1
|
Yepes M. Reprint of: Fibrinolytic and Non-fibrinolytic Roles of Tissue-type Plasminogen Activator in the Ischemic Brain. Neuroscience 2024; 550:21-29. [PMID: 38964373 DOI: 10.1016/j.neuroscience.2024.05.040] [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: 06/20/2023] [Accepted: 08/06/2023] [Indexed: 07/06/2024]
Abstract
The neurovascular unit (NVU) is assembled by endothelial cells (ECs) and pericytes, and encased by a basement membrane (BM) surveilled by microglia and surrounded by perivascular astrocytes (PVA), which in turn are in contact with synapses. Cerebral ischemia induces the rapid release of the serine proteinase tissue-type plasminogen activator (tPA) from endothelial cells, perivascular astrocytes, microglia and neurons. Owning to its ability to catalyze the conversion of plasminogen into plasmin, in the intravascular space tPA functions as a fibrinolytic enzyme. In contrast, the release of astrocytic, microglial and neuronal tPA have a plethora of effects that not always require the generation of plasmin. In the ischemic brain tPA increases the permeability of the NVU, induces microglial activation, participates in the recycling of glutamate, and has various effects on neuronal survival. These effects are mediated by different receptors, notably subunits of the N-methyl-D-aspartate receptor (NMDAR) and the low-density lipoprotein receptor-related protein-1 (LRP-1). Here we review data on the role of tPA in the NVU under non-ischemic and ischemic conditions, and analyze how this knowledge may lead to the development of potential strategies for the treatment of acute ischemic stroke patients.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology, Emory University, Atlanta, GA, USA; Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA; Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
2
|
Montecillo J, Pirker T, Pemberton C, Chew-Harris J. suPAR in cardiovascular disease. Adv Clin Chem 2024; 121:89-131. [PMID: 38797545 DOI: 10.1016/bs.acc.2024.04.005] [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] [Indexed: 05/29/2024]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR), the soluble counterpart of urokinase plasminogen activator receptor, is found in the circulation at various levels. suPAR and its parent molecule, cell surface uPAR, exhibit similar structure and extracellular functional roles facilitating fibrinolysis, cellular adhesion, and migration. Studies have assessed the correlation between suPAR in cardiovascular disease (CVD). It is postulated that suPAR may serve as an indicator of inflammatory activation and burden during CVD progression. Increased suPAR independently predicts poorer outcomes in acute coronary syndromes, in heart failure, as well as in coronary artery disease and atherosclerosis. To guide translation into clinical utization, suPAR has been assessed in numerous CVD settings for improved risk discrimination independently or in association with established traditional risk factors. Whilst the involvement of suPAR has been explored in other diseases such as kidney diseases and cancer, there is only emerging evidence of suPAR's mechanistic involvement in cardiovascular disease. In this review, we provide a background into suPAR and its potential role as a biomarker in CVD.
Collapse
Affiliation(s)
- Jaya Montecillo
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Thomas Pirker
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Janice Chew-Harris
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
3
|
Yepes M. Fibrinolytic and Non-fibrinolytic Roles of Tissue-type Plasminogen Activator in the Ischemic Brain. Neuroscience 2024; 542:69-80. [PMID: 37574107 DOI: 10.1016/j.neuroscience.2023.08.011] [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: 06/20/2023] [Revised: 08/03/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
The neurovascular unit (NVU) is assembled by endothelial cells (ECs) and pericytes, and encased by a basement membrane (BM) surveilled by microglia and surrounded by perivascular astrocytes (PVA), which in turn are in contact with synapses. Cerebral ischemia induces the rapid release of the serine proteinase tissue-type plasminogen activator (tPA) from endothelial cells, perivascular astrocytes, microglia and neurons. Owning to its ability to catalyze the conversion of plasminogen into plasmin, in the intravascular space tPA functions as a fibrinolytic enzyme. In contrast, the release of astrocytic, microglial and neuronal tPA have a plethora of effects that not always require the generation of plasmin. In the ischemic brain tPA increases the permeability of the NVU, induces microglial activation, participates in the recycling of glutamate, and has various effects on neuronal survival. These effects are mediated by different receptors, notably subunits of the N-methyl-D-aspartate receptor (NMDAR) and the low-density lipoprotein receptor-related protein-1 (LRP-1). Here we review data on the role of tPA in the NVU under non-ischemic and ischemic conditions, and analyze how this knowledge may lead to the development of potential strategies for the treatment of acute ischemic stroke patients.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology, Emory University, Atlanta, GA, USA; Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA; Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
4
|
Enhanced Expression of Plasminogen Activators and Inhibitor in the Healing of Tympanic Membrane Perforation in Rats. J Assoc Res Otolaryngol 2023; 24:159-170. [PMID: 36810718 PMCID: PMC10121974 DOI: 10.1007/s10162-023-00891-5] [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: 04/19/2022] [Accepted: 01/25/2023] [Indexed: 02/23/2023] Open
Abstract
The significance of plasminogen activation during the tympanic membrane (TM) healing is known mainly from studies performed on knock-out mice. In the previous study, we reported activation of genes coding proteins of plasminogen activation and inhibition system in rat's TM perforation healing. The aim of the present study was the evaluation of protein products expressed by these genes and their tissue distribution using Western blotting and immunofluorescent method, respectively, during 10-day observation period after injury. Otomicroscopical and histological evaluation were employed to assess the healing process. The expression of urokinase plasminogen activator (uPA) and its receptor (uPAR) were significantly upregulated in the proliferation phase, with subsequent gradual attenuation during remodeling phase of healing process, when keratinocyte migration was weakening. The expression of plasminogen activator inhibitor type 1 (PAI-1) also showed the highest levels during the proliferation phase. The increase of tissue plasminogen activator (tPA) expression was observed during the whole observation period, with the highest activity during the remodeling phase. Immunofluorescence of these proteins was present mainly in migrating epithelium. Our study found that plasminogen activation (uPA, uPAR, tPA) and inhibitory (PAI-1) molecules form a well-structured regulatory system of the epithelial migration that is critical to the healing of TM after its perforation.
Collapse
|
5
|
Kaur B, Kerbrat J, Kho J, Kaler M, Kanatsios S, Cirillo N. Mechanism-based therapeutic targets of pemphigus vulgaris: A scoping review of pathogenic molecular pathways. Exp Dermatol 2022; 31:154-171. [PMID: 34435386 DOI: 10.1111/exd.14453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/20/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022]
Abstract
Pemphigus vulgaris (PV) is a potentially fatal autoimmune blistering disease characterised by cell-cell detachment or acantholysis. The mechanisms which follow antibody (Ab) binding and culminate in acantholytic changes and skin/mucosal blistering have not been fully clarified. Current treatment strategies are not specific to PV pathophysiology and although life-saving, harbour considerable side effects. We aimed to systematically assess the molecules amenable to targeted treatments that follow Ab binding and are associated with PV acantholysis. The resulting scoping review was conducted under PRISMA-ScR guidelines with clear inclusion and exclusion criteria and focused specifically on kinases, caspases, proteases, hydrolytic enzymes and other molecules of interest postulated to take part in the pathophysiology of PV. The review process resulted in the identification of 882 articles, of which 56 were eligible for qualitative synthesis. From the included articles, the majority (n = 42) used PV-IgG as the pathogenic agent, mainly via in vitro (n = 16) and in vivo (n = 10) models. Twenty-five molecules were found to play a pathogenic role in PV, including uPA, ADAM10, EGFR, Src, PKC, cdk2, ERK, PLC, calmodulin, NOS, p38MAPK and caspase-3. Selective inhibition of these molecules resulted in varying degrees of reduction in acantholysis and blistering. The pathogenic molecules identified in this review represent potential candidates for clinical translation.
Collapse
Affiliation(s)
- Bavleen Kaur
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Jenna Kerbrat
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Jia Kho
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Manreet Kaler
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Stefanos Kanatsios
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| |
Collapse
|
6
|
Li S, Jia H, Liu Z, Wang N, Guo X, Cao M, Fang F, Yang J, Li J, He Q, Guo R, Zhang T, Kang K, Wang Z, Liu S, Cao Y, Jiang X, Ren G, Wang K, Yu B, Xiao W, Li D. Fibroblast growth factor-21 as a novel metabolic factor for regulating thrombotic homeostasis. Sci Rep 2022; 12:400. [PMID: 35013379 PMCID: PMC8748457 DOI: 10.1038/s41598-021-00906-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/12/2021] [Indexed: 11/24/2022] Open
Abstract
Fibroblast growth factor-21 (FGF-21) performs a wide range of biological functions in organisms. Here, we report for the first time that FGF-21 suppresses thrombus formation with no notable risk of bleeding. Prophylactic and therapeutic administration of FGF-21 significantly improved the degree of vascular stenosis and reduced the thrombus area, volume and burden. We determined the antithrombotic mechanism of FGF-21, demonstrating that FGF-21 exhibits an anticoagulant effect by inhibiting the expression and activity of factor VII (FVII). FGF-21 exerts an antiplatelet effect by inhibiting platelet activation. FGF-21 enhances fibrinolysis by promoting tissue plasminogen activator (tPA) expression and activation, while inhibiting plasminogen activator inhibitor 1 (PAI-1) expression and activation. We further found that FGF-21 mediated the expression and activation of tPA and PAI-1 by regulating the ERK1/2 and TGF-β/Smad2 pathways, respectively. In addition, we found that FGF-21 inhibits the expression of inflammatory factors in thrombosis by regulating the NF-κB pathway.
Collapse
Affiliation(s)
- Shuai Li
- College of Life Sciences and Agriculture and Forestry, Qiqihar University, Qiqihar, 161006, People's Republic of China
| | - Haibo Jia
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Zhihang Liu
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Nan Wang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xiaochen Guo
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Muhua Cao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Fang Fang
- Molecular Imaging Research Center, Harbin Medical University, TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, 150028, People's Republic of China
| | - Jiarui Yang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Junyan Li
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qi He
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Rui Guo
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Teng Zhang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Kai Kang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Zongbao Wang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shijie Liu
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yukai Cao
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xinghao Jiang
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Guiping Ren
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Kai Wang
- Molecular Imaging Research Center, Harbin Medical University, TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, 150028, People's Republic of China.
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China.
| | - Wei Xiao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical CO. LTD, Lianyungang, 222001, People's Republic of China.
| | - Deshan Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical CO. LTD, Lianyungang, 222001, People's Republic of China.
- Bio-Pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
7
|
Kearney KJ, Ariëns RAS, Macrae FL. The Role of Fibrin(ogen) in Wound Healing and Infection Control. Semin Thromb Hemost 2021; 48:174-187. [PMID: 34428799 DOI: 10.1055/s-0041-1732467] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fibrinogen, one of the most abundant plasma proteins playing a key role in hemostasis, is an important modulator of wound healing and host defense against microbes. In the current review, we address the role of fibrin(ogen) throughout the process of wound healing and subsequent tissue repair. Initially fibrin(ogen) acts as a provisional matrix supporting incoming leukocytes and acting as reservoir for growth factors. It later goes on to support re-epithelialization, angiogenesis, and fibroplasia. Importantly, removal of fibrin(ogen) from the wound is essential for wound healing to progress. We also discuss how fibrin(ogen) functions through several mechanisms to protect the host against bacterial infection by providing a physical barrier, entrapment of bacteria in fibrin(ogen) networks, and by directing immune cell function. The central role of fibrin(ogen) in defense against bacterial infection has made it a target of bacterial proteins, evolved to interact with fibrin(ogen) to manipulate clot formation and degradation for the purpose of promoting microbial virulence and survival. Further understanding of the dual roles of fibrin(ogen) in wound healing and infection could provide novel means of therapy to improve recovery from surgical or chronic wounds and help to prevent infection from highly virulent bacterial strains, including those resistant to antibiotics.
Collapse
Affiliation(s)
- Katherine J Kearney
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Fraser L Macrae
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
8
|
Ruiz-Cañada C, Bernabé-García Á, Liarte S, Rodríguez-Valiente M, Nicolás FJ. Chronic Wound Healing by Amniotic Membrane: TGF-β and EGF Signaling Modulation in Re-epithelialization. Front Bioeng Biotechnol 2021; 9:689328. [PMID: 34295882 PMCID: PMC8290337 DOI: 10.3389/fbioe.2021.689328] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022] Open
Abstract
The application of amniotic membrane (AM) on chronic wounds has proven very effective at resetting wound healing, particularly in re-epithelialization. Historically, several aspects of AM effect on wound healing have been evaluated using cell models. In keratinocytes, the presence of AM induces the activation of mitogen-activated protein (MAP) kinase and c-Jun N-terminal kinase (JNK) pathways, together with the high expression of c-Jun, an important transcription factor for the progression of the re-epithelialization tongue. In general, the levels of transforming growth factor (TGF)-β present in a wound are critical for the process of wound healing; they are elevated during the inflammation phase and remain high in some chronic wounds. Interestingly, the presence of AM, through epidermal growth factor (EGF) signaling, produces a fine-tuning of the TGF-β signaling pathway that re-conducts the stalled process of wound healing. However, the complete suppression of TGF-β signaling has proven negative for the AM stimulation of migration, suggesting that a minimal amount of TGF-β signaling is required for proper wound healing. Regarding migration machinery, AM contributes to the dynamics of focal adhesions, producing a high turnover and thus speeding up remodeling. This is clear because proteins, such as Paxillin, are activated upon treatment with AM. On top of this, AM also produces changes in the expression of Paxillin. Although we have made great progress in understanding the effects of AM on chronic wound healing, a long way is still ahead of us to fully comprehend its effects.
Collapse
Affiliation(s)
- Catalina Ruiz-Cañada
- Laboratorio de Regeneración, Oncología Molecular y TGF-β, IMIB-Arrixaca, Murcia, Spain
| | - Ángel Bernabé-García
- Laboratorio de Regeneración, Oncología Molecular y TGF-β, IMIB-Arrixaca, Murcia, Spain
| | - Sergio Liarte
- Laboratorio de Regeneración, Oncología Molecular y TGF-β, IMIB-Arrixaca, Murcia, Spain
| | - Mónica Rodríguez-Valiente
- Laboratorio de Regeneración, Oncología Molecular y TGF-β, IMIB-Arrixaca, Murcia, Spain.,Unidad de Heridas Crónicas y Úlcera de Pie Diabético, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | | |
Collapse
|
9
|
Westby MJ, Norman G, Watson REB, Cullum NA, Dumville JC. Protease activity as a prognostic factor for wound healing in complex wounds. Wound Repair Regen 2020; 28:631-644. [DOI: 10.1111/wrr.12835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/26/2020] [Accepted: 04/17/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Maggie J. Westby
- Division of Nursing, Midwifery and Social Work, Manchester Academic Health Science Centre University of Manchester Manchester UK
- NIHR Manchester Biomedical Research Centre Manchester University NHS Foundation Trust Manchester UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences University of Oxford Oxford UK
| | - Gill Norman
- Division of Nursing, Midwifery and Social Work, Manchester Academic Health Science Centre University of Manchester Manchester UK
- NIHR Manchester Biomedical Research Centre Manchester University NHS Foundation Trust Manchester UK
| | - Rachel E. B. Watson
- NIHR Manchester Biomedical Research Centre Manchester University NHS Foundation Trust Manchester UK
- Centre for Dermatology Research, Division of Musculoskeletal & Dermatological Sciences University of Manchester Manchester UK
| | - Nicky A. Cullum
- Division of Nursing, Midwifery and Social Work, Manchester Academic Health Science Centre University of Manchester Manchester UK
- NIHR Manchester Biomedical Research Centre Manchester University NHS Foundation Trust Manchester UK
- Research and Innovation Division Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre Manchester UK
| | - Jo C. Dumville
- Division of Nursing, Midwifery and Social Work, Manchester Academic Health Science Centre University of Manchester Manchester UK
- NIHR Manchester Biomedical Research Centre Manchester University NHS Foundation Trust Manchester UK
| |
Collapse
|
10
|
Opneja A, Kapoor S, Stavrou EX. Contribution of platelets, the coagulation and fibrinolytic systems to cutaneous wound healing. Thromb Res 2019; 179:56-63. [PMID: 31078121 DOI: 10.1016/j.thromres.2019.05.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/14/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
Wound healing is a complex process that consists of multiple phases, each of which are indispensable for adequate repair. Timely initiation and resolution of each of these phases namely, hemostasis, inflammation, proliferation and tissue remodeling, is critical for promoting healing and avoiding excess scar formation. While platelets have long been known to influence the healing process, other components of blood particularly coagulation factors and the fibrinolytic system also contribute to efficient wound repair. This review aims to summarize our current understanding of the role of platelets, the coagulation and fibrinolytic systems in cutaneous wound healing, with a focus on how these components communicate with immune and non-immune cells in the wound microenvironment. We also outline current and potential therapeutic strategies to improve the management of chronic, non-healing wounds.
Collapse
Affiliation(s)
- Aman Opneja
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sargam Kapoor
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Evi X Stavrou
- Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Medicine, Louis Stokes Veterans Administration Medical Center, VA Northeast Ohio Healthcare System, Cleveland, OH, USA.
| |
Collapse
|
11
|
Westby MJ, Dumville JC, Stubbs N, Norman G, Wong JKF, Cullum N, Riley RD, Cochrane Wounds Group. Protease activity as a prognostic factor for wound healing in venous leg ulcers. Cochrane Database Syst Rev 2018; 9:CD012841. [PMID: 30171767 PMCID: PMC6513613 DOI: 10.1002/14651858.cd012841.pub2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Venous leg ulcers (VLUs) are a common type of complex wound that have a negative impact on people's lives and incur high costs for health services and society. It has been suggested that prolonged high levels of protease activity in the later stages of the healing of chronic wounds may be associated with delayed healing. Protease modulating treatments have been developed which seek to modulate protease activity and thereby promote healing in chronic wounds. OBJECTIVES To determine whether protease activity is an independent prognostic factor for the healing of venous leg ulcers. SEARCH METHODS In February 2018, we searched the following databases: Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE, Ovid Embase and CINAHL. SELECTION CRITERIA We included prospective and retrospective longitudinal studies with any follow-up period that recruited people with VLUs and investigated whether protease activity in wound fluid was associated with future healing of VLUs. We included randomised controlled trials (RCTs) analysed as cohort studies, provided interventions were taken into account in the analysis, and case-control studies if there were no available cohort studies. We also included prediction model studies provided they reported separately associations of individual prognostic factors (protease activity) with healing. Studies of any type of protease or combination of proteases were eligible, including proteases from bacteria, and the prognostic factor could be examined as a continuous or categorical variable; any cut-off point was permitted. The primary outcomes were time to healing (survival analysis) and the proportion of people with ulcers completely healed; the secondary outcome was change in ulcer size/rate of wound closure. We extracted unadjusted (simple) and adjusted (multivariable) associations between the prognostic factor and healing. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies for inclusion at each stage, and undertook data extraction, assessment of risk of bias and GRADE assessment. We collected association statistics where available. No study reported adjusted analyses: instead we collected unadjusted results or calculated association measures from raw data. We calculated risk ratios when both outcome and prognostic factor were dichotomous variables. When the prognostic factor was reported as continuous data and healing outcomes were dichotomous, we either performed regression analysis or analysed the impact of healing on protease levels, analysing as the standardised mean difference. When both prognostic factor and outcome were continuous data, we reported correlation coefficients or calculated them from individual participant data.We displayed all results on forest plots to give an overall visual representation. We planned to conduct meta-analyses where this was appropriate, otherwise we summarised narratively. MAIN RESULTS We included 19 studies comprising 21 cohorts involving 646 participants. Only 11 studies (13 cohorts, 522 participants) had data available for analysis. Of these, five were prospective cohort studies, four were RCTs and two had a type of case-control design. Follow-up time ranged from four to 36 weeks. Studies covered 10 different matrix metalloproteases (MMPs) and two serine proteases (human neutrophil elastase and urokinase-type plasminogen activators). Two studies recorded complete healing as an outcome; other studies recorded partial healing measures. There was clinical and methodological heterogeneity across studies; for example, in the definition of healing, the type of protease and its measurement, the distribution of active and bound protease species, the types of treatment and the reporting of results. Therefore, meta-analysis was not performed. No study had conducted multivariable analyses and all included evidence was of very low certainty because of the lack of adjustment for confounders, the high risk of bias for all studies except one, imprecision around the measures of association and inconsistency in the direction of association. Collectively the research indicated complete uncertainty as to the association between protease activity and VLU healing. AUTHORS' CONCLUSIONS This review identified very low validity evidence regarding any association between protease activity and VLU healing and there is complete uncertainty regarding the relationship. The review offers information for both future research and systematic review methodology.
Collapse
Affiliation(s)
- Maggie J Westby
- University of Manchester, Manchester Academic Health Science CentreDivision of Nursing, Midwifery and Social Work, School of Health Sciences, Faculty of Biology, Medicine and HealthJean McFarlane BuildingOxford RoadManchesterUKM13 9PL
| | - Jo C Dumville
- University of Manchester, Manchester Academic Health Science CentreDivision of Nursing, Midwifery and Social Work, School of Health Sciences, Faculty of Biology, Medicine and HealthJean McFarlane BuildingOxford RoadManchesterUKM13 9PL
| | - Nikki Stubbs
- St Mary's HospitalLeeds Community Healthcare NHS Trust3 Greenhill RoadLeedsUKLS12 3QE
| | - Gill Norman
- University of Manchester, Manchester Academic Health Science CentreDivision of Nursing, Midwifery and Social Work, School of Health Sciences, Faculty of Biology, Medicine and HealthJean McFarlane BuildingOxford RoadManchesterUKM13 9PL
| | - Jason KF Wong
- Manchester University NHS Foundation TrustManchester Centre for Plastic Surgery and Burns, Wythenshawe HospitalSouthmoor Road, WythenshaweManchesterUKM23 9LT
| | - Nicky Cullum
- University of Manchester, Manchester Academic Health Science CentreDivision of Nursing, Midwifery and Social Work, School of Health Sciences, Faculty of Biology, Medicine and HealthJean McFarlane BuildingOxford RoadManchesterUKM13 9PL
| | - Richard D Riley
- Keele UniversityResearch Institute for Primary Care and Health SciencesDavid Weatherall Building, Keele University CampusKeeleStaffordshireUKST5 5BG
| | | |
Collapse
|
12
|
Westby MJ, Dumville JC, Stubbs N, Norman G, Wong JKF, Cullum N, Riley R. Protease activity as a prognostic factor for wound healing in venous leg ulcers. Hippokratia 2017. [DOI: 10.1002/14651858.cd012841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Maggie J Westby
- University of Manchester, Manchester Academic Health Science Centre; Division of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine & Health; Jean McFarlane Building Oxford Road Manchester UK M13 9PL
| | - Jo C Dumville
- University of Manchester, Manchester Academic Health Science Centre; Division of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine & Health; Jean McFarlane Building Oxford Road Manchester UK M13 9PL
| | - Nikki Stubbs
- Leeds Community Healthcare NHS Trust, St Mary's Hospital; Wound Prevention and Management Service; 3 Greenhill Road Leeds UK LS12 3QE
| | - Gill Norman
- University of Manchester, Manchester Academic Health Science Centre; Division of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine & Health; Jean McFarlane Building Oxford Road Manchester UK M13 9PL
| | - Jason KF Wong
- University Hospital South Manchester; Plastic and Reconstructive Surgery; Southmoor Road, Wythenshawe Manchester UK M23 9LT
| | - Nicky Cullum
- University of Manchester, Manchester Academic Health Science Centre; Division of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine & Health; Jean McFarlane Building Oxford Road Manchester UK M13 9PL
| | - Richard Riley
- Keele University; Research Institute for Primary Care and Health Sciences; David Weatherall Building, Keele University Campus Keele Staffordshire UK ST5 5BG
| |
Collapse
|
13
|
Li Y, Zhang J, Yue J, Gou X, Wu X. Epidermal Stem Cells in Skin Wound Healing. Adv Wound Care (New Rochelle) 2017; 6:297-307. [PMID: 28894637 DOI: 10.1089/wound.2017.0728] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/13/2017] [Indexed: 12/15/2022] Open
Abstract
Significance: Skin serves as a protective barrier for mammals. Epidermal stem cells are responsible for maintaining skin homeostasis. When cutaneous injuries occur, skin homeostasis and integrity are damaged, leading to dire consequences such as acute, chronic, or infected wounds. Skin wound healing is an intrinsic self-saving chain reaction, which is crucial to facilitating the replacement of damaged or lost tissue. Recent Advances: An immense amount of research has uncovered the underlying mechanisms behind the complex and highly regulated wound healing process. In this review, we will dissect the biological process of adult skin wound healing and emphasize the importance of epidermal stem cells during the wound healing. Critical Issues: We will comprehensively discuss the current clinical practices used on patients with cutaneous wounds, including both traditional skin grafting procedures and advanced grafting techniques with cultured skin stem cells. The majority of these leading techniques still retain some deficiencies during clinical use. Moreover, the regeneration of skin appendages after severe injuries remains a challenge in treatment. Future Directions: Understanding epidermal stem cells and their essential functions during skin wound healing are fundamental components behind the development of clinical treatment on patients with cutaneous wounds. It is important to improve the current standard of care and to develop novel techniques improving patient outcomes and long-term rehabilitation, which should be the goals of future endeavors in the field of skin wound healing.
Collapse
Affiliation(s)
- Yuanyuan Li
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Jamie Zhang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Jiping Yue
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Xuewen Gou
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| |
Collapse
|
14
|
Montesinos MC, Desai-Merchant A, Cronstein BN. Promotion of Wound Healing by an Agonist of Adenosine A2A Receptor Is Dependent on Tissue Plasminogen Activator. Inflammation 2016; 38:2036-41. [PMID: 25991438 DOI: 10.1007/s10753-015-0184-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Impaired wound healing, as it occurs in diabetes mellitus or long-term corticoid treatment, is commonly associated with disability, diminished quality of life, and high economic costs. Selective agonists of the A2A receptor subtype of adenosine, an endogenous regulator of inflammation, promote tissue repair in animal models, both healthy and with impaired healing. Plasmin-mediated proteolysis of fibrin and other matrix proteins is essential for cell migration at sites of injury. Since adenosine A2A receptor activation increases plasminogen activator release from macrophages and mast cells, we studied the effect of a selective agonist, CGS-21680, on full-thickness excisional wound closure in wild-type, urokinase plasminogen activator (uPA)-deficient, and tissue plasminogen activator (tPA)-deficient mice. Wound closure was impaired in tPA- and uPA-deficient mice as compared with wild-type mice, and topical application of CGS-21680 significantly increased the rate at which wounds closed in wild-type mice and uPA-deficient mice, but not in tPA-deficient mice. Immunostaining of tissue sections showed that tPA was present in endothelial cells and histiocytes by day 3 post-wound and also by day 6. In contrast, uPA was more prominent in these cell types only by day 6 post-wound. Our results confirm that plasminogen activation contributes to wound repair and are consistent with the hypothesis that adenosine A2A receptor activation promotes wound closure by a mechanism that depends upon tPA, but not uPA. Moreover, our results suggest that topical adenosine A2A receptor agonists may be useful in promotion of wound closure in patients with impaired wound healing.
Collapse
Affiliation(s)
- M Carmen Montesinos
- Department of Medicine, New York University School of Medicine, 550 First Avenue, MSB251, New York, NY, 10016, USA. .,Instituto de Reconocimiento Molecular y Desarrollo Tecnológico, Centro Mixto Universidad Politécnica de Valencia, Universidad de Valencia, Valencia, Spain. .,Department de Farmacologia, Universitat de València, Ave. Vicent Andrès Estellès s/n, 46100 Burjassot, Valencia, Spain.
| | - Avani Desai-Merchant
- Department of Medicine, New York University School of Medicine, 550 First Avenue, MSB251, New York, NY, 10016, USA
| | - Bruce N Cronstein
- Department of Medicine, New York University School of Medicine, 550 First Avenue, MSB251, New York, NY, 10016, USA
| |
Collapse
|
15
|
Prothrombin Complex Concentrate Is Effective in Treating the Anticoagulant Effects of Dabigatran in a Porcine Polytrauma Model. Anesthesiology 2015; 123:1350-61. [DOI: 10.1097/aln.0000000000000863] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
Background
In the event of trauma, emergency reversal of anticoagulation therapy may be required. However, no specific reversal agents are routinely available for the direct oral anticoagulants such as dabigatran. The authors investigated four-factor prothrombin complex concentrate (PCC) for treating dabigatran-induced anticoagulation in a porcine polytrauma model.
Methods
Dabigatran etexilate was given orally for 3 days and intravenously on day 4 to 32 pigs. Animals were randomized 1:1:1:1 to PCC (25, 50, or 100 U/kg) or saline. Study medication was administered 12 min after bilateral femur fractures and blunt liver injury. The primary endpoint was blood loss at 300 min.
Results
The mean plasma concentration of dabigatran was 487 ± 161 ng/ml after intravenous administration. Blood loss was 3,855 ± 258 ml in controls and 3,588 ± 241 ml in the PCC25 group. In the PCC50 and PCC100 groups, blood loss was significantly lower: 1,749 ± 47 ml and 1,692 ± 97 ml, respectively. PCC50 and PCC100 effectively reduced dabigatran’s effects on coagulation parameters, whereas control and (to a lesser extent) PCC25 animals developed severe coagulopathy. Sustained increases in endogenous thrombin potential occurred with PCC50 and PCC100.
Conclusion
Four-factor PCC (50 or 100 U/kg) is effective in reducing blood loss in dabigatran-anticoagulated pigs, but higher doses may induce a procoagulant state.
Collapse
|
16
|
Bhattacharya S, Aggarwal R, Singh VP, Ramachandran S, Datta M. Downregulation of miRNAs during Delayed Wound Healing in Diabetes: Role of Dicer. Mol Med 2015; 21:847-860. [PMID: 26602065 DOI: 10.2119/molmed.2014.00186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/02/2015] [Indexed: 12/11/2022] Open
Abstract
Delayed wound healing is a major complication associated with diabetes and is a result of a complex interplay among diverse deregulated cellular parameters. Although several genes and pathways have been identified to be mediating impaired wound closure, the role of microRNAs (miRNAs) in these events is not very well understood. Here, we identify an altered miRNA signature in the prolonged inflammatory phase in a wound during diabetes, with increased infiltration of inflammatory cells in the basal layer of the epidermis. Nineteen miRNAs were downregulated in diabetic rat wounds (as compared with normal rat wound, d 7 postwounding) together with inhibited levels of the central miRNA biosynthesis enzyme, Dicer, suggesting that in wounds of diabetic rats, the decreased levels of Dicer are presumably responsible for miRNA downregulation. Compared with unwounded skin, Dicer levels were significantly upregulated 12 d postwounding in normal rats, and this result was notably absent in diabetic rats that showed impaired wound closure. In a wound-healing specific quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) array, 10 genes were significantly altered in the diabetic rat wound and included growth factors and collagens. Network analyses demonstrated significant interactions and correlations between the miRNA predicted targets (regulators) and the 10 wound-healing specific genes, suggesting altered miRNAs might fine-tune the levels of these genes that determine wound closure. Dicer inhibition prevented HaCaT cell migration and affected wound closure. Altered levels of Dicer and miRNAs are critical during delayed wound closure and offer promising targets to address the issue of impaired wound healing.
Collapse
Affiliation(s)
- Sushant Bhattacharya
- Council of Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology, Delhi, India
| | - Rangoli Aggarwal
- Council of Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology, Delhi, India
| | - Vijay Pal Singh
- Council of Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology, Delhi, India
| | - Srinivasan Ramachandran
- Council of Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology, Delhi, India
| | - Malabika Datta
- Council of Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology, Delhi, India
| |
Collapse
|
17
|
Wada T, Nangaku M. A circulating permeability factor in focal segmental glomerulosclerosis: the hunt continues. Clin Kidney J 2015; 8:708-15. [PMID: 26613029 PMCID: PMC4655796 DOI: 10.1093/ckj/sfv090] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 08/11/2015] [Indexed: 01/23/2023] Open
Abstract
Primary focal segmental glomerulosclerosis (FSGS) is one of the major causes of steroid-resistant nephrotic syndrome, and renal prognosis in patients with steroid-resistant FSGS is poor. It has been long speculated that a circulating permeability factor should be implicated in the pathogenesis of the disease because a substantial portion of the patients with primary FSGS experience recurrence shortly after transplantation. Although molecules such as cardiotrophin-like cytokine 1 (CLC-1) and anti-CD40 antibody have been proposed to be potential circulating permeability factors, a definitive factor remains to be discovered. Soluble urokinase-type plasminogen activator receptor (suPAR) has attracted substantial attention and garnered scrutiny by renal researchers since Reiser's group suggested that it was linked to the pathogenesis of primary FSGS and that it might be useful as a diagnostic biomarker. A number of different cohort studies have shown that serum suPAR levels are negatively associated with renal function and can scarcely differentiate FSGS from the other glomerular/renal diseases. In contrast to initial studies, several in vivo studies investigating the effects of forced suPAR upregulation could not show the induction of proteinuria or podocyte injury. Currently it is suggested that a different form of suPAR, which cannot be measured by presently available enzyme-linked immunosorbent assay, might be the culprit; however, it remains to be determined whether this is the case. Because a circulating permeability factor might be a useful biomarker for diagnosing FSGS as well as a potent therapeutic target for primary and recurrent FSGS, further dedicated work will be needed.
Collapse
Affiliation(s)
- Takehiko Wada
- Division of Nephrology and Endocrinology , University of Tokyo School of Medicine , Tokyo 113-8655 , Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology , University of Tokyo School of Medicine , Tokyo 113-8655 , Japan
| |
Collapse
|
18
|
Ahmad A, Saha P, Evans C, Thurison T, Hoyer-Hansen G, Patel A, Modarai B, Smith A. The soluble urokinase plasminogen activator receptor and its fragments in venous ulcers. J Vasc Surg Venous Lymphat Disord 2014; 3:190-7. [PMID: 26993839 DOI: 10.1016/j.jvsv.2014.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/08/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Activation of proteolytic mechanisms at the cell surface through the activity of urokinase-type plasminogen activator (uPA) bound to its receptor, uPAR, is an important process in wound healing. The soluble forms of uPAR (suPAR and its fragments I, II, and III) have nonproteolytic functions that include chemotaxis, adhesion, and proliferation, which also have a role in wound healing. The aim of this study was to determine whether suPAR and its cleaved fragments are present in venous ulcers and whether their levels are associated with healing. METHODS Ulcer exudates were collected from patients with venous leg ulcers (n = 30). Healing was defined as complete re-epithelialization within 6 months of compression therapy. Time-resolved fluorescence immunoassays were validated for quantification of suPAR and its fragments in ulcer exudates. The effect of exudates on keratinocyte migration was analyzed by an in vitro scratch assay. RESULTS Ulcer exudates from patients who healed (n = 9) had approximately threefold higher levels of intact suPAR (P = .005), twofold higher levels of suPARI (P = .03), and approximately threefold higher levels suPARII-III (P < .0001) compared with nonhealers (n = 21). Exudate from healing ulcers stimulated keratinocyte migration (P = .02), whereas depletion of suPAR from exudates resulted in cell apoptosis. CONCLUSIONS We conclude that suPAR and its fragments are present in the environs of venous ulcers and may act as indicators of the propensity of venous ulcers to heal, with suPARII-III being the best discriminator. We speculate that suPAR and its fragments may have a role in the maintenance of an optimal ulcer-healing environment.
Collapse
Affiliation(s)
- Anwar Ahmad
- King's College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at GSTT and King's College London, Academic Department of Vascular Surgery, Cardiovascular Division, St Thomas' Hospital, London, United Kingdom
| | - Prakash Saha
- King's College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at GSTT and King's College London, Academic Department of Vascular Surgery, Cardiovascular Division, St Thomas' Hospital, London, United Kingdom
| | - Colin Evans
- King's College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at GSTT and King's College London, Academic Department of Vascular Surgery, Cardiovascular Division, St Thomas' Hospital, London, United Kingdom
| | - Tine Thurison
- Finsen Laboratory, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Ashish Patel
- King's College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at GSTT and King's College London, Academic Department of Vascular Surgery, Cardiovascular Division, St Thomas' Hospital, London, United Kingdom
| | - Bijan Modarai
- King's College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at GSTT and King's College London, Academic Department of Vascular Surgery, Cardiovascular Division, St Thomas' Hospital, London, United Kingdom
| | - Alberto Smith
- King's College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at GSTT and King's College London, Academic Department of Vascular Surgery, Cardiovascular Division, St Thomas' Hospital, London, United Kingdom.
| |
Collapse
|
19
|
Trimarchi H. Primary focal and segmental glomerulosclerosis and soluble factor urokinase-type plasminogen activator receptor. World J Nephrol 2013; 2:103-110. [PMID: 24255893 PMCID: PMC3832866 DOI: 10.5527/wjn.v2.i4.103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 09/24/2013] [Accepted: 10/20/2013] [Indexed: 02/06/2023] Open
Abstract
Primary focal and segmental glomerulosclerosis (FSGS) may be due to genetic or acquired etiologies and is a common cause of nephrotic syndrome with high morbidity that often leads to end-stage renal failure. The different available therapeutic approaches are unsuccessful, in part due to partially deciphered heterogeneous and complex pathophysiological mechanisms. Moreover, the term FSGS, even in its primary form, comprises a histological description shared by a number of different causes with completely different molecular pathways of disease. This review focuses on the latest developments regarding the pathophysiology of primary acquired FSGS caused by soluble factor urokinase type plasminogen activator receptor, a circulating permeability factor involved in proteinuria and edema formation, and describes recent advances with potential success in therapy.
Collapse
|
20
|
Shen Y, Guo Y, Du C, Wilczynska M, Hellström S, Ny T. Mice deficient in urokinase-type plasminogen activator have delayed healing of tympanic membrane perforations. PLoS One 2012; 7:e51303. [PMID: 23236466 PMCID: PMC3517469 DOI: 10.1371/journal.pone.0051303] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 10/31/2012] [Indexed: 11/18/2022] Open
Abstract
Mice deficient in plasminogen, the precursor of plasmin, show completely arrested healing of tympanic membrane (TM) perforations, indicating that plasmin plays an essential role in TM healing. The activation of plasminogen to plasmin is performed by two plasminogen activators (PAs), urokinase-type PA (uPA) and tissue-type PA (tPA). To elucidate the functional roles of PAs in the healing of TM perforations, we investigated the phenotypes of single gene-deficient mice lacking uPA (uPA(-/-)) or tPA (tPA(-/-)) after TM perforation. Delayed healing of TM perforations was observed in uPA(-/-) mice but not tPA(-/-) mice. The migration of keratinocytes was clearly delayed and seemed to be misoriented in uPA(-/-) mice. Furthermore, fibrin deposition and the inflammatory response were persistent in these mice. Our findings demonstrate that uPA plays a role in the healing of TM perforations. The observed phenotypes in uPA(-/-) mice are most likely due to the reduced generation of plasmin.
Collapse
Affiliation(s)
- Yue Shen
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Yongzhi Guo
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Chun Du
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | | | - Sten Hellström
- Department of Audiology and Neurotology, Karolinska University Hospital, Stockholm, Sweden
| | - Tor Ny
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
21
|
Botta A, Delteil F, Mettouchi A, Vieira A, Estrach S, Négroni L, Stefani C, Lemichez E, Meneguzzi G, Gagnoux-Palacios L. Confluence switch signaling regulates ECM composition and the plasmin proteolytic cascade in keratinocytes. J Cell Sci 2012; 125:4241-52. [PMID: 22641690 DOI: 10.1242/jcs.096289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In culture, cell confluence generates signals that commit actively growing keratinocytes to exit the cell cycle and differentiate to form a stratified epithelium. Using a comparative proteomic approach, we studied this 'confluence switch' and identified a new pathway triggered by cell confluence that regulates basement membrane (BM) protein composition by suppressing the uPA-uPAR-plasmin pathway. Indeed, confluence triggers adherens junction maturation and enhances TGF-β and activin A activity, resulting in increased deposition of PAI-1 and perlecan in the BM. Extracellular matrix (ECM)-accumulated PAI-1 suppresses the uPA-uPAR-plasmin pathway and further enhances perlecan deposition by inhibiting its plasmin-dependent proteolysis. We show that perlecan deposition in the ECM strengthens cell adhesion, inhibits keratinocyte motility and promotes additional accumulation of PAI-1 in the ECM at confluence. In agreement, during wound-healing, perlecan concentrates at the wound-margin, where BM matures to stabilize keratinocyte adhesion. Our results demonstrate that confluence-dependent signaling orchestrates not only growth inhibition and differentiation, but also controls ECM proteolysis and BM formation. These data suggest that uncontrolled integration of confluence-dependent signaling, might favor skin disorders, including tumorigenesis, not only by promoting cell hyperproliferation, but also by altering protease activity and deposition of ECM components.
Collapse
|
22
|
Plasminogen is a key proinflammatory regulator that accelerates the healing of acute and diabetic wounds. Blood 2012; 119:5879-87. [PMID: 22563086 DOI: 10.1182/blood-2012-01-407825] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Despite decades of research on wound healing, effective biologic agents for the treatment of chronic wounds, especially diabetic wounds, are still lacking. In the present study, we report that the inert plasma protein plasminogen (plg) acts as a key regulatory molecule that potentiates wound healing in mice. Early in the healing process, plg bound to inflammatory cells is transported to the wound area, where the level of plg is increased locally, leading to the induction of cytokines and intracellular signaling events and to a potentiation of the early inflammatory response. Systemic administration of additional plg not only accelerates the healing of acute burn wounds in wild-type mice, but also improves the healing of chronic diabetic wounds in a mouse model of diabetes. Our results suggest that the administration of plg may be a novel therapeutic strategy to treat many different types of wounds, especially chronic wounds such as those caused by diabetes.
Collapse
|
23
|
|
24
|
Concomitant lack of MMP9 and uPA disturbs physiological tissue remodeling. Dev Biol 2011; 358:56-67. [PMID: 21802414 DOI: 10.1016/j.ydbio.2011.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 07/07/2011] [Accepted: 07/08/2011] [Indexed: 01/15/2023]
Abstract
Urokinase-type plasminogen activator (uPA) and matrix metalloproteinase-9 (MMP9, gelatinase B) have separately been recognized to play important roles in various tissue remodeling processes. In this study, we demonstrate that deficiency for MMP9 in combination with ablation of either uPA- or tissue-type plasminogen activator (tPA)-catalyzed plasminogen activation is critical to accomplish normal gestation in mice. Gestation was also affected by simultaneous lack of MMP9 and the uPA receptor (uPAR). Interestingly, uPA-deficiency additionally exacerbated the effect of MMP9-deficiency on bone growth and an additive effect caused by combined lack in MMP9 and uPA was observed during healing of cutaneous wounds. By comparison, MMP9-deficiency combined with absence of either tPA or uPAR resulted in no significant effect on wound healing, indicating that the role of uPA during wound healing is independent of uPAR, when MMP9 is absent. Notably, compensatory upregulation of uPA activity was seen in wounds from MMP9-deficient mice. Taken together, these studies reveal essential functional dependency between MMP9 and uPA during gestation and tissue repair.
Collapse
|
25
|
Aalinkeel R, Nair BB, Reynolds JL, Sykes DE, Mahajan SD, Chadha KC, Schwartz SA. Overexpression of MMP-9 contributes to invasiveness of prostate cancer cell line LNCaP. Immunol Invest 2011; 40:447-64. [PMID: 21391788 DOI: 10.3109/08820139.2011.557795] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Matrix metallaprotinase-9 (MMP-9) is zinc-containing proteinase whose expression and trafficking are frequently altered in cancer. MMP-9 in the plasma membrane and the secreted forms are thought to contribute to the invasive and metastatic properties of malignant tumors. We have manipulated the expression of MMP-9 in prostate tumor cell line LNCaP and measured their capacity to invade through a basement membrane matrix. Stable expression of human MMP-9 in a poorly metastatic LNCaP prostate cancer cell line produced a 2-3-fold increase in MMP-9 activity and a comparable increase in invasiveness. Transient transfection of LNCaP stable clone expressing MMP-9 with MMP-9 antisense oligonucleotide (ASODN) produced 55-90% less MMP-9 than control cells and were proportionately less invasive. In contrast, manipulating MMP-9 levels had no effect on cell migration across an uncoated membrane. A standard MMP-9 inhibitor at a concentration ranging from 1-10 nM, caused a nearly quantitative inhibition of extracellular MMP-9 activity and had significant effect on basement membrane invasion. Collectively, these results confirm the role of MMP-9 in tissue remodeling associated with prostate tumor invasion.
Collapse
Affiliation(s)
- Ravikumar Aalinkeel
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, State University of New York at Buffalo and Buffalo General Hospital, Kaleida Health, New York 14203, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Neutralisation of uPA with a monoclonal antibody reduces plasmin formation and delays skin wound healing in tPA-deficient mice. PLoS One 2010; 5:e12746. [PMID: 20856796 PMCID: PMC2939870 DOI: 10.1371/journal.pone.0012746] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Accepted: 08/10/2010] [Indexed: 11/21/2022] Open
Abstract
Background Proteolytic degradation by plasmin and metalloproteinases is essential for epidermal regeneration in skin wound healing. Plasminogen deficient mice have severely delayed wound closure as have mice simultaneously lacking the two plasminogen activators, urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA). In contrast, individual genetic deficiencies in either uPA or tPA lead to wound healing kinetics with no or only slightly delayed closure of skin wounds. Methodology/Principal Findings To evaluate the therapeutic potential in vivo of a murine neutralizing antibody directed against mouse uPA we investigated the efficacy in skin wound healing of tPA-deficient mice. Systemic administration of the anti-mouse uPA monoclonal antibody, mU1, to tPA-deficient mice caused a dose-dependent delay of skin wound closure almost similar to the delayed kinetics observed in uPA;tPA double-deficient mice. Analysis of wound extracts showed diminished levels of plasmin in the mU1-treated tPA-deficent mice. Immunohistochemistry revealed that fibrin accumulated in the wounds of such mU1-treated tPA-deficent mice and that keratinocyte tongues were aberrant. Together these abnormalities lead to compromised epidermal closure. Conclusions/Significance Our findings demonstrate that inhibition of uPA activity with a monoclonal antibody in adult tPA-deficient mice mimics the effect of simultaneous genetic ablation of uPA and tPA. Thus, application of the murine inhibitory mU1 antibody provides a new and highly versatile tool to interfere with uPA-activity in vivo in mouse models of disease.
Collapse
|
27
|
Abstract
SummaryOur ability to heal wounds deteriorates with age, leading in many cases to a complete lack of repair and development of a chronic wound. Moreover, as the elderly population continues to grow the prevalence of non-healing chronic wounds is escalating. Cutaneous wound repair occurs through a combination of overlapping phases, including an initial inflammatory response, a proliferative phase and a final remodelling phase. In elderly subjects the inflammatory response is delayed, macrophage and fibroblast function compromised, angiogenesis reduced and re-epithelialization inhibited. Whilst a large body of historic research describes the defective processes that lead to delayed healing, only recently have the molecular mechanisms by which these defects arise begun to be elucidated. Current therapies available for treatment of chronic wounds in elderly people are surprisingly limited and generally ineffective. Thus there is an urgent need to develop new therapeutic strategies based on these recent molecular and cellular insights.
Collapse
|
28
|
Abstract
The involvement and role of the plasminogen activator-plasmin system in normal and pathological wound healing is reviewed. The methods currently available for demonstrating plasmin activity are briefly described. The article also reviews some other serine proteases potentially involved in pathological wound healing processes. The current ophthalmological therapeutic measures used to regulate tissue proteolysis are also described.
Collapse
Affiliation(s)
- T Tervo
- Department of Ophthalmology, University of Helsinki, Finland
| | | | | | | |
Collapse
|
29
|
Lee SG, Koh HY, Lee HK, Yim JH. Possible Roles of Antarctic Krill Proteases for Skin Regeneration. ACTA ACUST UNITED AC 2008. [DOI: 10.4217/opr.2008.30.4.467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
30
|
Zakrzewska M, Marcinkowska E, Wiedlocha A. FGF-1: From Biology Through Engineering to Potential Medical Applications. Crit Rev Clin Lab Sci 2008; 45:91-135. [DOI: 10.1080/10408360701713120] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
31
|
VAALAMO M, WECKROTH M, PUOLAKKAINEN P, KERE J, SAARINEN P, LAUHARANTA J, SAARIALHO-KERE U. Patterns of matrix metalloproteinase and TIMP-1 expression in chronic and normally healing human cutaneous wounds. Br J Dermatol 2008. [DOI: 10.1046/j.1365-2133.1996.d01-932.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
32
|
BECHTEL MJ, SCHAEFER BM, KRAMER MD. Plasminogen activator inhibitor type-2 in the lesional epidermis of lupus erythematosus. Br J Dermatol 2008. [DOI: 10.1046/j.1365-2133.1996.24759.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Profibrinolytic effects of metalloproteinases during skin wound healing in the absence of plasminogen. J Invest Dermatol 2008; 128:2092-101. [PMID: 18337830 DOI: 10.1038/jid.2008.54] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Genetic ablation of plasminogen (Plg) and pharmacological inhibition of metalloproteinase activity by galardin delay skin wound healing in mice, whereas the combined inhibition of these two enzyme systems completely prevents healing. In this study, the impact of plasmin and metalloproteinases as profibrinolytic enzymes has been investigated by comparing skin wound healing in the absence and presence of fibrin. Plg deficiency impairs skin wound healing kinetics, but this delay is only partially restored in the absence of fibrin. This suggests that plasmin-mediated fibrinolysis is the primary, but not the exclusive, requirement for healing of wounds in these mice. In addition, we observe that lack of fibrin reduces Plg activation significantly during wound healing. The profibrinolytic role of metalloproteinases is revealed by the finding that lack of fibrin partially restores the otherwise arrested healing of Plg-deficient wounds after metalloproteinase inhibition. In conclusion, the residual impairment of skin wound healing in the absence of fibrin suggests the existence of a fibrin-independent substrate(s) for plasmin and metalloproteinases. Furthermore, these in vivo data reveal that galardin-sensitive metalloproteinases mediate compensatory fibrinolysis to facilitate wound healing in the absence of plasmin.
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Acute coagulopathy of trauma has only been described relatively recently. Developing early in the postinjury phase, it is associated with increased transfusion requirements and poor outcomes. This review examines the possible initiators, mechanism and clinical importance of acute coagulopathy. RECENT FINDINGS Acute coagulopathy of trauma occurs in patients with shock and is characterized by a systemic anticoagulation and hyperfibrinolysis. Dilution, acidemia and consumption of coagulation proteases do not appear to be significant factors at this stage. There is evidence to implicate activation of the protein C pathway in this process. Diagnosis of acute coagulopathy currently relies on laboratory assessment of clotting times. These tests do not fully characterize the coagulopathy and have significant limitations, which reduce their clinical utility. SUMMARY Acute coagulopathy results in increased transfusion requirements, incidence of organ dysfunction, critical care stay and mortality. Recognition of an early coagulopathic state has implications for the care of shocked patients and the management of massive transfusion. Identification of novel mechanisms for traumatic coagulopathy may lead to new avenues for drug discovery and therapeutic intervention.
Collapse
|
35
|
Lian X, Yang L, Gao Q, Yang T. IL-1α is a potent stimulator of keratinocyte tissue plasminogen activator expression and regulated by TGF-β1. Arch Dermatol Res 2008; 300:185-93. [DOI: 10.1007/s00403-007-0828-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Revised: 12/11/2007] [Accepted: 12/21/2007] [Indexed: 01/24/2023]
|
36
|
Jensen PJ, Baird J, Belin D, Vassalli JD, Busso N, Gubler P, Lazarus GS. Tissue plasminogen activator in psoriasis. J Invest Dermatol 2007; 95:13S-14S. [PMID: 16788619 DOI: 10.1111/1523-1747.ep12505666] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Elevated levels of the serine proteinase plasminogen activator are observed in psoriatic lesions. In contrast to normal epidermis, lesional psoriatic epidermis contains primarily tissue type plasminogen activator (tPA) activity and much lower levels of urokinase type plasminogen activator (uPA) activity. Tissue plasminogen activator is also detectable immunocytochemically in lesional psoriatic but not normal epidermis. Similarly, mRNA for tPA is observed in lesional epidermis only. These results suggest that lesional psoriatic epidermis synthesizes enhanced levels of tPA compared to normal. Additional data support the hypothesis that enhanced tPA may be another marker common to psoriatic epidermis, epidermis during wound repair, and keratinocytes in culture. The significance of elevated tPA in psoriatic lesions is presently unclear, but its possible relationship to epidermal proliferation and cutaneous inflammation is under study.
Collapse
Affiliation(s)
- P J Jensen
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Pawar SC, Demetriou MC, Nagle RB, Bowden GT, Cress AE. Integrin alpha6 cleavage: a novel modification to modulate cell migration. Exp Cell Res 2007; 313:1080-9. [PMID: 17303120 PMCID: PMC1905853 DOI: 10.1016/j.yexcr.2007.01.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2006] [Revised: 12/19/2006] [Accepted: 01/08/2007] [Indexed: 01/25/2023]
Abstract
Integrins play a major role in cell adhesion and migration. Previous work reported that a cleaved form of integrin alpha6 (alpha6p) was detected in invasive human prostate cancer tissue, absent in normal prostate tissue and was produced by urokinase-type Plasminogen Activator (uPA) in a plasmin-independent manner. Using site-directed mutagenesis we identified amino acid residues R594 and R595, located in the "stalk" region of integrin alpha6, as essential for cleavage. The cleavage site is located on the extracellular region of the protein between the beta-barrel domain and the thigh domain. Prostate cancer cells (PC3N) were stably transfected to overexpress the cleavable, wild-type (PC3N-alpha6-WT) or the non-cleavable form of integrin alpha6 (PC3N-alpha6-RR). The number of cells invading laminin 111- and laminin 332-coated filters by PC3N-alpha6-WT cells increased by threefold as compared to PC3N-alpha6-RR cells. Plasminogen activator inhibitor-1 (PAI-1) reduced the invasion of PC3N-alpha6-WT cells by approximately 42% through laminin 332-coated filters and plasmin inhibitor aprotinin had no significant effect. Linear cell migration increased production of integrin alpha6p in the PC3N-alpha6-WT cells and not in the PC3N-alpha6-RR cells and 32% of the PC3N-alpha6-WT cells migrated on laminin 111 in the linear migration assay as compared to the 5% PC3N-alpha6-RR cells. These data taken together suggest that the uPA-mediated cell surface cleavage of the alpha6 integrin extracellular domain is involved in tumor cell invasion and migration on laminin.
Collapse
Affiliation(s)
- Sangita C. Pawar
- Dept. of Cell Biology & Anatomy, University of Arizona, Tucson, AZ
| | | | | | - G. Tim Bowden
- Dept. of Cell Biology & Anatomy, University of Arizona, Tucson, AZ
| | - Anne E. Cress
- Dept. of Cell Biology & Anatomy, University of Arizona, Tucson, AZ. Arizona Cancer Center, University of Arizona, Tucson, Arizona 85724
| |
Collapse
|
38
|
Nakao H, Yamazaki M, Tsuboi R, Ogawa H. Mixture of sugar and povidone--iodine stimulates wound healing by activating keratinocytes and fibroblast functions. Arch Dermatol Res 2006; 298:175-82. [PMID: 16862430 DOI: 10.1007/s00403-006-0683-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Revised: 06/19/2006] [Accepted: 06/30/2006] [Indexed: 10/24/2022]
Abstract
The topical application of a mixture of sugar and povidone--iodine (PI) has been reported to accelerate the healing of cutaneous wounds and ulcers by promoting re-epithelialization and granulation tissue formation as well as having an anti-microbial effect. To clarify the mechanisms accounting for the efficacy of a 70% sugar and 3% PI paste (U-PASTAtrade mark) (SP), various keratinocytes and fibroblasts functions, including proliferation, collagen synthesis, integrin expression, and cytokine and proteinase secretions in the presence of SP were investigated. Cultured human keratinocytes and fibroblasts were treated with various concentrations of SP, SU and PI. The secretion of urokinase-type plasminogen activator (u-PA), transforming growth factor (TGF)-alpha and interleukin-1alpha from keratinocytes, was detected by ELISA. Collagen synthesis of fibroblasts was examined by means of detecting proline uptake. Furthermore, integrin expressions of these cells were analyzed using a flow cytometer. SP and PI increased intra-cellular u-PA of keratinocytes and stimulated the secretion of u-PA and TGF-alpha. Sugar accelerated the extra-cellular u-PA level only. Both SP and sugar increased the collagen synthesis of fibroblasts. SP and PI also remarkably induced the expressions of extra-cellular matrix receptor integrins, alpha1, alpha2, alpha3, alpha4, alpha5 and beta1, on the surface of keratinocytes and fibroblasts. SP, the mixture of sugar and PI, is likely to act on wounds not only as an antibiotic agent, but also as a modulator for keratinocytes and fibroblasts.
Collapse
Affiliation(s)
- Hiroshi Nakao
- Pharmacology Group, Fuji Research Laboratories, Pharmaceutical Division, Kowa Company Ltd, 332-1 Ohno-shinden, Fuji, Shizuoka, Japan
| | | | | | | |
Collapse
|
39
|
Kanno Y, Hirade K, Ishisaki A, Nakajima K, Suga H, Into T, Matsushita K, Okada K, Matsuo O, Matsuno H. Lack of alpha2-antiplasmin improves cutaneous wound healing via over-released vascular endothelial growth factor-induced angiogenesis in wound lesions. J Thromb Haemost 2006; 4:1602-10. [PMID: 16839360 DOI: 10.1111/j.1538-7836.2006.01978.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND The fibrinolytic system is supposed to play an important role in the degradation of extracellular matrices for physiological and pathological tissue remodeling; however, the detailed mechanism regarding how this system affects cutaneous wound healing remains to be clarified. METHODS AND RESULTS We performed experimental cutaneous wounding in mice with a deficiency of alpha(2)-antiplasmin (alpha(2)AP), which is a potent and specific plasmin inhibitor. We found that an accelerated wound closure was observed in alpha(2)AP-deficient (alpha(2)AP-/-) mice in comparison with wild type (WT) mice. Moreover, we observed that a greater increase of angiogenesis occurred in the process of wound healing in alpha(2)AP-/- mice than in the WT mice. Intriguingly, mRNA expression of vascular endothelial growth factor (VEGF), which is the best characterized positive regulator of angiogenesis, in wound lesions was found to show a greater increase in the early phase of the healing process in alpha(2)AP-/- mice than in WT mice. In addition, the amount of released-VEGF from the explanted fibroblasts of alpha(2)AP-/- mice increased dramatically more than in the WT mice. Finally, the intra-jugular administration of anti-VEGF antibody clearly suppressed the increased angiogenesis and accelerated wound closure in the wound lesion of alpha(2)AP-/- mice. CONCLUSION The lack of alpha(2)AP markedly causes an over-release of VEGF from the fibroblasts in cutaneous wound lesions, thereby inducing angiogenesis around the area, and thus resulting in an accelerated-wound closure. CONCLUSIONS This is the first report to describe the crucial role that alpha(2)AP plays following angiogenesis in the process of wound healing. Our results provide new insight into the role of alpha(2)AP on cutaneous wound healing.
Collapse
Affiliation(s)
- Y Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, DWCLA, Kyoto, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Matsuzaki K, Inoue H, Kumagai N. Re-epithelialisation and the possible involvement of the transcription factor, basonuclin. Int Wound J 2006; 1:135-40. [PMID: 16722885 PMCID: PMC7951248 DOI: 10.1111/j.1742-4801.2004.00033.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
This article briefly summarises the basic mechanism of re-epithelialisation and discusses the possible role of the cell-type-specific transcription factor, basonuclin. Re-epithelialisation is initiated by a signal resulting from the absence of neighbouring cells at the wound edge. Basal cells at the wound edge become flattened and lose their intercellular desmosomes and substratum attachment. The amount of cytoplasmic actinomyosin filaments that insert into the new adhesion complexes is increased, and contraction of those filaments produces cell movement. The epithelial cells at the wound edge migrate on a provisional matrix using the newly expressed integrin receptors. Once re-epithelialisation is complete, the epithelial cells revert to the normal phenotype of basal epidermal cells, firmly attach to the newly developed basement membrane zone through hemidesmosomes and resume standard differentiation. Protein synthesis increases in the epidermal cells at the wound edge during re-epithelialisation. Active protein synthesis requires accelerated transcription of ribosomal RNA genes. The transcription factor basonuclin binds to the ribosomal RNA gene promoter and increases the transcription of the genes. Therefore, it is speculated that basonuclin in epithelial cells is required in the process of re-epithelialisation.
Collapse
Affiliation(s)
- Kyoichi Matsuzaki
- Department of Plastic and Reconstructive Surgery, St Marianna University School of Medicine, Miyamae-Ku, Kawasaki, Japan.
| | | | | |
Collapse
|
41
|
Kirfel G, Rigort A, Borm B, Herzog V. Cell migration: mechanisms of rear detachment and the formation of migration tracks. Eur J Cell Biol 2005; 83:717-24. [PMID: 15679116 DOI: 10.1078/0171-9335-00421] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cell migration is central to many biological and pathological processes, including embryogenesis, tissue repair and regeneration as well as cancer and the inflammatory response. In general, cell migration can be usefully conceptualized as a cyclic process. The initial response of a cell to a migration-promoting agent is to polarize and extend protrusions in the direction of migration. These protrusions can be large, broad lamellipodia or spike-like filopodia, are usually driven by actin polymerization, and are stabilized by adhering to the extracellular matrix (ECM) via transmembrane receptors of the integrin family linked to the actin cytoskeleton. These adhesions serve as traction sites for migration as the cell moves forward over them, and they must be disassembled at the cell rear, allowing it to detach. The mechanisms of rear detachment and the regulatory processes involved are not well understood. The disassembly of adhesions that is required for detachment depends on a coordinated interaction of actin and actin-binding proteins, signaling molecules and effector enzymes including proteases, kinases and phosphatases. Originally, the biochemically regulated processes leading to rear detachment of migrating cells were thought not to be necessarily accompanied by any loss of cell material. However, it has been shown that during rear detachment long tubular extensions, the retracting fibers, are formed and that "membrane ripping" occurs at the cell rear. By this process, a major fraction of integrin-containing cellular material is left behind forming characteristic migration tracks that exactly mark the way a cell has taken.
Collapse
Affiliation(s)
- Gregor Kirfel
- Institute of Cell Biology, University of Bonn, Bonn, Germany.
| | | | | | | |
Collapse
|
42
|
Weckroth M, Vaheri A, Virolainen S, Saarialho-Kere U, Jahkola T, Sirén V. Epithelial tissue-type plasminogen activator expression, unlike that of urokinase, its receptor, and plasminogen activator inhibitor-1, is increased in chronic venous ulcers. Br J Dermatol 2005; 151:1189-96. [PMID: 15606514 DOI: 10.1111/j.1365-2133.2004.06261.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The plasminogen activation system represents a potent mechanism of extracellular proteolysis and is an essential component of normal wound healing. It has also been implicated in the pathogenesis of chronic, nonhealing ulcers. Traditionally, urokinase-type plasminogen activator (uPA) has been associated with pericellular proteolytic activity involved in tissue remodelling processes, and tissue-type plasminogen activator (tPA) mainly with intravascular fibrinolysis. OBJECTIVES The present study was conducted to characterize the spatial distribution of the various plasminogen activation system components in chronic ulcers and acute, well-granulating wounds. METHODS The expression of uPA, tPA, urokinase receptor (uPAR), plasminogen activator inhibitor-1 (PAI-1), and vitronectin was investigated by immunohistochemical staining, in addition to uPA, tPA and PAI-1 expression by in-situ hybridization, in samples from eight chronic venous ulcers, five decubitus ulcers, five well-granulating acute wounds and five normal skin samples. RESULTS In chronic venous leg ulcers tPA mRNA was detected in basal and suprabasal keratinocytes at the leading wound edge, while in well-granulating wounds and in decubitus ulcers tPA mRNA was expressed only in a few keratinocytes. However, tPA was widely expressed in fibroblast- and macrophage-like cells in the stroma of well-granulating wounds, while less tPA was detected in the granulation tissue of chronic ulcers. tPA mRNA and protein were localized in the superficial granular layers in normal skin. Although no qualitative differences in expression of uPA, PAI-1 or uPAR in the wound edge keratinocytes in chronic ulcers vs. normally granulating wounds were found, their expressions were more pronounced in the granulation tissue of well-granulating wounds. CONCLUSIONS These results suggest that in poorly healing venous leg ulcers, the pattern of tPA expression is altered in keratinocytes at the leading edge of the wound, and the patterns of tPA, uPA and PAI-1 expression are altered in the granulation tissue.
Collapse
Affiliation(s)
- M Weckroth
- Department of Virology, Haartman Institute, P.O. 21, FI-00014, University of Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
43
|
Massaro-Giordano M, Marshall CM, Lavker RM, Jensen PJ, Risse Marsh BC. Plasminogen activator inhibitor type 2 (PAI-2) is present in normal human conjunctiva. J Cell Physiol 2005; 205:295-301. [PMID: 15887231 DOI: 10.1002/jcp.20398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The purpose was to characterize plasminogen activator inhibitor type 2 (PAI-2) expression in normal human conjunctiva in vivo and in vitro. PAI-2 antigen was assayed by immunostaining and immunoblotting of extracts from normal human conjunctival epithelial lysates and conditioned media (CM) of cultured human conjunctival keratinocytes. Immunostaining of normal human conjunctival epithelia revealed that PAI-2 was found consistently in the superficial keratinocytes and, in some biopsies, also in the lower keratinocyte layers. In all cases, PAI-2 was concentrated around the cell periphery. In extracts of conjunctival epithelia and cultured conjunctival keratinocytes, PAI-2 had an apparent molecular weight of 45 kDa, consistent with the non-glycosylated form. The majority of PAI-2, approximately 90%, was cell associated, however, a small percentage of PAI-2 was released into the CM in a linear manner with time. PAI-2 in the conditioned medium had a higher molecular weight, consistent with a glycosylated form. Conjunctival PAI-2 was active, as shown by its ability to complex with a target enzyme, urokinase plasminogen activator (uPA). Although PAI-2 was detectable both in monolayer (i.e., relatively undifferentiated) conjunctival keratinocyte cultures as well as in stratified (i.e., more differentiated) cultures, steady state levels of PAI-2 were greater in the latter. PAI-2 is constitutively expressed by normal human conjunctival epithelial cells. The expression of PAI-2 throughout all epithelial layers in some biopsies of conjunctiva in vivo contrasts with the previously established distribution of PAI-2 in corneal epithelia, where it is present exclusively in the most superficial (i.e. most highly differentiated) cells. The role of PAI-2 in either tissue is unclear. However, we speculate that its distinct distribution in conjunctival versus corneal epithelia underscores inherent differences between these tissues, and may reflect specific functions of this proteinase inhibitor in both conjunctival and corneal epithelial cells.
Collapse
Affiliation(s)
- M Massaro-Giordano
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | | | | | |
Collapse
|
44
|
Abstract
Cutaneous wounds inevitably heal with scars, which can be disfiguring and compromise function. In general, the greater the insult, the worse the scarring, although genetic make up, regional variations and age can influence the final result. Excessive scarring manifests as hypertrophic and keloid scars. At the other end of the spectrum are poorly healing chronic wounds, such as foot ulcers in diabetic patients and pressure sores. Current therapies to minimize scarring and accelerate wound healing rely on the optimization of systemic conditions, early wound coverage and closure of lacerations, and surgical incisions with minimal trauma to the surrounding skin. The possible benefits of topical therapies have also been assessed. Further major improvements in wound healing and scarring require an understanding of the molecular basis of this process. Promising strategies for modulating healing include the local administration of platelet derived growth factor (PDGF)-BB to accelerate the healing of chronic ulcers, and increasing the relative ratio of transforming growth factor (TGF)beta-3 to TGFbeta-1 and TGFbeta-2 in order to minimize scarring.
Collapse
|
45
|
Brudzynski K, Carlone R. Stage-dependent modulation of limb regeneration by caffeic acid phenethyl ester (CAPE)--immunocytochemical evidence of a CAPE-evoked delay in mesenchyme formation and limb regeneration. ACTA ACUST UNITED AC 2004; 301:389-400. [PMID: 15114646 DOI: 10.1002/jez.a.20064] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Caffeic acid phenethyl ester (CAPE), a natural compound of bee propolis, selectively inhibits proliferation of transformed cells in several cancer models in vitro. To examine in vivo CAPE function, we used the newt regeneration blastema as a model system wherein the processes of de-differentiation and subsequent proliferation of undifferentiated cells mimic changes associated with oncogenic transformation and tumorigenesis. We have shown that a single dose of CAPE significantly increased cell proliferation at the stages of blastema growth and re-differentiation. At the de-differentiation stage, CAPE significantly stimulated proliferation of wound epidermis keratinocytes, but decreased proliferation in the blastema mesenchyme. Immunohistochemistry with a mesenchymal cell marker, vimentin, revealed a highly significant reduction of vimentin staining in the mesenchyme of CAPE-treated regenerates (p<0.001). These results, together with morphological observations indicate that, at the de-differentiation stage, CAPE stimulated wound re-epithelization, increased keratinocyte proliferation and increased thickness of the wound epidermis. However, CAPE inhibited mesenchyme formation and proliferation. The functional consequence of the CAPE inhibitory action was a delay in limb regeneration.
Collapse
|
46
|
Takamiya M, Saigusa K, Kumagai R, Nakayashiki N, Aoki Y. Studies on mRNA expression of tissue-type plasminogen activator in bruises for wound age estimation. Int J Legal Med 2004; 119:16-21. [PMID: 15205965 DOI: 10.1007/s00414-004-0453-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2003] [Accepted: 04/14/2004] [Indexed: 12/18/2022]
Abstract
We investigated mRNA expression of tissue-type plasminogen activator (tPA) and inflammatory cell dynamics for wound age estimation of bruises in mice. Neutrophils were detected from 1 h post-injury. Up to 8 h, they accumulated in subcutaneous tissue and the lower part of the dermis, and thereafter they extended to all the layers. Macrophages became detectable 3 h post-injury, and moderate infiltration of lymphocytes was seen from 144 h. In addition, epidermal thickening was also seen from 72 h. tPA mRNA expression peaked at 1 h, and increased slightly at 72 h post-injury. tPA mRNA was detected in epidermal cells, fibroblasts, and endothelial cells before and after injury, from 3 h in neutrophils and from 72 h in macrophages, respectively. This study presents the time-dependent expression of tPA mRNA in bruises in relation to temporal histologic characteristics during wound healing, which was considered to be useful for wound age estimation. Furthermore, it is suggested that tPA plays an important role in the first step of tissue remodeling.
Collapse
Affiliation(s)
- Masataka Takamiya
- Department of Legal Medicine, Iwate Medical University School of Medicine, 19-1 Uchimaru, 020-8505 Morioka, Japan.
| | | | | | | | | |
Collapse
|
47
|
Abstract
OBJECTIVE Many aspects of the care and underlying pathologies in patients suffering critical illness can detrimentally influence the normal healing processes of skin and soft tissues. Although a great diversity of pathologies exists, some aspects of the diseases and their treatments are common in critically ill patients. We aimed to identify some features, both common and specific, that could influence wound healing and the mechanisms by which they may do so. DESIGN In this review, we first outline the biology of normal skin and muscle healing and then explore how critical illness may influence the normal healing cascade. FINDINGS The healing of skin and skeletal muscle in critical illness is influenced by both underlying disease processes and the intensive care environment. Local and systemic factors can contribute to impaired healing, with the potential to prolong functional disability and increase the likelihood of wound complications. The frequency and number of soft tissue injuries derived from accidental injury, surgical intervention, and the need for invasive monitoring and therapies in the intensive care unit setting are likely to compromise the innate immunity and potentially further jeopardize the patient's ability to heal. Alterations in coagulation, tissue perfusion, inflammation, immune functioning, metabolism, nutrition, and drug therapies will influence healing responses by modifying the biological responses to tissue disruption. Locally, wound contamination, sepsis, tissue hypoxia, edema, and excessive or prolonged local pressure all have the potential to compromise soft tissue healing. One or more of these factors may be present at any time. CONCLUSION The skin and soft tissues are vulnerable to both injury and compromised healing when a patient is critically ill and exposed to a critical care environment. The identification of risk factors may aid in forming and modifying treatment strategies when caring for the critically ill patient with soft tissue injuries.
Collapse
Affiliation(s)
- Dean Thomas Williams
- Wound Healing Research Unit, University of Wales College of Medicine, Cardiff, UK
| | | |
Collapse
|
48
|
Li WY, Chong SSN, Huang EY, Tuan TL. Plasminogen activator/plasmin system: a major player in wound healing? Wound Repair Regen 2003; 11:239-47. [PMID: 12846910 DOI: 10.1046/j.1524-475x.2003.11402.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The role of the plasminogen activator/plasmin system in fibrinolysis has been well established. Indeed, clinicians worldwide have successfully utilized recombinant tissue-type plasminogen activator as first-line treatment of acute myocardial infarction for almost 2 decades. Outside the field of cardiology, there has been increasing excitement regarding the possible contribution of this system in many other important biological processes, including cell adhesion, cell migration, cell-cell signaling, tumor invasion and metastasis, ovulation, and wound healing. In this review, we present evidence in the current literature that the plasminogen activator/plasmin system does have a role in wound healing, looking at both normal and abnormal healing. Furthermore, the invaluable insights provided by numerous transgenic animal experiments are summarized.
Collapse
Affiliation(s)
- Wai-Yee Li
- Department of Surgery, Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California 90027, USA
| | | | | | | |
Collapse
|
49
|
Witowsky J, Abell A, Johnson NL, Johnson GL, Cuevas BD. MEKK1 is required for inducible urokinase-type plasminogen activator expression. J Biol Chem 2003; 278:5941-6. [PMID: 12493778 DOI: 10.1074/jbc.m212363200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Urokinase-type plasminogen activator (uPA) regulates the remodeling of extracellular matrix and controls reparative processes such as wound healing and liver regeneration. Here we show inducible uPA expression is controlled by MEKK1, a MAPK kinase kinase that regulates the ERK1/2 and JNK pathways. MEKK1 is activated in response to growth factors and cytoskeletal changes. We have found MEKK1 to be necessary for uPA up-regulation in response to treatment with phorbol 12-myristate 13-acetate or basic fibroblast growth factor. We demonstrate that growth factor-treated MEKK1-deficient fibroblasts display greatly reduced uPA expression and activity compared with control fibroblasts. Further, we show that growth factor-induced uPA expression requires MEKK1-dependent MKK1 and JNK activity and that transfection of MEKK1 into knockout cells restores inducible uPA expression and activity. Importantly, disrupted expression of MEKK2, a related MAPK kinase kinase, had no effect on uPA activity. Therefore, we conclude that MEKK1 expression is required for PMA- or FGF-2-induced signals to control uPA expression and function.
Collapse
Affiliation(s)
- James Witowsky
- Department of Pharmacology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | | | |
Collapse
|
50
|
Daniel RJ, Groves RW. Increased migration of murine keratinocytes under hypoxia is mediated by induction of urokinase plasminogen activator. J Invest Dermatol 2002; 119:1304-9. [PMID: 12485432 DOI: 10.1046/j.1523-1747.2002.19533.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
One of the key consequences of cutaneous wounding is the development of tissue hypoxia. Recent data have suggested that this is a potent stimulus for increased keratinocyte migration and hence re-epithelialization, although the mechanisms responsible for this remain unclear. In this study we have investigated the relationship between hypoxia, plasminogen activation, and in vitro wound healing. Exposure of keratinocyte cultures to hypoxia resulted in upregulation of urokinase plasminogen activator mRNA and a subsequent increase in urokinase plasminogen activator-mediated plasminogen activation, as determined by indirect chromogenic peptide assay and plasminogen-linked zymography. Analysis of keratinocyte wound healing in vitro confirmed enhanced wound closure in hypoxic cultures compared with normoxic cultures after 16 h. Pretreatment of normoxic and hypoxic cultures with mitomycin C and cytochalasin B indicated that in this system wound closure was due to keratinocyte migration rather than proliferation. Addition of the broad-spectrum serine proteinase inhibitor, p-aminobenzamidine, or the specific urokinase plasminogen activator inhibitors, amiloride and WX-293, significantly reduced wound closure in hypoxic cultures and abrogated the hypoxic enhancement of wound closure. These data indicate a central role for urokinase plasminogen activators in hypoxic keratinocyte migration and suggest a potential mechanism for enhanced re-epithelialization of wounds under low oxygen tensions.
Collapse
Affiliation(s)
- Richard J Daniel
- Center for Dermatology, Department of Medicine, University College London, UK
| | | |
Collapse
|