1
|
Bektasoglu PK, Somay A, Hazneci J, Borekci A, Gurer B. The Antifibrotic Effects of Plasminogen Activator Inhibitor-1 Antagonists are Observed in Rats with Epidural Fibrosis. SISLI ETFAL HASTANESI TIP BULTENI 2025; 59:59-63. [PMID: 40226558 PMCID: PMC11983027 DOI: 10.14744/semb.2024.75301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 04/15/2025]
Abstract
Objectives Epidural fibrosis occurs after laminectomy as part of the local repair mechanisms. Adhesion around the nervous tissue could cause pain and disability. In the current study, we investigated the possible antifibrotic effects of the plasminogen activator inhibitor-1 (PAI-1) antagonists in a rat laminectomy model. Methods The rats were randomly assigned to the control, the TM5441, and the TM5484 groups (n=6 per group). In the control group, just a laminectomy was performed. In the treatment groups, intragastric administration of PAI-1 antagonists was done after skin closure. Epidural fibrosis was investigated macroscopically and histopathologically four weeks later. Results In the TM5441 and TM5484 groups, the macroscopic epidural fibrosis score was less than the control group (p<0.001 for both groups). Microscopic epidural fibrosis score was also decreased in the TM5441 and TM5484 groups (p>0.05 for both groups). Fibroblast cell density classification scores in the TM5441 and TM5484 groups were lower when compared to the control group (p>0.05 for both groups). Fibrosis thickness was lower in the TM5441 and TM5484 groups when compared to the control group (p<0.01 for both groups). Conclusion Plasminogen activator inhibitor-1 antagonists could be a treatment alternative for the prevention of epidural fibrosis.
Collapse
Affiliation(s)
- Pinar Kuru Bektasoglu
- Department of Neurosurgery, University of Health Sciences Türkiye, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Türkiye
| | - Adnan Somay
- Department of Pathology, University of Health Sciences Türkiye, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Türkiye
| | - Julide Hazneci
- Department of Neurosurgery, University of Health Sciences Türkiye, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Türkiye
| | - Ali Borekci
- Department of Neurosurgery, University of Health Sciences Türkiye, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Türkiye
| | - Bora Gurer
- Department of Neurosurgery, Istinye University Faculty of Medicine, Istanbul, Türkiye
| |
Collapse
|
2
|
Zhai YL, Sun SG, Zhang WH, Tian HJ, Zhao ZZ. Clinical Value of ABCB1 and PAI-1 Gene Polymorphisms in Predicting Glucocorticoid-induced Adverse Reactions in Nephrotic Syndrome Patients. Curr Med Sci 2024; 44:923-931. [PMID: 39285050 DOI: 10.1007/s11596-024-2887-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/01/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Glucocorticoid (GC)-induced adverse reactions (ARs) have been extensively studied due to their potential impact on patients' health. This study aimed to examine the potential correlation between two polymorphisms [adenosine triphosphate-binding cassette B1 (ABCB1) C3435T and plasminogen activator inhibitor-1 (PAI-1) 4G/5G] and various GC-induced ARs in nephrotic syndrome (NS) patients. METHODS In this study, 513 NS patients who underwent GC treatment were enrolled. Then, the patients were divided into two groups based on ABCB1 C3435T and PAI-1 4G/5G genotyping, and intergroup comparisons of clinicopathological data and GC-induced ARs were performed. Univariate and multivariate logistic analyses were subsequently conducted to identify potential risk factors for GC-induced ARs, and a nomogram was subsequently established and validated via the area under the ROC curve (AUC), calibration curve and decision curve analysis (DCA). RESULTS We identified ABCB1 C3435T as an independent risk factor for the development of steroid-associated avascular necrosis of the femoral head (SANFH) (OR: 2.191, 95% CI: 1.258-3.813, P=0.006) but not as a risk factor for the occurrence of steroid diabetes mellitus (S-DM). On the other hand, PAI-1 4G/5G was identified as an independent risk factor for the development of both SANFH (OR: 2.198, 95% CI: 1.267-3.812, P=0.005) and S-DM (OR: 2.080, 95% CI: 1.166-3.711, P=0.013). Notably, no significant correlation was found between the two gene polymorphisms and other GC-induced ARs. In addition, two nomograms were established and validated to demonstrate strong calibration capability and clinical utility. CONCLUSION Assessing ABCB1 C3435T and PAI-1 4G/5G before steroid treatment in NS patients could be useful for identifying patients at a high risk of developing SANFH and S-DM.
Collapse
Affiliation(s)
- Ya-Ling Zhai
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- The Renal Research Institution of Zhengzhou University, Zhengzhou, 450000, China
| | - Shuai-Gang Sun
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- The Renal Research Institution of Zhengzhou University, Zhengzhou, 450000, China
| | - Wen-Hui Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- The Renal Research Institution of Zhengzhou University, Zhengzhou, 450000, China
| | - Hui-Juan Tian
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- The Renal Research Institution of Zhengzhou University, Zhengzhou, 450000, China
| | - Zhan-Zheng Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- The Renal Research Institution of Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
3
|
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: 13] [Impact Index Per Article: 4.3] [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
|
4
|
Cancer-Associated Fibroblasts: Mechanisms of Tumor Progression and Novel Therapeutic Targets. Cancers (Basel) 2022; 14:cancers14051231. [PMID: 35267539 PMCID: PMC8909913 DOI: 10.3390/cancers14051231] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The tumor microenvironment plays an important role in determining the biological behavior of several of the more aggressive malignancies. Among the various cell types evident in the tumor “field”, cancer-associated fibroblasts (CAFs) are a heterogenous collection of activated fibroblasts secreting a wide repertoire of factors that regulate tumor development and progression, inflammation, drug resistance, metastasis and recurrence. Insensitivity to chemotherapeutics and metastatic spread are the major contributors to cancer patient mortality. This review discusses the complex interactions between CAFs and the various populations of normal and neoplastic cells that interact within the dynamic confines of the tumor microenvironment with a focus on the involved pathways and genes. Abstract Cancer-associated fibroblasts (CAFs) are a heterogenous population of stromal cells found in solid malignancies that coexist with the growing tumor mass and other immune/nonimmune cellular elements. In certain neoplasms (e.g., desmoplastic tumors), CAFs are the prominent mesenchymal cell type in the tumor microenvironment, where their presence and abundance signal a poor prognosis in multiple cancers. CAFs play a major role in the progression of various malignancies by remodeling the supporting stromal matrix into a dense, fibrotic structure while secreting factors that lead to the acquisition of cancer stem-like characteristics and promoting tumor cell survival, reduced sensitivity to chemotherapeutics, aggressive growth and metastasis. Tumors with high stromal fibrotic signatures are more likely to be associated with drug resistance and eventual relapse. Clarifying the molecular basis for such multidirectional crosstalk among the various normal and neoplastic cell types present in the tumor microenvironment may yield novel targets and new opportunities for therapeutic intervention. This review highlights the most recent concepts regarding the complexity of CAF biology including CAF heterogeneity, functionality in drug resistance, contribution to a progressively fibrotic tumor stroma, the involved signaling pathways and the participating genes.
Collapse
|
5
|
Kuru Bektaşoğlu P, Koyuncuoğlu T, Akbulut S, Akakın D, Eyüboğlu İP, Erzik C, Yüksel M, Kurtel H. Neuroprotective Effect of Plasminogen Activator Inhibitor-1 Antagonist in the Rat Model of Mild Traumatic Brain Injury. Inflammation 2021; 44:2499-2517. [PMID: 34460025 DOI: 10.1007/s10753-021-01520-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 10/20/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) antagonists are known for their neuroprotective effects. In this study, it was aimed to investigate the possible protective effects of PAI-1 antagonists in a rat mild traumatic brain injury (TBI) model. Sprague-Dawley male rats were grouped as sham (n = 7), TBI (n = 9), and TBI + PAI-1 antagonist (5 and 10 mg/kg TM5441 and TM5484; n = 6-7). Under anesthesia, TBI was induced by dropping a metal 300-g weight from a height of 1 m on the skull. Before and 24-h after trauma neurological examination, tail suspension, Y-maze, and novel object recognition tests were performed. Twenty-four hours after TBI, the rats were decapitated and activities of myeloperoxidase, nitric oxide release, luminol-, and lucigenin-enhanced chemiluminescence were measured. Also, interleukin-1β, interleukin-6, tumor necrosis factor, interleukin-10, tumor growth factor-β, caspase-3, cleaved caspase-3, and PAI levels were measured with the ELISA method in the brain tissue. Brain injury was graded histopathologically following hematoxylin-eosin staining. Western blot and immunohistochemical investigation for low-density lipoprotein receptor, matrix metalloproteinase-3, and nuclear factor-κB were also performed. Data were analyzed using GraphPad Prism 8.0 (GraphPad Software, San Diego, CA, USA) and expressed as means ± SEM. Values of p < 0.05 were considered to be statistically significant. Higher levels of myeloperoxidase activity in the TBI group (p < 0.05) were found to be suppressed in 5 and 10 mg/kg TM5441 treatment groups (p < 0.05-p < 0.01). The tail suspension test score was increased in the TBI group (p < 0.001) and decreased in all treatment groups (p < 0.05-0.001). The histologic damage score was increased statistically significantly in the cortex, dentate gyrus, and CA3 regions in the TBI group (p < 0.01-0.001), decreased in the treatment groups in the cortex and dentate gyrus (p < 0.05-0.001). PAI antagonists, especially TM5441, have antioxidant and anti-inflammatory properties against mild TBI in the acute period. Behavioral test results were also improved after PAI antagonist treatment after mild TBI.
Collapse
Affiliation(s)
- Pınar Kuru Bektaşoğlu
- Department of Physiology, Marmara University Institute of Health Sciences, Istanbul, Turkey.
- Department of Neurosurgery, University of Health Sciences, Fatih Sultan Mehmet Education and Research Hospital, Istanbul, Turkey.
- Department of Physiology, Marmara University Institute of Health Sciences, Istanbul, Turkey.
| | - Türkan Koyuncuoğlu
- Department of Physiology, Biruni University School of Medicine, Istanbul, Turkey
| | - Selin Akbulut
- Department of Histology and Embryology, Marmara University School of Medicine, Istanbul, Turkey
| | - Dilek Akakın
- Department of Histology and Embryology, Marmara University School of Medicine, Istanbul, Turkey
| | - İrem Peker Eyüboğlu
- Department of Medical Biology, Marmara University School of Medicine, Istanbul, Turkey
| | - Can Erzik
- Department of Medical Biology, Marmara University School of Medicine, Istanbul, Turkey
| | - Meral Yüksel
- Department of Medical Laboratory Techniques, Marmara University Vocational School of Health Services, Istanbul, Turkey
| | - Hızır Kurtel
- Department of Physiology, Marmara University Institute of Health Sciences, Istanbul, Turkey
| |
Collapse
|
6
|
Banfi C, Baetta R, Barbieri SS, Brioschi M, Guarino A, Ghilardi S, Sandrini L, Eligini S, Polvani G, Bergman O, Eriksson P, Tremoli E. Prenylcysteine oxidase 1, an emerging player in atherosclerosis. Commun Biol 2021; 4:1109. [PMID: 34548610 PMCID: PMC8455616 DOI: 10.1038/s42003-021-02630-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
The research into the pathophysiology of atherosclerosis has considerably increased our understanding of the disease complexity, but still many questions remain unanswered, both mechanistically and pharmacologically. Here, we provided evidence that the pro-oxidant enzyme Prenylcysteine Oxidase 1 (PCYOX1), in the human atherosclerotic lesions, is both synthesized locally and transported within the subintimal space by proatherogenic lipoproteins accumulating in the arterial wall during atherogenesis. Further, Pcyox1 deficiency in Apoe-/- mice retards atheroprogression, is associated with decreased features of lesion vulnerability and lower levels of lipid peroxidation, reduces plasma lipid levels and inflammation. PCYOX1 silencing in vitro affects the cellular proteome by influencing multiple functions related to inflammation, oxidative stress, and platelet adhesion. Collectively, these findings identify the pro-oxidant enzyme PCYOX1 as an emerging player in atherogenesis and, therefore, understanding the biology and mechanisms of all functions of this unique enzyme is likely to provide additional therapeutic opportunities in addressing atherosclerosis.
Collapse
Affiliation(s)
- C. Banfi
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - R. Baetta
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - S. S. Barbieri
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - M. Brioschi
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - A. Guarino
- grid.418230.c0000 0004 1760 1750Cardiovascular Tissue Bank of Milan, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - S. Ghilardi
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - L. Sandrini
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - S. Eligini
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - G. Polvani
- grid.418230.c0000 0004 1760 1750Cardiovascular Tissue Bank of Milan, Centro Cardiologico Monzino IRCCS, Milano, Italy ,grid.4708.b0000 0004 1757 2822Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Milano, Italy ,grid.418230.c0000 0004 1760 1750Department of Cardiovascular Disease, Development and Innovation Cardiac Surgery Unit, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - O. Bergman
- grid.4714.60000 0004 1937 0626Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - P. Eriksson
- grid.4714.60000 0004 1937 0626Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - E. Tremoli
- grid.418230.c0000 0004 1760 1750Centro Cardiologico Monzino IRCCS, Milano, Italy
| |
Collapse
|
7
|
Zhang H, Liu B, Jiang S, Wu JF, Qi CH, Mohammadtursun N, Li Q, Li L, Zhang H, Sun J, Dong JC. Baicalin ameliorates cigarette smoke-induced airway inflammation in rats by modulating HDAC2/NF-κB/PAI-1 signalling. Pulm Pharmacol Ther 2021; 70:102061. [PMID: 34314854 DOI: 10.1016/j.pupt.2021.102061] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease distinguished by airway remodelling and progressive inflammation. PAI-1 is an important regulator of fibrosis. Recent studies have shown that PAI-1 seems to be involved in COPD progression. Elevated levels of PAI-1 have been found in the lungs of patients with acute inflammation. PAI-1 has been shown to regulate the levels of proinflammatory cytokines in the lungs, such as tumour necrosis factor (TNF)-α and interleukin (IL)-6, indicating that PAI-1 may play a fundamental role during inflammation. In the present study, we investigated the anti-inflammatory role of baicalin, the main active component of Scutellaria baicalensis, against cigarette smoke (extract) (CS/CSE)-induced airway inflammation in vivo and in vitro. For the in vivo study, SD rats were exposed to CS for 1 h/day, 6 days/week, for 24 weeks and treated with baicalin (40, 80 and 160 mg/kg) or budesonide (0.2 mg/kg). For this study, HBE cells were pretreated with baicalin (10, 20, 40 μM) or dexamethasone (10-7 M) and then exposed to CSE. We found that baicalin treatment could ameliorate CS-induced airway inflammatory infiltration in rats and decrease PAI-1 expression. The ELISA results showed that baicalin significantly inhibited the levels of TNF-α and IL-1β in CS/CSE-exposed rats and cells. Mechanistic studies showed that baicalin enhanced histone deacetylase 2 (HDAC2) protein expression and inhibited the expression of NF-κB and its downstream target PAI-1, and these effects were reversed by the HDAC2 inhibitor CAY-10683. In conclusion, baicalin ameliorated CS-induced airway inflammation in rats, and these effects were partially attributed to the modulation of HDAC2/NF-κB/PAI-1 signalling.
Collapse
Affiliation(s)
- Hu Zhang
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Baojun Liu
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Shan Jiang
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jin-Feng Wu
- Huashan Hospital, Fudan University, Shanghai, China; Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Chun-Hui Qi
- Department of Respiratory Medicine, Qingpu District Traditional Chinese Medicine Hospital, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Nabijan Mohammadtursun
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Qiuping Li
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Lulu Li
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Hongying Zhang
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jing Sun
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| | - Jing-Cheng Dong
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| |
Collapse
|
8
|
Tzekaki EE, Tsolaki M, Pantazaki ΑA, Geromichalos G, Lazarou E, Kozori M, Sinakos Z. The pleiotropic beneficial intervention of olive oil intake on the Alzheimer's disease onset via fibrinolytic system. Exp Gerontol 2021; 150:111344. [PMID: 33836262 DOI: 10.1016/j.exger.2021.111344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 12/31/2022]
Abstract
The daily consumption of Extra Virgin Olive Oil (EVOO) in Mediterranean nutrition is tightly associated with lower frequency of many diseases' appearance, including Alzheimer's disease (AD). Fibrinolytic system is already assumed to be involved in AD pathophysiology through various factors, especially plasminogen activator inhibitor-1 (PAI-1), a2-antiplasmin (α2ΑP) and tissue plasminogen activator (tPA). We, here, present a biochemical study, as a continuation of a clinical trial of a cohort of 84 participants, focusing on the pleiotropic effect of the annual EVOO consumption on the fibrinolytic factors of Mild Cognitive Impairment (MCI) patients. The levels of all these fibrinolytic factors, measured by Enzyme-Linked Immunosorbent Assay (ELISA) method, were reduced in the serum of MCI patients annually administered with EVOO, versus not treated MCI patients, as well as AD patients. The well-established AD hallmarks (Aβ1-40 and Aβ1-42 species, tau, and p-tau) of MCI patients' group, annually administered with EVOO, were restored to levels equal to those of the cognitively-healthy group; in contrast to those patients not being administered, and their AD hallmarks levels increased at the end of the year. Moreover, one of the EVOO annual consumption multimodal effects on the MCI patients focused on the levels of an oxidative stress trademark, malondialdehyde (MDA), which displayed also a visible quenching; On the other hand, an increase exhibited in the MCI patients not consuming EVOO one year after, was attributed to the lack of the EVOO anti-oxidative properties. These outcomes are exploitable towards the establishment of natural products like EVOO, as a preventive remedy fighting this neurodegenerative disorder, AD. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT03362996 MICOIL gov Identifier: NCT03362996.
Collapse
Affiliation(s)
- Elena E Tzekaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece
| | - Magda Tsolaki
- 1(st) Department of Neurology, Medical School, "AHEPA" General Hospital Medical School, Aristotle University of Thessaloniki, Faculty of Health Sciences, 54124 Thessaloniki, Makedonia, Greece; Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece.
| | - Αnastasia A Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece.
| | - George Geromichalos
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece
| | - Eftychia Lazarou
- Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece
| | - Mahi Kozori
- Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece
| | - Zacharias Sinakos
- Emeritus Professor of Hematology, Medical School, Aristotle University of Thessaloniki, Faculty of Health Sciences, Greece
| |
Collapse
|
9
|
Kwak SY, Park S, Kim H, Lee SJ, Jang WS, Kim MJ, Lee S, Jang WI, Kim AR, Kim EH, Shim S, Jang H. Atorvastatin Inhibits Endothelial PAI-1-Mediated Monocyte Migration and Alleviates Radiation-Induced Enteropathy. Int J Mol Sci 2021; 22:ijms22041828. [PMID: 33673196 PMCID: PMC7917640 DOI: 10.3390/ijms22041828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/15/2023] Open
Abstract
Intestinal injury is observed in cancer patients after radiotherapy and in individuals exposed to radiation after a nuclear accident. Radiation disrupts normal vascular homeostasis in the gastrointestinal system by inducing endothelial damage and senescence. Despite advances in medical technology, the toxicity of radiation to healthy tissue remains an issue. To address this issue, we investigated the effect of atorvastatin, a commonly prescribed hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor of cholesterol synthesis, on radiation-induced enteropathy and inflammatory responses. We selected atorvastatin based on its pleiotropic anti-fibrotic and anti-inflammatory effects. We found that atorvastatin mitigated radiation-induced endothelial damage by regulating plasminogen activator inhibitor-1 (PAI-1) using human umbilical vein endothelial cells (HUVECs) and mouse model. PAI-1 secreted by HUVECs contributed to endothelial dysfunction and trans-endothelial monocyte migration after radiation exposure. We observed that PAI-1 production and secretion was inhibited by atorvastatin in irradiated HUVECs and radiation-induced enteropathy mouse model. More specifically, atorvastatin inhibited PAI-1 production following radiation through the JNK/c-Jun signaling pathway. Together, our findings suggest that atorvastatin alleviates radiation-induced enteropathy and supports the investigation of atorvastatin as a radio-mitigator in patients receiving radiotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sehwan Shim
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| | - Hyosun Jang
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| |
Collapse
|
10
|
Marquard S, Thomann S, Weiler SME, Bissinger M, Lutz T, Sticht C, Tóth M, de la Torre C, Gretz N, Straub BK, Marquardt J, Schirmacher P, Breuhahn K. Yes-associated protein (YAP) induces a secretome phenotype and transcriptionally regulates plasminogen activator Inhibitor-1 (PAI-1) expression in hepatocarcinogenesis. Cell Commun Signal 2020; 18:166. [PMID: 33097058 PMCID: PMC7583285 DOI: 10.1186/s12964-020-00634-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Background Overexpression and nuclear enrichment of the oncogene yes-associated protein (YAP) cause tumor initiation and support tumor progression in human hepatocellular carcinoma (HCC) via cell autonomous mechanisms. However, how YAP expression in tumor cells affects intercellular communication within the tumor microenvironment is not well understood. Methods To investigate how tumor cell-derived YAP is changing the paracrine communication network between tumor cells and non-neoplastic cells in hepatocarcinogenesis, the expression and secretion of cytokines, growth factors and chemokines were analyzed in transgenic mice with liver-specific and inducible expression of constitutively active YAP (YAPS127A). Transcriptomic and proteomic analyses were performed using primary isolated hepatocytes and blood plasma. In vitro, RNAinterference (RNAi), expression profiling, functional analyses and chromatin immunoprecipitation (ChIP) analyses of YAP and the transcription factor TEA domain transcription factor 4 (TEAD4) were performed using immortalized cell lines. Findings were confirmed in cohorts of HCC patients at the transcript and protein levels. Results YAP overexpression induced the expression and secretion of many paracrine-acting factors with potential impact on tumorous or non-neoplastic cells (e.g. plasminogen activator inhibitor-1 (PAI-1), C-X-C motif chemokine ligand 13 (CXCL13), CXCL16). Expression analyses of human HCC patients showed an overexpression of PAI-1 in human HCC tissues and a correlation with poor overall survival as well as early cancer recurrence. PAI-1 statistically correlated with genes typically induced by YAP, such as connective tissue growth factor (CTGF) and cysteine rich angiogenic inducer 61 (CYR61) or YAP-dependent gene signatures (CIN4/25). In vitro, YAP inhibition diminished the expression and secretion of PAI-1 in murine and human liver cancer cell lines. PAI-1 affected the expression of genes involved in cellular senescence and oncogene-induced senescence was confirmed in YAPS127A transgenic mice. Silencing of TEAD4 as well as treatment with the YAP/TEAD interfering substance Verteporfin reduced PAI-1 expression. ChIP analyses confirmed the binding of YAP and TEAD4 to the gene promoter of PAI-1 (SERPINE1). Conclusions These results demonstrate that the oncogene YAP changes the secretome response of hepatocytes and hepatocyte-derived tumor cells. In this context, the secreted protein PAI-1 is transcriptionally regulated by YAP in hepatocarcinogenesis. Perturbation of these YAP-dependent communication hubs including PAI-1 may represent a promising pharmacological approach in tumors with YAP overexpression. Video abstract
Supplementary information Supplementary information accompanies this paper at 10.1186/s12964-020-00634-6.
Collapse
Affiliation(s)
- Simone Marquard
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Thomann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sofia M E Weiler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michaela Bissinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Teresa Lutz
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Present address: Department of Medicine II, LMU Munich, Munich, Germany
| | - Carsten Sticht
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Marcell Tóth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carolina de la Torre
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Beate K Straub
- Institute of Pathology, Johannes Gutenberg University, Mainz, Germany
| | - Jens Marquardt
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany.,Present address: Department of Medicine I, University Hospital Lübeck, Lübeck, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
11
|
Postoperative peritoneal adhesion: an update on physiopathology and novel traditional herbal and modern medical therapeutics. Naunyn Schmiedebergs Arch Pharmacol 2020; 394:317-336. [PMID: 32979062 DOI: 10.1007/s00210-020-01961-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023]
Abstract
Postoperative peritoneal adhesion (PPA) is a serious clinical condition that affects the high percentage of patients after abdominal surgery. In this review, we have tried to focus on pathophysiology and different underlying signal pathways of adhesion formation based on recent progress in the molecular and cellular mechanisms. Also, the strategies, developed based on traditional herbal and modern medicines, to prevent and treat the PPA via regulation of the molecular mechanisms were investigated. The search engines such as Google Scholar, PubMed, Scopus, and Science Direct have been used to evaluate the current literature related to the pathogenesis of adhesion formation and novel products. Recently, different mechanisms have been defined for adhesion formation, mainly categorized in fibrin formation and adhesion fibroblast function, inflammation, and angiogenesis. Therefore, the suppression of these mechanisms via traditional and modern medicine has been suggested in several studies. While different strategies with encouraging findings have been developed, most of the studies showed contradictory results and were performed on animals. The herbal products have been introduced as safe and effective agent which can be considered in future preclinical and clinical studies. Although a wide range of therapeutics based on traditional and modern medicines have been suggested, there is no agreement in the efficacy of these methods to prevent or treat adhesion formation after surgeries. Further basic and clinical researches are still needed to propose the efficiency of recommended strategies for prevention and treatment of PPA.
Collapse
|
12
|
Khoukaz HB, Ji Y, Braet DJ, Vadali M, Abdelhamid AA, Emal CD, Lawrence DA, Fay WP. Drug Targeting of Plasminogen Activator Inhibitor-1 Inhibits Metabolic Dysfunction and Atherosclerosis in a Murine Model of Metabolic Syndrome. Arterioscler Thromb Vasc Biol 2020; 40:1479-1490. [PMID: 32268785 PMCID: PMC7255962 DOI: 10.1161/atvbaha.119.313775] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/23/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Enhanced expression of PAI-1 (plasminogen activator inhibitor-1) has been implicated in atherosclerosis formation in humans with obesity and metabolic syndrome. However, little is known about the effects of pharmacological targeting of PAI-1 on atherogenesis. This study examined the effects of pharmacological PAI-1 inhibition on atherosclerosis formation in a murine model of obesity and metabolic syndrome. Approach and Results: LDL receptor-deficient (ldlr-/-) mice were fed a Western diet high in cholesterol, fat, and sucrose to induce obesity, metabolic dysfunction, and atherosclerosis. Western diet triggered significant upregulation of PAI-1 expression compared with normal diet controls. Addition of a pharmacological PAI-1 inhibitor (either PAI-039 or MDI-2268) to Western diet significantly inhibited obesity and atherosclerosis formation for up to 24 weeks without attenuating food consumption. Pharmacological PAI-1 inhibition significantly decreased macrophage accumulation and cell senescence in atherosclerotic plaques. Recombinant PAI-1 stimulated smooth muscle cell senescence, whereas a PAI-1 mutant defective in LRP1 (LDL receptor-related protein 1) binding did not. The prosenescent effect of PAI-1 was blocked by PAI-039 and R2629, a specific anti-LRP1 antibody. PAI-039 significantly decreased visceral adipose tissue inflammation, hyperglycemia, and hepatic triglyceride content without altering plasma lipid profiles. CONCLUSIONS Pharmacological targeting of PAI-1 inhibits atherosclerosis in mice with obesity and metabolic syndrome, while inhibiting macrophage accumulation and cell senescence in atherosclerotic plaques, as well as obesity-associated metabolic dysfunction. PAI-1 induces senescence of smooth muscle cells in an LRP1-dependent manner. These results help to define the role of PAI-1 in atherosclerosis formation and suggest a new plasma-lipid-independent strategy for inhibiting atherogenesis.
Collapse
Affiliation(s)
- Hekmat B Khoukaz
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Yan Ji
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Drew J Braet
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Manisha Vadali
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Ahmed A Abdelhamid
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Cory D Emal
- Department of Chemistry, Eastern Michigan University, Ypsilanti (C.D.E.)
| | - Daniel A Lawrence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - William P Fay
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
- Department of Medical Pharmacology & Physiology (W.P.F.), University of Missouri School of Medicine
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (W.P.F.)
| |
Collapse
|
13
|
Abstract
The paradoxical pro-tumorigenic function of plasminogen activator inhibitor 1 (PAI-1, aka Serpin E1) in cancer progression and metastasis has been the subject of an abundant scientific literature that has pointed to a pro-angiogenic role, a growth and migration stimulatory function, and an anti-apoptotic activity, all directed toward promoting tumor growth, cancer cell survival, and metastasis. With uPA, PAI-1 is among the most reliable biomarkers and prognosticators in many cancer types. More recently, a novel pro-tumorigenic function of PAI-1 in cancer-related inflammation has been demonstrated. These multifaceted activities of PAI-1 in cancer progression are explained by the complex structure of PAI-1 and its multiple functions that go beyond its anti-fibrinolytic and anti-plasminogen activation activities. However, despite the multiple evidences supporting a pro-tumorigenic role of PAI-1 in cancer, and the development of several inhibitors, targeting PAI-1, has remained elusive. In this article, the various mechanisms responsible for the pro-tumorigenic functions of PAI-1 are reviewed with emphasis on its more recently described contribution to cancer inflammation. The challenges of targeting PAI-1 in cancer therapy are then discussed.
Collapse
Affiliation(s)
- Marta Helena Kubala
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA, 90033, USA
- The Saban Research Institute of Children's Hospital, Los Angeles, CA, 90027, USA
| | - Yves Albert DeClerck
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA, 90033, USA.
- The Saban Research Institute of Children's Hospital, Los Angeles, CA, 90027, USA.
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
14
|
Liu Y, Wang L, Luo M, Chen N, Deng X, He J, Zhang L, Luo P, Wu J. Inhibition of PAI-1 attenuates perirenal fat inflammation and the associated nephropathy in high-fat diet-induced obese mice. Am J Physiol Endocrinol Metab 2019; 316:E260-E267. [PMID: 30532990 DOI: 10.1152/ajpendo.00387.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is increasingly recognized as a mediator in extracellular matrix (ECM) accumulation in diabetic nephropathy. Previous studies have implicated PAI-1 in adipose tissue (AT) expansion, while also contributing to insulin resistance. As inflammation is also known to occur in perirenal AT during obesity, we hypothesized that in a high-fat diet (HFD)-induced obese mouse model, PAI-1 contributes to macrophage-mediated inflammation and diabetic nephropathy. The HFD mice showed increased expression of PAI-1 in perirenal fat and also displayed increased fat weight and macrophage numbers. We found that the macrophage polarization, proinflammatory macrophage-M1-phenotype, including CD11c, IL-6, and monocyte chemoattractant protein-1, were increased by an HFD and decreased by either the genetic depletion of PAI-1 or treatment with the PAI-1 inhibitor, PAI-039. Similarly, an enhanced anti-inflammatory M2-phenotype, including CD206 and IL-10, was accompanied by either the genetic deletion of PAI-1 or PAI-039 treatment. Furthermore, the inhibition of PAI-1 reduced HFD-induced renal histological lesions and abated profibrotic/extracellular-matrix protein. Collectively, our findings provide support that PAI-1 contributes to the development of inflammation in perirenal fat and correlates with the development of diabetic nephropathy in HFD-induced obesity.
Collapse
Affiliation(s)
- Yong Liu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, People's Republic of China
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Lin Wang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Mao Luo
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Ni Chen
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Xin Deng
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Jing He
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Liping Zhang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
| | - Pei Luo
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, People's Republic of China
| | - Jianbo Wu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, People's Republic of China
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Laboratory for Cardiovascular Pharmacology of the Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan , People's Republic of China
- Department of Medicine and Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine and Research Service , Columbia, Missouri
- Harry S. Truman Memorial Veterans Hospital , Columbia, Missouri
| |
Collapse
|
15
|
Wang L, Chen L, Liu Z, Liu Y, Luo M, Chen N, Deng X, Luo Y, He J, Zhang L, Hill MA, Li R, Wu J. PAI-1 Exacerbates White Adipose Tissue Dysfunction and Metabolic Dysregulation in High Fat Diet-Induced Obesity. Front Pharmacol 2018; 9:1087. [PMID: 30319420 PMCID: PMC6169321 DOI: 10.3389/fphar.2018.01087] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 09/06/2018] [Indexed: 01/29/2023] Open
Abstract
Background: Plasminogen activator inhibitor (PAI)-1 levels and activity are known to increase during metabolic syndrome and obesity. In addition, previous studies have implicated PAI-1 in adipose tissue (AT) expansion while also contributing to insulin resistance. As inflammation is also known to occur in AT during obesity, we hypothesized that in a high-fat diet (HFD)-induced obese mouse model PAI-1 contributes to macrophage-mediated inflammation and metabolic dysfunction. Methods: Four- to five-weeks-old male C57B6/6J mice were fed a HFD (45%) for 14 weeks, while age-matched control mice were fed a standard laboratory chow diet (10% fat). Additional studies were performed in PAI-1 knockout mice and wild type mice treated with an inhibitor (PAI-039) of PAI-1. Macrophage polarization were measured by real time PCR. Results: HFD mice showed increased expression of PAI-1 in visceral white AT (WAT) that also displayed increased macrophage numbers. PAI-1 deficient mice exhibited increased numbers of anti-inflammatory macrophages in WAT and were resistant to HFD-induced obesity. Similarly, pharmacological inhibition of PAI-1 using PAI-039 significantly decreased macrophage infiltration in WAT and improved metabolic status in HFD-induced wild-type mice. Importantly, the numbers of M1 macrophages appeared to be increased by the HFD and decreased by either genetic PAI-1 depletion or PAI-039 treatment. Conclusions: Collectively, our findings provide support for PAI-1 contributing to the development of inflammation in adipose tissue and explain the mechanism of inflammation modulated by PAI-1 in the disordered metabolism in HFD-induced obesity.
Collapse
Affiliation(s)
- Lin Wang
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liyuan Chen
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zheran Liu
- Queen Mary School, Medical College of Nanchang University, Nanchang, China
| | - Yaofang Liu
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mao Luo
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ni Chen
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xin Deng
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yulin Luo
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing He
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liping Zhang
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Michael A Hill
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Rong Li
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianbo Wu
- Drug Discovery Research Center, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
16
|
Mukherjee S, Das S, Chattopadhyay D, Sarkar S, Chatterjee SK, Talukdar D, Mukherjee S, Majumdar SS, Mukhopadhyay S, Chaudhuri MK, Bhattacharya S. Attenuation of macrophage accumulation and polarisation in obese diabetic mice by a small molecule significantly improved insulin sensitivity. Biochem Biophys Res Commun 2018; 501:771-778. [PMID: 29763604 DOI: 10.1016/j.bbrc.2018.05.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 05/11/2018] [Indexed: 10/16/2022]
Abstract
Accumulation and polarization of anti-inflammatory M2 to proinflammatory M1 macrophage in the adipose tissue of obese diabetic mice is an important pathogenic signature. It worsens lipid induced inflammation and insulin resistance. Here we demonstrate that a small molecule, a peroxyvanadate compound i.e. DmpzH [VO(O2)2 (dmpz)] or dmp, could robustly decrease macrophage infiltration, accumulation and their polarization in high fat diet (HFD) induced obese diabetic mice. In searching the underlying mechanism it was revealed that SIRT1 level was strikingly low in the inflamed adipose tissue of HFD mice as compared to mice fed with standard diet (SD). Administration of dmp markedly increased SIRT1 level by inducing its gene expression with a consequent decrease in macrophage population. Elevation of SIRT1 coincided with the decrease of MCP1, Fetuin-A (FetA) and IFNγ. Since MCP1 and FetA drive macrophage to inflamed adipose tissue and IFNγ promotes M2 to M1 transformation, both recruitment and M1 induced inflammation were found to be significantly repressed by dmp. In addressing the question about how dmp induced excess SIRT1 could reduce MCP1, FetA and IFNγ levels, we found that it was due to the inactivation of NFκB because of its deacetylation by SIRT1. Since NFκB is the transcriptional regulator of these molecules, their expressions were significantly suppressed and that caused sharp decline in macrophage recruitment and their polarity to M1. This effected a marked fall in proinflammatory cytokine level which significantly improved insulin sensitivity. dmp is likely to be the first molecule that rescues inflammatory burden contributed by macrophage in obese diabetic mice adipose tissue which causes significant increase in insulin sensitivity therefore it may be a meaningful choice to treat type 2 diabetes.
Collapse
Affiliation(s)
- Sandip Mukherjee
- Department of Zoology, Visva-Bharati (A Central University), Santiniketan, India
| | - Snehasis Das
- Department of Zoology, Visva-Bharati (A Central University), Santiniketan, India
| | | | - Shuvasree Sarkar
- Department of Botany, Visva-Bharati (A Central University), Santiniketan, India
| | | | | | - Sutapa Mukherjee
- Department of Zoology, Visva-Bharati (A Central University), Santiniketan, India
| | | | - Satinath Mukhopadhyay
- Department of Endrocrinology & Metabolism, Institute of Post-graduate Medical Education & Research-SSKM Hospital, Kolkata, India
| | | | - Samir Bhattacharya
- Department of Zoology, Visva-Bharati (A Central University), Santiniketan, India.
| |
Collapse
|
17
|
Nakagawa M, Uno S, Iriyama N, Matsunawa M, Makishima M, Takeuchi J, Tsuboi I, Hatta Y, Takei M. Combined treatment with benzo[a]pyrene and 1α,25-dihydroxyvitamin D 3 induces expression of plasminogen activator inhibitor 1 in monocyte/macrophage-derived cells. Toxicol Appl Pharmacol 2018. [PMID: 29524502 DOI: 10.1016/j.taap.2018.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Benzo[a]pyrene (BaP) is an environmental pollutant found in cigarette smoke and is implicated as a causative agent of tobacco-related diseases, such as arteriosclerosis. In contrast, vitamin D signaling, which is principally mediated by conversion of vitamin D to the active form, 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], decreases cardiovascular disease risk. However, combined treatment with BaP and 1,25(OH)2D3 enhances BaP toxicity, including BaP-DNA adduct formation. We further investigated the cross-talk between BaP and 1,25(OH)2D3 signaling pathways, and found that combined treatment with these compounds induces mRNA and protein expression of plasminogen activator inhibitor 1 (PAI-1) in monocyte/macrophage-derived THP-1 and U937 cells. Protein synthesis inhibitor treatment did not inhibit induction of the PAI-1 gene (SERPINE1) in these cells. BaP plus 1,25(OH)2D3 induced differentiation markers, inhibited cellular proliferation, and induced apoptosis and oxidative stress in these cells. Reactive oxygen species scavenger treatment suppressed apoptosis but not SERPINE1 induction in cells treated with BaP plus 1,25(OH)2D3. Thus, combined treatment with BaP and 1,25(OH)2D3 induced SERPINE1 mRNA expression in these cells through a mechanism that does not require de novo protein synthesis or reactive oxygen species production. These findings suggest that induction of the proinflammatory factor PAI-1 plays a role in BaP toxicity. Interestingly, PAI-1 knockdown decreased expression of the cell surface antigen CD14, a monocytic differentiation marker, in THP-1 cells treated with BaP plus 1,25(OH)2D3. PAI-1 induction may also be related to a function of monocytes/macrophages in response to xenobiotic and vitamin D signaling.
Collapse
Affiliation(s)
- Masaru Nakagawa
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Shigeyuki Uno
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Noriyoshi Iriyama
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Manabu Matsunawa
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan.
| | - Jin Takeuchi
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Isao Tsuboi
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Yoshihiro Hatta
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Masami Takei
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| |
Collapse
|
18
|
Tsuge M, Osaki M, Sasaki R, Hirahata M, Okada F. SK-216, a Novel Inhibitor of Plasminogen Activator Inhibitor-1, Suppresses Lung Metastasis of Human Osteosarcoma. Int J Mol Sci 2018; 19:ijms19030736. [PMID: 29510576 PMCID: PMC5877597 DOI: 10.3390/ijms19030736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/17/2018] [Accepted: 03/02/2018] [Indexed: 11/16/2022] Open
Abstract
Lung metastasis constitutes the leading cause of the death in patients with osteosarcoma. We have previously reported that plasminogen activator inhibitor-1 (PAI-1) regulates the invasion and lung metastasis of osteosarcoma cells in a mouse model and as well as in clinical samples. In the present study, we examined the anti-metastatic effect of SK-216, a small compound PAI-1 inhibitor, in human 143B osteosarcoma cells. An in vitro study showed that SK-216 treatment suppressed invasion activity by inhibiting PAI-1 expression in 143B cells, but had no influence on their proliferation or migration. 143B cells treated with SK-216 exhibited reduced matrix metalloproteinase-13 (MMP-13) secretion in a dose-dependent manner. Moreover, intraperitoneal injection of SK-216 into mouse models resulted in downregulation of PAI-1 expression levels in the primary tumors and showed suppression of lung metastases without influencing the proliferative activity of the tumor cells in the primary lesions. These results indicate that SK-216, a PAI-1 inhibitor, may serve as a novel drug to prevent lung metastasis in human osteosarcoma.
Collapse
Affiliation(s)
- Minori Tsuge
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| | - Mitsuhiko Osaki
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| | - Ryo Sasaki
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| | - Mio Hirahata
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| | - Futoshi Okada
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| |
Collapse
|
19
|
Plasma Hemopexin ameliorates murine spinal cord injury by switching microglia from the M1 state to the M2 state. Cell Death Dis 2018; 9:181. [PMID: 29415995 PMCID: PMC5833847 DOI: 10.1038/s41419-017-0236-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/28/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) is a devastating type of central nervous system (CNS) trauma with limited therapeutic treatments. The polarization of microglia into the M1 or M2 state has been documented to play important roles in the pathogenesis of SCI, although the complete repertoire of underlying factors has not been identified. Interestingly, the time point at which hematomyelia (intramedullary spinal cord hemorrhage) is alleviated coincides with a decrease in the number of M2 microglia. Here the function of Hemopexin (Hpx), a hematogenous glycoprotein, was examined in the crush model of SCI. Hpx levels were elevated at the lesion site during hematomyelia and were synchronously correlated with the level of the M2 marker Arginase-1 (Arg-1). Ablation of Hpx in vivo affected the polarization state of lipopolysaccharide (LPS)-stimulated microglia, as mirrored by a lower percentage of M2 microglia and a higher percentage of M1 microglia in the lesion site, which delayed the recovery and exacerbated the behavioral dysfunction after SCI. However, Hpx induced a rapid switch from the M1 to M2 phenotype in LPS-stimulated primary cultured microglia in a heme scavenging-independent manner. The supernant of Hpx-treated microglia ameliorated neuronal degeneration, alleviated demyelination, and promoted oligodendrocyte precursor cell (OPC) maturation. This modulatory effect of Hpx on microglia polarization was at least partially mediated by the LRP-1 receptor. Based on these results, Hpx is considered a novel modulator of the polarization of microglia during the pathogenesis of SCI and may play a crucial role in the recovery from SCI.
Collapse
|
20
|
Fang WF, Chen YM, Lin CY, Huang HL, Yeh H, Chang YT, Huang KT, Lin MC. Histone deacetylase 2 (HDAC2) attenuates lipopolysaccharide (LPS)-induced inflammation by regulating PAI-1 expression. JOURNAL OF INFLAMMATION-LONDON 2018; 15:3. [PMID: 29344006 PMCID: PMC5763578 DOI: 10.1186/s12950-018-0179-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
Background Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to infection, and is primarily characterized by an uncontrolled systemic inflammatory response. In the present study, we developed an effective adjunct therapy mediated by a novel mechanism, to attenuate overt inflammation. LPS-treated macrophages were adopted as an in vitro model of endotoxin-induced inflammation during sepsis. Experiments were carried out using primary mouse peritoneal macrophages and the murine macrophage cell line RAW264.7, to elucidate the mechanisms by which HDAC2 modulates endotoxin-induced inflammation. Results Results revealed that PAI-1, TNF, and MIP-2 expression were inhibited by theophylline, an HDAC2 enhancer, in a RAW macrophage cell line, following LPS-induced inflammation. Thus, HDAC2 plays an important role in immune defense by regulating the expression of inflammatory genes via the c-Jun/PAI-1 pathway. During LPS-induced inflammation, overexpression of HDAC2 was found to inhibit PAI-1, TNF, and MIP-2 expression. Following LPS stimulation, HDAC2 knockdown increased nuclear translocation and DNA binding of c-Jun to the PAI-1 gene promoter, thereby activating PAI-1 gene transcription. Furthermore, inhibition of PAI-1 by TM5275 alone or in combination with theophylline notably suppressed TNF and MIP-2 expression. Conclusion HDAC2 can attenuate lipopolysaccharide-induced inflammation by regulating c-Jun and PAI-1 expression in macrophages.
Collapse
Affiliation(s)
- Wen-Feng Fang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan.,2Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta-Pei Rd, Niao-Sung Dist, Kaohsiung, 833 Taiwan.,3Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, 813 Taiwan
| | - Yu-Mu Chen
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Chiung-Yu Lin
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Hui-Lin Huang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Hua Yeh
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Ya-Ting Chang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Kuo-Tung Huang
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan
| | - Meng-Chih Lin
- 1Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 833 Taiwan.,2Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta-Pei Rd, Niao-Sung Dist, Kaohsiung, 833 Taiwan
| |
Collapse
|
21
|
Yamaoka N, Murano K, Kodama H, Maeda A, Dan T, Nakabayashi T, Miyata T, Meguro K. Identification of novel plasminogen activator inhibitor-1 inhibitors with improved oral bioavailability: Structure optimization of N-acylanthranilic acid derivatives. Bioorg Med Chem Lett 2018; 28:809-813. [PMID: 29366646 DOI: 10.1016/j.bmcl.2017.11.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/03/2017] [Accepted: 11/08/2017] [Indexed: 12/20/2022]
Abstract
Novel plasminogen activator inhibitor-1 (PAI-1) inhibitors with highly improved oral bioavailability were discovered by structure-activity relationship studies on N-acyl-5-chloroanthranilic acid derivatives. Because lipophilic N-acyl groups seemed to be important for the anthranilic acid derivatives to strongly inhibit PAI-1, synthesis of compounds in which 5-chloroanthranilic acid was bound to a variety of highly lipophilic moieties with appropriate linkers was investigated. As the result it appeared that some of the derivatives possessing aryl- or heteroaryl-substituted phenyl groups in the acyl chain had potent in vitro PAI-1 inhibitory activity. Oral absorbability of typical compounds was also evaluated in rats, and compounds 40, 55, 60 and 76 which have diverse chemical structure with each other were selected for further pharmacological evaluation.
Collapse
Affiliation(s)
- Nagahisa Yamaoka
- CT Laboratory, Hamari Chemicals, Ltd., 1-4-29 Kunijima, Higashiyodogawa-ku, Osaka 533-0024, Japan.
| | - Kenji Murano
- CT Laboratory, Hamari Chemicals, Ltd., 1-4-29 Kunijima, Higashiyodogawa-ku, Osaka 533-0024, Japan
| | - Hidehiko Kodama
- CT Laboratory, Hamari Chemicals, Ltd., 1-4-29 Kunijima, Higashiyodogawa-ku, Osaka 533-0024, Japan
| | - Akihisa Maeda
- CT Laboratory, Hamari Chemicals, Ltd., 1-4-29 Kunijima, Higashiyodogawa-ku, Osaka 533-0024, Japan
| | - Takashi Dan
- United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Tetsuo Nakabayashi
- United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kanji Meguro
- CT Laboratory, Hamari Chemicals, Ltd., 1-4-29 Kunijima, Higashiyodogawa-ku, Osaka 533-0024, Japan
| |
Collapse
|
22
|
Lee SM, Dorotea D, Jung I, Nakabayashi T, Miyata T, Ha H. TM5441, a plasminogen activator inhibitor-1 inhibitor, protects against high fat diet-induced non-alcoholic fatty liver disease. Oncotarget 2017; 8:89746-89760. [PMID: 29163785 PMCID: PMC5685706 DOI: 10.18632/oncotarget.21120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/03/2017] [Indexed: 12/16/2022] Open
Abstract
Recent evidences showed that elevation of plasminogen activator inhibitor 1 (PAI-1) was responsible in mediating obesity-induced non-alcoholic fatty liver disease (NAFLD) and metabolic disorders. Here, we investigated the effect of TM5441, an oral PAI-1 inhibitor that lacks of bleeding risk, on high-fat diet (HFD)-induced NAFLD. HFD-fed C57BL/6J mice was daily treated with 20 mg/kg TM5441. To examine the preventive effect, 10-week-treatment was started along with initiation of HFD; alternatively, 4-week-treatment was started in mice with glucose intolerance in the interventional strategy. In vivo study showed that early and delayed treatment decreased hepatic steatosis. Particularly, early treatment prevented the progression of hepatic inflammation and fibrosis in HFD mice. Interestingly, both strategies abrogated hepatic insulin resistance and mitochondrial dysfunction, presented by enhanced p-Akt and p-GSK3β, reduced p-JNK signaling, along with p-AMPK and PGC-1α activation. Consistently, TM5441 treatment in the presence of either PAI-1 exposure or TNF-α stimulated-PAI-1 activity showed a restoration of mitochondrial biogenesis related genes expression on HepG2 cells. Thus, improvement of insulin sensitivity and mitochondrial function was imperative to partially explain the therapeutic effects of TM5441, a novel agent targeting HFD-induced NAFLD.
Collapse
Affiliation(s)
- Seon Myeong Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Inji Jung
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Tetsuo Nakabayashi
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
23
|
TGF-β-induced intracellular PAI-1 is responsible for retaining hematopoietic stem cells in the niche. Blood 2017; 130:2283-2294. [PMID: 28821477 DOI: 10.1182/blood-2017-02-767384] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 08/10/2017] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) reside in the supportive stromal niche in bone marrow (BM); when needed, however, they are rapidly mobilized into the circulation, suggesting that HSPCs are intrinsically highly motile but usually stay in the niche. We questioned what determines the motility of HSPCs. Here, we show that transforming growth factor (TGF)-β-induced intracellular plasminogen activator inhibitor (PAI)-1 activation is responsible for keeping HSPCs in the BM niche. We found that the expression of PAI-1, a downstream target of TGF-β signaling, was selectively augmented in niche-residing HSPCs. Functional inhibition of the TGF-β-PAI-1 signal increased MT1-MMP-dependent cellular motility, causing a detachment of HSPCs from the TGF-β-expressing niche cells, such as megakaryocytes. Furthermore, consistently high motility in PAI-1-deficient HSPCs was demonstrated by both a transwell migration assay and reciprocal transplantation experiments, indicating that intracellular, not extracellular, PAI-1 suppresses the motility of HSPCs, thereby causing them to stay in the niche. Mechanistically, intracellular PAI-1 inhibited the proteolytic activity of proprotein convertase Furin, diminishing MT1-MMP activity. This reduced expression of MT1-MMP in turn affected the expression levels of several adhesion/deadhesion molecules for determination of HSPC localization, such as CD44, VLA-4, and CXCR4, which then promoted the retention of HSPCs in the niche. Our findings open up a new field for the study of intracellular proteolysis as a regulatory mechanism of stem cell fate, which has the potential to improve clinical HSPC mobilization and transplantation protocols.
Collapse
|
24
|
Honjo K, Munakata S, Tashiro Y, Salama Y, Shimazu H, Eiamboonsert S, Dhahri D, Ichimura A, Dan T, Miyata T, Takeda K, Sakamoto K, Hattori K, Heissig B. Plasminogen activator inhibitor‐1 regulates macrophage‐dependent postoperative adhesion by enhancing EGF‐HER1 signaling in mice. FASEB J 2017; 31:2625-2637. [DOI: 10.1096/fj.201600871rr] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/21/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Kumpei Honjo
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Shinya Munakata
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Yoshihiko Tashiro
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Yousef Salama
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Hiroshi Shimazu
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Salita Eiamboonsert
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Douaa Dhahri
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Atsuhiko Ichimura
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Takashi Dan
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Kazuyoshi Takeda
- Department of Immunology and Atopy CenterGraduate School of Medicine, Juntendo University Tokyo Japan
| | | | - Koichi Hattori
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Center for Genomic and Regenerative MedicineFaculty of Medicine Tokyo Japan
| | - Beate Heissig
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Immunology and Atopy CenterGraduate School of Medicine, Juntendo University Tokyo Japan
| |
Collapse
|
25
|
Ji Y, Weng Z, Fish P, Goyal N, Luo M, Myears SP, Strawn TL, Chandrasekar B, Wu J, Fay WP. Pharmacological Targeting of Plasminogen Activator Inhibitor-1 Decreases Vascular Smooth Muscle Cell Migration and Neointima Formation. Arterioscler Thromb Vasc Biol 2016; 36:2167-2175. [PMID: 27659097 DOI: 10.1161/atvbaha.116.308344] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/09/2016] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Plasminogen activator inhibitor-1 (PAI-1), a serine protease inhibitor that promotes and inhibits cell migration, plays a complex and important role in adverse vascular remodeling. Little is known about the effects of pharmacological PAI-1 inhibitors, an emerging drug class, on migration of vascular smooth muscle cells (SMCs) and endothelial cells (ECs), crucial mediators of vascular remodeling. We investigated the effects of PAI-039 (tiplaxtinin), a specific PAI-1 inhibitor, on SMC and EC migration in vitro and vascular remodeling in vivo. APPROACH AND RESULTS PAI-039 inhibited SMC migration through collagen gels, including those supplemented with vitronectin and other extracellular matrix proteins, but did not inhibit migration of PAI-1-deficient SMCs, suggesting that its antimigratory effects were PAI-1-specific and physiologically relevant. However, PAI-039 did not inhibit EC migration. PAI-039 inhibited phosphorylation and nuclear translocation of signal transducers and activators of transcription-1 in SMCs, but had no discernable effect on signal transducer and activator of transcription-1 signaling in ECs. Expression of low-density lipoprotein receptor-related protein 1, a motogenic PAI-1 receptor that activates Janus kinase/signal transducers and activators of transcription-1 signaling, was markedly lower in ECs than in SMCs. Notably, PAI-039 significantly inhibited intimal hyperplasia and inflammation in murine models of adverse vascular remodeling, but did not adversely affect re-endothelialization after endothelium-denuding mechanical vascular injury. CONCLUSIONS PAI-039 inhibits SMC migration and intimal hyperplasia, while having no inhibitory effect on ECs, which seems to be because of differences in PAI-1-dependent low-density lipoprotein receptor-related protein 1/Janus kinase/signal transducer and activator of transcription-1 signaling between SMCs and ECs. These findings suggest that PAI-1 may be an important therapeutic target in obstructive vascular diseases characterized by neointimal hyperplasia.
Collapse
Affiliation(s)
- Yan Ji
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Zhen Weng
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Philip Fish
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Neha Goyal
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Mao Luo
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Samantha P Myears
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Tammy L Strawn
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Bysani Chandrasekar
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - Jianbo Wu
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO
| | - William P Fay
- From the Departments of Medicine and Medical Pharmacology & Physiology, University of Missouri School of Medicine (Y.J., Z.W., P.F., N.G., M.L., S.P.M., T.L.S., B.C., J.W., W.P.F.), and the Research Service, Harry S. Truman Memorial Veterans Hospital (B.C., W.P.F.), Columbia, MO.
| |
Collapse
|
26
|
Piao L, Jung I, Huh JY, Miyata T, Ha H. A novel plasminogen activator inhibitor-1 inhibitor, TM5441, protects against high-fat diet-induced obesity and adipocyte injury in mice. Br J Pharmacol 2016; 173:2622-32. [PMID: 27339909 DOI: 10.1111/bph.13541] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/11/2016] [Accepted: 06/16/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Obesity is one of the most prevalent chronic diseases worldwide, and dysregulated adipocyte function plays an important role in obesity-associated metabolic disorder. The level of plasma plasminogen activator inhibitor-1 (PAI-1) is increased in obese subjects, and PAI-1 null mice show improved insulin sensitivity when subjected to high-fat and high-sucrose diet-induced metabolic stress, suggesting that a best-in-class PAI-1 inhibitor may become a novel therapeutic agent for obesity-associated metabolic syndrome. TM5441 is a novel orally active PAI-1 inhibitor that does not cause bleeding episodes. Hence, in the present study we examined the preventive effect of TM5441 on high-fat diet (HFD)-induced adipocyte dysfunction. EXPERIMENTAL APPROACH Ten-week-old C57BL/6J mice were fed a normal diet (18% of total calories from fat) or HFD (60% of total calories from fat) for 10 weeks, and TM5441 (20 mg·kg(-1) oral gavage) was administered daily with the initiation of HFD. KEY RESULTS TM5441 prevented HFD-induced body weight gain and systemic insulin resistance. TM5441 normalized HFD-induced dysregulated JNK and Akt phosphorylation, suggesting that it prevents the insulin resistance of adipocytes. TM5441 also attenuated the macrophage infiltration and increased expression of pro-inflammatory cytokines, such as inducible nitric oxide synthase, induced by the HFD. In addition, TM5441 prevented the HFD-induced down-regulation of genes involved in mitochondrial biogenesis and function, suggesting that it may prevent adipocyte inflammation and dysregulation by maintaining mitochondrial fitness. CONCLUSION AND IMPLICATIONS Our data suggest that TM5441 may become a novel therapeutic agent for obesity and obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Lingjuan Piao
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Inji Jung
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Joo Young Huh
- Colleges of Pharmacy, Chonnam National University, Gwang-Ju, Korea
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
27
|
Novel Plasminogen Activator Inhibitor-1 Inhibitors Prevent Diabetic Kidney Injury in a Mouse Model. PLoS One 2016; 11:e0157012. [PMID: 27258009 PMCID: PMC4892642 DOI: 10.1371/journal.pone.0157012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 05/22/2016] [Indexed: 01/13/2023] Open
Abstract
Diabetic nephropathy is the leading cause of end-stage renal disease worldwide, but no effective therapeutic strategy is available. Because plasminogen activator inhibitor-1 (PAI-1) is increasingly recognized as a key factor in extracellular matrix (ECM) accumulation in diabetic nephropathy, this study examined the renoprotective effects of TM5275 and TM5441, two novel orally active PAI-1 inhibitors that do not trigger bleeding episodes, in streptozotocin (STZ)-induced diabetic mice. TM5275 (50 mg/kg) and TM5441 (10 mg/kg) were administered orally for 16 weeks to STZ-induced diabetic and age-matched control mice. Relative to the control mice, the diabetic mice showed significantly increased (p < 0.05) plasma glucose and creatinine levels, urinary albumin excretion, kidney-to-bodyweight ratios, glomerular volume, and fractional mesangial area. Markers of fibrosis and inflammation along with PAI-1 were also upregulated in the kidney of diabetic mice, and treatment with TM5275 and TM5441 effectively inhibited albuminuria, mesangial expansion, ECM accumulation, and macrophage infiltration in diabetic kidneys. Furthermore, in mouse proximal tubular epithelial (mProx24) cells, both TM5275 and TM5441 effectively inhibited PAI-1-induced mRNA expression of fibrosis and inflammation markers and also reversed PAI-1-induced inhibition of plasmin activity, which confirmed the efficacy of the TM compounds as PAI-1 inhibitors. These data suggest that TM compounds could be used to prevent diabetic kidney injury.
Collapse
|
28
|
Liu RM, Eldridge S, Watanabe N, Deshane J, Kuo HC, Jiang C, Wang Y, Liu G, Schwiebert L, Miyata T, Thannickal VJ. Therapeutic potential of an orally effective small molecule inhibitor of plasminogen activator inhibitor for asthma. Am J Physiol Lung Cell Mol Physiol 2015; 310:L328-36. [PMID: 26702150 DOI: 10.1152/ajplung.00217.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/19/2015] [Indexed: 11/22/2022] Open
Abstract
Asthma is one of the most common respiratory diseases. Although progress has been made in our understanding of airway pathology and many drugs are available to relieve asthma symptoms, there is no cure for chronic asthma. Plasminogen activator inhibitor 1 (PAI-1), a primary inhibitor of tissue-type and urokinase-type plasminogen activators, has pleiotropic functions besides suppression of fibrinolysis. In this study, we show that administration of TM5275, an orally effective small-molecule PAI-1 inhibitor, 25 days after ovalbumin (OVA) sensitization-challenge, significantly ameliorated airway hyperresponsiveness in an OVA-induced chronic asthma model. Furthermore, we show that TM5275 administration significantly attenuated OVA-induced infiltration of inflammatory cells (neutrophils, eosinophils, and monocytes), the increase in the levels of OVA-specific IgE and Th2 cytokines (IL-4 and IL-5), the production of mucin in the airways, and airway subepithelial fibrosis. Together, the results suggest that the PAI-1 inhibitor TM5275 may have therapeutic potential for asthma through suppressing eosinophilic allergic response and ameliorating airway remodeling.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Stephanie Eldridge
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Nobuo Watanabe
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Tohoku, Japan
| | - Jessy Deshane
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hui-Chien Kuo
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Chunsun Jiang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yong Wang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lisa Schwiebert
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Tohoku, Japan
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
29
|
Zakaria E, Al-Din MH, Ghanem NS, Sadik NA, Assem M, Taha F. Urinary albumin excretion and progression of renal disease with impaired fibrinolytic activity in type 2 diabetes mellitus. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2015. [DOI: 10.4103/1110-7782.165450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
30
|
Rebalka IA, Raleigh MJ, D'Souza DM, Coleman SK, Rebalka AN, Hawke TJ. Inhibition of PAI-1 Via PAI-039 Improves Dermal Wound Closure in Diabetes. Diabetes 2015; 64:2593-602. [PMID: 25754958 DOI: 10.2337/db14-1174] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/02/2015] [Indexed: 11/13/2022]
Abstract
Diabetes impairs the ability to heal cutaneous wounds, leading to hospitalization, amputations, and death. Patients with diabetes experience elevated levels of plasminogen activator inhibitor 1 (PAI-1), regardless of their glycemic control. It has been demonstrated that PAI-1-deficient mice exhibit improved cutaneous wound healing, and that PAI-1 inhibition improves skeletal muscle repair in mice with type 1 diabetes mellitus, leading us to hypothesize that pharmacologically mediated reductions in PAI-1 using PAI-039 would normalize cutaneous wound healing in streptozotocin (STZ)-induced diabetic (STZ-diabetic) mice. To simulate the human condition of variations in wound care, wounds were aggravated or minimally handled postinjury. Following cutaneous injury, PAI-039 was orally administered twice daily for 10 days. Compared with nondiabetic mice, wounds in STZ-diabetic mice healed more slowly. Wound site aggravation exacerbated this deficit. PAI-1 inhibition had no effect on dermal collagen levels or wound bed size. PAI-039 treatment failed to improve angiogenesis in the wounds of STZ-diabetic mice and blunted angiogenesis in the wounds of nondiabetic mice. Importantly, PAI-039 treatment significantly improved epidermal cellular migration and wound re-epithelialization compared with vehicle-treated STZ-diabetic mice. These findings support the use of PAI-039 as a novel therapeutic agent to improve diabetic wound closure and demonstrate the primary mechanism of its action to be related to epidermal closure.
Collapse
Affiliation(s)
- Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Matthew J Raleigh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Donna M D'Souza
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Samantha K Coleman
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alexandra N Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
31
|
Pelisch N, Dan T, Ichimura A, Sekiguchi H, Vaughan DE, van Ypersele de Strihou C, Miyata T. Plasminogen Activator Inhibitor-1 Antagonist TM5484 Attenuates Demyelination and Axonal Degeneration in a Mice Model of Multiple Sclerosis. PLoS One 2015; 10:e0124510. [PMID: 25915660 PMCID: PMC4411110 DOI: 10.1371/journal.pone.0124510] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/15/2015] [Indexed: 11/20/2022] Open
Abstract
Multiple sclerosis (MS) is characterized by inflammatory demyelination and deposition of fibrinogen in the central nervous system (CNS). Elevated levels of a critical inhibitor of the mammalian fibrinolitic system, plasminogen activator inhibitor 1 (PAI-1) have been demonstrated in human and animal models of MS. In experimental studies that resemble neuroinflammatory disease, PAI-1 deficient mice display preserved neurological structure and function compared to wild type mice, suggesting a link between the fibrinolytic pathway and MS. We previously identified a series of PAI-1 inhibitors on the basis of the 3-dimensional structure of PAI-1 and on virtual screening. These compounds have been reported to provide a number of in vitro and in vivo benefits but none was tested in CNS disease models because of their limited capacity to penetrate the blood-brain barrier (BBB). The existing candidates were therefore optimized to obtain CNS-penetrant compounds. We performed an in vitro screening using a model of BBB and were able to identify a novel, low molecular PAI-1 inhibitor, TM5484, with the highest penetration ratio among all other candidates. Next, we tested the effects on inflammation and demyelination in an experimental allergic encephalomyelitis mice model. Results were compared to either fingolimod or 6α-methylprednisolone. Oral administration of TM5484 from the onset of signs, ameliorates paralysis, attenuated demyelination, and axonal degeneration in the spinal cord of mice. Furthermore, it modulated the expression of brain-derived neurotrophic factor, which plays a protective role in neurons against various pathological insults, and choline acetyltransferase, a marker of neuronal density. Taken together, these results demonstrate the potential benefits of a novel PAI-1 inhibitor, TM5484, in the treatment of MS.
Collapse
Affiliation(s)
- Nicolas Pelisch
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- * E-mail:
| | - Takashi Dan
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Atsuhiko Ichimura
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroki Sekiguchi
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Douglas E. Vaughan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | | | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
32
|
Affiliation(s)
- Ziad Mallat
- From the Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; and Institut National de la Santé et de la Recherche Médicale, U970, Paris, France.
| |
Collapse
|
33
|
Kobayashi N, Ueno T, Ohashi K, Yamashita H, Takahashi Y, Sakamoto K, Manabe S, Hara S, Takashima Y, Dan T, Pastan I, Miyata T, Kurihara H, Matsusaka T, Reiser J, Nagata M. Podocyte injury-driven intracapillary plasminogen activator inhibitor type 1 accelerates podocyte loss via uPAR-mediated β1-integrin endocytosis. Am J Physiol Renal Physiol 2015; 308:F614-26. [PMID: 25587125 PMCID: PMC4360033 DOI: 10.1152/ajprenal.00616.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/08/2015] [Indexed: 11/22/2022] Open
Abstract
Podocyte-endothelial cell cross-talk is paramount for maintaining the filtration barrier. The present study investigated the endothelial response to podocyte injury and its subsequent role in glomerulosclerosis using the podocyte-specific injury model of NEP25/LMB2 mice. NEP25/LMB2 mice showed proteinuria and local podocyte loss accompanied by thrombotic microangiopathy on day 8. Mice showed an increase of glomerular plasminogen activator inhibitor type 1 (PAI-1) mRNA and aberrant endothelial PAI-1 protein already on day 1, before thrombosis and proteinuria. A PAI-1-specific inhibitor reduced proteinuria and thrombosis and preserved podocyte numbers in NEP25/LMB2 mice by stabilization of β1-integrin translocation. Heparin loading significantly reduced thrombotic formation, whereas proteinuria and podocyte numbers were unchanged. Immortalized podocytes treated with PAI-1 and the urokinase plasminogen activator (uPA) complex caused significant cell detachment, whereas podocytes treated with PAI-1 or uPA alone or with the PAI-1/uPA complex pretreated with an anti-uPA receptor (uPAR) antibody failed to cause detachment. Confocal microscopy and cell surface biotinylation experiments showed that internalized β1-integrin was found together with uPAR in endocytotic vesicles. The administration of PAI-1 inhibitor or uPAR-blocking antibody protected cultured podocytes from cell detachment. In conclusion, PAI-1/uPA complex-mediated uPAR-dependent podocyte β1-integrin endocytosis represents a novel mechanism of glomerular injury leading to progressive podocytopenia. This aberrant cross-talk between podocytes and endothelial cells represents a feedforward injury response driving podocyte loss and progressive glomerulosclerosis.
Collapse
Affiliation(s)
- Namiko Kobayashi
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Toshiharu Ueno
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kumi Ohashi
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Hanako Yamashita
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Yukina Takahashi
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kazuo Sakamoto
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Shun Manabe
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Satoshi Hara
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Yasutoshi Takashima
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Takashi Dan
- Center for Translational and Advanced Research, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Toshio Miyata
- Center for Translational and Advanced Research, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hidetake Kurihara
- Department of Anatomy, Juntendo University School of Medicine, Bunkyo, Tokyo, Japan
| | - Taiji Matsusaka
- Department of Internal Medicine, Institute of Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan; and
| | - Jochen Reiser
- Department of Medicine, Rush University, Chicago, Illinois
| | - Michio Nagata
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan;
| |
Collapse
|
34
|
Inhibition of SERPINE1 Function Attenuates Wound Closure in Response to Tissue Injury: A Role for PAI-1 in Re-Epithelialization and Granulation Tissue Formation. J Dev Biol 2015. [DOI: 10.3390/jdb3010011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
35
|
Mashiko S, Kitatani K, Toyoshima M, Ichimura A, Dan T, Usui T, Ishibashi M, Shigeta S, Nagase S, Miyata T, Yaegashi N. Inhibition of plasminogen activator inhibitor-1 is a potential therapeutic strategy in ovarian cancer. Cancer Biol Ther 2015; 16:253-60. [PMID: 25587663 PMCID: PMC4623014 DOI: 10.1080/15384047.2014.1001271] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/14/2014] [Accepted: 12/18/2014] [Indexed: 10/22/2022] Open
Abstract
Plasminogen activator inhibitor (PAI)-1 is predictive of poor outcome in several types of cancer. The present study investigated the biological role for PAI-1 in ovarian cancer and potential of targeted pharmacotherapeutics. In patients with ovarian cancer, PAI-1 mRNA expression in tumor tissues was positively correlated with poor prognosis. To determine the role of PAI-1 in cell proliferation in ovarian cancer, the effects of PAI-1 inhibition were examined in PAI-1-expressing ovarian cancer cells. PAI-1 knockdown by small interfering RNA resulted in significant suppression of cell growth accompanied with G2/M cell cycle arrest and intrinsic apoptosis. Similarly, treatment with the small molecule PAI-1 inhibitor TM5275 effectively blocked cell proliferation of ovarian cancer cells that highly express PAI-1. Together these results suggest that PAI-1 promotes cell growth in ovarian cancer. Interestingly, expression of PAI-1 was increased in ovarian clear cell carcinoma compared with that in serous tumors. Our results suggest that PAI-1 inhibition promotes cell cycle arrest and apoptosis in ovarian cancer and that PAI-1 inhibitors potentially represent a novel class of anti-tumor agents.
Collapse
Affiliation(s)
- Satsuki Mashiko
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Kazuyuki Kitatani
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; Sendai, Japan
- Tohoku Medical Megabank Organization; Tohoku University; Sendai, Japan
| | - Masafumi Toyoshima
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Atsuhiko Ichimura
- Division of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Dan
- Division of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshinori Usui
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; Sendai, Japan
- Tohoku Medical Megabank Organization; Tohoku University; Sendai, Japan
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Shogo Shigeta
- Department of Obstetrics and Gynecology; Yamagata University Faculty of Medicine; Yamagata, Japan
| | - Satoru Nagase
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Toshio Miyata
- Division of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; Sendai, Japan
| |
Collapse
|
36
|
Huang WT, Akhter H, Jiang C, MacEwen M, Ding Q, Antony V, Thannickal VJ, Liu RM. Plasminogen activator inhibitor 1, fibroblast apoptosis resistance, and aging-related susceptibility to lung fibrosis. Exp Gerontol 2014; 61:62-75. [PMID: 25451236 DOI: 10.1016/j.exger.2014.11.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/24/2014] [Accepted: 11/27/2014] [Indexed: 12/31/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disorder with unknown cause and no effective treatment. The incidence of and mortality from IPF increase with age, suggesting that advanced age is a major risk factor for IPF. The mechanism underlying the increased susceptibility of the elderly to IPF, however, is unknown. In this study, we show for the first time that the protein level of plasminogen activator inhibitor 1 (PAI-1), a protease inhibitor which plays an essential role in the control of fibrinolysis, was significantly increased with age in mouse lung homogenate and lung fibroblasts. Upon bleomycin challenge, old mice experienced augmented PAI-1 induction and lung fibrosis as compared to young mice. Most interestingly, we show that fewer (myo)fibroblasts underwent apoptosis and more (myo)fibroblasts with increased level of PAI-1 accumulated in the lung of old than in young mice after bleomycin challenge. In vitro studies further demonstrate that fibroblasts isolated from lungs of old mice were resistant to H2O2 and tumor necrosis factor alpha-induced apoptosis and had augmented fibrotic responses to TGF-β1, compared to fibroblasts isolated from young mice. Inhibition of PAI-1 activity with a PAI-1 inhibitor, on the other hand, eliminated the aging-related apoptosis resistance and TGF-β1 sensitivity in isolated fibroblasts. Moreover, we show that knocking down PAI-1 in human lung fibroblasts with PAI-1 siRNA significantly increased their sensitivity to apoptosis and inhibited their responses to TGF-β1. Together, the results suggest that increased PAI-1 expression may underlie the aging-related sensitivity to lung fibrosis in part by protecting fibroblasts from apoptosis.
Collapse
Affiliation(s)
- Wen-Tan Huang
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, USA
| | - Hasina Akhter
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, USA
| | - Chunsun Jiang
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Mark MacEwen
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, USA
| | - Qiang Ding
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Veena Antony
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Victor John Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Rui-Ming Liu
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, USA.
| |
Collapse
|
37
|
Miyata T, Ando T, Hiragi H, Watanabe K, Yamamoto F, Vaughan DE, Kurokawa T, Oshima Y, van Ypersele de Strihou C, Takeuchi M. Drug discovery in renal disease—towards a more efficient framework. Nat Rev Nephrol 2014; 10:290-6. [DOI: 10.1038/nrneph.2014.36] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Zhang Y, Wang R, Li S, Kong X, Wang Z, Chen W, Lin N. Genetic polymorphisms in plasminogen activator inhibitor-1 predict susceptibility to steroid-induced osteonecrosis of the femoral head in Chinese population. Diagn Pathol 2013; 8:169. [PMID: 24135164 PMCID: PMC4016530 DOI: 10.1186/1746-1596-8-169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/08/2013] [Indexed: 12/11/2022] Open
Abstract
Background Steroid usage has been considered as a leading cause of non-traumatic osteonecrosis of the femoral head (ONFH), which is involved in hypo-fibrinolysis and blood supply interruption. Genetic polymorphisms in plasminogen activator inhibitor-1 (PAI-1) have been demonstrated to be associated with ONFH risk in several populations. However, this relationship has not been established in Chinese population. The aim of this study was to investigate the association of PAI-1 gene polymorphisms with steroid-induced ONFH in a large cohort of Chinese population. Methods A case–control study was conducted, which included 94 and 106 unrelated patients after steroid administration recruited from 14 provinces in China, respectively. Two SNPs (rs11178 and rs2227631) within PAI-1 were genotyped using Sequenom MassARRAY system. Results rs2227631 SNP was significantly associated with steroid-induced ONFH group in codominant (P = 0.04) and recessive (P = 0.02) models. However, there were no differences found in genotype frequencies of rs11178 SNP between controls and patients with steroid-induced ONFH (all P > 0.05). Conclusions Our data offer the convincing evidence for the first time that rs2227631 SNP of PAI-1 may be associated with the risk of steroid-induced ONFH, suggesting that the genetic variations of this gene may play an important role in the disease development. Virtual slides The virtual slide(s) for this article can be found here:
http://www.diagnosticpathology.diagnomx.eu/vs/1569909986109783.
Collapse
Affiliation(s)
| | | | | | | | | | - Weiheng Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No, 16, Nanxiaojie, Dongzhimennei, Beijing 100700, China.
| | | |
Collapse
|