-dependent coronary arterial thrombosis in mice transgenic for human PAI-1. This is the first study of a murine model of coronary thrombosis that occurs in the absence of severe hypercholesterolemia or multiple genetic manipulations. These findings provide new evidence to support the hypothesis that PAI-1 excess contributes to the development of coronary arterial thrombosis.


Journal of Molecular and Cellular Cardiology | 2003

Regulation of the PAI-1 promoter by circadian clock components: differential activation by BMAL1 and BMAL2.

John A. Schoenhard; Layton H. Smith; Corrie A. Painter; Mesut Eren; Carl Hirschie Johnson; Douglas E. Vaughan

Circadian variation in plasminogen activator inhibitor-1 (PAI-1) production likely contributes to increased risk of myocardial infarction and decreased efficacy of thrombolytic therapy during the morning. In this study, we characterize the abilities of fundamental molecular components of intrinsic circadian clocks to regulate the human PAI-1 promoter in transfected endothelial cells. Both CLOCK:BMAL1 and CLOCK:BMAL2 heterodimers activate the PAI-1 promoter through requisite proximal (-565 to -560 bp) and distal (-680 to -675 bp) E-box enhancers. Although the distal E-box overlaps the 4G/5G polymorphism of the PAI-1 promoter, allelic variation at this site does not influence CLOCK:BMAL1-and CLOCK:BMAL2-mediated transactivation. Together, CLOCK:BMAL1 and CLOCK:BMAL2 make additive contributions to PAI-1 gene transcription. While the abilities of these heterodimers to activate gene expression differ by twofold, the susceptibilities of these circadian activators to inhibition by period and cryptochrome proteins are equivalent and redox independent. Given that BMAL1 and BMAL2 differ in their spatiotemporal distributions, such distinctions may allow intrinsic circadian clocks to modulate the amplitudes of their oscillators, while maintaining circadian periodicity. In this way, fundamental circadian clock components may drive circadian variation in PAI-1, which in turn influences the pathogenesis, timing, and treatment of acute atherothrombotic events.


Proceedings of the National Academy of Sciences of the United States of America | 2014

PAI-1–regulated extracellular proteolysis governs senescence and survival in Klotho mice

Mesut Eren; Amanda E. Boe; Sheila B. Murphy; Aaron T. Place; Varun Nagpal; Luisa Morales-Nebreda; Daniela Urich; Susan E. Quaggin; G. R. Scott Budinger; Gökhan M. Mutlu; Toshio Miyata; Douglas E. Vaughan

Significance Plasminogen activator inhibitor-1 (PAI-1) is an essential mediator of cellular senescence in vitro and is one of the biochemical fingerprints of senescence in vivo. Klotho-deficient (kl/kl) mice display a complex phenotype reminiscent of human aging and exhibit age-dependent increases in PAI-1 in tissues and in plasma. Thus, we hypothesized that PAI-1 contributes to the aging-like phenotype of kl/kl mice. We observed that either genetic deficiency or pharmacological inhibition of PAI-1 in kl/kl mice was associated with reduced evidence of senescence, preserved organ structure and function, and a fourfold increase in median lifespan. These findings indicate that PAI-1 is a critical mediator of senescence in vivo and defines a novel target for the prevention and treatment of age-related disorders in man. Cellular senescence restricts the proliferative capacity of cells and is accompanied by the production of several proteins, collectively termed the “senescence-messaging secretome” (SMS). As senescent cells accumulate in tissue, local effects of the SMS have been hypothesized to disrupt tissue regenerative capacity. Klotho functions as an aging-suppressor gene, and Klotho-deficient (kl/kl) mice exhibit an accelerated aging-like phenotype that includes a truncated lifespan, arteriosclerosis, and emphysema. Because plasminogen activator inhibitor-1 (PAI-1), a serine protease inhibitor (SERPIN), is elevated in kl/kl mice and is a critical determinant of replicative senescence in vitro, we hypothesized that a reduction in extracellular proteolytic activity contributes to the accelerated aging-like phenotype of kl/kl mice. Here we show that PAI-1 deficiency retards the development of senescence and protects organ structure and function while prolonging the lifespan of kl/kl mice. These findings indicate that a SERPIN-regulated cell-nonautonomous proteolytic cascade is a critical determinant of senescence in vivo.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2006

Antiproliferative Agents Alter Vascular Plasminogen Activator Inhibitor-1 Expression. A Potential Prothrombotic Mechanism of Drug-Eluting Stents

James A.S. Muldowney; John R. Stringham; Shawn Levy; Linda A. Gleaves; Mesut Eren; Robert N. Piana; Douglas E. Vaughan

Objectives—Drug eluting stents (DES) reduce the incidence of restenosis after coronary angioplasty. Enthusiasm has been tempered by a possible increased risk of in-stent thrombosis. We examined the effects of paclitaxel and rapamycin on the endothelial transcriptome to identify alterations in gene expression associated with thrombosis. Methods and Results—Gene expression profiling was performed on human coronary artery endothelial cells treated with rapamycin or paclitaxel. Plasminogen activator inhibitor-1 (PAI-1) was the most consistently induced transcript in rapamycin-treated human coronary artery endothelial cells. RT-PCR and ELISA were performed to confirm positive findings. Transgenic mice engineered to express enhanced green fluorescent protein under control of the human PAI-1 promoter were also treated. Rapamycin and paclitaxel treated endothelial cells produced dose-dependent increases in PAI-1. There was a variable effect on endothelial tissue-type plasminogen activator (t-PA) expression. Enhanced expression of PAI-1 and enhanced green fluorescent protein were detected in coronary arteries, the aorta, and kidney of the mice. Conclusion—Antiproliferative agents stimulate the expression of prothrombotic genes. PAI-1 expression may also play a role in the prevention of restenosis through an antimigratory mechanism. The effects of antiproliferatives on vascular gene expression deserve further scrutiny in view of the increasing utilization of drug-eluting stents.


Circulation | 2013

Plasminogen Activator Inhibitor-1 Antagonist TM5441 Attenuates Nω-Nitro-l-Arginine Methyl Ester–Induced Hypertension and Vascular Senescence

Amanda E. Boe; Mesut Eren; Sheila B. Murphy; Christine Kamide; Atsuhiko Ichimura; David B. Terry; Danielle McAnally; Layton H. Smith; Toshio Miyata; Douglas E. Vaughan

Background Long-term inhibition of nitric oxide synthase (NOS) by L-arginine analogues such as Nω-nitro-L-arginine methyl ester (L-NAME) has been shown to induce senescence in vitro and systemic hypertension and arteriosclerosis in vivo. We previously reported that PAI-1-deficient mice (PAI-1−/−) are protected against L-NAME-induced pathologies. In this study, we investigated whether a novel, orally active PAI-1 antagonist (TM5441) has a similar protective effect against L-NAME treatment. Additionally, we studied whether L-NAME can induce vascular senescence in vivo and investigated the role of PAI-1 in this process.Background— Long-term inhibition of nitric oxide synthase by L-arginine analogues such as N&ohgr;-nitro-L-arginine methyl ester (L-NAME) has been shown to induce senescence in vitro and systemic hypertension and arteriosclerosis in vivo. We previously reported that plasminogen activator inhibitor-1 (PAI-1)–deficient mice (PAI-1−/−) are protected against L-NAME-induced pathologies. In this study, we investigated whether a novel, orally active PAI-1 antagonist (TM5441) has a similar protective effect against L-NAME treatment. Additionally, we studied whether L-NAME can induce vascular senescence in vivo and investigated the role of PAI-1 in this process. Methods and Results— Wild-type mice received either L-NAME or L-NAME and TM5441 for 8 weeks. Systolic blood pressure was measured every 2 weeks. We found that TM5441 attenuated the development of hypertension and cardiac hypertrophy compared with animals that had received L-NAME alone. Additionally, TM5441-treated mice had a 34% reduction in periaortic fibrosis relative to animals on L-NAME alone. Finally, we investigated the development of vascular senescence by measuring p16Ink4a expression and telomere length in aortic tissue. We found that L-NAME increased p16Ink4a expression levels and decreased telomere length, both of which were prevented with TM5441 cotreatment. Conclusions— Pharmacological inhibition of PAI-1 is protective against the development of hypertension, cardiac hypertrophy, and periaortic fibrosis in mice treated with L-NAME. Furthermore, PAI-1 inhibition attenuates the arterial expression of p16Ink4a and maintains telomere length. PAI-1 appears to play a pivotal role in vascular senescence, and these findings suggest that PAI-1 antagonists may provide a novel approach in preventing vascular aging and hypertension.


Circulation Research | 2004

Differential and Opposing Regulation of PAI-1 Promoter Activity by Estrogen Receptor α and Estrogen Receptor β in Endothelial Cells

Layton H. Smith; Stephen R. Coats; Hao Qin; Matthew S. Petrie; Joseph W. Covington; Ming Su; Mesut Eren; Douglas E. Vaughan

To investigate the molecular mechanisms involved in the estrogen-dependent control of plasminogen activator inhibitor-1 (PAI-1) gene expression in vascular cells, we compared the transactivation properties of estrogen receptors (ER&agr; and ER&bgr;) in regulating the activity of a human PAI-1 promoter reporter construct in transfected bovine aortic endothelial cells (BAECs). ER&agr; increased PAI-1 promoter activity in BAECs by an estrogen-dependent mechanism, whereas ER&bgr; suppressed PAI-1 promoter activity by an estrogen-independent mechanism. The suppressive activity of ER&bgr; was dominant over the inductive activity of ER&agr;. Mutation of a putative estrogen response element (ERE) located at position −427 in the proximal promoter abolished the ER&agr; action without influencing the suppressive effects of ER&bgr;. Mutation of either AP1-like site did not eliminate the ER&agr; or ER&bgr; actions at the PAI-1 promoter, suggesting that other promoter elements are involved in these responses. These mutations significantly reduced the −3.4kbp PAI-1 promoter response to serum. We concluded that ER&agr; and ER&bgr; exert differential effects on the PAI-1 promoter activity in transfected BAECs. ER&agr; activated the PAI-1 promoter through a proximal ERE (−427) and possibly additional EREs located within the PAI-1 promoter, whereas ER&bgr; suppressed the promoter construct via an unidentified mechanism. This is the first demonstration of the differential regulation of a vascular gene promoter by ER&agr; and ER&bgr;.


Current Drug Targets | 2007

PAI-1 antagonists: Predictable indications and unconventional applications

Douglas E. Vaughan; Bart De Taeye; Mesut Eren

At present, thrombolytic agents represent the only direct way of augmenting fibrinolytic activity in humans. While these agents are proven to be efficacious in the treatment of acute thrombotic events, they are not a viable option for long-term administration. There are numerous drugs available that indirectly to increase fibrinolytic activity by reducing plasma levels of plasminogen activator inhibitor-1 (PAI-1), including ACE inhibitors, insulin-sensitizing agents, and hormone replacement therapy in women. At present, efforts are underway to develop and test synthetic, selective PAI-1 antagonists. The potential applications of PAI-1 antagonists include thrombotic disorders (arterial and venous), amyloidosis, obesity, polycystic ovarian syndrome, and perhaps even type 2 diabetes mellitus. The availability of specific PAI-1 antagonists promises to expand the limits of understanding the role the fibrinolytic system plays in human disease and break through the current confines of therapeutic options that can effectively restore and augment the activity of the fibrinolytic system.


Journal of Molecular Endocrinology | 2007

Transgenic overexpression of plasminogen activator inhibitor-1 promotes the development of polycystic ovarian changes in female mice

Jessica K. Devin; Joyce E. Johnson; Mesut Eren; Linda A. Gleaves; William Bradham; John R. Bloodworth; Douglas E. Vaughan

Reproductive age women (5-10%) are affected by the polycystic ovarian syndrome (PCOS), a diagnosis which confers lifelong cardiovascular and reproductive health implications. Plasminogen activator inhibitor-1 (PAI-1), the main physiological inhibitor of plasminogen activation, is consistently elevated in women with PCOS, regardless of metabolic status. Interestingly, the plasminogen system has long been implicated in proteolytic processes within the dynamic ovary. A non-physiologic elevation in PAI-1 may thus contribute systemically to endothelial dysfunction and locally to abnormal ovarian phenotype and function. We herein characterize the phenotypic alterations in ovaries from transgenic mice, which constitutively express a stable form of human PAI-1 and determine the plasma testosterone level in these mice as opposed to their unaffected counterparts. Over half of the ovaries from transgenic mice were found to contain large cystic structures, in contrast to wild-type controls of the same genetic background (53% (N = 17) vs 5% (N = 22); P = 0.001). Plasma testosterone was nearly twofold elevated in transgenic female mice versus wild-type females (0.312 ng/ml +/- 0.154 (N = 10) vs 0.181 ng/ml +/- 0.083 (N = 8); P = 0.014). An elevation in PAI-1 therefore appears to predispose mice to the development of this abnormal architecture, which in turn is associated with an increase in plasma testosterone. Therefore, we propose that an inappropriate elevation in PAI-1 contributes to the development of polycystic structures; these findings may thus reorient the efforts aimed at the development of therapeutic agents for the treatment of this increasingly common syndrome.


Circulation Research | 2006

Foxc2 Is a Common Mediator of Insulin and Transforming Growth Factor β Signaling to Regulate Plasminogen Activator Inhibitor Type I Gene Expression

Hideo Fujita; Myengmo Kang; Mesut Eren; Linda A. Gleaves; Douglas E. Vaughan; Tsutomu Kume

Elevated plasma levels of plasminogen activator inhibitor type I (PAI-1), a significant risk factor of ischemic heart disease, are associated with insulin resistance in which insulin and transforming growth factor (TGF)-&bgr; play a pivotal role in regulating PAI-1 production. Forkhead transcription factor FOXC2 is an important regulator of insulin resistance. However, the underlying molecular mechanisms to link FOXC2 to PAI-1 levels in insulin resistance remain to be elucidated. Here, we demonstrate that Foxc2 is a common transcriptional activator of insulin and TGF-&bgr; signaling to directly regulate PAI-1 expression via 2 distinct target sites, an insulin response element (IRE) and a novel forkhead-binding element (FBE), adjacent to a Smad-binding site. We found that in adipocytes and endothelial cells Foxc2 mediates insulin action competing with another Forkhead protein, FOXO1, via the insulin response element, and simultaneously cooperate with the TGF-&bgr;/Smad pathway to transactivate PAI-1. Importantly, Foxc2 haploinsufficiency in mice significantly attenuates TGF-&bgr;1–induced PAI-1 expression in the cardiovascular system and adipose tissue. Taken together, we propose that Foxc2 is a key molecule to regulate PAI-1 gene expression.


Journal of Thrombosis and Haemostasis | 2007

Reactive site-dependent phenotypic alterations in plasminogen activator inhibitor-1 transgenic mice

Mesut Eren; Linda A. Gleaves; James B. Atkinson; Lloyd E. King; Paul Declerck; Douglas E. Vaughan

Summary.  Background: Plasminogen activator inhibitor‐1 (PAI‐1) is the major physiological inhibitor of plasminogen activators (PAs) and plays a role in the regulation of a number of physiological processes including the degradation of extracellular matrix proteins, cell proliferation and migration, and intracellular signaling. Aim: To characterize the effects of durable expression of a stable form of human PAI‐1 and to characterize important structure–function relationships in PAI‐1 in vivo.Methods: We developed transgenic mice lines overexpressing stable variants of human PAI‐1 under the control of the murine preproendothelin‐1 promoter and characterized the phenotypic alterations displayed by transgenic mice. Results: Transgenic mice expressing an active form of human PAI‐1 (PAI‐1‐stab) display complex phenotypic abnormalities including alopecia and hepatosplenomegaly. Reactive site mutant transgenic mice expressing inactive PAI‐1 exhibit complete phenotypic rescue, while transgenic mice expressing PAI‐1 with reduced affinity for vitronectin manifest all of the phenotypic abnormalities present in PAI‐1‐stab transgenic mice. Conclusions: The protease inhibitory activity of PAI‐1 toward PAs and/or other serine proteases is necessary and sufficient to promote complex phenotypic abnormalities and mediates many of the physiological effects of PAI‐1 in vivo.

Collaboration


Dive into the Mesut Eren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Corrie A. Painter

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James B. Atkinson

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge