1
|
Rodriguez M, Zheng Z. Connecting impaired fibrinolysis and dyslipidemia. Res Pract Thromb Haemost 2024; 8:102394. [PMID: 38706781 PMCID: PMC11066549 DOI: 10.1016/j.rpth.2024.102394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/07/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
A State of the Art lecture entitled "Connecting Fibrinolysis and Dyslipidemia" was presented at the International Society on Thrombosis and Haemostasis Congress 2023. Hemostasis balances the consequences of blood clotting and bleeding. This balance relies on the proper formation of blood clots, as well as the breakdown of blood clots. The primary mechanism that breaks down blood clots is fibrinolysis, where the fibrin net becomes lysed and the blood clot dissolves. Dyslipidemia is a condition where blood lipid and lipoprotein levels are abnormal. Here, we review studies that observed connections between impaired fibrinolysis and dyslipidemia. We also summarize the different correlations between thrombosis and dyslipidemia in different racial and ethnic groups. Finally, we summarize relevant and new findings on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress. More studies are needed to investigate the mechanistic connections between impaired fibrinolysis and dyslipidemia and whether these mechanisms differ in racially and ethnically diverse populations.
Collapse
Affiliation(s)
- Maya Rodriguez
- Thrombosis & Hemostasis Program, Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - Ze Zheng
- Thrombosis & Hemostasis Program, Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
2
|
Badran M, Gozal D. PAI-1: A Major Player in the Vascular Dysfunction in Obstructive Sleep Apnea? Int J Mol Sci 2022; 23:5516. [PMID: 35628326 PMCID: PMC9141273 DOI: 10.3390/ijms23105516] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Obstructive sleep apnea is a chronic and prevalent condition that is associated with endothelial dysfunction, atherosclerosis, and imposes excess overall cardiovascular risk and mortality. Despite its high prevalence and the susceptibility of CVD patients to OSA-mediated stressors, OSA is still under-recognized and untreated in cardiovascular practice. Moreover, conventional OSA treatments have yielded either controversial or disappointing results in terms of protection against CVD, prompting the need for the identification of additional mechanisms and associated adjuvant therapies. Plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of tissue-type plasminogen activator (tPA) and urinary-type plasminogen activator (uPA), is a key regulator of fibrinolysis and cell migration. Indeed, elevated PAI-1 expression is associated with major cardiovascular adverse events that have been attributed to its antifibrinolytic activity. However, extensive evidence indicates that PAI-1 can induce endothelial dysfunction and atherosclerosis through complex interactions within the vasculature in an antifibrinolytic-independent matter. Elevated PAI-1 levels have been reported in OSA patients. However, the impact of PAI-1 on OSA-induced CVD has not been addressed to date. Here, we provide a comprehensive review on the mechanisms by which OSA and its most detrimental perturbation, intermittent hypoxia (IH), can enhance the transcription of PAI-1. We also propose causal pathways by which PAI-1 can promote atherosclerosis in OSA, thereby identifying PAI-1 as a potential therapeutic target in OSA-induced CVD.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
3
|
Afroz R, Kumarapperuma H, Nguyen QVN, Mohamed R, Little PJ, Kamato D. Lipopolysaccharide acting via toll-like receptor 4 transactivates the TGF-β receptor in vascular smooth muscle cells. Cell Mol Life Sci 2022; 79:121. [PMID: 35122536 PMCID: PMC8817999 DOI: 10.1007/s00018-022-04159-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLRs) recognise pathogen‑associated molecular patterns, which allow the detection of microbial infection by host cells. Bacterial-derived toxin lipopolysaccharide activates TLR4 and leads to the activation of the Smad2 transcription factor. The phosphorylation of the Smad2 transcription factor is the result of the activation of the transforming growth factor-β receptor 1 (TGFBR1). Therefore, we sought to investigate LPS via TLR4-mediated Smad2 carboxy terminal phosphorylation dependent on the transactivation of the TGFBR1. The in vitro model used human aortic vascular smooth muscle cells to assess the implications of TLR4 transactivation of the TGFBR1 in vascular pathophysiology. We show that LPS-mediated Smad2 carboxy terminal phosphorylation is inhibited in the presence of TGFBR1 inhibitor, SB431542. Treatment with MyD88 and TRIF pathway antagonists does not affect LPS-mediated phosphorylation of Smad2 carboxy terminal; however, LPS-mediated Smad2 phosphorylation was inhibited in the presence of MMP inhibitor, GM6001, and unaffected in the presence of ROCK inhibitor Y27632 or ROS/NOX inhibitor DPI. LPS via transactivation of the TGFBR1 stimulates PAI-1 mRNA expression. TLRs are first in line to respond to exogenous invading substances and endogenous molecules; our findings characterise a novel signalling pathway in the context of cell biology. Identifying TLR transactivation of the TGFBR1 may provide future insight into the detrimental implications of pathogens in pathophysiology.
Collapse
Affiliation(s)
- Rizwana Afroz
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD, 4111, Australia
| | - Hirushi Kumarapperuma
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Quang V N Nguyen
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Raafat Mohamed
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Peter J Little
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-Sen University, Tianhe District, Guangzhou, 510520, China.,Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, 4575, Australia
| | - Danielle Kamato
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
4
|
Li S, Jia H, Liu Z, Wang N, Guo X, Cao M, Fang F, Yang J, Li J, He Q, Guo R, Zhang T, Kang K, Wang Z, Liu S, Cao Y, Jiang X, Ren G, Wang K, Yu B, Xiao W, Li D. Fibroblast growth factor-21 as a novel metabolic factor for regulating thrombotic homeostasis. Sci Rep 2022; 12:400. [PMID: 35013379 PMCID: PMC8748457 DOI: 10.1038/s41598-021-00906-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/12/2021] [Indexed: 11/24/2022] Open
Abstract
Fibroblast growth factor-21 (FGF-21) performs a wide range of biological functions in organisms. Here, we report for the first time that FGF-21 suppresses thrombus formation with no notable risk of bleeding. Prophylactic and therapeutic administration of FGF-21 significantly improved the degree of vascular stenosis and reduced the thrombus area, volume and burden. We determined the antithrombotic mechanism of FGF-21, demonstrating that FGF-21 exhibits an anticoagulant effect by inhibiting the expression and activity of factor VII (FVII). FGF-21 exerts an antiplatelet effect by inhibiting platelet activation. FGF-21 enhances fibrinolysis by promoting tissue plasminogen activator (tPA) expression and activation, while inhibiting plasminogen activator inhibitor 1 (PAI-1) expression and activation. We further found that FGF-21 mediated the expression and activation of tPA and PAI-1 by regulating the ERK1/2 and TGF-β/Smad2 pathways, respectively. In addition, we found that FGF-21 inhibits the expression of inflammatory factors in thrombosis by regulating the NF-κB pathway.
Collapse
Affiliation(s)
- Shuai Li
- College of Life Sciences and Agriculture and Forestry, Qiqihar University, Qiqihar, 161006, People's Republic of China
| | - Haibo Jia
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Zhihang Liu
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Nan Wang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xiaochen Guo
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Muhua Cao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Fang Fang
- Molecular Imaging Research Center, Harbin Medical University, TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, 150028, People's Republic of China
| | - Jiarui Yang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Junyan Li
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qi He
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Rui Guo
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Teng Zhang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Kai Kang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Zongbao Wang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shijie Liu
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yukai Cao
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xinghao Jiang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Guiping Ren
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Kai Wang
- Molecular Imaging Research Center, Harbin Medical University, TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, 150028, People's Republic of China.
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China.
| | - Wei Xiao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical CO. LTD, Lianyungang, 222001, People's Republic of China.
| | - Deshan Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical CO. LTD, Lianyungang, 222001, People's Republic of China.
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
5
|
The Roles and Associated Mechanisms of Adipokines in Development of Metabolic Syndrome. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020334. [PMID: 35056647 PMCID: PMC8781412 DOI: 10.3390/molecules27020334] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is a cluster of metabolic indicators that increase the risk of diabetes and cardiovascular diseases. Visceral obesity and factors derived from altered adipose tissue, adipokines, play critical roles in the development of metabolic syndrome. Although the adipokines leptin and adiponectin improve insulin sensitivity, others contribute to the development of glucose intolerance, including visfatin, fetuin-A, resistin, and plasminogen activator inhibitor-1 (PAI-1). Leptin and adiponectin increase fatty acid oxidation, prevent foam cell formation, and improve lipid metabolism, while visfatin, fetuin-A, PAI-1, and resistin have pro-atherogenic properties. In this review, we briefly summarize the role of various adipokines in the development of metabolic syndrome, focusing on glucose homeostasis and lipid metabolism.
Collapse
|
6
|
Sillen M, Declerck PJ. A Narrative Review on Plasminogen Activator Inhibitor-1 and Its (Patho)Physiological Role: To Target or Not to Target? Int J Mol Sci 2021; 22:ijms22052721. [PMID: 33800359 PMCID: PMC7962805 DOI: 10.3390/ijms22052721] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the main physiological inhibitor of plasminogen activators (PAs) and is therefore an important inhibitor of the plasminogen/plasmin system. Being the fast-acting inhibitor of tissue-type PA (tPA), PAI-1 primarily attenuates fibrinolysis. Through inhibition of urokinase-type PA (uPA) and interaction with biological ligands such as vitronectin and cell-surface receptors, the function of PAI-1 extends to pericellular proteolysis, tissue remodeling and other processes including cell migration. This review aims at providing a general overview of the properties of PAI-1 and the role it plays in many biological processes and touches upon the possible use of PAI-1 inhibitors as therapeutics.
Collapse
|
7
|
Plasminogen activator inhibitor-1 reduces cardiac fibrosis and promotes M2 macrophage polarization in inflammatory cardiomyopathy. Basic Res Cardiol 2021; 116:1. [PMID: 33432417 PMCID: PMC7801308 DOI: 10.1007/s00395-020-00840-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/15/2020] [Indexed: 11/01/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) has a cardioprotective function in mice by repressing cardiac fibrosis through TGF-β and plasminogen-mediated pathways. In addition it is known to be involved in the recruitment and polarization of monocytes/macrophages towards a M2 phenotype in cancer. Here, we investigated the expression of PAI-1 in human dilated cardiomyopathy (DCM) and inflammatory dilated cardiomyopathy (DCMi) and its effect on cardiac fibrosis and macrophage polarization. We retrospectively analyzed endomyocardial biopsies (EMBs) of patients with DCM or DCMi for PAI-1 expression by immunohistochemistry. Furthermore, EMBs were evaluated for the content of fibrotic tissue, number of activated myofibroblasts, TGF-β expression, as well as for M1 and M2 macrophages. Patients with high-grade DCMi (DCMi-high, CD3+ lymphocytes > 30 cells/mm2) had significantly increased PAI-1 levels compared to DCM and low-grade DCMi patients (DCMi-low, CD3+ lymphocytes = 14-30 cells/mm2) (15.5 ± 0.4% vs. 1.0 ± 0.1% and 4.0 ± 0.1%, p ≤ 0.001). Elevated PAI-1 expression in DCMi-high subjects was associated with a diminished degree of cardiac fibrosis, decreased levels of TGF-β and reduced number of myofibroblasts. In addition, DCMi-high patients revealed an increased proportion of non-classical M2 macrophages towards classical M1 macrophages, indicating M2 macrophage-favoring properties of PAI-1 in inflammatory cardiomyopathies. Our findings give evidence that elevated expression of cardiac PAI-1 in subjects with high-grade DCMi suppresses fibrosis by inhibiting TGF-β and myofibroblast activation. Moreover, our data indicate that PAI-1 is involved in the polarization of M2 macrophages in the heart. Thus, PAI-1 could serve as a potential prognostic biomarker and as a possible therapeutic target in inflammatory cardiomyopathies.
Collapse
|
8
|
Sillen M, Declerck PJ. Targeting PAI-1 in Cardiovascular Disease: Structural Insights Into PAI-1 Functionality and Inhibition. Front Cardiovasc Med 2020; 7:622473. [PMID: 33415130 PMCID: PMC7782431 DOI: 10.3389/fcvm.2020.622473] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/03/2020] [Indexed: 01/31/2023] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1), a member of the serine protease inhibitor (serpin) superfamily with antiprotease activity, is the main physiological inhibitor of tissue-type (tPA) and urokinase-type (uPA) plasminogen activators (PAs). Apart from being crucially involved in fibrinolysis and wound healing, PAI-1 plays a pivotal role in various acute and chronic pathophysiological processes, including cardiovascular disease, tissue fibrosis, cancer, and age-related diseases. In the prospect of treating the broad range of PAI-1-related pathologies, many efforts have been devoted to developing PAI-1 inhibitors. The use of these inhibitors, including low molecular weight molecules, peptides, antibodies, and antibody fragments, in various animal disease models has provided ample evidence of their beneficial effect in vivo and moved forward some of these inhibitors in clinical trials. However, none of these inhibitors is currently approved for therapeutic use in humans, mainly due to selectivity and toxicity issues. Furthermore, the conformational plasticity of PAI-1, which is unique among serpins, poses a real challenge in the identification and development of PAI-1 inhibitors. This review will provide an overview of the structural insights into PAI-1 functionality and modulation thereof and will highlight diverse approaches to inhibit PAI-1 activity.
Collapse
Affiliation(s)
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Miller HA, Dean ES, Pletcher SD, Leiser SF. Cell non-autonomous regulation of health and longevity. eLife 2020; 9:62659. [PMID: 33300870 PMCID: PMC7728442 DOI: 10.7554/elife.62659] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/28/2022] Open
Abstract
As the demographics of the modern world skew older, understanding and mitigating the effects of aging is increasingly important within biomedical research. Recent studies in model organisms demonstrate that the aging process is frequently modified by an organism’s ability to perceive and respond to changes in its environment. Many well-studied pathways that influence aging involve sensory cells, frequently neurons, that signal to peripheral tissues and promote survival during the presence of stress. Importantly, this activation of stress response pathways is often sufficient to improve health and longevity even in the absence of stress. Here, we review the current landscape of research highlighting the importance of cell non-autonomous signaling in modulating aging from C. elegans to mammals. We also discuss emerging concepts including retrograde signaling, approaches to mapping these networks, and development of potential therapeutics.
Collapse
Affiliation(s)
- Hillary A Miller
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, United States
| | - Elizabeth S Dean
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States
| | - Scott D Pletcher
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States
| | - Scott F Leiser
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| |
Collapse
|
10
|
Coskun B, Ercan CM, Togrul C, Ozhamam EU, Coskun B, Eren M, Vaughan DE. Effects of lisinopril treatment on the pathophysiology of PCOS and plasminogen activator inhibitor-1 concentrations in rats. Reprod Biomed Online 2020; 42:S1472-6483(20)30516-2. [PMID: 34756399 DOI: 10.1016/j.rbmo.2020.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
RESEARCH QUESTION Angiotensin-converting enzyme inhibition results in a significant reduction in plasma concentrations of plasminogen activator inhibitor-1 (PAI-1). What are the effects of lisinopril treatment on PAI-1 concentrations and the morphology and function of the ovaries in the letrozole-induced polycystic ovary syndrome (PCOS) rat model? DESIGN This prospective randomized controlled animal study involved female Wistar albino rats. Twelve rats were assigned as controls (group I). In the study group (n = 48), letrozole (an aromatase inhibitor) was administered for PCOS modelling for 9 weeks. After confirming disrupted oestrous cycles, the study group was randomized into two groups: group II (n = 24; letrozole only) and group III (n = 24; letrozole + lisinopril 15 mg/kg per day). After 12 weeks, each group was divided randomly into two. Biochemical, histopathological and immunohistochemical analyses was performed in subgroups designated A, and fertilization rates were studied in subgroups designated B. RESULTS Lisinopril treatment reduced the weight and area of the ovaries, the number and wall thickness of cystic follicles, and serum concentrations of LH and testosterone, relative to group II (P < 0.001). Circulating PAI-1 concentrations were significantly different among three groups (7.7 ± 0.9 ng/ml, 9.8 ± 0.7 ng/ml and 8.6 ± 0.7 ng/ml for groups IA, IIA and IIIA; P < 0.001). Pregnancy rates were 100%, 0% and 16.7% in groups IB, IIB and IIIB. CONCLUSIONS In the letrozole-induced rodent PCOS model, lisinopril modifies the action of letrozole, possibly by inhibition of systemic and ovarian production of PAI-1. The use of PAI-1 inhibitors deserves further investigation in understanding the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Bugra Coskun
- Department of Obstetrics and Gynecology, Yuksek Ihtisas University, Ankara, Turkey
| | - Cihangir M Ercan
- Department of Obstetrics and Gynecology, University of Health Sciences, Gulhane Education and Research Hospital, Ankara, Turkey.
| | - Cihan Togrul
- Department of Obstetrics and Gynecology, Hitit University, Çorum, Turkey
| | - Esra U Ozhamam
- Department of Pathology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Bora Coskun
- Department of Obstetrics and Gynecology, Yuksek Ihtisas University, Ankara, Turkey
| | - Mesut Eren
- Department of Medicine, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Douglas E Vaughan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| |
Collapse
|
11
|
Slack MA, Gordon SM. Protease Activity in Vascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:e210-e218. [PMID: 31553665 PMCID: PMC6764587 DOI: 10.1161/atvbaha.119.312413] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Megan A. Slack
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
12
|
Graßler A, Bauernschmitt R, Guthoff I, Kunert A, Hoenicka M, Albrecht G, Liebold A. Effects of pulsatile minimal invasive extracorporeal circulation on fibrinolysis and organ protection in adult cardiac surgery-a prospective randomized trial. J Thorac Dis 2019; 11:S1453-S1463. [PMID: 31293794 DOI: 10.21037/jtd.2019.02.66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Minimal invasive extracorporeal circulation (MiECC) reduces the impact of cardiopulmonary bypass during cardiac surgery on inflammation and hemostasis. Pulsatile perfusion may enhance organ perfusion and help to prevent renal and neuronal damage. The present study investigated the impact of pulsatile MiECC in low-risk coronary artery bypass grafting (CABG) patients. Methods CABG patients were prospectively randomized for non-pulsatile (np: n=19) and pulsatile (p: n=21) MiECC. Blood and urine samples were collected at several time points until 72 h post-operative and analyzed for biochemical markers of fibrinolytic capacity, renal damage, and neuronal damage. Results Although intraoperative tissue plasminogen activator (tPA) levels tended to be higher in the p group, none of the fibrinolysis markers including plasminogen activator inhibitor (PAI-1) and the PAI-1/tPA ratio were significantly affected by pulsation. Hemolysis and markers of renal and neuronal damage were comparable between groups. Intraoperative urinary excretion [np: 400 mL (355 to 680) vs. p: 530 mL (360 to 900)] and cumulative 24 h volume intake [np: 7,090 mL (5,492 to 7,544) vs. p: 7,155 mL (6,682 to 8,710)] were increased by pulsation whereas blood losses up to 12 h post-operative [np: 365 mL (270 to 515) vs. p: 310 mL (225 to 470)] and up to 24 h post-operative [np: 760 mL (555 to 870) vs. p: 520 mL (460 to 670)] were attenuated. Conclusions The present study did not find evidence for a beneficial effect of pulsation on markers of fibrinolysis, renal damage, and neuronal damage. However, pulsatile perfusion increased intraoperative urinary secretion and reduced post-operative blood losses.
Collapse
Affiliation(s)
- Angelika Graßler
- Department of Cardiothoracic and Vascular Surgery, University of Ulm Medical Center, Ulm, Germany.,RoMed Clinical Center Rosenheim, Department of Internal Medicine III, Rosenheim, Germany
| | - Robert Bauernschmitt
- Department of Cardiothoracic and Vascular Surgery, University of Ulm Medical Center, Ulm, Germany.,MediClin Herzzentrum, Department of Cardiothoracic and Vascular Surgery, Lahr, Germany
| | - Irene Guthoff
- Department of Cardiothoracic and Vascular Surgery, University of Ulm Medical Center, Ulm, Germany
| | - Andreas Kunert
- Department of Cardiothoracic and Vascular Surgery, University of Ulm Medical Center, Ulm, Germany
| | - Markus Hoenicka
- Department of Cardiothoracic and Vascular Surgery, University of Ulm Medical Center, Ulm, Germany
| | - Günter Albrecht
- Department of Cardiothoracic and Vascular Surgery, University of Ulm Medical Center, Ulm, Germany
| | - Andreas Liebold
- Department of Cardiothoracic and Vascular Surgery, University of Ulm Medical Center, Ulm, Germany
| |
Collapse
|
13
|
The endothelial tumor suppressor p53 is essential for venous thrombus formation in aged mice. Blood Adv 2019; 2:1300-1314. [PMID: 29891592 DOI: 10.1182/bloodadvances.2017014050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/11/2018] [Indexed: 12/24/2022] Open
Abstract
Venous thromboembolism (VTE) is a leading cause of morbidity and mortality in elderly people. Increased expression of tumor suppressor protein 53 (p53) has been implicated in vascular senescence. Here, we examined the importance of endothelial p53 for venous thrombosis and whether endothelial senescence and p53 overexpression are involved in the exponential increase of VTE with age. Mice with conditional, endothelial-specific deletion of p53 (End.p53-KO) and their wild-type littermates (End.p53-WT) underwent subtotal inferior vena cava (IVC) ligation to induce venous thrombosis. IVC ligation in aged (12-month-old) End.p53-WT mice resulted in higher rates of thrombus formation and greater mean thrombus size vs adult (12-week-old) End.p53-WT mice, whereas aged End.p53-KO mice were protected from vein thrombosis. Analysis of primary endothelial cells from aged mice or human vein endothelial cells after induction of replicative senescence revealed significantly increased early growth response gene-1 (Egr1) and heparanase expression, and plasma factor Xa levels were elevated in aged End.p53-WT, but not in End.p53-KO mice. Increased endothelial Egr1 and heparanase expression also was observed after doxorubicin-induced p53 overexpression, whereas p53 inhibition using pifithrin-α reduced tissue factor (TF) expression. Importantly, inhibition of heparanase activity using TF pathway inhibitor-2 (TFPI2) peptides prevented the enhanced venous thrombus formation in aged mice and restored it to the thrombotic phenotype of adult mice. Our findings suggest that p53 accumulation and heparanase overexpression in senescent endothelial cells are critically involved in mediating the increased risk of venous thrombosis with age and that heparanase antagonization may be explored as strategy to ameliorate the prothrombotic endothelial phenotype with age.
Collapse
|
14
|
Mutch NJ. Regulation of Fibrinolysis by Platelets. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
15
|
Jung RG, Simard T, Labinaz A, Ramirez FD, Di Santo P, Motazedian P, Rochman R, Gaudet C, Faraz MA, Beanlands RS, Hibbert B. Role of plasminogen activator inhibitor-1 in coronary pathophysiology. Thromb Res 2018; 164:54-62. [DOI: 10.1016/j.thromres.2018.02.135] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/03/2018] [Accepted: 02/15/2018] [Indexed: 01/13/2023]
|
16
|
Bao CX, Zhang DX, Wang NN, Zhu XK, Zhao Q, Sun XL. MicroRNA-335-5p suppresses lower extremity deep venous thrombosis by targeted inhibition of PAI-1 via the TLR4 signalingpathway. J Cell Biochem 2018; 119:4692-4710. [PMID: 29278662 DOI: 10.1002/jcb.26647] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022]
Abstract
This study aims to investigate the effects of microRNA-335-5p (miR-335-5p) on lower-extremity deep vein thrombosis (LEDVT) by targeting PAI-1 through the TLR4 signaling pathway in rat models. siRNA, mimic, and inhibitor were used for transfection. The miR-335-5p expression was detected by in situ hybridization. CCK-8 assay and flow cytometry were adopted to detect proliferation, cell cycle, and apoptosis, respectively. Scratch test and Matrigel-based tube formation assay were used to detect the effect of miR-335-5p on cell migration ability and tube formation ability. A miR-335-5p lentivirus plasmid was constructed and injected into LEDVT rats. The length and weight of thrombus were measured, changes of thrombus recanalization were observed by CD34 immunohistochemistry, and levels of PAI-1 and inflammatory factors in femoral vein blood were detected by ELISA. LEDVT rats showed a higher AOD value of PAI-1, higher expression of PAI-1, NF-κB, Rac1, IL-1β, and TLR4 and a lower miR-335-5p expression. PAI-1 and miR-335-5p were negatively correlated. Compared to the blank and siRNA-NC groups, the miR-335-5p mimic and siRNA-PAI-1 groups showed declined expression of PAI-1, TLR4, NF-κB, Rac1, and IL-1β, increased proliferation and tube formation abilities, less cells in G0/G1 phase, and decreased apoptosis, decreased length and weight of thrombus, organized thrombus, increased new blood vessels, and decreased levels of PAI-1, IL-1, IL-6, and Tnf-a. miR-335-5p may suppress the occurrence and development of LEDVT in rats by repressing the activation of the TLR4 signaling pathway by targeted inhibition of PAI-1.
Collapse
Affiliation(s)
- Cui-Xia Bao
- Clinical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P. R. China
| | - Dong-Xia Zhang
- Department of Cardiovascular Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P. R. China
| | - Na-Na Wang
- Clinical Laboratory, Yantai Yeda Hospital, Yantai, P. R. China
| | - Xiang-Kui Zhu
- Department of Radiology, Yantai Stomatological Hospital, Yantai, P. R. China
| | - Qi Zhao
- Clinical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P. R. China
| | - Xiao-Lei Sun
- Department of Cardiology, Yantai Hospital of Traditional Chinese Medicine, Yantai, P. R. China
| |
Collapse
|
17
|
Praetner M, Zuchtriegel G, Holzer M, Uhl B, Schaubächer J, Mittmann L, Fabritius M, Fürst R, Zahler S, Funken D, Lerchenberger M, Khandoga A, Kanse S, Lauber K, Krombach F, Reichel CA. Plasminogen Activator Inhibitor-1 Promotes Neutrophil Infiltration and Tissue Injury on Ischemia-Reperfusion. Arterioscler Thromb Vasc Biol 2018; 38:829-842. [PMID: 29371242 DOI: 10.1161/atvbaha.117.309760] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/15/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Ischemia-reperfusion (I/R) injury significantly contributes to organ dysfunction and failure after myocardial infarction, stroke, and transplantation. In addition to its established role in the fibrinolytic system, plasminogen activator inhibitor-1 has recently been implicated in the pathogenesis of I/R injury. The underlying mechanisms remain largely obscure. APPROACH AND RESULTS Using different in vivo microscopy techniques as well as ex vivo analyses and in vitro assays, we identified that plasminogen activator inhibitor-1 rapidly accumulates on microvascular endothelial cells on I/R enabling this protease inhibitor to exhibit previously unrecognized functional properties by inducing an increase in the affinity of β2 integrins in intravascularly rolling neutrophils. These events are mediated through low-density lipoprotein receptor-related protein-1 and mitogen-activated protein kinase-dependent signaling pathways that initiate intravascular adherence of these immune cells to the microvascular endothelium. Subsequent to this process, extravasating neutrophils disrupt endothelial junctions and promote the postischemic microvascular leakage. Conversely, deficiency of plasminogen activator inhibitor-1 effectively reversed leukocyte infiltration, microvascular dysfunction, and tissue injury on experimental I/R without exhibiting side effects on microvascular hemostasis. CONCLUSIONS Our experimental data provide novel insights into the nonfibrinolytic properties of the fibrinolytic system and emphasize plasminogen activator inhibitor-1 as a promising target for the prevention and treatment of I/R injury.
Collapse
Affiliation(s)
- Marc Praetner
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Gabriele Zuchtriegel
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Martin Holzer
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Bernd Uhl
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Johanna Schaubächer
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Laura Mittmann
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Matthias Fabritius
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Robert Fürst
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Stefan Zahler
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Dominik Funken
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Maximilian Lerchenberger
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Andrej Khandoga
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Sandip Kanse
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Kirsten Lauber
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Fritz Krombach
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.)
| | - Christoph A Reichel
- From the Walter Brendel Centre of Experimental Medicine (M.P., G.Z., M.H., B.U., J.S., L.M., M.F., D.F., M.L., A.K., F.K., C.A.R.), Department of Otorhinolaryngology (G.Z., M.H., B.U., C.A.R.), Head and Neck Surgery (M.P.), Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research (S.Z.), Department of Surgery (D.F., M.L., A.K.), and Department of Radiation Oncology (K.L.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany (M.P); Institute of Pharmaceutical Biology, Goethe University Frankfurt, Germany (R.F.); and Institute of Basic Medical Sciences, University of Oslo, Norway (S.K.).
| |
Collapse
|
18
|
Peng S, Xue G, Gong L, Fang C, Chen J, Yuan C, Chen Z, Yao L, Furie B, Huang M. A long-acting PAI-1 inhibitor reduces thrombus formation. Thromb Haemost 2017; 117:1338-1347. [DOI: 10.1160/th16-11-0891] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/25/2017] [Indexed: 01/05/2023]
Abstract
SummaryPlasminogen activator inhibitor 1 (PAI-1) is the main inhibitor of tissue-type and urokinase-type plasminogen activators (t/uPA) and plays an important role in fibrinolysis. Inhibition of PAI-1 activity prevents thrombosis and accelerates fibrinolysis, indicating that PAI-1 inhibitors may be used as effective antithrombotic agents. We previously designed a PAI-1 inhibitor (PAItrap) which is a variant of inactivated urokinase protease domain. In the present study, we fused PAItrap with human serum albumin (HSA) to develop a long-acting PAI-1 inhibitor. Unfortunately, the fusion protein PAItrap-HSA lost some potency compared to PAItrap (33 nM vs 10 nM). Guided by computational method, we carried out further optimisation to enhance inhibitory potency for PAI-1. The new PAItrap, denominated PAItrap(H37R)-HSA, which was the H37R variant of PAItrap fused to HSA, gave a six-fold improvement of IC50 (5 nM) for human active PAI-1 compared to PAItrap-HSA, and showed much longer plasma half-life (200-fold) compared to PAItrap. We further demonstrated that the PAItrap(H37R)-HSA inhibited exogenous or endogenous PAI-1 to promote fibrinolysis in fibrin-clot lysis assay. PAItrap(H37R)-HSA inhibits murine PAI-1 with IC50 value of 12 nM, allowing the inhibitor to be evaluated in murine models. Using an intravital microscopy, we demonstrated that PAItrap(H37R)-HSA blocks thrombus formation and platelet accumulation in vivo in a laser-induced vascular injury mouse model. Additionally, mouse tail bleeding assay showed that PAItrap(H37R)-HSA did not affect the global haemostasis. These results suggest that PAItrap(H37R)-HSA have the potential benefit to prevent thrombosis and accelerates fibrinolysis.
Collapse
|
19
|
Eren M, Place AT, Thomas PM, Flevaris P, Miyata T, Vaughan DE. PAI-1 is a critical regulator of FGF23 homeostasis. SCIENCE ADVANCES 2017; 3:e1603259. [PMID: 28924605 PMCID: PMC5597312 DOI: 10.1126/sciadv.1603259] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 08/11/2017] [Indexed: 05/29/2023]
Abstract
Elevated levels of fibroblast growth factor 23 (FGF23), a bone-derived phosphaturic hormone, are associated with a number of pathologic conditions including chronic kidney disease, cardiac hypertrophy, and congestive heart failure. Currently, there are no specific treatments available to lower plasma FGF23 levels. We have recently reported that genetic plasminogen activator inhibitor-1 (PAI-1) deficiency provided a significant reduction in circulating FGF23 levels while simultaneously prolonging the life span of Klotho-deficient mice. We extend our investigations into the effect of PAI-1 on FGF23 homeostasis. Transgenic overexpression of PAI-1 resulted in threefold increase in FGF23 levels compared to wild-type littermates. Moreover, pharmacological modulation of PAI-1 activity with the small-molecule PAI-1 antagonist TM5441 significantly reduced FGF23 levels in PAI-1 transgenic and Klotho-deficient mice. In addition, TM5441 treatment or PAI-1 deficiency significantly accelerated the clearance of endogenous FGF23 and recombinant human FGF23 from circulation in mice with acute kidney injury. On the basis of these observations, we studied the effects of plasminogen activators (PAs), tissue-type PA (tPA) and urokinase-type PA (uPA), on FGF23. We demonstrate that both PAs directly cleave FGF23; however, it is not known whether the PA-generated FGF23 peptides retain or acquire functions that affect binding and/or signaling properties of intact FGF23. PAI-1 inhibits the PA-dependent cleavage of FGF23, and TM5441 inhibition of PAI-1 restores the proteolysis of FGF23. Furthermore, top-down proteomic analysis indicates that tPA cleaves FGF23 at multiple arginines including the proconvertase sensitive site R176. In summary, our results indicate that PAI-1 prevents the PA-driven proteolysis of FGF23 and PAI-1 inhibition provides a novel therapeutic approach to prevent the pathologic consequences of increased FGF23.
Collapse
Affiliation(s)
- Mesut Eren
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aaron T. Place
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul M. Thomas
- Proteomics Center of Excellence, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Panagiotis Flevaris
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Douglas E. Vaughan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
20
|
Peng H, Yeh F, Lin J, Best LG, Cole SA, Lee ET, Howard BV, Zhao J. Plasminogen activator inhibitor-1 is associated with leukocyte telomere length in American Indians: findings from the Strong Heart Family Study. J Thromb Haemost 2017; 15:1078-1085. [PMID: 28378522 PMCID: PMC5500969 DOI: 10.1111/jth.13689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Indexed: 01/28/2023]
Abstract
Essentials Plasminogen activator inhibitor-1 (PAI-1) advanced cellular senescence in experiment studies. No population study exists on the association between PAI-1 and biological aging in American Indians. We found cross-sectional and longitudinal associations between higher PAI-1 and shorter telomere length. Our findings suggest a pathway linking PAI-1 with biological aging beyond metabolic factors. SUMMARY Background Plasminogen activator inhibitor-1 (PAI-1) promotes cellular aging both in vitro and in vivo. Telomere length is a marker of biological aging. Objectives To examine the cross-sectional and longitudinal associations between plasma PAI-1 and leukocyte telomere length in a large-scale epidemiological study of American Indians. Methods We measured leukocyte telomere length (LTL) and plasma PAI-1 in 2560 American Indians who were free of overt cardiovascular disease (CVD) and participated in the Strong Heart Family Study (SHFS) clinical examination in 2001-2003. LTL and PAI-1 were repeatedly measured in 475 participants who attended SHFS clinical visits in both 2001-2003 and 1998-1999. A generalized estimating equation model was used to examine the cross-sectional and longitudinal associations between PAI-1 and LTL, adjusting for known risk factors. Results A higher level of plasma PAI-1 was negatively associated with shorter age-adjusted LTL (β = -0.023; 95% CI, -0.034 to -0.013). This association was attenuated (β = -0.015; 95% CI, -0.029 to -0.002) after adjustments for demographics, study site, lifestyle (smoking, drinking and physical activity) and metabolic factors (obesity, blood pressure, fasting glucose, insulin, lipids and kidney function). Further adjustment for hsCRP did not change this association (β = -0.015; 95% CI, -0.029 to -0.001). Longitudinal analysis revealed that change in plasma PAI-1 was also inversely associated with change in LTL after adjusting for demographics, follow-up years, lifestyle factors, changes in metabolic factors, baseline levels of PAI-1 and LTL (β = -0.0005; 95% CI, -0.0009 to -0.0001). Conclusions A higher level of plasma PAI-1 was associated with shorter LTL in American Indians. This finding may suggest a potential role of PAI-1 in biological aging among American Indians.
Collapse
Affiliation(s)
- Hao Peng
- Department of Epidemiology, College of Public Health and Health Professions, College of Medicine, University of Florida, Gainesville, FL
| | - Fawn Yeh
- Center for American Indian Health Research, University of Oklahoma Health Science Center, Oklahoma City, OK
| | - Jue Lin
- Department of Biochemistry and Biophysics at the University of California, San Francisco
| | - Lyle G. Best
- Missouri Breaks Industries Research Inc, Eagle Butte, SD
| | - Shelley A. Cole
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX
| | - Elisa T. Lee
- Center for American Indian Health Research, University of Oklahoma Health Science Center, Oklahoma City, OK
| | | | - Jinying Zhao
- Department of Epidemiology, College of Public Health and Health Professions, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
21
|
Vaughan DE, Rai R, Khan SS, Eren M, Ghosh AK. Plasminogen Activator Inhibitor-1 Is a Marker and a Mediator of Senescence. Arterioscler Thromb Vasc Biol 2017; 37:1446-1452. [PMID: 28572158 DOI: 10.1161/atvbaha.117.309451] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/15/2017] [Indexed: 01/23/2023]
Abstract
PAI-1 (plasminogen activator inhibitor-1) is a member of the evolutionarily conserved serine protease inhibitor family and a potent and rapid-acting inhibitor of both of the mammalian plasminogen activators. Organismal homeostasis requires physiological levels of endogenous PAI-1, and increased PAI-1 production guides the onset and progression of numerous human diseases and contributes to the multimorbidity of aging. Both chronological and stress-induced accelerated aging are associated with cellular senescence and accompanied by marked increases in PAI-1 expression in tissues. Recent studies suggest that PAI-1 is not only a marker but also a key mediator of cellular senescence and organismal aging. Here, we review the significance of PAI-1 as a bonafide marker, as well as a critical mediator, of cellular senescence associated with aging and aging-related pathologies.
Collapse
Affiliation(s)
- Douglas E Vaughan
- From the Department of Medicine, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL.
| | - Rahul Rai
- From the Department of Medicine, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Sadiya S Khan
- From the Department of Medicine, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Mesut Eren
- From the Department of Medicine, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Asish K Ghosh
- From the Department of Medicine, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
22
|
Kaji H. Adipose Tissue‐Derived Plasminogen Activator Inhibitor‐1 Function and Regulation. Compr Physiol 2016; 6:1873-1896. [DOI: 10.1002/cphy.c160004] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Rather RA, Dhawan V. Genetic markers: Potential candidates for cardiovascular disease. Int J Cardiol 2016; 220:914-23. [PMID: 27416153 DOI: 10.1016/j.ijcard.2016.06.251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 06/22/2016] [Accepted: 06/26/2016] [Indexed: 02/07/2023]
Abstract
The effective prevention of cardiovascular disease depends upon the ability to recognize the high-risk individuals at an early stage of the disease or long before the development of adverse events. Evolving technologies in the fields of proteomics, metabolomics, and genomics have played a significant role in the discovery of cardiovascular biomarkers, but so far these methods have achieved the modest success. Hence, there is a crucial need for more reliable, suitable, and lasting diagnostic and therapeutic markers to screen the disease well in time to start the clinical aid to the patients. Gene polymorphisms associated with the cardiovascular disease play a decisive role in the disease onset. Therefore, the genetic marker evaluation to classify high-risk patients from low-risk patients trends an effective approach to patient management and care. Currently, there are no genetic markers available for extensive adoption as risk factors for coronary vascular disease, yet, there are numerous promising, biologically acceptable candidates. Many of these gene biomarkers, alone or in combination, can play an essential role in the prediction of cardiovascular risk. The present review highlights some putative emerging genetic biomarkers that could facilitate more authentic and fast diagnosis of CVD. This review also briefly describes few technological approaches employed in the biomarker search.
Collapse
Affiliation(s)
- Riyaz Ahmad Rather
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
24
|
Shimizu T, Uematsu M, Yoshizaki T, Obata JE, Nakamura T, Fujioka D, Watanabe K, Watanabe Y, Kugiyama K. Myocardial Production of Plasminogen Activator Inhibitor-1 is Associated with Coronary Endothelial and Ventricular Dysfunction after Acute Myocardial Infarction. J Atheroscler Thromb 2015; 23:557-66. [PMID: 26632165 DOI: 10.5551/jat.32300] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Although plasminogen activator inhibitor-1 (PAI-1) is abundantly expressed in infarcted myocardium, the pathogenic role of myocardial PAI-1 remains unknown. This study examined whether PAI-1 in the infarcted lesion contributes to coronary endothelial dysfunction and left ventricular (LV) dysfunction in patients with acute myocardial infarction (AMI). METHODS Plasma levels of PAI-1 activity and tissue-plasminogen activator (tPA) antigen were measured 2 weeks and 6 months after MI by ELISA in plasma obtained from the aortic root (AO) and anterior interventricular vein (AIV) in 28 patients with a first AMI due to occlusion of the left anterior descending coronary artery (LAD). Coronary blood flow responses in LAD to intracoronary infusion of acetylcholine (ACh) and left ventriculography were measured at the same time points: 2 weeks and 6 months after MI. RESULTS The trans-myocardial gradient of PAI-1 from AO to AIV, reflecting production/release of PAI-1 in the infarcted lesion, was inversely correlated with the coronary blood flow response to ACh 6 months after MI (r=-0.43, p=0.02) and with the percentage change in LV regional motion in the LAD territory from 2 weeks to 6 months after MI (r=-0.38, p=0.04). The trans-myocardial gradient of tPA level showed no significant correlations. CONCLUSIONS PAI-1 produced in the infarcted myocardium and released into the coronary circulation is associated with endothelial dysfunction in resistance vessels of the infarct-related coronary arteries and with progressive dysfunction of the infarcted region of the left ventricle in AMI survivors.
Collapse
Affiliation(s)
- Takuya Shimizu
- University of Yamanashi, Department of Internal Medicine II
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Stanley FM, Linder KM, Cardozo TJ. Statins Increase Plasminogen Activator Inhibitor Type 1 Gene Transcription through a Pregnane X Receptor Regulated Element. PLoS One 2015; 10:e0138097. [PMID: 26379245 PMCID: PMC4574702 DOI: 10.1371/journal.pone.0138097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022] Open
Abstract
Plasminogen activator inhibitor type 1 (PAI-1) is a multifunctional protein that has important roles in inflammation and wound healing. Its aberrant regulation may contribute to many disease processes such as heart disease. The PAI-1 promoter is responsive to multiple inputs including cytokines, growth factors, steroids and oxidative stress. The statin drugs, atorvastatin, mevastatin and rosuvastatin, increased basal and stimulated expression of the PAI-1 promoter 3-fold. A statin-responsive, nuclear hormone response element was previously identified in the PAI-1 promoter, but it was incompletely characterized. We characterized this direct repeat (DR) of AGGTCA with a 3-nucleotide spacer at -269/-255 using deletion and directed mutagenesis. Deletion or mutation of this element increased basal transcription from the promoter suggesting that it repressed PAI-1 transcription in the unliganded state. The half-site spacing and the ligand specificity suggested that this might be a pregnane X receptor (PXR) responsive element. Computational molecular docking showed that atorvastatin, mevastatin and rosuvastatin were structurally compatible with the PXR ligand-binding pocket in its agonist conformation. Experiments with Gal4 DNA binding domain fusion proteins showed that Gal4-PXR was activated by statins while other DR + 3 binding nuclear receptor fusions were not. Overexpression of PXR further enhanced PAI-1 transcription in response to statins. Finally, ChIP experiments using Halo-tagged PXR and RXR demonstrated that both components of the PXR-RXR heterodimer bound to this region of the PAI-1 promoter.
Collapse
Affiliation(s)
- Frederick M. Stanley
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York, United States of America
| | - Kathryn M. Linder
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Timothy J. Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York, United States of America
| |
Collapse
|
26
|
Agirbasli M, Eren M, Yasar S, Delil K, Goktay F, Oner ET, Vaughan DE. Functionally stable plasminogen activator inhibitor-1 in a family with cardiovascular disease and vitiligo. J Thromb Thrombolysis 2015; 38:50-6. [PMID: 24197654 DOI: 10.1007/s11239-013-1021-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vitiligo is a common skin condition with a complex pathophysiology characterized by the lack of pigmentation due to melanocyte degeneration. In this study, we investigated PAI-1 antigen (Ag) and activity levels in a 34 year old male with extensive vascular disease, alopecia areata and vitiligo. Fasting PAI-1 Ag and activity levels were measured at 9 a.m. in the subject and family members. Both PAI-1 Ag (67 ± 38 vs. 18.6 ± 6.5 ng/ml, P < 0.001) and specific activity (15.8 ± 10.0 vs. 7.6 ± 6.0 IU/pmol, P < 0.04) levels of PAI-1 were moderately elevated in subjects compared to the controls. PAI-1 kinetic studies demonstrated a markedly enhanced stability of plasma PAI-1 activity in the family members. Specific activity at 16 h was significantly higher than expected activity levels (0.078 ± 0.072 vs. 0.001 ± 0.001 IU/ng/ml, P < 0.001). While the exact mechanism of increased stability of PAI-1 activity in vitiligo is not known, it is likely due to post-translational modifications or increased binding affinity for a stabilizing cofactor. In conclusion, enhanced stability of PAI-1 may contribute to the pathophysiology of vascular disease and associated melanocyte degeneration. Systemic or local treatment with PAI-1 inhibitors may offer a potential treatment alternative to the near orphan status for vitiligo drug development.
Collapse
Affiliation(s)
- Mehmet Agirbasli
- Department of Cardiology, Faculty of Medicine, Marmara University Hospital, Yeşilbahar Sok 68/14 Palmiye Apt., Goztepe Kadikoy, 34726, Istanbul, Turkey,
| | | | | | | | | | | | | |
Collapse
|
27
|
Engoren M, Schwann TA, Jewell E, Neill S, Benedict P, Likosky DS, Habib RH. Is Transfusion Associated With Graft Occlusion After Cardiac Operations? Ann Thorac Surg 2015; 99:502-8. [DOI: 10.1016/j.athoracsur.2014.09.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/10/2014] [Accepted: 09/19/2014] [Indexed: 02/01/2023]
|
28
|
Functional stability of plasminogen activator inhibitor-1. ScientificWorldJournal 2014; 2014:858293. [PMID: 25386620 PMCID: PMC4214104 DOI: 10.1155/2014/858293] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/17/2014] [Indexed: 12/23/2022] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the main inhibitor of plasminogen activators, such as tissue-type plasminogen activator (t-PA) and urokinase-type plasminogen activator (u-PA), and a major regulator of the fibrinolytic system. PAI-1 plays a pivotal role in acute thrombotic events such as deep vein thrombosis (DVT) and myocardial infarction (MI). The biological effects of PAI-1 extend far beyond thrombosis including its critical role in fibrotic disorders, atherosclerosis, renal and pulmonary fibrosis, type-2 diabetes, and cancer. The conversion of PAI-1 from the active to the latent conformation appears to be unique among serpins in that it occurs spontaneously at a relatively rapid rate. Latency transition is believed to represent a regulatory mechanism, reducing the risk of thrombosis from a prolonged antifibrinolytic action of PAI-1. Thus, relying solely on plasma concentrations of PAI-1 without assessing its function may be misleading in interpreting the role of PAI-1 in many complex diseases. Environmental conditions, interaction with other proteins, mutations, and glycosylation are the main factors that have a significant impact on the stability of the PAI-1 structure. This review provides an overview on the current knowledge on PAI-1 especially importance of PAI-1 level and stability and highlights the potential use of PAI-1 inhibitors for treating cardiovascular disease.
Collapse
|
29
|
Abstract
The ever growing prevalence of childhood obesity is being accompanied by an increase in the pediatric population of diseases once believed to be exclusive of the adulthood such as the metabolic syndrome (MS). The MS has been defined as the link between insulin resistance, hypertension, dyslipidemia, impaired glucose tolerance, and other metabolic abnormalities associated with an increased risk of atherosclerotic cardiovascular diseases in adults. In this review, we will discuss the peculiar aspects of the pediatric MS and the role of novel molecules and biomarkers in its pathogenesis.
Collapse
|
30
|
Eren M, Boe AE, Klyachko EA, Vaughan DE. Role of plasminogen activator inhibitor-1 in senescence and aging. Semin Thromb Hemost 2014; 40:645-51. [PMID: 25173500 DOI: 10.1055/s-0034-1387883] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The average age of the US population continues to increase. Age is the most important determinant of disease and disability in humans, but the fundamental mechanisms of aging remain largely unknown. Many age-related diseases are associated with an impaired fibrinolytic system. Elevated plasminogen activator inhibitor-1 (PAI-1) levels are reported in age-associated clinical conditions including cardiovascular diseases, type 2 diabetes, obesity and inflammation. PAI-1 levels are also elevated in animal models of aging. While the association of PAI-1 with physiological aging is well documented, it is only recently that its critical role in the regulation of aging and senescence has become evident. PAI-1 is synthesized and secreted in senescent cells and contributes directly to the development of senescence by acting downstream of p53 and upstream of insulin-like growth factor binding protein-3. Pharmacologic inhibition or genetic deficiency of PAI-1 was shown to be protective against senescence and the aging-like phenotypes in kl/kl and N(ω)-nitro-l-arginine methyl ester-treated wild-type mice. Further investigation into PAI-1's role in senescence and aging will likely contribute to the prevention and treatment of aging-related pathologies.
Collapse
Affiliation(s)
- Mesut Eren
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amanda E Boe
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ekaterina A Klyachko
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Douglas E Vaughan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
31
|
PAI-1-regulated extracellular proteolysis governs senescence and survival in Klotho mice. Proc Natl Acad Sci U S A 2014; 111:7090-5. [PMID: 24778222 DOI: 10.1073/pnas.1321942111] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
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.
Collapse
|
32
|
Mohebali D, Kaplan D, Carlisle M, Supiano MA, Rondina MT. Alterations in platelet function during aging: clinical correlations with thromboinflammatory disease in older adults. J Am Geriatr Soc 2014; 62:529-35. [PMID: 24512275 PMCID: PMC3959242 DOI: 10.1111/jgs.12700] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Platelets have a dynamic functional repertoire that mediates hemostatic and inflammatory responses. Many of these functions are altered in older adults, promoting a prothrombotic, proinflammatory milieu and contributing to risk of adverse clinical events. Drawing primarily from human studies, this review summarizes important aspects of aging-related changes in platelets. The relationship between altered platelet functions and thrombotic and inflammatory disorders in older adults is highlighted. Established and developing antiplatelet therapies for the treatment of thrombotic and inflammatory disorders are also discussed in light of these data.
Collapse
Affiliation(s)
- Donya Mohebali
- Divisions of General Internal Medicine, Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, 84132
| | - David Kaplan
- Divisions of General Internal Medicine, Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, 84132
| | - McKenzie Carlisle
- Program in Molecular Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, 84132
| | - Mark A. Supiano
- Divisions of Geriatrics, Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, 84132
| | - Matthew T. Rondina
- Divisions of General Internal Medicine, Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, 84132
- Program in Molecular Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, 84132
| |
Collapse
|
33
|
Davidson L, Wilcox J, Kim D, Benton S, Fredi J, Vaughan D. Clinical features of precocious acute coronary syndrome. Am J Med 2014; 127:140-4. [PMID: 24332726 DOI: 10.1016/j.amjmed.2013.09.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 09/30/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Acute coronary syndrome due to acute plaque rupture has been well described and is associated with established risk factors, including hypertension, diabetes mellitus, hyperlipidemia, and smoking. The prevalence of these risk factors in very young patients (aged ≤35 years) is not well known, and they may have other nontraditional risk factors. We hypothesized that acute coronary syndrome in very young patients may represent a thrombotic event independent of underlying atherosclerotic disease. METHODS We performed a dual-institution, retrospective study of consecutive patients aged ≤35 years who presented with acute coronary syndrome and underwent coronary angiography from January 2000 to December 2011. Standard demographics, risk factors, and detailed angiographic information were obtained. RESULTS A total of 124 patients met inclusion criteria. The mean age was 31 ± 4 years for both sexes. Approximately half (49%) of the patients were obese (body mass index ≥30 kg/m(2)); 90% of patients had at least 1 traditional risk factor, most commonly hyperlipidemia (63%) and smoking (60%); 52% of patients underwent re-vascularization, of which 94% were by percutaneous coronary intervention, and 42.9% of patients had intracoronary thrombus, of whom approximately one third had no detectable underlying coronary disease. CONCLUSIONS Very young patients with acute coronary syndrome tend to be obese, with a high prevalence of smoking and hyperlipidemia. The presence of thrombus in the absence of underlying coronary disease suggests a thromboembolic event or de novo thrombotic occlusion, which may reflect primary hemostatic dysfunction in a considerable number of these patients.
Collapse
Affiliation(s)
- Laura Davidson
- Northwestern University Feinberg School of Medicine, Chicago, Ill.
| | - Jane Wilcox
- Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - David Kim
- Vanderbilt University Medical Center, Nashville, Tenn
| | | | - Joseph Fredi
- Vanderbilt University Medical Center, Nashville, Tenn
| | - Douglas Vaughan
- Northwestern University Feinberg School of Medicine, Chicago, Ill
| |
Collapse
|
34
|
Twomley KM, Rao SV, Becker RC. Proinflammatory, immunomodulating, and prothrombotic properties of anemia and red blood cell transfusions. J Thromb Thrombolysis 2014; 21:167-74. [PMID: 16622613 DOI: 10.1007/s11239-006-5206-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
For many years, the traditional treatment for hospitalized patients in the United States who have developed anemia, whether associated with medical illness, surgical procedures or trauma, has been red blood cell transfusion, despite the absence of supporting data in many patient populations. Emerging evidence suggests that transfusions may, in fact, be associated with risk beyond commonly held concerns of microbial transmission and acute antigen-antibody reactions. The following overview represents a biological paradigm for understanding the relationship between medical illness, surgical procedures, inflammatory states, anemia, red blood cell transfusion and immunothrombotic phenomena among hospitalized patients.
Collapse
Affiliation(s)
- Katie M Twomley
- Department of Medicine, Duke University Medical Center, Cardiovascular Thrombosis Center, Duke Clinical Research Institute, Durham, NC 27715, USA
| | | | | |
Collapse
|
35
|
Genetic dissection of quantitative trait Loci for hemostasis and thrombosis on mouse chromosomes 11 and 5 using congenic and subcongenic strains. PLoS One 2013; 8:e77539. [PMID: 24147020 PMCID: PMC3798288 DOI: 10.1371/journal.pone.0077539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 09/06/2013] [Indexed: 12/25/2022] Open
Abstract
Susceptibility to thrombosis varies in human populations as well as many inbred mouse strains. Only a small portion of this variation has been identified, suggesting that there are unknown modifier genes. The objective of this study was to narrow the quantitative trait locus (QTL) intervals previously identified for hemostasis and thrombosis on mouse distal chromosome 11 (Hmtb6) and on chromosome 5 (Hmtb4 and Hmtb5). In a tail bleeding/rebleeding assay, a reporter assay for hemostasis and thrombosis, subcongenic strain (6A-2) had longer clot stability time than did C57BL/6J (B6) mice but a similar time to the B6-Chr11A/J consomic mice, confirming the Hmtb6 phenotype. Six congenic and subcongenic strains were constructed for chromosome 5, and the congenic strain, 2A-1, containing the shortest A/J interval (16.6 cM, 26.6 Mbp) in the Hmtb4 region, had prolonged clot stability time compared to B6 mice. In the 3A-2 and CSS-5 mice bleeding time was shorter than for B6, mice confirming the Hmtb5 QTL. An increase in bleeding time was identified in another congenic strain (3A-1) with A/J interval (24.8 cM, 32.9 Mbp) in the proximal region of chromosome 5, confirming a QTL for bleeding previously mapped to that region and designated as Hmtb10. The subcongenic strain 4A-2 with the A/J fragment in the proximal region had a long occlusion time of the carotid artery after ferric chloride injury and reduced dilation after injury to the abdominal aorta compared to B6 mice, suggesting an additional locus in the proximal region, which was designated Hmtb11 (5 cM, 21.4 Mbp). CSS-17 mice crossed with congenic strains, 3A-1 and 3A-2, modified tail bleeding. Using congenic and subcongenic analysis, candidate genes previously identified and novel genes were identified as modifiers of hemostasis and thrombosis in each of the loci Hmtb6, Hmtb4, Hmtb10, and Hmtb11.
Collapse
|
36
|
Boe AE, Eren M, Murphy SB, Kamide CE, Ichimura A, Terry D, McAnally D, Smith LH, Miyata T, Vaughan DE. Plasminogen activator inhibitor-1 antagonist TM5441 attenuates Nω-nitro-L-arginine methyl ester-induced hypertension and vascular senescence. Circulation 2013; 128:2318-24. [PMID: 24092817 DOI: 10.1161/circulationaha.113.003192] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Long-term inhibition of nitric oxide synthase by L-arginine analogues such as N(ω)-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 p16(Ink4a) expression and telomere length in aortic tissue. We found that L-NAME increased p16(Ink4a) 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 p16(Ink4a) 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.
Collapse
Affiliation(s)
- Amanda E Boe
- Department of Medicine (A.E.B., M.E., S.B.M., C.E.K., D.E.V.) and Feinberg Cardiovascular Research Institute (A.E.B., S.B.M., C.E.K., D.E.V.), Northwestern University Feinberg School of Medicine, Chicago, IL; United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Miyagi, Japan (A.I., T.M.); and Conrad Prebys Center for Chemical Genomics (D.T., D.M., L.H.S.) and Cardiopathobiology Program, Diabetes and Obesity Research Center (D.M., L.H.S.), Sanford Burnham Medical Research Institute at Lake Nona, Orlando FL
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lin Z, Jensen JK, Hong Z, Shi X, Hu L, Andreasen PA, Huang M. Structural insight into inactivation of plasminogen activator inhibitor-1 by a small-molecule antagonist. ACTA ACUST UNITED AC 2013; 20:253-61. [PMID: 23438754 DOI: 10.1016/j.chembiol.2013.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 12/24/2012] [Accepted: 12/27/2012] [Indexed: 12/19/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1), a serpin, is the physiological inhibitor of tissue-type and urokinase-type plasminogen activators and thus also an inhibitor of fibrinolysis and tissue remodeling. It is a potential therapeutic target in many pathological conditions, including thrombosis and cancer. Several types of PAI-1 antagonist have been developed, but the structural basis for their action has remained largely unknown. Here we report X-ray crystal structure analysis of PAI-1 in complex with a small-molecule antagonist, embelin. We propose a mechanism for embelin-induced rapid conversion of PAI-1 into a substrate for its target proteases and the subsequent slow conversion of PAI-1 into an irreversibly inactivated form. Our work provides structural clues to an understanding of PAI-1 inactivation by small-molecule antagonists and an important step toward the design of drugs targeting PAI-1.
Collapse
Affiliation(s)
- Zhonghui Lin
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | | | | | | | | | | | | |
Collapse
|
38
|
Fortenberry YM. Plasminogen activator inhibitor-1 inhibitors: a patent review (2006-present). Expert Opin Ther Pat 2013; 23:801-15. [PMID: 23521527 DOI: 10.1517/13543776.2013.782393] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Plasminogen activator inhibitor-1 (PAI-1), the serine protease inhibitor (serpin), binds to and inhibits the plasminogen activators-tissue-type plasminogen activator (tPA) and the urokinase-type plasminogen activator (uPA). This results in both a decrease in plasmin production and a decrease in the dissolution of fibrin clots. Elevated levels of PAI-1 are correlated with an increased risk for cardiovascular disease and have been linked to obesity and metabolic syndrome. Consequently, the pharmacological suppression of PAI-1 might prevent or treat vascular disease. AREAS COVERED This article provides an overview of the patenting activity on PAI-1 inhibitors. Patents filed by pharmaceutical companies or individual research groups are described, and the biological and biochemical evaluation of the inhibitors, including in vitro and in vivo studies, is discussed. An overview of patents pertaining to using these inhibitors for treating various diseases is also included. EXPERT OPINION Although there is still no PAI-1 inhibitor being evaluated in a clinical setting or approved for human therapy, research in this field has progressed, and promising new compounds have been designed. Most research has focused on improving the pharmacological profile of these compounds, which will hopefully allow them to proceed to clinical studies. Despite the need for further testing and research, the potential use of PAI-1 inhibitors for treating cardiovascular disease appears quite promising.
Collapse
Affiliation(s)
- Yolanda M Fortenberry
- Johns Hopkins University School of Medicine, Division of Hematology/Department of Pediatrics, 720 Rutland Avenue Ross 1120, Baltimore, MD 21205, USA.
| |
Collapse
|
39
|
Van De Craen B, Declerck PJ, Gils A. The Biochemistry, Physiology and Pathological roles of PAI-1 and the requirements for PAI-1 inhibition in vivo. Thromb Res 2012; 130:576-85. [DOI: 10.1016/j.thromres.2012.06.023] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/12/2012] [Accepted: 06/27/2012] [Indexed: 12/16/2022]
|
40
|
Glycosylation influences the stability of human plasminogen activator inhibitor-1. Blood Coagul Fibrinolysis 2012; 23:570-2. [DOI: 10.1097/mbc.0b013e3283551163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a common disorder that affects both quality of life and cardiovascular health. The causal link between OSA and cardiovascular morbidity⁄mortality remains elusive. One possible explanation is that repeated episodes of nocturnal hypoxia lead to a hypercoagulable state that predisposes patients to thrombotic events. There is evidence supporting a wide array of hematological changes that affect hemostasis (eg, increased hematocrit, blood viscosity, platelet activation, clotting factors and decreased fibrinolytic activity). OBJECTIVE To provide a comprehensive review of the current evidence associating OSA with increased coagulability, and to highlight areas for future research. METHODS Keyword searches in Ovid Medline were used to identify relevant articles; all references in the articles were searched for relevant titles. The Web of Science was used to identify articles citing the relevant articles found using the Ovid Medline search. All original peer-reviewed articles, meta-analyses and systematic reviews regarding the pertinent topics between 1990 and present were selected for review. RESULTS Hematocrit, blood viscosity, certain clotting factors, tissue factor, platelet activity and whole blood coagulability are increased in patients with OSA, while fibrinolysis is impaired. CONCLUSION There is considerable evidence that OSA is associated with a procoagulant state. Several factors are involved in the procoagulant state associated with OSA. There is a need for adequately powered clinical studies involving well-matched control groups to address potential confounding variables, and to accurately delineate the individual factors involved in the procoagulant state associated with OSA and their response to treatment.
Collapse
|
42
|
Ekström M, Liska J, Eriksson P, Sverremark-Ekström E, Tornvall P. Stimulated in vivo synthesis of plasminogen activator inhibitor-1 in human adipose tissue. Thromb Haemost 2012; 108:485-92. [PMID: 22740034 DOI: 10.1160/th11-11-0822] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 05/07/2012] [Indexed: 11/05/2022]
Abstract
Plasminogen activator inhibitor type-1 (PAI-1) is one of the most important inhibitors of endogenous fibrinolysis. Adipose tissue is a suggested source of the elevated plasma levels of PAI-1 in obesity. The relation between PAI-1 and inflammation is of particular interest, but current knowledge regarding regulation of PAI-1 in adipose tissue is mainly based on animal studies or ex vivo experiments on human cultured adipocytes. So far, no study has described stimulated gene expression and protein synthesis of PAI-1 in vivo in human adipose tissue. We used open heart surgery as a model of acute systemic inflammation. Twenty-two male patients underwent blood sampling and omental and subcutaneous adipose tissue biopsies for gene expression studies before and after surgery. Expression and localisation of PAI-1 antigen was evaluated by immunohistochemistry. After surgery gene expression of PAI-1 increased 27-fold in omental adipose tissue and three-fold in subcutaneous adipose tissue, but no differences were found in tissue-type plasminogen activator (t-PA) mRNA. PAI-1 antigen was localised within endothelial cells and in the adipose tissue interstitium close to vessels. The upregulated gene expression and protein synthesis in adipose tissue was followed by increased concentrations of PAI-1 antigen in plasma. In conclusion, we present for the first time that an acute systemic inflammation in humans increased gene expression and protein synthesis of PAI-1 in adipose tissue and that this increase was most prominent in omental adipose tissue. PAI-1 synthesis in adipose tissue due to acute systemic inflammation may be a link between inflammation and impaired endogenous fibrinolysis.
Collapse
Affiliation(s)
- Mattias Ekström
- Cardiology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
43
|
Simone TM, Higgins PJ. Low Molecular Weight Antagonists of Plasminogen Activator Inhibitor-1: Therapeutic Potential in Cardiovascular Disease. ACTA ACUST UNITED AC 2012; 1:101. [PMID: 23936868 DOI: 10.4172/2324-8769.1000102] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Plasminogen activator inhibitor-1 (PAI-1; SERPINE1) is the major physiologic regulator of the plasmin-based pericellular proteolytic cascade, a modulator of vascular smooth muscle cell (VSMC) migration and a causative factor in cardiovascular disease and restenosis, particularly in the context of increased vessel transforming growth factor- β1 (TGF-β1) levels. PAI-1 limits conversion of plasminogen to plasmin (and, thereby, fibrin degradation) by inhibiting its protease targets urokinase and tissue-type plasminogen activators (uPA, tPA). PAI-1 also has signaling functions and binds to the low density lipoprotein receptor-related protein 1 (LRP1) to regulate LRP1-dependent cell motility that, in turn, contributes to neointima formation. PAI-1/uPA/uPA receptor/LRPI/integrin complexes are endocytosed with subsequent uPAR/LRP1/integrin redistribution to the leading edge, initiating an "adhesion-detachment-readhesion" cycle to promote cell migration. PAI-1 also interacts with LRP1 in a uPA/uPAR-independent manner triggering Jak/Stat1 pathway activation to stimulate cell motility. PAI-1 itself is a substrate for extracellular proteases and exists in a "cleaved" form which, while unable to interact with uPA and tPA, retains LRP1-binding and migratory activity. These findings suggest that there are multiple mechanisms through which inhibition of PAI-1 may promote cardiovascular health. Several studies have focused on the design, synthesis and preclinical assessment of PAI-1 antagonists including monoclonal antibodies, peptides and low molecular weight (LMW) antagonists. This review discusses the translational impact of LMW PAI-1 antagonists on cardiovascular disease addressing PAI-1-initiated signaling, PAI-1 structure, the design and characteristics of PAI-1-targeting drugs, results of in vitro and in vivo studies, and their clinical implications.
Collapse
Affiliation(s)
- Tessa M Simone
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, New York 12208, USA
| | | |
Collapse
|
44
|
Maximal PAI-1 inhibition in vivo requires neutralizing antibodies that recognize and inhibit glycosylated PAI-1. Thromb Res 2011; 129:e126-33. [PMID: 22178065 DOI: 10.1016/j.thromres.2011.11.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 11/21/2011] [Accepted: 11/22/2011] [Indexed: 11/23/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) regulates the activity of t-PA and u-PA and is an important inhibitor of the plasminogen activator system. Elevated PAI-1 levels have been implicated in the pathogenesis of several diseases. Prior to the evaluation of PAI-1 inhibitors in humans, there is a strong need to study the effect of PAI-1 inhibition in mouse models. In the current study, four monoclonal antibodies previously reported to inhibit recombinant PAI-1 in vitro, were evaluated in an LPS-induced endotoxemia model in mice. Both MA-33H1F7 and MA-MP2D2 exerted a strong PAI-1 inhibitory effect, whereas for MA-H4B3 and MA-124K1 no reduced PAI-1 activity was observed in vivo. Importantly, the lack of PAI-1 inhibition observed for MA-124K1 and MA-H4B3 in vivo corresponded with the absence of inhibition toward glycosylated mouse PAI-1 in vitro. Three potential N-glycosylation sites were predicted for mouse PAI-1 (i.e. N209, N265 and N329). Electrophoretic mobility analysis of glycosylation knock-out mutants before and after deglycosylation indicates the presence of glycan chains at position N265. These data demonstrate that an inhibitory effect toward glycosylated PAI-1 is a prerequisite for efficient PAI-1 inhibition in mice. Our data also suggest that PAI-1 inhibitors for use in humans must preferably be screened on glycosylated PAI-1 and not on recombinant non-glycosylated PAI-1.
Collapse
|
45
|
Enhanced functional stability of plasminogen activator inhibitor-1 in patients with livedoid vasculopathy. J Thromb Thrombolysis 2011; 32:59-63. [PMID: 21311952 DOI: 10.1007/s11239-011-0556-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Livedoid vasculopathy (LV) is a chronic, recurrent, painful cutaneous disease with distinctive clinical features and an uncertain etiology. The skin lesions are recognizable by focal purpura, depigmentation and shallow ulcers. Thrombophilic conditions occur frequently in patients with LV. While no definitive treatment exists for LV, smoking cessation, antiplatelet therapy, immunosuppressive treatment, and anabolic steroids are often included in the therapeutic ladder. Recently, a possible link between LV and impaired fibrinolysis was established as cutaneous LV lesions responded to tissue plasminogen activator (t-PA) infusion suggesting that inhibition of the fibrinolysis through plasminogen activator inhibitor-1 (PAI-1) activity may determine the disease course in patients with LV. In this study, we investigated PAI-1 antigen (Ag) and activity levels in 20 patients with biopsy proven LV (mean age 26 ± 11, M/F = 7/13, median disease duration 3.5 years). All patients received antiplatelet treatment with aspirin and/or dipyrimadole and 14 patients received anabolic steroids or immunosuppressive treatment. Fasting PAI-1 Ag and activity levels were measured at 9 AM in all patients. Both Ag (34 (26) ng/ml) (median (interquartile range)) and specific activity (17 (23) IU/fmole) levels of PAI-1 were moderately elevated in LV patients compared to the controls, however, PAI-1 kinetic studies demonstrated markedly enhanced stability of PAI-1 activity in plasma from patients with LV. Specific activity at 16 h was significantly higher than expected specific activity levels (7 (11) vs. 0.07 (0.09) IU/fmole, P < 0.01). While the exact mechanism of increased stability of PAI-1 activity is not known, it may be due to post-translational modifications or increased binding affinity for a stabilizing cofactor. In conclusion, enhanced stability of PAI-1 may contribute to the pathophysiology of LV, and systemic or local treatment with PAI-1 inhibitors may offer a potential treatment alternative in patients with LV.
Collapse
|
46
|
Samarakoon R, Chitnis SS, Higgins SP, Higgins CE, Krepinsky JC, Higgins PJ. Redox-induced Src kinase and caveolin-1 signaling in TGF-β1-initiated SMAD2/3 activation and PAI-1 expression. PLoS One 2011; 6:e22896. [PMID: 21829547 PMCID: PMC3145778 DOI: 10.1371/journal.pone.0022896] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 07/08/2011] [Indexed: 11/29/2022] Open
Abstract
Background Plasminogen activator inhibitor-1 (PAI-1), a major regulator of the plasmin-based pericellular proteolytic cascade, is significantly increased in human arterial plaques contributing to vessel fibrosis, arteriosclerosis and thrombosis, particularly in the context of elevated tissue TGF-β1. Identification of molecular events underlying to PAI-1 induction in response to TGF-β1 may yield novel targets for the therapy of cardiovascular disease. Principal Findings Reactive oxygen species are generated within 5 minutes after addition of TGF-β1 to quiescent vascular smooth muscle cells (VSMCs) resulting in pp60c-src activation and PAI-1 expression. TGF-β1-stimulated Src kinase signaling sustained the duration (but not the initiation) of SMAD3 phosphorylation in VSMC by reducing the levels of PPM1A, a recently identified C-terminal SMAD2/3 phosphatase, thereby maintaining SMAD2/3 in an active state with retention of PAI-1 transcription. The markedly increased PPM1A levels in triple Src kinase (c-Src, Yes, Fyn)-null fibroblasts are consistent with reductions in both SMAD3 phosphorylation and PAI-1 expression in response to TGF-β1 compared to wild-type cells. Activation of the Rho-ROCK pathway was mediated by Src kinases and required for PAI-1 induction in TGF-β1-stimulated VSMCs. Inhibition of Rho-ROCK signaling blocked the TGF-β1-mediated decrease in nuclear PPM1A content and effectively attenuated PAI-1 expression. TGF-β1-induced PAI-1 expression was undetectable in caveolin-1-null cells, correlating with the reduced Rho-GTP loading and SMAD2/3 phosphorylation evident in TGF-β1-treated caveolin-1-deficient cells relative to their wild-type counterparts. Src kinases, moreover, were critical upstream effectors of caveolin-1Y14 phosphoryation and initiation of downstream signaling. Conclusions TGF-β1-initiated Src-dependent caveolin-1Y14 phosphorylation is a critical event in Rho-ROCK-mediated suppression of nuclear PPM1A levels maintaining, thereby, SMAD2/3-dependent transcription of the PAI-1 gene.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Aorta/metabolism
- Blotting, Western
- Caveolin 1/metabolism
- Cells, Cultured
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Humans
- Immunoenzyme Techniques
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Oxidation-Reduction
- Phosphoprotein Phosphatases/antagonists & inhibitors
- Phosphoprotein Phosphatases/genetics
- Phosphoprotein Phosphatases/metabolism
- Phosphorylation
- Plasminogen Activator Inhibitor 1/metabolism
- Protein Phosphatase 2C
- Proto-Oncogene Proteins pp60(c-src)/metabolism
- RNA, Small Interfering/genetics
- Rats
- Reactive Oxygen Species
- Signal Transduction
- Smad2 Protein/genetics
- Smad2 Protein/metabolism
- Smad3 Protein/antagonists & inhibitors
- Smad3 Protein/genetics
- Smad3 Protein/metabolism
- Transforming Growth Factor beta1/pharmacology
Collapse
Affiliation(s)
- Rohan Samarakoon
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York, United States of America
| | - Subhanir S. Chitnis
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York, United States of America
| | - Stephen P. Higgins
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York, United States of America
| | - Craig E. Higgins
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York, United States of America
| | - Joan C. Krepinsky
- Division of Nephrology, McMaster University, Hamilton, Ontario, Canada
| | - Paul J. Higgins
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
47
|
Sa Q, Hoover-Plow JL. EMILIN2 (Elastin microfibril interface located protein), potential modifier of thrombosis. Thromb J 2011; 9:9. [PMID: 21569335 PMCID: PMC3113922 DOI: 10.1186/1477-9560-9-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 05/11/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Elastin microfibril interface located protein 2 (EMILIN2) is an extracellular glycoprotein associated with cardiovascular development. While other EMILIN proteins are reported to play a role in elastogenesis and coagulation, little is known about EMILIN2 function in the cardiovascular system. The objective of this study was to determine whether EMILIN2 could play a role in thrombosis. RESULTS EMILIN2 mRNA was expressed in 8 wk old C57BL/6J mice in lung, heart, aorta and bone marrow, with the highest expression in bone marrow. In mouse cells, EMILIN2 mRNA expression in macrophages was higher than expression in endothelial cells and fibroblasts. EMILIN2 was identified with cells and extracellular matrix by immunohistochemistry in the carotid and aorta. After carotid ferric chloride injury, EMILIN2 was abundantly expressed in the thrombus and inhibition of EMILIN2 increased platelet de-aggregation after ADP-stimulated platelet aggregation. CONCLUSIONS These results suggest EMILIN2 could play a role in thrombosis as a constituent of the vessel wall and/or a component of the thrombus.
Collapse
Affiliation(s)
- Qila Sa
- Joseph J, Jacobs Center For Thrombosis and Vascular Biology, Department of Cardiovascular Medicine and Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | |
Collapse
|
48
|
Ogawa M, Suzuki JI, Yamaguchi Y, Muto S, Itai A, Hirata Y, Isobe M, Nagai R. The effects of pharmacologic plasminogen activator inhibitor-1 inhibition in acute and chronic rejection in murine cardiac allografts. Transplantation 2011; 91:21-6. [PMID: 21452408 DOI: 10.1097/tp.0b013e3181fd3c0f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Acute rejection and graft arterial disease (GAD) in cardiac transplantation limit the long-term survival of recipients; these processes are enhanced by inflammation and thrombus formation. Plasminogen activator inhibitor (PAI)-1 is critical in the inflammation and thrombus formation. However, little is known about the effect of PAI-1 in heart transplantation. Thus, the objective was to clarify the role of PAI-1 in the progression of cardiac rejection. METHODS Murine hearts were heterotopically transplanted using major mismatch combinations for evaluation of acute rejection and class II mismatch combinations for the GAD. We administered the specific PAI-1 inhibitor (IMD-1622) into murine recipients after cardiac allografts. RESULTS Nontreated allografts of the major mismatch group were acutely rejected, whereas the PAI-1 inhibitor prolonged their survival. Although severe cell infiltration and intimal thickening with enhancement of inflammatory factors were observed in untreated allografts of class II mismatch group on day 60, the PAI-1 inhibitor attenuated these changes. CONCLUSION The PAI-1 inhibitor is potent for the suppression of both acute rejection and GAD.
Collapse
Affiliation(s)
- Masahito Ogawa
- Department of Advanced Clinical Science and Therapeutics, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Riehl TE, He L, Zheng L, Greco S, Tollefsen DM, Stenson WF. COX-1(+/-)COX-2(-/-) genotype in mice is associated with shortened time to carotid artery occlusion through increased PAI-1. J Thromb Haemost 2011; 9:350-60. [PMID: 21138526 PMCID: PMC3687774 DOI: 10.1111/j.1538-7836.2010.04156.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND We found a high incidence of thrombotic deaths in COX-1(+/-)COX-2(-/-) mice and sought to define the mechanism of these events. The cyclooxygenase products thromboxane A(2) and prostacyclin are important in the regulation of coagulation but their role in fibrinolysis is largely unexplored. PAI-1 blocks fibrinolysis by inhibiting plasminogen activator. AIM Our objective was to explain the mechanism of increased thrombosis associated with the COX-1(+/-)COX-2(-/-) genotype. METHODS Carotid artery occlusion times were measured after photochemical injury. PAI-1 levels were measured in the plasma by ELISA. PAI-1 levels in the aorta were measured by RT-PCR and Western blotting. Urinary metabolites of Thromboxane A(2) and prostacyclin were measured by ELISA. RESULTS The COX-1(+/-)COX-2(-/-) genotype is associated with a decreased time to occlusion in the carotid artery thrombosis model (30 ± 5 minutes vs 60 ± minutes in wild type, p<.001). The COX-1(-/-)COX-2(+/+), COX-1(+/-)COX-2(+/-) and COX-1(+/-)COX-2(+/+) all had occlusion times similar to wild type. COX-1(+/+)COX-2(-/-) had a prolonged occlusion time. COX-1(+/-)COX-2(-/-) had increased PAI-1 levels in the plasma and aorta and with a prolonged euglobulin lysis time (37.4 ± 10.2 hours vs 15.6 ± 9.8 hours in wild type, p<.004). The decreased time to occlusion in the COX-1(+/-)COX2(-/-) mice was normalized by an inhibitory antibody to PAI-1 whereas the antibody had no effect on the time to occlusion in wild type mice. CONCLUSION The COX-1(+/-)COX-2(-/-) genotype is associated with a shortened time to occlusion in the carotid thrombosis model and the shortened time to occlusion is mediated through increased PAI-1 levels resulting in decreased fibrinolysis.
Collapse
Affiliation(s)
- T E Riehl
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the main inhibitor of tissue-type plasminogen activator (t-PA) and urokinase-type plasminogen activator (u-PA) and therefore plays an important role in the plasminogen/plasmin system. PAI-1 is involved in a variety of cardiovascular diseases (mainly through inhibition of t-PA) as well as in cell migration and tumor development (mainly through inhibition of u-PA and interaction with vitronectin). PAI-1 is a unique member of the serpin superfamily, exhibiting particular unique conformational and functional properties. Since its involvement in various biological and pathophysiological processes PAI-1 has been the subject of many in vivo studies in mouse models. We briefly discuss structural and physiological differences between human and mouse PAI-1 that should be taken into account prior to extrapolation of data obtained in mouse models to the human situation. The current review provides an overview of the various models, with a focus on cardiovascular disease and cancer, using wild-type mice or genetically modified mice, either deficient in PAI-1 or overexpressing different variants of PAI-1.
Collapse
|