1
|
Lin M, Wang L, Guan B, Tang S, Lin L, Wu K, Huang Q, He G, Zhang Z, Gao R, Liu X, Liu X, Chen Z, Liu L. Effect of PAI-1 inhibitor on pancreatic islet function and hepatic insulin resistance in db/db mice. Biochem Pharmacol 2025; 237:116906. [PMID: 40158817 DOI: 10.1016/j.bcp.2025.116906] [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: 10/30/2024] [Revised: 02/27/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a global health challenge, necessitating innovative antidiabetic treatments. Levels of plasminogen activator inhibitor-1 (PAI-1) are elevated in patients with T2DM and may be an important but underappreciated risk factor for diabetes. However, its relationship with T2DM remains unclear. To this end, we developed a potent and highly specific PAI-1 inhibitor named PAItrap3. We aimed to elucidate the metabolic effects of PAItrap3 using a preclinical db/db mouse model. PAItrap3 was administered to mice intravenously, followed by an assessment of biochemical markers, histopathological examination of the liver and pancreas, and evaluation of the expression of hepatic proteins integral to insulin signaling. PAItrap3 demonstrated potent efficacy in alleviating hyperglycemia and enhancing glycemic control. This therapeutic action was supported by its ability to enhance β-cell function, consequently mitigating β-cell apoptosis and preserving their integrity. Furthermore, PAItrap3 alleviated hepatic insulin resistance through the regulation of lipid and glucose metabolism, thereby maintaining the delicate homeostasis of systemic lipid and glucose metabolism. These findings suggest that PAItrap3 is a promising therapeutic candidate for T2DM. The multifaceted benefits of PAItrap3 highlight its potential to vastly improve the effectiveness and specificity of T2DM treatment paradigms.
Collapse
Affiliation(s)
- Menghua Lin
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Lijing Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Binbin Guan
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Shuzhi Tang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian Academy, University of Chinese Academy of Sciences, Fuzhou, Fujian 350108, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lu Lin
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Kejun Wu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Qintao Huang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Guanlian He
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Zhouyangyang Zhang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Ruonan Gao
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Xiaoying Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - XiaoHong Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Zhuo Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian Academy, University of Chinese Academy of Sciences, Fuzhou, Fujian 350108, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Libin Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China.
| |
Collapse
|
2
|
Qu F, Brough SC, Michno W, Madubata CJ, Hartmann GG, Puno A, Drainas AP, Bhattacharya D, Tomasich E, Lee MC, Yang D, Kim J, Peiris-Pagès M, Simpson KL, Dive C, Preusser M, Toland A, Kong C, Das M, Winslow MM, Pasca AM, Sage J. Crosstalk between small-cell lung cancer cells and astrocytes mimics brain development to promote brain metastasis. Nat Cell Biol 2023; 25:1506-1519. [PMID: 37783795 PMCID: PMC11230587 DOI: 10.1038/s41556-023-01241-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/25/2023] [Indexed: 10/04/2023]
Abstract
Brain metastases represent an important clinical problem for patients with small-cell lung cancer (SCLC). However, the mechanisms underlying SCLC growth in the brain remain poorly understood. Here, using intracranial injections in mice and assembloids between SCLC aggregates and human cortical organoids in culture, we found that SCLC cells recruit reactive astrocytes to the tumour microenvironment. This crosstalk between SCLC cells and astrocytes drives the induction of gene expression programmes that are similar to those found during early brain development in neurons and astrocytes. Mechanistically, the brain development factor Reelin, secreted by SCLC cells, recruits astrocytes to brain metastases. These astrocytes in turn promote SCLC growth by secreting neuronal pro-survival factors such as SERPINE1. Thus, SCLC brain metastases grow by co-opting mechanisms involved in reciprocal neuron-astrocyte interactions during brain development. Targeting such developmental programmes activated in this cancer ecosystem may help prevent and treat brain metastases.
Collapse
Affiliation(s)
- Fangfei Qu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Siqi C Brough
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Wojciech Michno
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Chioma J Madubata
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Griffin G Hartmann
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alyssa Puno
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexandros P Drainas
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Debadrita Bhattacharya
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Erwin Tomasich
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Myung Chang Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dian Yang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jun Kim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria Peiris-Pagès
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Kathryn L Simpson
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Caroline Dive
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Angus Toland
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Millie Das
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anca M Pasca
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
3
|
Mashayekhi M, Beckman JA, Nian H, Garner EM, Mayfield D, Devin JK, Koethe JR, Brown JD, Cahill KN, Yu C, Silver H, Niswender K, Luther JM, Brown NJ. Comparative effects of weight loss and incretin-based therapies on vascular endothelial function, fibrinolysis and inflammation in individuals with obesity and prediabetes: A randomized controlled trial. Diabetes Obes Metab 2023; 25:570-580. [PMID: 36306151 PMCID: PMC10306232 DOI: 10.1111/dom.14903] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Accepted: 10/24/2022] [Indexed: 02/02/2023]
Abstract
AIM To test the hypothesis that glucagon-like peptide-1 receptor (GLP-1R) agonists have beneficial effects on vascular endothelial function, fibrinolysis and inflammation through weight loss-independent mechanisms. MATERIALS AND METHODS Individuals with obesity and prediabetes were randomized to 14 weeks of the GLP-1R agonist liraglutide, hypocaloric diet or the dipeptidyl peptidase-4 inhibitor sitagliptin in a 2:1:1 ratio. Treatment with drug was double blind and placebo-controlled. Measurements were made at baseline, after 2 weeks prior to significant weight loss and after 14 weeks. The primary outcomes were measures of endothelial function: flow-mediated vasodilation (FMD), plasminogen activator inhibitor-1 (PAI-1) and urine albumin-to-creatinine ratio (UACR). RESULTS Eighty-eight individuals were studied (liraglutide N = 44, diet N = 22, sitagliptin N = 22). Liraglutide and diet reduced weight, insulin resistance and PAI-1, while sitagliptin did not. There was no significant effect of any treatment on endothelial vasodilator function measured by FMD. Post hoc subgroup analyses in individuals with baseline FMD below the median, indicative of greater endothelial dysfunction, showed an improvement in FMD by all three treatments. GLP-1R antagonism with exendin (9-39) increased fasting blood glucose but did not change FMD or PAI-1. There was no effect of treatment on UACR. Finally, liraglutide, but not sitagliptin or diet, reduced the chemokine monocyte chemoattractant protein-1 (MCP-1). CONCLUSION Liraglutide and diet reduce weight, insulin resistance and PAI-1. Liraglutide, sitagliptin and diet do not change FMD in obese individuals with prediabetes with normal endothelial function. Liraglutide alone lowers the pro-inflammatory and pro-atherosclerotic chemokine MCP-1, indicating that this beneficial effect is independent of weight loss.
Collapse
Affiliation(s)
- Mona Mashayekhi
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN
| | - Joshua A. Beckman
- Vanderbilt University Medical Center, Department of Medicine, Division of Cardiovascular Medicine, Nashville, TN
| | - Hui Nian
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, TN
| | - Erica M. Garner
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN
| | - Dustin Mayfield
- Vanderbilt University Medical Center, Department of Medicine, Division of Clinical Pharmacology, Nashville, TN
| | - Jessica K. Devin
- UCHealth Endocrinology, Yampa Valley Medical Center, Steamboat Springs, CO
| | - John R. Koethe
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN
| | - Jonathan D. Brown
- Vanderbilt University Medical Center, Department of Medicine, Division of Cardiovascular Medicine, Nashville, TN
| | - Katherine N. Cahill
- Vanderbilt University Medical Center, Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Nashville, TN
| | - Chang Yu
- NYU Grossman School of Medicine, Department of Population Health, New York, NY
| | - Heidi Silver
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Nashville, TN
| | - Kevin Niswender
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
| | - James M. Luther
- Vanderbilt University Medical Center, Department of Medicine, Division of Clinical Pharmacology, Nashville, TN
| | | |
Collapse
|
4
|
A Possible Role for PAI-1 Blockade in Melanoma Immunotherapy. J Invest Dermatol 2021; 141:2566-2568. [PMID: 34688409 DOI: 10.1016/j.jid.2021.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 11/23/2022]
Abstract
In their new article in the Journal of Investigative Dermatology, Tseng et al. (2021) confirm that the sensitivity of melanoma cells to anti‒PD-L1 checkpoint inhibitor therapy is correlated with high PD-L1 surface expression. By blocking PD-L1 membrane clearing, controlled by LRP1 and PAI-1, the expression of high-cell-surface levels of PD-L1 was maintained.
Collapse
|
5
|
Wang WJ, Chen XM, Cai GY. Cellular senescence and the senescence-associated secretory phenotype: Potential therapeutic targets for renal fibrosis. Exp Gerontol 2021; 151:111403. [PMID: 33984448 DOI: 10.1016/j.exger.2021.111403] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022]
Abstract
Renal fibrosis plays a crucial role in the progression of chronic kidney disease and end-stage renal disease. However, because the aetiology of this pathological process is complex and remains unclear, there is still no effective treatment. Cellular senescence and the senescence-associated secretory phenotype (SASP) have been reported to lead to renal fibrosis. This review first discusses the relationships among cellular senescence, the SASP and renal fibrosis. Then, the key role of the SASP in irreversible renal fibrosis, including fibroblast activation and abnormal extracellular matrix accumulation, is discussed, with the results of studies having indicated that inhibiting cellular senescence and the SASP might be a potential preventive and therapeutic strategy for renal fibrosis. Finally, we summarize promising therapeutic strategies revealed by existing research on senescent cells and the SASP, including emerging interventions targeting the SASP, caloric restriction and mimetics, and novel regeneration therapies with stem cells.
Collapse
Affiliation(s)
- Wen-Juan Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiang-Mei Chen
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| | - Guang-Yan Cai
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| |
Collapse
|
6
|
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: 31] [Impact Index Per Article: 6.2] [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
|
7
|
Liu RM, Liu G. Cell senescence and fibrotic lung diseases. Exp Gerontol 2020; 132:110836. [PMID: 31958492 PMCID: PMC7036279 DOI: 10.1016/j.exger.2020.110836] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fatal lung disorder with an unknown etiology and very limited therapeutic options. The incidence and severity of IPF increase with advanced age, suggesting that aging is a major risk factor for IPF. The mechanism underlying the aging-related susceptibility to IPF, however, remains unclear. Cellular senescence, a permanent arrest of cell growth, has been increasingly recognized as an important contributor to aging and aging-related diseases, including IPF. Senescent cells have been identified in IPF lungs and in experimental lung fibrosis models. Removal of senescent cells pharmacologically or genetically improves lung function and reverses pulmonary fibrosis induced by different stimuli in experimental fibrosis models. Treatment with senolytic drugs also improves clinical symptoms in IPF patients. These intriguing findings suggest that cellular senescence contributes importantly to the pathogenesis of fibrotic lung diseases and targeting senescent cells may represent a novel approach for the treatment of fibrotic lung disorders. In this mini review, we summarize the recent advance in the field regarding the role of cellular senescence in fibrotic lung diseases, with a focus on IPF.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
8
|
Lin H, Xu L, Yu S, Hong W, Huang M, Xu P. Therapeutics targeting the fibrinolytic system. Exp Mol Med 2020; 52:367-379. [PMID: 32152451 PMCID: PMC7156416 DOI: 10.1038/s12276-020-0397-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/08/2019] [Accepted: 01/01/2020] [Indexed: 02/07/2023] Open
Abstract
The function of the fibrinolytic system was first identified to dissolve fibrin to maintain vascular patency. Connections between the fibrinolytic system and many other physiological and pathological processes have been well established. Dysregulation of the fibrinolytic system is closely associated with multiple pathological conditions, including thrombosis, inflammation, cancer progression, and neuropathies. Thus, molecules in the fibrinolytic system are potent therapeutic and diagnostic targets. This review summarizes the currently used agents targeting this system and the development of novel therapeutic strategies in experimental studies. Future directions for the development of modulators of the fibrinolytic system are also discussed. The fibrinolytic system was originally identified to dissolve blood clots, and is shown to have important roles in other pathological processes, including cancer progression, inflammation, and thrombosis. Molecules or therapeutics targeting fibrinolytic system have been successfully used in the clinical treatments of cancer and thrombotic diseases. The clinical studies and experimental models targeting fibrinolytic system are reviewed by Haili Lin at Sanming First Hosipital, Mingdong Huang at Fuzhou University in China, and Peng Xu at A*STAR in Singapore to demonstrate fibrinolytic system as novel therapeutic targets. As an example, the inhibition of fibrinolytic system protein can be used to suppress cancer prolifieration and metastasis. This review also discusses the potential therapeutic effects of inhibitiors of fibrinolytic system on inflammatory disorders.
Collapse
Affiliation(s)
- Haili Lin
- Department of Pharmacy, Sanming First Hospital, 365000, Sanming, Fujian, People's Republic of China
| | - Luning Xu
- Department of Pharmacy, Sanming First Hospital, 365000, Sanming, Fujian, People's Republic of China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, 350116, Fuzhou, Fujian, People's Republic of China
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, 138673, Singapore
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, 350116, Fuzhou, Fujian, People's Republic of China.
| | - Peng Xu
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, 138673, Singapore.
| |
Collapse
|
9
|
Li W, Lu Y, Lou Y, Zhao S, Cui W, Wang Y, Luo M, Sun J, Miao L. FFNT25 ameliorates unilateral ureteral obstruction-induced renal fibrosis. Ren Fail 2019; 41:419-426. [PMID: 31140898 PMCID: PMC6566665 DOI: 10.1080/0886022x.2019.1612430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 01/06/2023] Open
Abstract
Renal fibrosis is a common pathological feature of chronic kidney disease (CKD) patients who progress to end-stage renal disease (ESRD). With the increasing incidence of CKD, it is of importance to develop effective therapies that blunt development of renal fibrosis. FFNT25 is a newly developed molecular compound that could be used to prevent fibrosis. In this study, we administered FFNT25 to rats following unilateral ureteral obstruction (UUO) to investigate its anti-fibrosis mechanism. Thirty-two Sprague-Dawley rats were randomly divided into four groups: (1) control (normal rats), (2) sham-operated, (3) UUO-operated + vehicle, and (4) UUO-operated + FFNT25. Two weeks after UUO, the rats were gavaged with either FFNT25 (20.6 mg/kg/day) or vehicle for two weeks. Serum, urine, and kidney samples were collected at the end of the study. FFNT25 reduced levels of renal fibrosis and decreased mRNA and protein levels of extracellular matrix (ECM) markers α-smooth muscle actin (α-SMA) and plasminogen activator inhibitor-1 (PAI-1) following UUO compared to vehicle treatment (n = 8, p<.05). The current results indicate that FFNT25 can affect both the production and degradation of collagen fibers to reduce fibrosis.
Collapse
Affiliation(s)
- Wen Li
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Yue Lu
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Yan Lou
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Shiyue Zhao
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Wenpeng Cui
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Yangwei Wang
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Manyu Luo
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Jing Sun
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| | - Lining Miao
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
PAI-1 inhibition by simvastatin as a positive adjuvant in cell therapy. Mol Biol Rep 2019; 46:1511-1517. [DOI: 10.1007/s11033-018-4562-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/07/2018] [Indexed: 01/05/2023]
|
11
|
Xu P, Huang M. Small Peptides as Modulators of Serine Proteases. Curr Med Chem 2018; 27:3686-3705. [PMID: 30332941 DOI: 10.2174/0929867325666181016163630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/26/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023]
Abstract
Serine proteases play critical roles in many physiological and pathological processes, and are proven diagnostic and therapeutic targets in a number of clinical indications. Suppression of the aberrant proteolytic activities of these proteases has been clinically used for the treatments of relevant diseases. Polypeptides with 10-20 residues are of great interests as medicinal modulators of serine proteases, because these peptides demonstrate the characteristics of both small molecule drugs and macromolecular drugs. In this review, we summarized the recent development of peptide-based inhibitors against serine proteases with potent inhibitory and high specificity comparable to monoclonal antibodies. In addition, we also discussed the strategies of enhancing plasma half-life and bioavailability of peptides in vivo, which is the main hurdle that limits the clinical translation of peptide-based drugs. This review advocates new avenue for the development of effective serine protease inhibitors and highlights the prospect of the medicinal use of these inhibitors.
Collapse
Affiliation(s)
- Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| |
Collapse
|
12
|
Jeffries CD, Perkins DO, Fournier M, Do KQ, Cuenod M, Khadimallah I, Domenici E, Addington J, Bearden CE, Cadenhead KS, Cannon TD, Cornblatt BA, Mathalon DH, McGlashan TH, Seidman LJ, Tsuang M, Walker EF, Woods SW. Networks of blood proteins in the neuroimmunology of schizophrenia. Transl Psychiatry 2018; 8:112. [PMID: 29875399 PMCID: PMC5990539 DOI: 10.1038/s41398-018-0158-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/06/2018] [Accepted: 04/15/2018] [Indexed: 02/05/2023] Open
Abstract
Levels of certain circulating cytokines and related immune system molecules are consistently altered in schizophrenia and related disorders. In addition to absolute analyte levels, we sought analytes in correlation networks that could be prognostic. We analyzed baseline blood plasma samples with a Luminex platform from 72 subjects meeting criteria for a psychosis clinical high-risk syndrome; 32 subjects converted to a diagnosis of psychotic disorder within two years while 40 other subjects did not. Another comparison group included 35 unaffected subjects. Assays of 141 analytes passed early quality control. We then used an unweighted co-expression network analysis to identify highly correlated modules in each group. Overall, there was a striking loss of network complexity going from unaffected subjects to nonconverters and thence to converters (applying standard, graph-theoretic metrics). Graph differences were largely driven by proteins regulating tissue remodeling (e.g. blood-brain barrier). In more detail, certain sets of antithetical proteins were highly correlated in unaffected subjects (e.g. SERPINE1 vs MMP9), as expected in homeostasis. However, for particular protein pairs this trend was reversed in converters (e.g. SERPINE1 vs TIMP1, being synthetical inhibitors of remodeling of extracellular matrix and vasculature). Thus, some correlation signals strongly predict impending conversion to a psychotic disorder and directly suggest pharmaceutical targets.
Collapse
Affiliation(s)
- Clark D Jeffries
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, NC, USA.
| | - Diana O Perkins
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Margot Fournier
- Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Kim Q Do
- Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Michel Cuenod
- Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Ines Khadimallah
- Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Enrico Domenici
- Laboratory of Neurogenomic Biomarkers, Centre for Integrative Biology, and Microsoft Research, Centre for Computational Systems Biology, University of Trento, Trento, Italy
| | - Jean Addington
- Hotchkiss Brain Institute, Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Carrie E Bearden
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, UCLA, Los Angeles, CA, USA
| | | | - Tyrone D Cannon
- Department of Psychology, Yale University, New Haven, CT, USA
| | | | - Daniel H Mathalon
- Department of Psychiatry, UCSF and San Francisco VA Healthcare System, San Francisco, CA, USA
| | | | - Larry J Seidman
- Department of Psychiatry, Harvard Medical School at Beth Israel Deaconess Medical Center and Massachusetts General Hospital, Boston, MA, USA
| | - Ming Tsuang
- Department of Psychiatry, Center for Behavioral Genomics UCSD, San Diego, CA, USA
| | - Elaine F Walker
- Departments of Psychology and Psychiatry, Emory University, Atlanta, GA, USA
| | - Scott W Woods
- Department of Psychology, Yale University, New Haven, CT, USA
| |
Collapse
|
13
|
Yusova OI, Savchuk OV, Grinenko TV, Kuchmenko OB. Determination of plasminogen/plasmin system components and indicators of lipoproteins oxidative modification under arterial hypertension. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.01.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
14
|
Ohkura N, Oishi K, Kihara-Negishi F, Atsumi GI, Tatefuji T. Effects of a diet containing Brazilian propolis on lipopolysaccharide-induced increases in plasma plasminogen activator inhibitor-1 levels in mice. JOURNAL OF COMPLEMENTARY MEDICINE RESEARCH 2016; 5:439-443. [PMID: 27757277 PMCID: PMC5061490 DOI: 10.5455/jice.20160814112735] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/23/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND Brazilian propolis has many biological activities including the ability to help prevent thrombotic diseases, but this particular effect has not been proven. Plasma levels of plasminogen activator inhibitor-1 (PAI-1), an inhibitor of fibrinolysis, increase under inflammatory conditions such as infection, obesity and atherosclerosis and such elevated levels predispose individuals to a risk of developing thrombotic diseases. AIM This study aimed to determine the effects of a diet containing Brazilian propolis on lipopolysaccharide (LPS)-induced increases in plasma PAI-1 levels. MATERIALS AND METHODS Mice were fed with a diet containing 0.5% (w/w) Brazilian propolis for 8 weeks. Thereafter, the mice were subcutaneously injected with saline containing 0.015 mg/kg of LPS and sacrificed 4 h later. RESULTS Orally administered Brazilian propolis significantly suppressed the LPS-induced increase in PAI-1 antigen and its activity in mouse plasma. CONCLUSION This study indicated that Brazilian propolis contains natural products that can decrease thrombotic tendencies in mice.
Collapse
Affiliation(s)
- Naoki Ohkura
- Department of Molecular Physiology and Pathology, School of Pharma-Sciences, Teikyo University, Tokyo, Itabashi, Japan
| | - Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Japan
| | - Fumiko Kihara-Negishi
- Department of Life and Health Science, School of Pharma-Sciences, Teikyo University, Tokyo, Itabashi, Japan
| | - Gen-Ichi Atsumi
- Department of Molecular Physiology and Pathology, School of Pharma-Sciences, Teikyo University, Tokyo, Itabashi, Japan
| | - Tomoki Tatefuji
- Institute for Bee Products and Health Science, Yamada Apiculture Center, Kagamino, Okayama, Japan
| |
Collapse
|
15
|
Gu C, Zhang J, Noble NA, Peng XR, Huang Y. An additive effect of anti-PAI-1 antibody to ACE inhibitor on slowing the progression of diabetic kidney disease. Am J Physiol Renal Physiol 2016; 311:F852-F863. [PMID: 27511457 DOI: 10.1152/ajprenal.00564.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 07/05/2016] [Indexed: 02/03/2023] Open
Abstract
While angiotensin II blockade slows the progression of diabetic nephropathy, current data suggest that it alone cannot stop the disease process. New therapies or drug combinations will be required to further slow or halt disease progression. Inhibition of plasminogen activator inhibitor type 1 (PAI-1) aimed at enhancing ECM degradation has shown therapeutic potential in diabetic nephropathy. Here, using a mouse model of type diabetes, the maximally therapeutic dose of the PAI-1-neutralizing mouse monoclonal antibody (MEDI-579) was determined and compared with the maximally effective dose of enalapril. We then examined whether addition of MEDI-579 to enalapril would enhance the efficacy in slowing the progression of diabetic nephropathy. Untreated uninephrectomized diabetic db/db mice developed progressive albuminuria and glomerulosclerosis associated with increased expression of transforming growth factor (TGF)-β1, PAI-1, type IV collagen, and fibronectin from weeks 18 to 22, which were reduced by MEDI-579 at 3 mg/kg body wt, similar to enalapril given alone from weeks 12 to 22 Adding MEDI-579 to enalapril from weeks 18 to 22 resulted in further reduction in albuminuria and markers of renal fibrosis. Renal plasmin generation was dramatically reduced by 57% in diabetic mice, a decrease that was partially reversed by MEDI-579 or enalapril given alone but was further restored by these two treatments given in combination. Our results suggest that MEDI-579 is effective in slowing the progression of diabetic nephropathy in db/db mice and that the effect is additive to ACEI. While enalapril is renal protective, the add-on PAI-1 antibody may offer additional renoprotection in progressive diabetic nephropathy via enhancing ECM turnover.
Collapse
Affiliation(s)
- Chunyan Gu
- Department of Pathology, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China.,Division of Nephrology, University of Utah, Salt Lake City, Utah; and
| | - Jiandong Zhang
- Division of Nephrology, University of Utah, Salt Lake City, Utah; and
| | - Nancy A Noble
- Division of Nephrology, University of Utah, Salt Lake City, Utah; and
| | - Xiao-Rong Peng
- Bioscience, AstraZeneca R&D, Pepparredsleden 1, Molndal SE-43183, Sweden
| | - Yufeng Huang
- Division of Nephrology, University of Utah, Salt Lake City, Utah; and
| |
Collapse
|
16
|
Tofler GH, Massaro J, O'Donnell CJ, Wilson PWF, Vasan RS, Sutherland PA, Meigs JB, Levy D, D'Agostino RB. Plasminogen activator inhibitor and the risk of cardiovascular disease: The Framingham Heart Study. Thromb Res 2016; 140:30-35. [PMID: 26896607 DOI: 10.1016/j.thromres.2016.02.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/25/2016] [Accepted: 02/02/2016] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Although plasminogen activator inhibitor (PAI-1) plays a key regulatory role in fibrinolysis, it has not been clearly shown to independently predict cardiovascular disease (CVD) among individuals without prior CVD. We investigated, in the Framingham Heart Study offspring cohort, whether PAI-1 predicted CVD risk among individuals without prior CVD. METHODS Plasma PAI-1 antigen and tissue plasminogen activator (TPA) antigen were measured in 3203 subjects without prior CVD between 1991 and 1995; average follow-up of 10 years. PAI-1 was remeasured 4 years after baseline, to determine the effect of serial change on risk. RESULTS PAI-1 levels (mean ± SD) were 29.1 ng/ml (19.2) versus 22.1 (16.5) for those and without incident CVD; p<0.001, and TPA levels were 12.0 ng/ml (5.7) versus 9.0 (4.7); p<0.001. PAI-1 and TPA antigen levels had a strong unadjusted linear relation with incident CVD (p<0.001). After adjustment for conventional risk factors, the hazard ratios (HRs) for higher quartiles of PAI-1, compared with the lowest, were 1.9, 1.9, 2.6 (linear trend p=0.006), and 1.6, 1.6, 2.9 (p<0.001) for TPA antigen. The adjusted HRs for increasing quartiles of serial change in PAI-1 at 4 years, compared with the lowest, were 0.9, 0.8, 1.3 (p=0.050). C statistic assessment showed that adding PAI-1 or TPA to conventional risk factors resulted in small increases in discrimination and modest reclassification of risk, which was statistically significant for TPA (net reclassification 6.8%, p=0.037) but not PAI-1 (4.8%, p=0.113). CONCLUSION PAI-1 and TPA antigen levels are predictive of CVD events after accounting for established risk factors. A serial increase in PAI-1 is associated with a further increase in risk. These findings support the importance of fibrinolytic potential in CVD.
Collapse
Affiliation(s)
- G H Tofler
- Royal North Shore Hospital, Sydney University, Australia.
| | | | - C J O'Donnell
- The Framingham Heart Study of the National Heart, Lung, and Blood Institute of the National Institutes of Health, United States; The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States; Massachusetts General Hospital, United States
| | | | - R S Vasan
- The Framingham Heart Study of the National Heart, Lung, and Blood Institute of the National Institutes of Health, United States
| | - P A Sutherland
- The Framingham Heart Study of the National Heart, Lung, and Blood Institute of the National Institutes of Health, United States; The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - J B Meigs
- Massachusetts General Hospital, United States
| | - D Levy
- The Framingham Heart Study of the National Heart, Lung, and Blood Institute of the National Institutes of Health, United States; The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | | |
Collapse
|
17
|
Placencio VR, DeClerck YA. Plasminogen Activator Inhibitor-1 in Cancer: Rationale and Insight for Future Therapeutic Testing. Cancer Res 2015; 75:2969-74. [PMID: 26180080 DOI: 10.1158/0008-5472.can-15-0876] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/28/2015] [Indexed: 12/19/2022]
Abstract
Despite its function as an inhibitor of urokinase and tissue-type plasminogen activator (PA), PA inhibitor-1 (PAI-1) has a paradoxical protumorigenic role in cancer, promoting angiogenesis and tumor cell survival. In this review, we summarize preclinical evidence in support of the protumorigenic function of PAI-1 that has led to the testing of small-molecule PAI-1 inhibitors, initially developed as antithrombotic agents, in animal models of cancer. The review discusses the challenges and the opportunities that lay ahead to the development of efficacious and nontoxic PAI-1 inhibitors as anticancer agents.
Collapse
Affiliation(s)
- Veronica R Placencio
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, California. The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, California
| | - Yves A DeClerck
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, California. The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, California. Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California.
| |
Collapse
|
18
|
WYGANOWSKA-ŞWIĄTKOWSKA MARZENA, JANKUN JERZY. Plasminogen activation system in oral cancer: Relevance in prognosis and therapy (Review). Int J Oncol 2015; 47:16-24. [DOI: 10.3892/ijo.2015.3021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/04/2015] [Indexed: 11/06/2022] Open
|
19
|
Rouch A, Vanucci-Bacqué C, Bedos-Belval F, Baltas M. Small molecules inhibitors of plasminogen activator inhibitor-1 - an overview. Eur J Med Chem 2015; 92:619-36. [PMID: 25615797 DOI: 10.1016/j.ejmech.2015.01.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 12/14/2022]
Abstract
PAI-1, a glycoprotein from the serpin family and the main inhibitor of tPA and uPA, plays an essential role in the regulation of intra and extravascular fibrinolysis by inhibiting the formation of plasmin from plasminogen. PAI-1 is also involved in pathological processes such as thromboembolic diseases, atherosclerosis, fibrosis and cancer. The inhibition of PAI-1 activity by small organic molecules has been observed in vitro and with some in vivo models. Based on these findings, PAI-1 appears as a potential therapeutic target for several pathological conditions. Over the past decades, many efforts have therefore been devoted to developing PAI-1 inhibitors. This article provides an overview of the publishing activity on small organic molecules used as PAI-1 inhibitors. The chemical synthesis of the most potent inhibitors as well as their biological and biochemical evaluations is also presented.
Collapse
Affiliation(s)
- Anne Rouch
- Université Paul Sabatier Toulouse III, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France; CNRS, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France
| | - Corinne Vanucci-Bacqué
- Université Paul Sabatier Toulouse III, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France; CNRS, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France
| | - Florence Bedos-Belval
- Université Paul Sabatier Toulouse III, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France; CNRS, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France.
| | - Michel Baltas
- Université Paul Sabatier Toulouse III, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France; CNRS, UMR 5068, Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, 118, Route de Narbonne, F-31062 Toulouse Cedex 9, France.
| |
Collapse
|
20
|
Abstract
Protease inhibition by serpins requires a large conformational transition from an active, metastable state to an inactive, stable state. Similar reactions can also occur in the absence of proteases, and these latency transitions take hours, making their time scales many orders of magnitude larger than are currently accessible using conventional molecular dynamics simulations. Using a variational path sampling algorithm, we simulated the entire serpin active-to-latent transition in all-atom detail with a physically realistic force field using a standard computing cluster. These simulations provide a unifying picture explaining existing experimental data for the latency transition of the serpin plasminogen activator inhibitor-1 (PAI-1). They predict a long-lived intermediate that resembles a previously proposed, partially loop-inserted, prelatent state; correctly predict the effects of PAI-1 mutations on the kinetics; and provide a potential means to identify ligands able to accelerate the latency transition. Interestingly, although all of the simulated PAI-1 variants readily access the prelatent intermediate, this conformation is not populated in the active-to-latent transition of another serpin, α1-antitrypsin, which does not readily go latent. Thus, these simulations also help elucidate why some inhibitory serpin families are more conformationally labile than others.
Collapse
|
21
|
Hoirisch-Clapauch S, Nardi AE. Markers of low activity of tissue plasminogen activator/plasmin are prevalent in schizophrenia patients. Schizophr Res 2014; 159:118-23. [PMID: 25205258 DOI: 10.1016/j.schres.2014.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/03/2014] [Accepted: 08/06/2014] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Clot buster tissue plasminogen activator (tPA) and its end-product plasmin play a well-defined role in neurochemistry. They mediate a number of events that culminate in tolerance against excitotoxicity, hippocampal neurogenesis, synaptic remodeling, neuronal plasticity, cognitive and emotional processing. Abnormalities in these processes have been implicated in schizophrenia pathogenesis. METHODS Laboratory markers of low activity of tPA/plasmin were analyzed in 70 schizophrenia adults (DSM-IV), and 98 age-matched controls, consecutively selected at university hospitals. RESULTS All but two patients had positive markers (1-6, mean 2.1). Twenty-nine patients and 11 controls had hyperinsulinemia (44% vs. 11%) and 20 patients and 11 controls had hypertriglyceridemia (29% vs. 11%). Both insulin and triglycerides stimulate production of plasminogen activator inhibitor (PAI)-1, a major tPA inhibitor. Nineteen patients and six controls had hyperhomocysteinemia (27% vs. 6%), a condition that impairs tPA catalytic activity. Fifteen patients (22%) but no controls had free-protein S deficiency, a condition that reduces PAI-1 inhibition. Twenty-one patients (30%) but no controls had 1-3 antiphospholipid antibodies in medium or/high levels. Such antibodies are able to inhibit tPA/plasmin activity. Both PAI-1 polymorphism 4G/5G and heterozygous prothrombin G20210A were more prevalent in patients (60% vs. 48% and 2% vs. 1%, respectively), but difference lacked significance. PAI-1 polymorphism was synergistic with hyperinsulinemia. Protein C deficiency was not detected in patients or controls. CONCLUSION We have found a high prevalence of markers of low tPA/plasmin activity in a sample of schizophrenia patients. Our findings should be validated in large studies, preferably in medication-naïve patients.
Collapse
Affiliation(s)
- Silvia Hoirisch-Clapauch
- Department of Hematology, Hospital Federal dos Servidores do Estado, Ministry of Health, Rio de Janeiro, Brazil.
| | - Antonio E Nardi
- Institute of Psychiatry, Federal University of Rio de Janeiro, National Institute for Translational Medicine, INCT-TM, CNPq, Brazil
| |
Collapse
|
22
|
Damare J, Brandal S, Fortenberry YM. Inhibition of PAI-1 antiproteolytic activity against tPA by RNA aptamers. Nucleic Acid Ther 2014; 24:239-49. [PMID: 24922319 DOI: 10.1089/nat.2013.0475] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1; SERPINE1) inhibits the plasminogen activators: tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). Elevated levels of PAI-1 have been correlated with an increased risk for cardiovascular disease. Pharmacologically suppressing PAI-1 might prevent, or successfully treat PAI-1 related vascular diseases. This can potentially be accomplished by using small RNA molecules (aptamers). This study's goal is to develop RNA aptamers to a region of PAI-1 that will prevent the ability of PAI-1 to interact with the plasminogen activators. The aptamers were generated through a systematic evolution of ligands via exponential enrichment approach that ensures the creation of RNA molecules that bind to our target protein, PAI-1. In vitro assays were used to determine the effect of these aptamers on PAI-1's inhibitory activity. Three aptamers that bind to PAI-1 with affinities in the nanomolar range were isolated. The aptamer clones R10-4 and R10-2 inhibited PAI-1's antiproteolytic activity against tPA and disrupted PAI-1's ability to form a stable covalent complex with tPA. Increasing aptamer concentrations correlated positively with an increase in cleaved PAI-1. To the best of our knowledge, this is the first report of RNA molecules that inhibit the antiproteolytic activity of PAI-1.
Collapse
Affiliation(s)
- Jared Damare
- 1 Department of Pediatric Hematology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | | |
Collapse
|
23
|
Overstreet JM, Samarakoon R, Meldrum KK, Higgins PJ. Redox control of p53 in the transcriptional regulation of TGF-β1 target genes through SMAD cooperativity. Cell Signal 2014; 26:1427-36. [PMID: 24613410 DOI: 10.1016/j.cellsig.2014.02.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) regulates the tissue response to injury and is the principal driver of excessive scarring leading to fibrosis and eventual organ failure. The TGF-β1 effectors SMAD3 and p53 are major contributors to disease progression. While SMAD3 is an established pro-fibrotic factor, the role of p53 in the TGF-β1-induced fibrotic program is not clear. p53 gene silencing, genetic ablation/subsequent rescue, and pharmacological inhibition confirmed that p53 was required for expression of plasminogen activator inhibitor-1 (PAI-1), a major TGF-β1 target gene and a key causative element in fibrotic disorders. TGF-β1 regulated p53 activity by stimulating p53(Ser15 and 9) phosphorylation and acetylation, promoting interactions with activated SMADs and subsequent binding of p53/SMAD3 to the PAI-1 promoter in HK-2 human renal tubular epithelial cells and HaCaT human keratinocytes. Immunohistochemistry revealed prominent co-induction of SMAD3, p53 and PAI-1 in the tubular epithelium of the obstructed kidney consistent with a potential in vivo role for p53 and SMADs in TGF-β1-driven renal fibrosis. TGF-β1-initiated phosphorylation of p53(Ser15) and up-regulation of expression of several pro-fibrotic genes, moreover, was dependent on the rapid generation of reactive oxygen species (ROS). shRNA silencing of the p22(Phox) subunit of NADP(H) oxidases in HK-2 cells partially attenuated (over 50%) p53(Ser15) phosphorylation and PAI-1 induction. These studies highlight the role of free radicals in p53 activation and subsequent pro-fibrotic reprogramming by TGF-β1 via the SMAD3-p53 transcriptional axis. Present findings provide a rationale for therapeutic targeting of SMAD3-p53 in aberrant TGF-β1 signaling associated with renal fibrosis.
Collapse
Affiliation(s)
- Jessica M Overstreet
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, United States
| | - Rohan Samarakoon
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, United States
| | - Kirstan K Meldrum
- Department of Urology, University of Florida School of Medicine, Gainesville, FL 32611, United States
| | - Paul J Higgins
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, United States.
| |
Collapse
|
24
|
Wyganowska-Świątkowska M, Surdacka A, Skrzypczak-Jankun E, Jankun J. The plasminogen activation system in periodontal tissue (Review). Int J Mol Med 2014; 33:763-8. [PMID: 24535478 DOI: 10.3892/ijmm.2014.1653] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/28/2014] [Indexed: 11/05/2022] Open
Abstract
The plasminogen activation system (PAS) plays an essential role in tissue proteolysis in physiological and pathological processes. Periodontitis is a chronic infection associated with increased proteolysis driven by plasminogen activation. In this comprehensive review, we summarise the effects of PAS in wound healing, tissue remodelling, inflammation, bacterial infection, and in the initiation and progression of periodontal disease. Specifically, we discuss the role of plasminogen activators (PAs), including urokinase PA (uPA), tissue-type PA (tPA), PA inhibitor type 1 (PAI-1) and 2 (PAI-2) and activated plasminogen in periodontal tissue, where their concentrations can reach much higher values than those found in other parts of the body. We also discuss whether PA deficiencies can have effects on periodontal tissue. We conclude that in periodontal disease, PAS is unbalanced and equalizing its function can improve the clinical periodontal tissue condition.
Collapse
Affiliation(s)
| | - Anna Surdacka
- Department of Conservative Dentistry and Periodontology, Poznań University of Medical Sciences, Poznań 60-820, Poland
| | - Ewa Skrzypczak-Jankun
- Urology Research Center, Department of Urology, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| | - Jerzy Jankun
- Protein Research Chair, Department of Biochemistry, College of Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
25
|
Zhang J, Gu C, Lawrence DA, Cheung AK, Huang Y. A plasminogen activator inhibitor type 1 mutant retards diabetic nephropathy in db/db mice by protecting podocytes. Exp Physiol 2014; 99:802-15. [PMID: 24443353 DOI: 10.1113/expphysiol.2013.077610] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A mutant non-inhibiting plasminogen activator inhibitor type 1 (PAI-1), termed PAI-1R, which reduces endogenous PAI-1 activity, has been shown to inhibit albuminuria and reduce glomerulosclerosis in experimental diabetes. The mechanism of the reduction of albuminuria is unclear. This study sought to determine whether the administration of PAI-1R protected podocytes from injury directly, thereby reducing albuminuria in the db/db mouse, a model of type 2 diabetes. Untreated uninephrectomized db/db mice developed significant mesangial matrix expansion and albuminuria at week 22 of age, associated with segmental podocyte foot-process effacement, reduction of renal nephrin, podocin and zonula occludin-1 production and induction of renal desmin and B7-1 generation. In contrast, treatment with PAI-1R at 0.5 mg (kg body weight)(-1) i.p., twice daily from week 20 to 22, reduced glomerular matrix accumulation, fibronectin and collagen production and albuminuria by 36, 62, 65 and 31%, respectively (P < 0.05), without affecting blood glucose level or body weight. Podocyte morphology and protein markers were also significantly attenuated by PAI-1R administration. Importantly, recombinant PAI-1 downregulated nephrin and zonula occludin-1 but increased desmin and B7-1 mRNA expression and protein production by podocytes in vitro, similar to the effects of transforming growth factor-β1. These observations provide evidence that PAI-1, in a manner similar to transforming growth factor-β1, directly induces podocyte injury, particularly in the setting of diabetes, where elevated PAI-1 may contribute to the progression of albuminuria. Reducing the increased PAI-1 activity by administration of PAI-1R, in fact, reduces podocyte injury, thereby reducing albuminuria. Therefore, PAI-1R provides an additional therapeutic effect in slowing the progression of diabetic nephropathy via the protection of podocytes.
Collapse
Affiliation(s)
- Jiandong Zhang
- Division of Nephrology & Hypertension, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Chunyan Gu
- Division of Nephrology & Hypertension, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Daniel A Lawrence
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Alfred K Cheung
- Division of Nephrology & Hypertension, University of Utah School of Medicine, Salt Lake City, UT, USA Medical Care Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Yufeng Huang
- Division of Nephrology & Hypertension, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
26
|
Małgorzewicz S, Skrzypczak-Jankun E, Jankun J. Plasminogen activator inhibitor-1 in kidney pathology (Review). Int J Mol Med 2013; 31:503-10. [PMID: 23314920 DOI: 10.3892/ijmm.2013.1234] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 11/09/2012] [Indexed: 11/06/2022] Open
Abstract
Plasminogen activator inhibitor type-1 (PAI-1) inhibits tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA), which convert plasminogen to plasmin, a strong proteolytic enzyme. Thus, PAI-1 is a primary and negative regulator of plasmin-driven proteolysis. In addition to its main role as an inhibitor of fibrinolysis, PAI‑1 has been implicated as a mediator in other processes, including fibrosis, rheumatoid arthritis, atherosclerosis, tumor angiogenesis and bacterial infections. It also significantly modulates cellular adhesion or migration, wound healing, angiogenesis and tumor cell metastasis. However, in the present study, we have reviewed the literature in relation to different kidney diseases where PAI-1 regulates fibrinolysis and acts independently of proteolysis. PAI-1 is normally produced in trace amounts in healthy kidneys but is synthesized in a wide variety of both acute and chronic diseased kidneys. We reviewed the role of PAI-1 in diabetic kidney nephropathy, chronic kidney disease, hemodialysis, peritoneal dialysis and in kidney transplantation. Increased PAI-1 expression results in accumulation of extracellular matrix (ECM) leading to numerous kidney diseases. Predisposition to some diseases is due to the genetic role of PAI-1 in their development. A number of studies demonstrated that the inhibition of PAI-1 activity or therapy with a mutant PAI-1 increases matrix turnover and reduces glomerulosclerosis by competing with endogenous PAI-1. This strongly suggests that PAI-1 is a valid target in the treatment of fibrotic renal disease. However, net proteolytic activity depends on the delicate balance between its negative regulation by PAI-1 and activation by uPA and tPA. Also, plasmin activated by its inhibitors upregulates activity of other enzymes. Thus, assessment of prognosis for the diseased kidney should include a variety of proteolysis regulators and enzymes.
Collapse
Affiliation(s)
- Sylwia Małgorzewicz
- Department of Clinical Nutrition, Medical University of Gdańsk, Gdańsk 80-211, Poland
| | | | | |
Collapse
|
27
|
Fang H, Placencio VR, DeClerck YA. Protumorigenic activity of plasminogen activator inhibitor-1 through an antiapoptotic function. J Natl Cancer Inst 2012; 104:1470-84. [PMID: 22984202 PMCID: PMC3529616 DOI: 10.1093/jnci/djs377] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 07/20/2012] [Accepted: 07/24/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1) is a protease inhibitor but is paradoxically associated with poor outcomes in cancer patients. However, the mechanisms of its effects on tumor cells have not been explored. METHODS Endogenous PAI-1 in human tumor cell lines (HT-1080, A549, HCT-116, and MDA-MB-231) was suppressed by small interfering RNAs (siRNAs) and PAI-039, a small molecule inhibitor of PAI-1, and the effects on apoptosis were examined. Tumorigenicity of PAI-1 knockdown (KD) tumor cells was examined in immunodeficient PAI-1 wild-type and knockout (KO) mice (9-15 per group), and event-free survival was analyzed by the Kaplan-Meier method. The effect of PAI-1 suppression on HT-1080 xenotransplanted tumors was evaluated for cell proliferation, apoptosis, and angiogenesis. All statistical tests were two-sided. RESULTS Genetic and pharmacological inhibition of PAI-1 in the four tumor cell lines increased spontaneous apoptosis (mean fold increase relative to control: HT-1080, siRNA#1, mean = 4.0, 95% CI = 2.6 to 5.3, P < .001; siRNA#2, mean = 2.6, 95% CI = 2.4 to 2.9, P < .001, Student t test), which was blocked in the presence of recombinant PAI-1, a caspase-8 inhibitor, or Fas/FasL neutralizing antibodies and was partially attenuated by a plasmin inhibitor-aprotinin. PAI-1 KO mice implanted with PAI-1 KD HT-1080 cells had decreased tumorigenesis and prolonged survival compared with control mice (P = .002, log-rank test), and their tumors exhibited decreased cell proliferation and angiogenesis and increased apoptosis. Furthermore, five of 15 PAI-1 KO mice implanted with PAI-1 KD HT-1080 cells never developed tumors. CONCLUSIONS These data suggest that PAI-1 exerts a protective effect against tumor cell apoptosis by a mechanism that, in part, involves plasmin activation and inhibition of Fas/Fas-L-mediated apoptosis and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Hua Fang
- Division of Hematology-Oncology, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | |
Collapse
|
28
|
Xiao YH, Yang LF, Feng XC, Yang H, Ma TH. Tanshinones increase fibrinolysis through inhibition of plasminogen activator inhibitor-1. CNS Neurosci Ther 2012; 18:436-8. [PMID: 22533728 DOI: 10.1111/j.1755-5949.2012.00307.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
29
|
Shirakawa J, Togashi Y, Tajima K, Orime K, Kikuchi K, Miyazaki T, Sato K, Kimura M, Goshima Y, Terauchi Y. Plasminogen activator inhibitor-1 is associated with renal dysfunction independent of BMI and serum lipid levels in patients with type 2 diabetes. Diabetes Res Clin Pract 2012; 97:e9-12. [PMID: 22497969 DOI: 10.1016/j.diabres.2012.03.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 03/04/2012] [Accepted: 03/19/2012] [Indexed: 11/27/2022]
Abstract
We investigated a possible association between serum plasminogen activator inhibitor-1 (PAI-1) levels and renal dysfunction in 124 type 2 diabetes patients. Multiple linear regression analyses indicated that the PAI-1 levels were significantly inversely correlated with estimated glomerular filtration rate (eGFR) independent of albuminuria, BMI, LDL-C, and triglyceride.
Collapse
Affiliation(s)
- Jun Shirakawa
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Jeon H, Kim JH, Kim JH, Lee WH, Lee MS, Suk K. Plasminogen activator inhibitor type 1 regulates microglial motility and phagocytic activity. J Neuroinflammation 2012; 9:149. [PMID: 22747686 PMCID: PMC3418576 DOI: 10.1186/1742-2094-9-149] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 06/29/2012] [Indexed: 01/05/2023] Open
Abstract
Background Plasminogen activator inhibitor type 1 (PAI-1) is the primary inhibitor of urokinase type plasminogen activators (uPA) and tissue type plasminogen activators (tPA), which mediate fibrinolysis. PAI-1 is also involved in the innate immunity by regulating cell migration and phagocytosis. However, little is known about the role of PAI-1 in the central nervous system. Methods In this study, we identified PAI-1 in the culture medium of mouse mixed glial cells by liquid chromatography and tandem mass spectrometry. Secretion of PAI-1 from glial cultures was detected by ELISA and western blotting analysis. Cell migration was evaluated by in vitro scratch-wound healing assay or Boyden chamber assay and an in vivo stab wound injury model. Phagocytic activity was measured by uptake of zymosan particles. Results The levels of PAI-1 mRNA and protein expression were increased by lipopolysaccharide and interferon-γ stimulation in both microglia and astrocytes. PAI-1 promoted the migration of microglial cells in culture via the low-density lipoprotein receptor-related protein (LRP) 1/Janus kinase (JAK)/signal transducer and activator of transcription (STAT)1 axis. PAI-1 also increased microglial migration in vivo when injected into mouse brain. PAI-1-mediated microglial migration was independent of protease inhibition, because an R346A mutant of PAI-1 with impaired PA inhibitory activity also promoted microglial migration. Moreover, PAI-1 was able to modulate microglial phagocytic activity. PAI-1 inhibited microglial engulfment of zymosan particles in a vitronectin- and Toll-like receptor 2/6-dependent manner. Conclusion Our results indicate that glia-derived PAI-1 may regulate microglial migration and phagocytosis in an autocrine or paracrine manner. This may have important implications in the regulation of brain microglial activities in health and disease.
Collapse
Affiliation(s)
- Hyejin Jeon
- Department of Pharmacology, Brain Science & Engineering Institute, CMRI, Kyungpook National University School of Medicine, 101 Dong-In, Daegu, Joong-gu, 700-422, South Korea
| | | | | | | | | | | |
Collapse
|
31
|
Elevated Tumor Expression of PAI-1 and SNAI2 in Obese Esophageal Adenocarcinoma Patients and Impact on Prognosis. Clin Transl Gastroenterol 2012; 3:e12. [PMID: 23238211 PMCID: PMC3365676 DOI: 10.1038/ctg.2012.5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES: Obesity is linked to increased mortality from many cancer types, and esophageal adenocarcinoma (EAC) displays one of the strongest epidemiological associations. The aims of this study are to dissect molecular pathways linking obesity with EAC and to determine if obesity is linked to increased aggressiveness of this disease. METHODS: Affymetrix microarrays identified altered signaling pathways in an EAC cell line following coculture with visceral adipose tissue or isolated adipocytes from viscerally obese EAC patients (n=6). Differentially expressed genes were subsequently investigated in patient tumor biopsies by quantitative reverse transcriptase PCR and examined with respect to obesity status, tumor biology, and patient survival. RESULTS: Visceral adipose tissue induced expression of genes involved in epithelial mesenchymal transition (EMT), plasminogen activator inhibitor (PAI)-1, and transcription factor SNAI2, in an EAC cell line. In EAC patient tumor biopsies from obese patients, we noted elevated expression of these genes, together with reduced expression of epithelial marker E-cadherin. SNAI2 was associated with EAC prognosis. CONCLUSIONS: Expression of EMT genes, PAI-1 and SNAI2, was elevated in tumors of obese EAC patients, and SNAI2 was associated with poor survival. Genes deregulated in obesity and associated with prognosis may represent potential targets for treatment stratification of obese EAC patients.
Collapse
|
32
|
Lee SH, Eren M, Vaughan DE, Schleimer RP, Cho SH. A plasminogen activator inhibitor-1 inhibitor reduces airway remodeling in a murine model of chronic asthma. Am J Respir Cell Mol Biol 2012; 46:842-6. [PMID: 22323366 DOI: 10.1165/rcmb.2011-0369oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We previously reported that plasminogen activator inhibitor (PAI)-1 deficiency prevents collagen deposition in the airways of ovalbumin (OVA)-challenged mice. In this study, we explored the therapeutic utility of blocking PAI-1 in preventing airway remodeling, using a specific PAI-1 inhibitor, tiplaxtinin. C57BL/6J mice were immunized with intraperitoneal injections of OVA on Days 0, 3, and 6. Starting on Day 11, mice were challenged with phosphate-buffered saline or OVA by nebulization three times per week for 4 weeks. Tiplaxtinin was mixed with chow and administered orally from 1 day before the phosphate-buffered saline or OVA challenge. Lung tissues were harvested after challenge and characterized histologically for infiltrating inflammatory cells, mucus-secreting goblet cells, and collagen deposition. Airway hyperresponsiveness was measured using whole-body plethysmography. Tiplaxtinin treatment significantly decreased levels of PAI-1 activity in bronchoalveolar lavage fluids, which indicates successful blockage of PAI-1 activity in the airways. The number of infiltrated inflammatory cells was reduced by tiplaxtinin treatment in the lungs of the OVA-challenged mice. Furthermore, oral administration of tiplaxtinin significantly attenuated the degree of goblet cell hyperplasia and collagen deposition in the airways of the OVA-challenged mice, and methacholine-induced airway hyperresponsiveness was effectively reduced by tiplaxtinin in these animals. This study supports our previous findings that PAI-1 promotes airway remodeling in a murine model of chronic asthma, and suggests that PAI-1 may be a novel target of treatment of airway remodeling in asthma.
Collapse
Affiliation(s)
- Sun H Lee
- Division of Allergy-Immunology, Department of Medicine, Northwestern Feinberg School of Medicine, 676 North St. Clair Street, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
33
|
DiMusto PD, Lu G, Ghosh A, Roelofs KJ, Su G, Zhao Y, Lau CL, Sadiq O, McEvoy B, Laser A, Diaz JA, Wakefield TW, Henke PK, Eliason JL, Upchurch GR. Increased PAI-1 in females compared with males is protective for abdominal aortic aneurysm formation in a rodent model. Am J Physiol Heart Circ Physiol 2012; 302:H1378-86. [PMID: 22307671 DOI: 10.1152/ajpheart.00620.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The serine proteases, along with their inhibitor plasmin activator inhibitor-1 (PAI-1), have been shown to play a role in abdominal aortic aneurysm (AAA) formation. The aim of this study is to determine if PAI-1 may be a protective factor for AAA formation and partially responsible for the gender difference observed in AAAs. Male and female wild-type (WT) C57BL/6 and PAI-1(-/-) mice 8-12 wk of age underwent aortic perfusion with porcine pancreatic elastase. Animals were harvested 14 days following perfusion and analyzed for phenotype, PAI-1 protein levels, and matrix metalloproteinase (MMP)-9 and -2 activity. WT males had an average increase in aortic diameter of 80%, whereas females only increased 32% (P < 0.001). PAI-1(-/-) males increased 204% and females 161%, significantly more than their WT counterparts (P < 0.001). Western blot revealed 61% higher PAI-1 protein levels in the WT females compared with the WT males (P = 0.01). Zymography revealed higher levels of pro-MMP-2 and active MMP-2 in the PAI-1(-/-) males and females compared with their WT counterparts. PAI-1(-/-) females had significantly higher serum plasmin levels compared with WT females (P = 0.003). In conclusion, WT female mice are protected from aneurysm formation and have higher levels of PAI-1 compared with males during experimental aneurysm formation. Additionally, both male and female PAI-1(-/-) animals develop significantly larger aneurysms than WT animals, correlating with higher pro- and active MMP-2 levels. These findings suggest that PAI-1 is protective for aneurysm formation in the elastase model of AAA and plays a role in the gender differences seen in AAA formation.
Collapse
Affiliation(s)
- Paul D DiMusto
- Jobst Vascular Research Laboratory, Department of Surgery, University of Michigan, Ann Arbor, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Thapa B, Kim YH, Kwon HJ, Kim DS. Novel regulatory mechanism and functional implication of plasminogen activator inhibitor-1 (PAI-1) expression in CpG-ODN-stimulated macrophages. Mol Immunol 2011; 49:572-81. [PMID: 22078208 DOI: 10.1016/j.molimm.2011.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 10/08/2011] [Accepted: 10/11/2011] [Indexed: 12/17/2022]
Abstract
Macrophages are activated by recognizing bacterial DNA and CpG-oligodeoxynucleotides (CpG-ODNs) through Toll-like receptor-9 (TLR-9). Plasminogen activator inhibitor-1 (PAI-1) has been shown to be an important factor in inflammation-induced macrophage migration which is essential for defense functions. The aim of this study was to demonstrate the molecular mechanism associated with the regulation of PAI-1 expression and its biological significance in CpG-ODN-stimulated mouse macrophages. Our results clearly show that PAI-1 expression in macrophages was highly up-regulated by CpG-ODN-stimulation in vitro and in vivo. The TLR-9-mediated stimulation of PAI-1 expression was independent of the NF-κB pathway and involved the synergistic activation of Sp1 and Elk-1 by the MEK1/2-ERK and JNK signaling pathways. The elevated PAI-1 expression resulted in significantly enhanced transmigration of RAW264.7 cells through vitronectin but not through fibronectin. We suggest that CpG-ODN plays a role in regulating macrophage migration by stimulating the expression of PAI-1, and the migration is modulated depending on the microenvironmental extracellular matrix components.
Collapse
Affiliation(s)
- Bikash Thapa
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 134 Sinchon-Dong, Seodaemun-Gu, 120-749 Seoul, Republic of Korea
| | | | | | | |
Collapse
|
35
|
Discovery of inhibitors of plasminogen activator inhibitor-1: Structure–activity study of 5-nitro-2-phenoxybenzoic acid derivatives. Bioorg Med Chem Lett 2011; 21:5701-6. [DOI: 10.1016/j.bmcl.2011.08.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 07/22/2011] [Accepted: 08/05/2011] [Indexed: 11/22/2022]
|
36
|
Chua KN, Poon KL, Lim J, Sim WJ, Huang RYJ, Thiery JP. Target cell movement in tumor and cardiovascular diseases based on the epithelial-mesenchymal transition concept. Adv Drug Deliv Rev 2011; 63:558-67. [PMID: 21335038 DOI: 10.1016/j.addr.2011.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 02/07/2011] [Accepted: 02/09/2011] [Indexed: 01/04/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a fundamental mechanism in development driving body plan formation. EMT describes a transition process wherein polarized epithelial cells lose their characteristics and acquire a mesenchymal phenotype. The apico-basal polarity of epithelial cells is replaced by a front-rear polarity in mesenchymal cells which favor cell-extracellular matrix than intercellular adhesion. These events serve as a prerequisite to the context-dependent migratory and invasive functions of mesenchymal cells. In solid tumors, carcinoma cells undergoing EMT not only invade and metastasize but also exhibit cancer stem cell-like properties, providing resistance to conventional and targeted therapies. In cardiovascular systems, epicardial cells engaged in EMT contribute to myocardial regeneration. Conversely, cardiovascular endothelial cells undergoing EMT cause cardiac fibrosis. Growing evidence has shed light on the potential development of novel therapeutics that target cell movement by applying the EMT concept, and this may provide new therapeutic strategies for the treatment of cancer and heart diseases.
Collapse
Affiliation(s)
- Kian-Ngiap Chua
- Institute of Molecular Cell Biology, Experimental Therapeutic Centre, Biopolis A*STAR, Cancer Science Institute National University of Singapore and Department of Obstetrics and Gynaecology, National University Hospital, Singapore, Republic of Singapore
| | | | | | | | | | | |
Collapse
|