1
|
Fanaee S, Austin W, Filiaggi M, Adibnia V. External Bleeding and Advanced Biomacromolecules for Hemostasis. Biomacromolecules 2024. [PMID: 39463174 DOI: 10.1021/acs.biomac.4c00952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Hemorrhage is a significant medical problem that has been an active area of research over the past few decades. The human body has a complex response to bleeding that leads to blood clot formation and hemostasis. Many biomaterials based on various biomacromolecules have been developed to either accelerate or improve the body's natural response to bleeding. This review examines the mechanisms of hemostasis, types of bleeding, and the in vitro or in vivo models and techniques used to study bleeding and hemostatic materials. It provides a detailed overview of the diverse hemostatic materials, including those that are highly absorbent, wet adhesives, and those that accelerate the biochemical cascade of blood clotting. These materials are currently marketed, under preclinical testing, or being researched. In exploring the latest advancements in hemostatic technologies, this paper highlights the potential of these materials to significantly improve bleeding control in clinical and emergency situations.
Collapse
Affiliation(s)
- Sajjad Fanaee
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - William Austin
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Mark Filiaggi
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Vahid Adibnia
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Chemistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
2
|
Liu J, Solanki A, White MJV, Hubbell JA, Briquez PS. Therapeutic use of α2-antiplasmin as an antifibrinolytic and hemostatic agent in surgery and regenerative medicine. NPJ Regen Med 2022; 7:34. [PMID: 35773290 PMCID: PMC9246914 DOI: 10.1038/s41536-022-00230-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 06/06/2022] [Indexed: 11/14/2022] Open
Abstract
The biomaterial fibrin is widely used as a clinical tissue sealant in surgery. In preclinical research, fibrin is also extensively studied as a carrier material for growth factor delivery. In these applications, premature fibrin degradation leads to recurrent bleeding, tissue dehiscence and limited regenerative efficacy. Therefore, fibrinolysis inhibitors have been added to clinical fibrin formulations, for example the bovine-derived serine protease inhibitor aprotinin. Aprotinin is additionally used as a hemostatic agent to prevent excessive bleeding during surgery, in this case protecting endogenous fibrin clots. Nevertheless, aprotinin use has been associated with serious safety issues. Here, we explore the use the human physiological fibrinolysis inhibitor α2-antiplasmin (α2PI) as a substitute for aprotinin. We evaluate the efficacy of α2PI in the three main applications of aprotinin. We first showed that recombinant α2PI can successfully prolong the durability of fibrin biomaterials as compared to aprotinin in a model of subcutaneous implantation in mice mimicking application as a tissue sealant. We then used α2PI to enhance the delivery of engineered vascular endothelial growth factor (VEGF)-A and platelet-derived growth factor (PDGF)-BB in fibrin in promoting diabetic wound healing, which lead to improved wound closure, granulation tissue formation and angiogenesis. Lastly, we demonstrated that α2PI can be as effective as aprotinin as an intravenous hemostatic agent to prevent blood loss, using a tail-vein bleeding model in mice. Therefore, we believe that engineering fibrin biomaterials or endogenous fibrin with α2PI can have a strong impact in surgery and regenerative medicine by providing a competitive substitute to aprotinin that is of human origin.
Collapse
Affiliation(s)
- Jialu Liu
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, USA
| | - Michael J V White
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA.
| | - Priscilla S Briquez
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Abstract
Intraoperative bleeding and postoperative bleeding are major surgical complications. Tissue sealants, hemostats, and adhesives provide the armamentarium for establishing hemostatic balance, including the tissue sealant fibrin. Fibrin sealants combine advantages including instantaneous effect, biocompatibility, and biodegradability. However, several challenges remain. This review summarizes current fibrin product generations and highlights new trends and potential strategies for future improvement.
Collapse
Affiliation(s)
- Matthias Beudert
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-University Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Marcus Gutmann
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-University Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-University Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-University Würzburg, Am Hubland, 97074 Würzburg, Germany.,Helmholtz Institute for RNA-based Infection Research, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
| |
Collapse
|
4
|
Bodega F, Sironi C, Zocchi L, Porta C. Optimization of Fibrin Scaffolds to Study Friction in Cultured Mesothelial Cells. Int J Mol Sci 2022; 23:4980. [PMID: 35563371 PMCID: PMC9104594 DOI: 10.3390/ijms23094980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022] Open
Abstract
To study the friction of cell monolayers avoiding damage due to stress concentration, cells can be cultured on fibrin gels, which have a structure and viscoelasticity similar to that of the extracellular matrix. In the present research, we studied different gel compositions and surface coatings in order to identify the best conditions to measure friction in vitro. We examined the adhesion and growth behavior of mesothelial cell line MET-5A on fibrin gels with different fibrinogen concentrations (15, 20, and 25 mg/mL) and with different adhesion coatings (5 μg/mL fibronectin, 10 μg/mL fibronectin, or 10 μg/mL fibronectin + 10 μg/mL collagen). We also investigated whether different substrates influenced the coefficient of friction and the ability of cells to stick to the gel during sliding. Finally, we studied the degradation rates of gels with and without cells. All substrates tested provided a suitable environment for the adherence and proliferation of mesothelial cells, and friction measurements did not cause significant cell damage or detachment. However, in gels with a lower fibrinogen concentration, cell viability was higher and cell detachment after friction measurement was lower. Fibrinolysis was negligible in all the substrates tested.
Collapse
Affiliation(s)
- Francesca Bodega
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Sezione di Fisiologia Umana, Università degli Studi di Milano, 20100 Milan, Italy; (C.S.); (L.Z.); (C.P.)
| | | | | | | |
Collapse
|
5
|
Zhou ZF, Zhai W, Yu LN, Sun K, Sun LH, Xing XF, Yan M. Comparison of the in-vivo effect of two tranexamic acid doses on fibrinolysis parameters in adults undergoing valvular cardiac surgery with cardiopulmonary bypass - a pilot investigation. BMC Anesthesiol 2021; 21:33. [PMID: 33530942 PMCID: PMC7852217 DOI: 10.1186/s12871-021-01234-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/04/2021] [Indexed: 11/24/2022] Open
Abstract
Background The blood saving efficacy of TXA in cardiac surgery has been proved in several studies, but TXA dosing regimens were varied in those studies. Therefore, we performed this study to investigate if there is a dose dependent in-vivo effect of TXA on fibrinolysis parameters by measurement of fibrinolysis markers in adults undergoing cardiac surgery with CPB. Methods A double-blind, randomized, controlled prospective trial was conducted from February 11, 2017 to May 05, 2017. Thirty patients undergoing cardiac valve surgery were identified and randomly divided into a placebo group, low-dose group and high-dose group by 1: 1: 1. Fibrinolysis parameters were measured by plasma levels of D-Dimers, plasminogen activator inhibitor-1 (PAI-1), thrombin activatable fibrinolysis inhibitor (TAFI), plasmin-antiplasmin complex (PAP), tissue plasminogen activator (tPA) and thrombomodulin (TM). Those proteins were measured at five different sample times: preoperatively before the TXA injection (T1), 5 min after the TXA bolus (T2), 5 min after the initiation of CPB (T3), 5 min before the end of CPB (T4) and 5 min after the protamine administration (T5). A Thrombelastography (TEG) and standard coagulation test were also performed. Results Compared with the control group, the level of the D-Dimers decreased in the low-dose and high-dose groups when the patients arrived at the ICU and on the first postoperative morning. Over time, the concentrations of PAI-1, TAFI, and TM, but not PAP and tPA, showed significant differences between the three groups (P < 0.05). Compared with the placebo group, the plasma concentrations of PAI-1 and TAFI decreased significantly at the T3 and T4 (P < 0.05); TAFI concentrations also decreased at the T5 in low-dose group (P < 0.05). Compared with the low-dose group, the concentration of TM increased significantly at the T4 in high-dose group. Conclusions The in-vivo effect of low dose TXA is equivalent to high dose TXA on fibrinolysis parameters in adults with a low bleeding risk undergoing valvular cardiac surgery with cardiopulmonary bypass, and a low dose TXA regimen might be equivalent to high dose TXA for those patients. Trial registration ChiCTR-IPR-17010303, Principal investigator: Zhen-feng ZHOU, Date of registration: January 1, 2017. Supplementary Information The online version contains supplementary material available at 10.1186/s12871-021-01234-8.
Collapse
Affiliation(s)
- Zhen-Feng Zhou
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, China.,Department of Anesthesiology, Hangzhou Women's Hospital (The Affiliated Women's Hospital of Hangzhou Normal University), Hangzhou, China
| | - Wen Zhai
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, China.,Department of Anesthesiology, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medicine College), Hangzhou, China
| | - Li-Na Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, China
| | - Kai Sun
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, China
| | - Li-Hong Sun
- Department of Anesthesiology, Women's Hospital, School of Medicine Zhejiang University, Hangzhou, China
| | - Xiu-Fang Xing
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, China
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Computational Model for Hyperfibrinolytic Onset of Acute Traumatic Coagulopathy. Ann Biomed Eng 2018; 46:1173-1182. [PMID: 29675813 DOI: 10.1007/s10439-018-2031-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/16/2018] [Indexed: 12/16/2022]
Abstract
The onset of acute traumatic coagulopathy in trauma patients exacerbates hemorrhaging and dramatically increases mortality. The disease is characterized by increased localized bleeding, and the mechanism for its onset is not yet known. We propose that the fibrinolytic response, specifically the release of tissue-plasminogen activator (t-PA), within vessels of different sizes leads to a variable susceptibility to local coagulopathy through hyperfibrinolysis which can explain many of the clinical observations in the early stages from severely injured coagulopathic patients. We use a partial differential equation model to examine the consequences of vessel geometry and extent of injury on fibrinolysis profiles. In addition, we simulate the efficacy of tranexamic acid treatment on coagulopathy initiated through endothelial t-PA release, and are able to reproduce the time-sensitive nature of the efficacy of this treatment as observed in clinical studies.
Collapse
|
7
|
The role of fibrinolysis inhibition in engineered vascular networks derived from endothelial cells and adipose-derived stem cells. Stem Cell Res Ther 2018; 9:35. [PMID: 29433579 PMCID: PMC5809876 DOI: 10.1186/s13287-017-0764-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 11/22/2017] [Accepted: 12/26/2017] [Indexed: 01/05/2023] Open
Abstract
Background Co-cultures of endothelial cells with mesenchymal stem cells currently represent one of the most promising approaches in providing oxygen and nutrient supply for microvascular tissue engineering. Still, to translate this model into clinics several in vitro parameters including growth medium and scaffold degradation need to be fine-tuned. Methods We recently described the co-culture of adipose-derived stem cells with endothelial cells in fibrin, resulting in capillary formation in vitro as well as their perfusion in vivo. Here, we aimed to further characterise microvascular tube formation in fibrin by determining the role of scaffold degradation, thrombin concentration and culture conditions on vascularisation. Results We observed that inhibition of cell-mediated fibrin degradation by the commonly used inhibitor aprotinin resulted in impaired vascular network formation. Aprotinin had no effect on laminin and collagen type IV deposition or formation of tube-like structures in scaffold-free co-culture, indicating that poor vascularisation of fibrin clots is primarily caused by inhibition of plasminogen-driven fibrinolysis. Co-culture in plasminogen- and factor XIII-depleted fibrin did not result in different vascular network density compared to controls. Furthermore, we demonstrate that thrombin negatively affects vascular network density at high concentrations. However, only transient activation of incorporated endothelial cells by thrombin could be observed, thus excluding a long-term inflammatory response in tissue-engineered micro-capillaries. Finally, we show that vascularisation of fibrin scaffolds in basal medium is undermined because of increased fibrinolytic activity leading to scaffold destabilisation without aprotinin. Conclusions Taken together, our data reveal a critical role of fibrinolysis inhibition in in vitro cell-mediated vascularisation of fibrin scaffolds. Electronic supplementary material The online version of this article (10.1186/s13287-017-0764-2) contains supplementary material, which is available to authorized users.
Collapse
|
8
|
Briquez PS, Lorentz KM, Larsson HM, Frey P, Hubbell JA. Human Kunitz-type protease inhibitor engineered for enhanced matrix retention extends longevity of fibrin biomaterials. Biomaterials 2017; 135:1-9. [PMID: 28477492 DOI: 10.1016/j.biomaterials.2017.04.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/26/2017] [Accepted: 04/26/2017] [Indexed: 12/18/2022]
Abstract
Aprotinin is a broad-spectrum serine protease inhibitor used in the clinic as an anti-fibrinolytic agent in fibrin-based tissue sealants. However, upon re-exposure, some patients suffer from hypersensitivity immune reactions likely related to the bovine origin of aprotinin. Here, we aimed to develop a human-derived substitute to aprotinin. Based on sequence homology analyses, we identified the Kunitz-type protease inhibitor (KPI) domain of human amyloid-β A4 precursor protein as being a potential candidate. While KPI has a lower intrinsic anti-fibrinolytic activity than aprotinin, we reasoned that its efficacy is additionally limited by its fast release from fibrin material, just as aprotinin's is. Thus, we engineered KPI variants for controlled retention in fibrin biomaterials, using either covalent binding through incorporation of a substrate for the coagulation transglutaminase Factor XIIIa or through engineering of extracellular matrix protein super-affinity domains for sequestration into fibrin. We showed that both engineered KPI variants significantly slowed plasmin-mediated fibrinolysis in vitro, outperforming aprotinin. In vivo, our best engineered KPI variant (incorporating the transglutaminase substrate) extended fibrin matrix longevity by 50%, at a dose at which aprotinin did not show efficacy, thus qualifying it as a competitive substitute of aprotinin in fibrin sealants.
Collapse
Affiliation(s)
- Priscilla S Briquez
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Kristen M Lorentz
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Anokion, Inc., Cambridge MA 02139, United States
| | - Hans M Larsson
- Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
| | - Peter Frey
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Jeffrey A Hubbell
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, United States.
| |
Collapse
|
9
|
Williams B, McNeil J, Crabbe A, Tanaka KA. Practical Use of Thromboelastometry in the Management of Perioperative Coagulopathy and Bleeding. Transfus Med Rev 2017; 31:11-25. [DOI: 10.1016/j.tmrv.2016.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 08/23/2016] [Accepted: 08/23/2016] [Indexed: 12/23/2022]
|
10
|
Seyedhassantehrani N, Li Y, Yao L. Dynamic behaviors of astrocytes in chemically modified fibrin and collagen hydrogels. Integr Biol (Camb) 2016; 8:624-34. [PMID: 27079938 PMCID: PMC4868780 DOI: 10.1039/c6ib00003g] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Astrocytes play a critical role in supporting the normal physiological function of neurons in the central nervous system (CNS). Astrocyte transplantation can potentially promote axonal regeneration and functional recovery after spinal cord injury (SCI). Fibrin and collagen hydrogels provide growth-permissive substrates and serve as carriers for therapeutic cell transplantation into an injured spinal cord. However, the application of fibrin and collagen hydrogels may be limited due to their relatively rapid degradation rate in vivo. In this study, immature astrocytes isolated from neonatal rats were grown in fibrin hydrogels containing aprotinin and collagen hydrogels crosslinked with poly(ethylene glycol) ether tetrasuccinimidyl glutarate (4S-StarPEG), and the cell behavior in these hydrogels was studied. The cell viability of astrocytes in the hydrogels was tested using the LIVE/DEAD® assay and the AlamarBlue® assay, and this study showed that astrocytes maintained good viability in these hydrogels. The cell migration study showed that astrocytes migrated in the fibrin and collagen hydrogels, and the migration speed is similar in these hydrogels. The crosslinking of collagen hydrogels with 4S-StarPEG did not change the astrocyte migration speed. However, the addition of aprotinin in the fibrin hydrogel inhibited astrocyte migration. The expression of chondroitin sulfate proteoglycan (CSPG), including NG2, neurocan, and versican, by astrocytes grown in the hydrogels was analyzed by quantitative RT-PCR. The expression of NG2, neurocan, and versican by the cells in these hydrogels was not significantly different.
Collapse
Affiliation(s)
- Negar Seyedhassantehrani
- Department of Biological Sciences, Wichita State University, Fairmount 1845, Wichita, KS 67260, USA.
| | | | | |
Collapse
|
11
|
Samal J, Hoban DB, Naughton C, Concannon R, Dowd E, Pandit A. Fibrin-based microsphere reservoirs for delivery of neurotrophic factors to the brain. Nanomedicine (Lond) 2015; 10:765-83. [DOI: 10.2217/nnm.14.221] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: The in vivo therapeutic potential of neurotrophic factors to modify neuronal dysfunctions is limited by their short half-life. A biomaterials-based intervention, which protects these factors and allows a controlled release, is required. Materials & methods: Hollow fibrin microspheres were fabricated by charge manipulation using polystyrene templates and were loaded with NGF. Bioactivity of released NGF was demonstrated by neuronal outgrowth assay in PC-12 cells followed by in vivo assessment for NGF release and host response. Results: Fibrin-based hollow spheres showed high loading efficiency (>80%). Neurotrophin encapsulation into the microspheres did not alter its bioactivity and controlled release of NGF was observed in the in vivo study. Conclusion: Fibrin hollow microspheres act as a suitable delivery platform for neurotrophic factors with tunable loading efficiency and maintaining their bioactive form after release in vivo.
Collapse
Affiliation(s)
- Juhi Samal
- Network of Excellence for Functional Biomaterials
| | - Deirdre B Hoban
- Pharmacology & Therapeutics, National University of Ireland, Galway, Ireland
| | - Carol Naughton
- Pharmacology & Therapeutics, National University of Ireland, Galway, Ireland
| | - Ruth Concannon
- Pharmacology & Therapeutics, National University of Ireland, Galway, Ireland
| | - Eilis Dowd
- Network of Excellence for Functional Biomaterials
- Pharmacology & Therapeutics, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials
| |
Collapse
|
12
|
Extracellular proteolysis of reelin by tissue plasminogen activator following synaptic potentiation. Neuroscience 2014; 274:299-307. [PMID: 24892761 DOI: 10.1016/j.neuroscience.2014.05.046] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/21/2014] [Indexed: 01/19/2023]
Abstract
The secreted glycoprotein reelin plays an indispensable role in neuronal migration during development and in regulating adult synaptic functions. The upstream mechanisms responsible for initiating and regulating the duration and magnitude of reelin signaling are largely unknown. Here we report that reelin is cleaved between EGF-like repeats 6-7 (R6-7) by tissue plasminogen activator (tPA) under cell-free conditions. No changes were detected in the level of reelin and its fragments in the brains of tPA knockouts, implying that other unknown proteases are responsible for generating reelin fragments found constitutively in the adult brain. Induction of NMDAR-independent long-term potentiation with the potassium channel blocker tetraethylammonium chloride (TEA-Cl) led to a specific up-regulation of reelin processing at R6-7 in wild-type mice. In contrast, no changes in reelin expression and processing were observed in tPA knockouts following TEA-Cl treatment. These results demonstrate that synaptic potentiation results in tPA-dependent reelin processing and suggest that extracellular proteolysis of reelin may regulate reelin signaling in the adult brain.
Collapse
|
13
|
Esper SA, Subramaniam K, Tanaka KA. Pathophysiology of Cardiopulmonary Bypass. Semin Cardiothorac Vasc Anesth 2014; 18:161-76. [DOI: 10.1177/1089253214532375] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The techniques and equipment of cardiopulmonary bypass (CPB) have evolved over the past 60 years, and numerous numbers of cardiac surgical procedures are conducted around the world using CPB. Despite more widespread applications of percutaneous coronary and valvular interventions, the need for cardiac surgery using CPB remains the standard approach for certain cardiac pathologies because some patients are ineligible for percutaneous procedures, or such procedures are unsuccessful in some. The ageing patient population for cardiac surgery poses a number of clinical challenges, including anemia, decreased cardiopulmonary reserve, chronic antithrombotic therapy, neurocognitive dysfunction, and renal insufficiency. The use of CPB is associated with inductions of systemic inflammatory responses involving both cellular and humoral interactions. Inflammatory pathways are complex and redundant, and thus, the reactions can be profoundly amplified to produce a multiorgan dysfunction that can manifest as capillary leak syndrome, coagulopathy, respiratory failure, myocardial dysfunction, renal insufficiency, and neurocognitive decline. In this review, pathophysiological aspects of CPB are considered from a practical point of view, and preventive strategies for hemodilutional anemia, coagulopathy, inflammation, metabolic derangement, and neurocognitive and renal dysfunction are discussed.
Collapse
|
14
|
Cheng AC, Tsai IH. Functional characterization of a slow and tight-binding inhibitor of plasmin isolated from Russell's viper venom. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1840:153-9. [PMID: 23999090 DOI: 10.1016/j.bbagen.2013.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/22/2013] [Accepted: 08/26/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Snake venoms are rich in Kunitz-type protease inhibitors that may have therapeutic applications. However, apart from trypsin or chymotrypsin inhibition, the functions of most of these inhibitors have not been elucidated. A detailed functional characterization of these inhibitors may lead to valuable drug candidates. METHODS A Kunitz-type protease inhibitor, named DrKIn-II, was tested for its ability to inhibit plasmin using various approaches such as far western blotting, kinetic analyses, fibrin plate assay and euglobulin clot lysis assay. In addition, the antifibrinolytic activity of DrKIn-II was demonstrated in vivo. RESULTS DrKIn-II potently decreased the amidolytic activity of plasmin in a dose-dependent manner, with a global inhibition constant of 0.2nM. Inhibition kinetics demonstrated that the initial binding of DrKIn-II causes the enzyme to isomerize, leading to the formation of a much tighter enzyme-inhibitor complex. DrKIn-II also demonstrated antifibrinolytic activity in fibrin plate assay and significantly prolonged the lysis of the euglobulin clot. Screening of DrKIn-II against a panel of serine proteases indicated that plasmin is the preferential target of DrKIn-II. Furthermore, DrKIn-II treatment prevented the increase of FDP in coagulation-stimulated mice and significantly reduced the bleeding time in a murine tail bleeding model. CONCLUSION DrKIn-II is a potent, slow and tight-binding plasmin inhibitor that demonstrates antifibrinolytic activity both in vitro and in vivo. GENERAL SIGNIFICANCE This is the first in-depth functional characterization of a plasmin inhibitor from a viperid snake. The potent antifibrinolytic activity of DrKIn-II makes it a potential candidate for the development of novel antifibrinolytic agents.
Collapse
Affiliation(s)
- An-Chun Cheng
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan.
| | | |
Collapse
|
15
|
A Protein from Aloe vera that Inhibits the Cleavage of Human Fibrin(ogen) by Plasmin. Appl Biochem Biotechnol 2013; 170:2034-45. [DOI: 10.1007/s12010-013-0356-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/17/2013] [Indexed: 11/25/2022]
|
16
|
Cheng AC, Wu HL, Shi GY, Tsai IH. A novel heparin-dependent inhibitor of activated protein C that potentiates consumptive coagulopathy in Russell's viper envenomation. J Biol Chem 2012; 287:15739-48. [PMID: 22416129 PMCID: PMC3346151 DOI: 10.1074/jbc.m111.323063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/17/2012] [Indexed: 11/06/2022] Open
Abstract
The activation of coagulation factors V and X by Russell's viper venom (RVV) has been implicated in the development of consumptive coagulopathies in severely envenomed patients. However, factor Va is prone to inactivation by activated protein C (APC), an important serine protease that negatively regulates blood coagulation. It is therefore hypothesized that APC may be down-regulated by some of the venom components. In this study, we managed to isolate a potent Kunitz-type APC inhibitor, named DrKIn-I. Using chromogenic substrate, DrKIn-I dose-dependently inhibited the activity of APC. Heparin potentiated the inhibition and reduced the IC(50) of DrKIn-I by 25-fold. DrKIn-I, together with heparin, also protected factor Va from APC-mediated inactivation. Using surface plasmon resonance, DrKIn-I exhibited fast binding kinetics with APC (association rate constant = 1.7 × 10(7) M(-1) s(-1)). Direct binding assays and kinetic studies revealed that this inhibition (K(i) = 53 pM) is due to the tight binding interactions of DrKIn-I with both heparin and APC. DrKIn-I also effectively reversed the anticoagulant activity of APC and completely restored the thrombin generation in APC-containing plasma. Furthermore, although the injection of either DrKIn-I or RVV-X (the venom factor X-activator) into ICR mice did not significantly deplete the plasma fibrinogen concentration, co-administration of DrKIn-I with RVV-X resulted in complete fibrinogen consumption and the deposition of fibrin thrombi in the glomerular capillaries. Our results provide new insights into the pathogenesis of RVV-induced coagulopathies and indicate that DrKIn-I is a novel APC inhibitor that is associated with potentially fatal thrombotic complications in Russell's viper envenomation.
Collapse
Affiliation(s)
- An-Chun Cheng
- From the Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Hua-Lin Wu
- the Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- the Center for Bioscience and Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan, and
| | - Guey-Yueh Shi
- the Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- the Center for Bioscience and Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan, and
| | - Inn-Ho Tsai
- From the Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
- the Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
17
|
Sniecinski RM, Chandler WL. Activation of the Hemostatic System During Cardiopulmonary Bypass. Anesth Analg 2011; 113:1319-33. [DOI: 10.1213/ane.0b013e3182354b7e] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Continuous localized monitoring of plasmin activity identifies differential and regional effects of the serine protease inhibitor aprotinin: relevance to antifibrinolytic therapy. J Cardiovasc Pharmacol 2011; 57:400-6. [PMID: 21502925 DOI: 10.1097/fjc.0b013e31820b7df1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Antifibrinolytic therapy, such as the use of the serine protease inhibitor aprotinin, was a mainstay for hemostasis after cardiac surgery. However, aprotinin was empirically dosed, and although the pharmacological target was the inhibition of plasmin activity (PLact), this was never monitored, off-target effects occurred, and led to withdrawn from clinical use. The present study developed a validated fluorogenic microdialysis method to continuously measure PLact and tested the hypothesis that standardized clinical empirical aprotinin dosing would impart differential and regional effects on PLact. METHODS/RESULTS Pigs (30 kg) were instrumented with microdialysis probes to continuously measure PLact in myocardial, kidney, and skeletal muscle compartments (deltoid) and then randomized to high-dose aprotinin administration (2 mKIU load/0.5 mKIU/hr infusion; n = 7), low-dose aprotinin administration (1 mKIU load/0.250 mKIU/hr infusion; n = 6). PLact was compared with time-matched vehicle (n = 4), and PLact was also measured in plasma by an in vitro fluorogenic method. Aprotinin suppressed PLact in the myocardium and kidney at both high and low doses, indicative that both doses exceeded a minimal concentration necessary for PLact inhibition. However, differential effects of aprotinin on PLact were observed in the skeletal muscle, indicative of different compartmentalization of aprotinin. CONCLUSIONS Using a large animal model and a continuous method to monitor regional PLact, these unique results demonstrated that an empirical aprotinin dosing protocol causes maximal and rapid suppression in the myocardium and kidney and in turn would likely increase the probability of off-target effects and adverse events. Furthermore, this proof of principle study demonstrated that continuous monitoring of determinants of fibrinolysis might provide a novel approach for managing fibrinolytic therapy.
Collapse
|
19
|
Lorentz KM, Kontos S, Frey P, Hubbell JA. Engineered aprotinin for improved stability of fibrin biomaterials. Biomaterials 2010; 32:430-8. [PMID: 20864171 DOI: 10.1016/j.biomaterials.2010.08.109] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 08/30/2010] [Indexed: 01/06/2023]
Abstract
Fibrin has been long used clinically for hemostasis and sealing, yet extension of use in other applications has been limited due to its relatively rapid resorption in vivo, even with addition of aprotinin or other protease inhibitors. We report an engineered aprotinin variant that can be immobilized within fibrin and thus provide extended longevity. When recombinantly fused to a transglutaminase substrate domain from α(2)-plasmin inhibitor (α(2)PI(1-8)), the resulting variant, aprotinin-α(2)PI(1-8), was covalently crosslinked into fibrin matrices during normal thrombin/factor XIIIa-mediated polymerization. Challenge with physiological plasmin concentrations revealed that aprotinin-α(2)PI(1-8)-containing matrices retained 78% of their mass after 3 wk, whereas matrices containing wild type (WT) aprotinin degraded completely within 1 wk. Plasmin challenge of commercial sealants Omrixil and Tisseel, supplemented with aprotinin-α(2)PI(1-8) or WT aprotinin, showed extended longevity as well. When seeded with human dermal fibroblasts, aprotinin-α(2)PI(1-8)-supplemented matrices supported cell growth for at least 33% longer than those containing WT aprotinin. Subcutaneously implanted matrices containing aprotinin-α(2)PI(1-8) were detectable in mice for more than twice as long as those containing WT aprotinin. We conclude that our engineered recombinant aprotinin variant can confer extended longevity to fibrin matrices more effectively than WT aprotinin in vitro and in vivo.
Collapse
Affiliation(s)
- Kristen M Lorentz
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | | | | |
Collapse
|
20
|
Martínez-Rizo A, Bueno-Topete M, González-Cuevas J, Armendáriz-Borunda J. Plasmin plays a key role in the regulation of profibrogenic molecules in hepatic stellate cells. Liver Int 2010; 30:298-310. [PMID: 19889106 DOI: 10.1111/j.1478-3231.2009.02155.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Plasmin role in transforming growth factor-beta (TGF-beta)-responsive gene regulation remains to be elucidated. Also, plasmin action on co-repressor Ski-related novel protein N (SnoN) and differential activation of matrix metalloproteinases (MMPs) are unknown. Thus, the role of plasmin on profibrogenic molecule expression, SnoN transcriptional kinetics and gelatinase activation was investigated. METHODS Hepatic stellate cells (HSC) were transduced with adenovirus-mediated human urokinase plasminogen activator (Ad-huPA) (4 x 10(9) viral particles/ml). Overexpression of urokinase plasminogen activator and therefore of plasmin, was blocked by tranexamic acid (TA) in transduced HSC. Gene expression was monitored by reverse transcriptase polymerase chain reaction. HSC-free supernatants were used to evaluate MMP-2 and MMP-9 by zymography. SnoN, TGF-beta and tissue inhibitor of metalloproteinase (TIMP)-1 were analysed by Western blot. Plasmin and SnoN expression kinetics were evaluated in bile duct-ligated (BDL) rats. RESULTS Plasmin overexpression in Ad-huPA-transduced HSC significantly decreased gene expression of profibrogenic molecules [alpha1(I)collagen 66%, TIMP-1 59%, alpha-smooth muscle actin 90% and TGF-beta 55%]. Interestingly, both SnoN gene and protein expression increased prominently. Plasmin inhibition by TA upregulated the profibrogenic genes, which respond to TGF-beta-intracellular signalling. In contrast, SnoN mRNA and protein dropped importantly. Plasmin-activated MMP-9 and MMP-2 in HSC supernatants. Taken together, these findings indicate that MMP-9 activation is totally plasmin dependent. SnoN levels significantly decreased in cholestatic-BDL rats (82%) as compared with control animals. Interestingly, hepatic plasmin levels dropped 46% in BDL rats as compared with control. CONCLUSION Plasmin plays a key role in regulating TGF-beta-responding genes. In particular, regulation of TGF-beta-co-repressor (SnoN) is greatly affected, which suggests SnoN as a cardinal player in cholestasis-induced fibrogenesis.
Collapse
Affiliation(s)
- Abril Martínez-Rizo
- Department of Molecular Biology and Genomics, CUCS, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, and OPD Hospital Civil de Guadalajara, Guadalajara, Jalisco, Mexico
| | | | | | | |
Collapse
|
21
|
Madlener K, Pötzsch B. Fibrinolysetests. Hamostaseologie 2010. [DOI: 10.1007/978-3-642-01544-1_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
22
|
Lessons from the aprotinin saga: current perspective on antifibrinolytic therapy in cardiac surgery. J Anesth 2009; 24:96-106. [DOI: 10.1007/s00540-009-0866-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 06/04/2009] [Indexed: 11/26/2022]
|
23
|
Bertram H, Boeuf S, Wachters J, Boehmer S, Heisel C, Hofmann MW, Piecha D, Richter W. Matrix metalloprotease inhibitors suppress initiation and progression of chondrogenic differentiation of mesenchymal stromal cells in vitro. Stem Cells Dev 2009; 18:881-92. [PMID: 19327011 DOI: 10.1089/scd.2008.0306] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stromal cells (MSC) are an attractive source for cell therapy and tissue engineering of joint cartilage. Common chondrogenic in vitro protocols, however, induce hypertrophic markers like COL10A1, matrix metalloproteinase 13 (MMP13), and alkaline phosphatase (ALP) reminiscent of endochondral bone formation. To direct MSC toward articular chondrocytes more specifically, a better understanding of the regulatory steps is desirable. Proteases are important players in matrix remodeling, display inhibitory effects on growth plate development and MMP13 inhibition prevented hypertrophy of bovine chondrocytes. The aim of this study was to evaluate whether the activity of proteases and MMPs, especially MMP13, is crucial for the transition of MSC toward mature chondrocytes and could allow to selectively influence aspects of early and late chondrogenic differentiation. Protease inhibitors were added during MSC chondrogenesis and stage-specific markers were assessed by histology, qPCR, and ALP quantification. Chondrogenesis was little affected by leupeptin, pepstatin, or aprotinin. In contrast, broad spectrum pan-MMP inhibitors dose dependently suppressed proteoglycan deposition, collagen type II and type X staining, ALP activity, and reduced SOX9 and COL2A1 expression. A selective MMP13 inhibitor allowed chondrogenesis and showed only weak effects on ALP activity. In conclusion, transition of MSC toward mature chondrocytes in vitro depended on molecules suppressed by pan-MMP inhibitors identifying chondrogenic differentiation of MSC as a sophistically regulated process in which catabolic enzymes are capable to directly influence cellular fate. In future therapeutic applications of diseased joints, the tested MMP13-specific inhibitor promises suppression of collagen type II degradation without imposing a risk to impair MSC-driven regeneration processes.
Collapse
Affiliation(s)
- Helge Bertram
- Division of Experimental Orthopaedics, Orthopaedic University Clinic Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
From the first description of the “systemic inflammatory response” in the early 1990s, it has been recognized that this is a multifaceted response of the body to the combined insult of cardiothoracic surgery with bypass, involving causation by “activation of complement, coagulation, fibrinolytic, and kallikrein cascades, activation of neutrophils with degranulation and protease enzyme release, oxygen radical production, and the synthesis of various cytokines from mononuclear cells.” Yet the intervening 15 years have seen a narrowing of research into individual systems and interventions naively targeted at single pathways without achieving clinically meaningful benefits. The time has come to redefine the systemic inflammatory response so that research can be more productively focused on objectively measuring and interdicting this multisystem disorder. A key concept of this new understanding is that translation into a hard adverse event occurs when the systemic imbalance is combined with a localized trigger. Triggers might be inadvertently provided by transient episodes of ischemia/malperfusion to vulnerable organs or handling trauma to major vessels. Future research should be directed at suppressing systemic activation with combinations of drugs and improved circuit coating, whereas changes in clinical practice and continuous monitoring of perfusion parameters can help eliminate localized triggering events.
Collapse
Affiliation(s)
- R. Clive Landis
- Edmund Cohen Laboratory for Vascular Research, Chronic Disease Research Centre, University of the West Indies, Bridgetown, Barbados, West Indies
| |
Collapse
|
25
|
Park SH, Cui JH, Park SR, Min BH. Potential of Fortified Fibrin/Hyaluronic Acid Composite Gel as a Cell Delivery Vehicle for Chondrocytes. Artif Organs 2009; 33:439-47. [DOI: 10.1111/j.1525-1594.2009.00744.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
26
|
PAI-1 gene: pharmacogenetic association of 4G/4G genotype with bleeding after cardiac surgery – pilot study. Eur J Anaesthesiol 2009; 26:404-11. [DOI: 10.1097/eja.0b013e3283240412] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Head BP, Patel HH, Niesman IR, Drummond JC, Roth DM, Patel PM. Inhibition of p75 neurotrophin receptor attenuates isoflurane-mediated neuronal apoptosis in the neonatal central nervous system. Anesthesiology 2009; 110:813-25. [PMID: 19293698 PMCID: PMC2767332 DOI: 10.1097/aln.0b013e31819b602b] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Exposure to anesthetics during synaptogenesis results in apoptosis and subsequent cognitive dysfunction in adulthood. Probrain-derived neurotrophic factor (proBDNF) is involved in synaptogenesis and can induce neuronal apoptosis via p75 neurotrophic receptors (p75). proBDNF is cleaved into mature BDNF (mBDNF) by plasmin, a protease converted from plasminogen by tissue plasminogen activator (tPA) that is released with neuronal activity; mBDNF supports survival and stabilizes synapses through tropomyosin receptor kinase B. The authors hypothesized that anesthetics suppress tPA release from neurons, enhance p75 signaling, and reduce synapses, resulting in apoptosis. METHODS Primary neurons (DIV5) and postnatal day 5-7 (PND5-7) mice were exposed to isoflurane (1.4%, 4 h) in 5% CO2, 95% air. Apoptosis was assessed by cleaved caspase-3 (Cl-Csp3) immunoblot and immunofluorescence microscopy. Dendritic spine changes were evaluated with the neuronal spine marker, drebrin. Changes in synapses in PND5-7 mouse hippocampi were assessed by electron microscopy. Primary neurons were exposed to tPA, plasmin, or pharmacologic inhibitors of p75 (Fc-p75 or TAT-Pep5) 15 min before isoflurane. TAT-Pep5 was administered by intraperitoneal injection to PND5-7 mice 15 min before isoflurane. RESULTS Exposure of neurons in vitro (DIV5) to isoflurane decreased tPA in the culture medium, reduced drebrin expression (marker of dendritic filopodial spines), and enhanced Cl-Csp3. tPA, plasmin, or TAT-Pep5 stabilized dendritic filopodial spines and decreased Cl-Csp3 in neurons. TAT-Pep5 blocked isoflurane-mediated increase in Cl-Csp3 and reduced synapses in PND5-7 mouse hippocampi. CONCLUSION tPA, plasmin, or p75 inhibition blocked isoflurane-mediated reduction in dendritic filopodial spines and neuronal apoptosis in vitro. Isoflurane reduced synapses and enhanced Cl-Csp3 in the hippocampus of PND5-7 mice, the latter effect being mitigated by p75 inhibition in vivo. These data support the hypothesis that isoflurane neurotoxicity in the developing rodent brain is mediated by reduced synaptic tPA release and enhanced proBDNF/p75-mediated apoptosis.
Collapse
Affiliation(s)
- Brian P. Head
- Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093
| | - Hemal H. Patel
- Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093
| | - Ingrid R. Niesman
- Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093
| | - John C. Drummond
- Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093
- VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| | - David M. Roth
- Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093
- VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| | - Piyush M. Patel
- Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093
- VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| |
Collapse
|
28
|
McEvoy MD, Reeves ST, Reves JG, Spinale FG. Aprotinin in Cardiac Surgery: A Review of Conventional and Novel Mechanisms of Action. Anesth Analg 2007; 105:949-62. [PMID: 17898372 DOI: 10.1213/01.ane.0000281936.04102.9f] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Induction of the coagulation and inflammatory cascades can cause multiorgan dysfunction after cardiopulmonary bypass (CPB). In light of these observations, strategies that can stabilize the coagulation process as well as attenuate the inflammatory response during and after cardiac surgery are important. Aprotinin has effects on hemostasis. In addition, aprotinin may exert multiple biologically relevant effects in the context of cardiac surgery and CPB. For example, it decreases neutrophil and macrophage activation and chemotaxis, attenuates release and activation of proinflammatory cytokines, and reduces oxidative stress. Despite these perceived benefits, the routine use of aprotinin in cardiac surgery with CPB has been called into question. In this review, we examined this controversial drug by discussing the classical and novel pathways in which aprotinin may be operative in the context of cardiac surgery.
Collapse
Affiliation(s)
- Matthew D McEvoy
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.
| | | | | | | |
Collapse
|
29
|
Sperzel M, Huetter J. Evaluation of aprotinin and tranexamic acid in different in vitro and in vivo models of fibrinolysis, coagulation and thrombus formation. J Thromb Haemost 2007; 5:2113-8. [PMID: 17666018 DOI: 10.1111/j.1538-7836.2007.02717.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The serine protease inhibitor aprotinin and plasminogen inhibitor tranexamic acid are used in coronary artery bypass graft (CABG) surgery to reduce bleeding. Clinicians may consider these agents as readily substitutable regarding their pharmacological profiles. OBJECTIVE These agents were evaluated in assays of hemostasis to elucidate their underlying mechanism(s) of action. METHODS In human plasma, effects on both clot fibrinolysis and coagulation were spectrophotometrically quantified in vitro. Rat-tail bleeding and arteriovenous shunt thrombus formation models were conducted in vivo. RESULTS Fibrinolysis was inhibited by aprotinin (IC(50), 0.16 +/- 0.02 micromol L(-1)) and tranexamic acid (IC(50), 24.1 +/-1.1 micromol L(-1)). In vivo, aprotinin dose-dependently reduced rat-tail bleeding time (minimal effective dose, 3 mg kg(-1) bolus plus 6 mg kg(-1 )h(-1) infusion); tranexamic acid reduced bleeding time (minimal effective dose, 100 mg kg(-1) h(-1)). In vitro, coagulation time was doubled by aprotinin at 3.2 +/- 0.2 micromol L(-1), while tranexamic acid showed no effect at concentrations up to 3 mmol L(-1). Aprotinin inhibited thrombus formation in vivo in a dose-dependent manner (minimal effective dose, 3 mg kg(-1) bolus plus 6 mg kg(-1) h(-1) infusion). Conversely, tranexamic acid dose-dependently increased thrombus formation and thrombus weight (minimal effective dose, 100 mg kg(-1 )h(-1) infusion). CONCLUSIONS These data show that aprotinin and tranexamic acid have differential effects on hemostasis and are not necessarily substitutable with respect to mechanism of action. Although both agents have been shown to reduce bleeding in patients undergoing CABG, their divergent effects on thrombus formation observed in vitro and in vivo should be critically evaluated clinically.
Collapse
Affiliation(s)
- M Sperzel
- Product Related Research, Bayer Healthcare AG, Wuppertal, Germany.
| | | |
Collapse
|
30
|
Ahmed TAE, Griffith M, Hincke M. Characterization and inhibition of fibrin hydrogel-degrading enzymes during development of tissue engineering scaffolds. ACTA ACUST UNITED AC 2007; 13:1469-77. [PMID: 17518706 DOI: 10.1089/ten.2006.0354] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The goal of articular cartilage tissue engineering is to provide cartilaginous constructs to replace abnormal cartilage. We have evaluated the chondroprogenitor clonal cell line RCJ3.1C5.18 (C5.18) as a model to guide the development of appropriate scaffolds for tissue engineering. Rapid degradation of fibrin hydrogels was observed after encapsulation of C5.18 cells. The enzymes responsible for this fibrin gel breakdown were characterized to control their activity and regulate gel stability. Western blotting, confirming zymography, revealed bands due to matrix metalloproteinases (MMP-2, MMP-3) that are secreted concomitantly with fibrin hydrogels breakdown. High plasmin activity was detected in conditioned media during hydrogel breakdown but not in the confluent cells before encapsulation. Reverse transcriptase polymerase chain reaction indicated the expression of MMP-2, -3, and -9 and plasminogen in the cells. MMP-9 was 100 times higher at day 1, whereas MMP-2 started to increase and reached its maximum level by day 7. Aprotinin, a known serine protease inhibitor, and galardin (GM6001), a potent MMP inhibitor, in combination or separately, prevented the breakdown of fibrin-C5.18 hydrogels, whereas only the combination of both promoted the accumulation of extracellular matrix. These findings suggest that plasmin and MMPs contribute independently to fibrin hydrogel breakdown, but that either enzyme can achieve extracellular matrix breakdown.
Collapse
Affiliation(s)
- Tamer A E Ahmed
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | | | | |
Collapse
|
31
|
Abstract
The reimplantation method of cultured chondrocytes broadly has been offered as an
alternative for articular cartilage repair. A variety of biologically derived and synthetic polymeric
and hydrogel materials also have been investigated for good cell delivery efficiency. Preciously, we
examined the feasibility of fibrin gel, mixed with hyaluronic acid(HA) as a cell delivery carrier. In
order to reinforce the material, hybrid biomaterials of fibrin/HA composite gels with fibrinolysis
inhibition factors(FIFs: aprotinin, DI101, EACA) have been investigated in the present work
because we did not satisfy a little progress. These fibrin/HA composite gels added FIFs maintained
their structural integrity in long-term culture over 4th weeks. Contrary to our expectation the mass of
the fibrin/HA composite with DI 101 was significantly superior to the ones of other combinations.
In histological evidence, all of them are showed good positive result of stain of Safranin-O and
alcian blue during the culture period. In gross examination, samples of all groups grossly resembled
cartilage in color and were resistant to external compression. Our study demonstrates that most
favorable polymer can be used good quality tissue engineered cartilage and in this culture systems
have been useful for studying the basic biology of chondrocyte biosynthesis of ECM and new
cartilage matrix formation without a loss of volume. After all, we proved the safety of inhibitors of
the fibrinolytic system without hazardous effect on cell behavior and found out that DI 101 would
be the most effective agent.
Collapse
|
32
|
Nielsen VG, Cankovic L, Steenwyk BL. ε-Aminocaproic acid inhibition of fibrinolysis in vitro: should the ‘therapeutic’ concentration be reconsidered? Blood Coagul Fibrinolysis 2007; 18:35-9. [PMID: 17179824 DOI: 10.1097/mbc.0b013e328010a359] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The therapeutic concentration of epsilon-aminocaproic acid (EACA) has been 130 microg/ml or greater for nearly 50 years. We tested the effects on clot growth/disintegration of EACA with a plasmatic model of hyperfibrinolysis in vitro. Human plasma was exposed to 1000 U/ml tissue-type plasminogen activator containing 0, 13, 65 or 130 microg/ml EACA, with clot growth/disintegration kinetics quantified via thrombelastography. Data were analyzed with one-way analysis of variance or Kruskal-Wallis analysis of variance as appropriate. Exposure of plasma to 1000 U/ml tissue-type plasminogen activator resulted in a brief-lived clot, lasting 2 min. EACA at all concentrations tested significantly increased the rate of clot growth compared with samples with 0 microg/ml EACA. Clot strength was significantly increased by EACA in a concentration-dependent fashion. Similarly, EACA significantly prolonged the time of onset of clot lysis and decreased the rate of lysis. Samples with 130 microg/ml EACA had no sign of lysis present for 30 min. Subtherapeutic to therapeutic concentrations of EACA significantly attenuated or abolished fibrinolysis in the presence of a concentration of tissue-type plasminogen activator more than 2000-fold that encountered systemically during cardiopulmonary bypass. Further clinical investigation is warranted to determine whether smaller concentrations of EACA could provide a reduction in bleeding with a concomitant decrease in thrombotic complications.
Collapse
Affiliation(s)
- Vance G Nielsen
- Department of Anesthesiology, The University of Alabama, Birmingham, Alabama, USA.
| | | | | |
Collapse
|
33
|
Willerth SM, Arendas KJ, Gottlieb DI, Sakiyama-Elbert SE. Optimization of fibrin scaffolds for differentiation of murine embryonic stem cells into neural lineage cells. Biomaterials 2006; 27:5990-6003. [PMID: 16919326 PMCID: PMC1794024 DOI: 10.1016/j.biomaterials.2006.07.036] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Accepted: 07/27/2006] [Indexed: 12/28/2022]
Abstract
The objective of this research was to determine the appropriate cell culture conditions for embryonic stem (ES) cell proliferation and differentiation in fibrin scaffolds by examining cell seeding density, location, and the optimal concentrations of fibrinogen, thrombin, and aprotinin (protease inhibitor). Mouse ES cells were induced to become neural progenitors by adding retinoic acid for 4 days to embryoid body (EB) cultures. For dissociated EBs, the optimal cell seeding density and location was determined to be 250,000 cells/cm(2) seeded on top of fibrin scaffolds. For intact EBs, three-dimensional (3D) cultures with one EB per 400 microL fibrin scaffold resulted in greater cell proliferation and differentiation than two-dimensional (2D) cultures. Optimal concentrations for scaffold polymerization were 10mg/mL of fibrinogen and 2 NIH units/mL of thrombin. The optimal aprotinin concentration was determined to be 50 microg/mL for dissociated EBs (2D) and 5 microg/mL for intact EBs in 3D fibrin scaffolds. Additionally, after 14 days in 3D culture EBs differentiated into neurons and astrocytes as indicated by immunohistochemisty. These conditions provide an optimal fibrin scaffold for evaluating ES cell differentiation and proliferation in culture, and for use as a platform for neural tissue engineering applications, such as the treatment for spinal cord injury.
Collapse
Affiliation(s)
- Stephanie M Willerth
- Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, Saint Louis, MO 63130, USA
| | | | | | | |
Collapse
|
34
|
Jurasz P, Santos-Martinez MJ, Radomska A, Radomski MW. Generation of platelet angiostatin mediated by urokinase plasminogen activator: effects on angiogenesis. J Thromb Haemost 2006; 4:1095-106. [PMID: 16689764 DOI: 10.1111/j.1538-7836.2006.01878.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Angiogenesis, the growth of new capillaries from pre-existing blood vessels, is regulated by a balance between its promoters and inhibitors. Platelets are an important circulating store of angiogenesis regulators. We have previously identified the angiogenesis inhibitor angiostatin in human platelets. AIM To identify the mechanism of platelet angiostatin generation and its pharmacological regulation. METHODS Platelet aggregometry, flow cytometry, Western blot, zymography, immunofluorescence microscopy, matrigel-induced angiogenesis of human umbilical vein endothelial cells (HUVECs), and a panel of selective proteinase inhibitors were used to study the mechanism of angiostatin generation by platelets, its pharmacological regulation, and effects on angiogenesis. Release of pro-MMP-2 by HUVECs was also used to quantify angiogenesis. RESULTS Platelet membranes were identified as the site of angiostatin generation from plasminogen. Generation of angiostatin by platelet membranes was not affected by a matrix metalloproteinase (MMP) inhibitor, phenanthroline, but was inhibited by serine proteinase inhibitors aprotinin, leupeptin, plasminogen activator inhibitor-1, and selective inhibitor of urokinase plasminogen activator (uPA), uPA-STOP(TM). Angiostatin generation by intact platelets was inhibited by aprotinin, and the resulting incubate promoted angiogenesis to a greater extent than incubate where angiostatin generation occurred. Furthermore, HUVECs incubated with reaction mixture, where angiostatin generation was inhibited, released more pro-MMP-2 than HUVECs incubated with supernatants, where angiostatin generation occurred. CONCLUSIONS We conclude that; (i) platelets constitutively generate angiostatin on their membranes; (ii) this mechanism is dependent on uPA, but not, MMPs; and (iii) inhibition of platelet angiostatin generation can further promote angiogenesis.
Collapse
Affiliation(s)
- P Jurasz
- Institute of Molecular Medicine for the Prevention of Human Diseases, Vascular Biology Section, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|