1
|
Tomo S, Kiran Kumar PVSN, Yadav D, Sankanagoudar S, Charan J, Purohit A, Nag VL, Bhatia PK, Singh K, Dutt N, Garg MK, Misra S, Sharma P, Purohit P. Association of Serum Complement C3 Levels with Severity and Mortality in COVID 19. Indian J Clin Biochem 2023; 38:447-456. [PMID: 37746543 PMCID: PMC10516839 DOI: 10.1007/s12291-023-01148-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/25/2023] [Indexed: 09/26/2023]
Abstract
The severe acute respiratory distress syndrome-associated coronavirus-2 infection can activate innate and adaptive immune responses which may lead to harmful tissue damage, both locally and systemically. C3, a member of complement system of serum proteins, is a major component of innate immune and inflammatory responses. This study is aimed to assess serum C3 as a marker of COVID-19 severity and a predictor of disease progression. A total of 150 COVID-19 patients, confirmed by RT-PCR, and 50 healthy controls were recruited. Serum C3 levels were determined by using direct colorimetric method. Median levels of serum C3 in total cases and controls were 157.8 and 165.7 mg/dL respectively. Serum C3 although not significantly decreased, they were lower in cases when compared to controls. Similarly, significant differences were found between the groups, with severe group (140.6 mg/dL) having low levels of serum C3 protein when compared to mild (161.0 mg/dL) and moderate group (167.1 mg/dL). Interestingly, during hospitalization, significant difference between baseline (admission) and follow-up (discharge) was observed only in patients with moderate disease. Based on our results, lower levels of C3, with an increase in IL-6 and d-dimer levels, are associated with higher odds of mortality. Therefore, we would like to emphasize that measuring serum C3 levels along with other inflammatory markers might give an added advantage in early identification of patients who are prone to having a severe disease course and can help in a more effective follow-up of disease progression. Supplementary Information The online version contains supplementary material available at 10.1007/s12291-023-01148-x.
Collapse
Affiliation(s)
- Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Jodhpur, Basni Phase 2, Jodhpur, Rajasthan 342005 India
| | - PVSN Kiran Kumar
- Department of Biochemistry, Andhra Medical College, Visakhapatnam, India
| | - Dharamveer Yadav
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Jodhpur, Basni Phase 2, Jodhpur, Rajasthan 342005 India
| | - Shrimanjunath Sankanagoudar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Jodhpur, Basni Phase 2, Jodhpur, Rajasthan 342005 India
| | - Jayakaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, India
| | - Abhishek Purohit
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Jodhpur, India
| | - Vijaya Lakshmi Nag
- Department of Microbiology, All India Institute of Medical Sciences, Jodhpur, India
| | - Pradeep Kumar Bhatia
- Department of Anaesthesiology and Critical Care, All India Institute of Medical Sciences, Jodhpur, India
| | - Kuldeep Singh
- Department of Paediatrics, All India Institute of Medical Sciences, Jodhpur, India
| | - Naveen Dutt
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, Jodhpur, India
| | - Mahendra Kumar Garg
- Department of General Medicine, All India Institute of Medical Sciences, Jodhpur, India
| | - Sanjeev Misra
- Department of Surgical Oncology, All India Institute of Medical Sciences, Jodhpur, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Jodhpur, Basni Phase 2, Jodhpur, Rajasthan 342005 India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Jodhpur, Basni Phase 2, Jodhpur, Rajasthan 342005 India
| |
Collapse
|
2
|
Abstract
The complement and hemostatic systems are complex systems, and both involve enzymatic cascades, regulators, and cell components-platelets, endothelial cells, and immune cells. The two systems are ancestrally related and are defense mechanisms that limit infection by pathogens and halt bleeding at the site of vascular injury. Recent research has uncovered multiple functional interactions between complement and hemostasis. On one side, there are proteins considered as complement factors that activate hemostasis, and on the other side, there are coagulation proteins that modulate complement. In addition, complement and coagulation and their regulatory proteins strongly interact each other to modulate endothelial, platelet and leukocyte function and phenotype, creating a potentially devastating amplifying system that must be closely regulated to avoid unwanted damage and\or disseminated thrombosis. In view of its ability to amplify all complement activity through the C3b-dependent amplification loop, the alternative pathway of complement may play a crucial role in this context. In this review, we will focus on available and emerging evidence on the role of the alternative pathway of complement in regulating hemostasis and vice-versa, and on how dysregulation of either system can lead to severe thromboinflammatory events.
Collapse
Affiliation(s)
- Marina Noris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Miriam Galbusera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
3
|
Song S, Regan B, Ereifej ES, Chan ER, Capadona JR. Neuroinflammatory Gene Expression Analysis Reveals Pathways of Interest as Potential Targets to Improve the Recording Performance of Intracortical Microelectrodes. Cells 2022; 11:2348. [PMID: 35954192 PMCID: PMC9367362 DOI: 10.3390/cells11152348] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Intracortical microelectrodes are a critical component of brain-machine interface (BMI) systems. The recording performance of intracortical microelectrodes used for both basic neuroscience research and clinical applications of BMIs decreases over time, limiting the utility of the devices. The neuroinflammatory response to the microelectrode has been identified as a significant contributing factor to its performance. Traditionally, pathological assessment has been limited to a dozen or so known neuroinflammatory proteins, and only a few groups have begun to explore changes in gene expression following microelectrode implantation. Our initial characterization of gene expression profiles of the neuroinflammatory response to mice implanted with non-functional intracortical probes revealed many upregulated genes that could inform future therapeutic targets. Emphasis was placed on the most significant gene expression changes and genes involved in multiple innate immune sets, including Cd14, C3, Itgam, and Irak4. In previous studies, inhibition of Cluster of Differentiation 14 (Cd14) improved microelectrode performance for up to two weeks after electrode implantation, suggesting CD14 can be explored as a potential therapeutic target. However, all measures of improvements in signal quality and electrode performance lost statistical significance after two weeks. Therefore, the current study investigated the expression of genes in the neuroinflammatory pathway at the tissue-microelectrode interface in Cd14-/- mice to understand better how Cd14 inhibition was connected to temporary improvements in recording quality over the initial 2-weeks post-surgery, allowing for the identification of potential co-therapeutic targets that may work synergistically with or after CD14 inhibition to improve microelectrode performance.
Collapse
Affiliation(s)
- Sydney Song
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Cleveland, OH 44106, USA; (S.S.); (E.S.E.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Brianna Regan
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA;
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evon S. Ereifej
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Cleveland, OH 44106, USA; (S.S.); (E.S.E.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA;
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - E. Ricky Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Cleveland, OH 44106, USA; (S.S.); (E.S.E.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
4
|
Polysaccharide-based layer-by-layer nanoarchitectonics with sulfated chitosan for tuning anti-thrombogenic properties. Colloids Surf B Biointerfaces 2022; 213:112359. [PMID: 35144082 DOI: 10.1016/j.colsurfb.2022.112359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 11/23/2022]
Abstract
The development of blood-interacting surfaces is critical to fabricate biomaterials for medical use, such as prostheses, implants, biosensors, and membranes. For instance, thrombosis is one of the leading clinical problems when polymer-based materials interact with blood. To overcome this limitation is necessary to develop strategies that limit platelets adhesion and activation. In this work, hyaluronan (HA)/chitosan (Chi) based-films, recently reported in the literature as platforms for tumor cell capture, were developed and, subsequently, functionalized with sulfated chitosan (ChiS) using a layer-by-layer technique. ChiS, when compared to native Chi, presents the unique abilities to confer anti-thrombogenic properties, to reduce protein adsorption, and also to limit calcification. Film physicochemical characterization was carried out using FTIR and XPS for chemical composition assessment, AFM for the surface morphology, and contact angle for hydrophilicity evaluation. The deposition of ChiS monolayer promoted a decrease in both roughness and hydrophilicity of the HA/Chi films. In addition, the appearance of sulfur in the chemical composition of ChiS-functionalized films confirmed the film modification. Biological assay indicated that the incorporation of sulfated groups limited platelet adhesion, mainly because a significant reduction of platelets adhesion to ChiS-functionalized films was observed compared to HA/Chi films. On balance, this work provides a new insight for the development of novel antithrombogenic biomaterials, opening up new possibilities for devising blood-interaction surfaces.
Collapse
|
5
|
Zang Y, Vlcek JR, Cuchiaro J, Popat KC, Olver CS, Kipper MJ, Reynolds MM. Ex vivo evaluation of blood coagulation on endothelial glycocalyx-inspired surfaces using thromboelastography. IN VITRO MODELS 2022; 1:59-71. [PMID: 39872977 PMCID: PMC11749744 DOI: 10.1007/s44164-021-00001-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 01/30/2025]
Abstract
Purpose Present blood-contacting materials have not yet demonstrated to be effective in reducing blood coagulation without causing additional side effects clinically. We have developed an endothelial glycocalyx-inspired biomimetic surface that combines nanotopography, heparin presentation, and nitric oxide (NO)-releasing features. The resulting modified surfaces have already shown promise in reducing unfavorable blood-material interactions using platelet-rich plasma. In this study, the efficacy of modified surfaces for reducing coagulation of human whole blood was measured. In addition, the effects of leached polysaccharides and chemical modification of the modified surfaces were evaluated. Methods Leached polysaccharides in the incubation solution were detected by a refractive index method to determine the potential influences of these modified surfaces on the blood coagulation observation. Chemical modifications by the nitrosation process on the polysaccharides in the modified surfaces were detected using attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR). Clot formation parameters were measured using thromboelastography (TEG), a clinically relevant technique to evaluate whole blood coagulation. Results No polysaccharides were detected in the heparinized polyelectrolyte multilayer-coated titania nanotube array surface (TiO2NT + PEM) incubation solution; however, polysaccharides were detected from NO-releasing TiO2NT + PEM surface (TiO2NT + PEM + NO) incubation solution both after the nitrosation process and after all NO was released. The structures of thiolated chitosan and heparin were altered by t-butyl nitrite. All heparin-containing surfaces were shown to slow or inhibit clot formation. Conclusion This study is the first to evaluate these endothelial glycocalyx-inspired surfaces using clinically relevant parameters, as well as proposing potential influences of these modified surfaces on the inhibition of clot formation. Supplementary Information The online version contains supplementary material available at 10.1007/s44164-021-00001-w.
Collapse
Affiliation(s)
- Yanyi Zang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO USA
- Present Address: Autonomous Reanimation and Evacuation Research Program, The Geneva Foundation, Brook City-Base, San Antonio, TX USA
| | - Jessi R. Vlcek
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO USA
| | - Jamie Cuchiaro
- Department of Chemistry, Colorado State University, Fort Collins, CO USA
| | - Ketul C. Popat
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO USA
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, CO USA
| | - Christine S. Olver
- Veterinary Clinical Pathology Section, Colorado State University, Fort Collins, CO USA
| | - Matt J. Kipper
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO USA
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, CO USA
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO USA
| | - Melissa M. Reynolds
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO USA
- Department of Chemistry, Colorado State University, Fort Collins, CO USA
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, CO USA
| |
Collapse
|
6
|
Ma J, Liu Q, White JR. Novel methods to determine complement activation in human serum induced by the complex of Dezamizumab and serum amyloid P. J Biol Chem 2021; 297:101136. [PMID: 34461096 PMCID: PMC8463879 DOI: 10.1016/j.jbc.2021.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 12/02/2022] Open
Abstract
Lack of simple and robust methods to determine complement activation in human serum induced by antigen–antibody complexes is a major hurdle for monitoring therapeutic antibody drug quality and stability. Dezamizumab is a humanized IgG1 monoclonal antibody that binds to serum amyloid P component (SAP) for potential treatment of systemic amyloidosis. The mechanism of action of Dezamizumab includes the binding of SAP, complement activation through classical pathway, and phagocytosis; however, the steps in this process cannot be easily monitored. We developed two novel methods to determine Dezamizumab-SAP complex-induced complement activation. Complement component 3 (C3) depletion was detected by homogeneous time-resolved fluorescence (HTRF), and C3a desArg fragment, formed after the cleavage of C3 to yield C3a followed by removal of its C-terminal arginine residue, was determined using Meso Scale Discovery (MSD) technology. We found that the presence of both Dezamizumab and SAP was required for complement activation via both methods. The optimal molar ratio of Dezamizumab:SAP was 6:1 in order to obtain maximal complement activation. The relative potency from both methods showed a good correlation to Dezamizumab-SAP-dependent complement component 1q (C1q) binding activity in Dezamizumab thermal-stressed samples. Both SAP and C1q binding, as determined by surface plasmon resonance and the two complement activation potency methods described here, reflect the mechanism of action of Dezamizumab. We conclude that these methods can be used to monitor Dezamizumab quality for drug release and stability testing, and the novel potency methods reported here can be potentially used to evaluate complement activity induced by other antigen–antibody complexes.
Collapse
Affiliation(s)
- Jianhong Ma
- GlaxoSmithKline, Structure Function Characterization, CMCA, Collegeville, Pennsylvania, USA.
| | - Qi Liu
- GlaxoSmithKline, Structure Function Characterization, CMCA, Collegeville, Pennsylvania, USA
| | - John R White
- GlaxoSmithKline, Structure Function Characterization, CMCA, Collegeville, Pennsylvania, USA
| |
Collapse
|
7
|
Kalm F, Mansouri L, Russom A, Lundahl J, Nopp A. Adhesion molecule cross-linking and cytokine exposure modulate IgE- and non-IgE-dependent basophil activation. Immunology 2020; 162:92-104. [PMID: 32955733 PMCID: PMC7730031 DOI: 10.1111/imm.13268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/26/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
Basophils are known for their role in allergic inflammation, which makes them suitable targets in allergy diagnostics such as the basophil activation test (BAT) and the microfluidic immunoaffinity basophil activation test (miBAT). Beside their role in allergy, basophils have an immune modulatory role in both innate immunity and adaptive immunity. To accomplish this mission, basophils depend on the capability to migrate from blood to extravascular tissues, which includes interactions with endothelial cells, extracellular matrix and soluble mediators. Their receptor repertoire is well known, but less is known how these receptor–ligand interactions impact the degranulation process and the responsiveness to subsequent activation. As the consequences of these interactions are crucial to fully appreciate the role of basophils in immune modulation and to enable optimization of the miBAT, we explored how basophil activation status is regulated by cytokines and cross‐linking of adhesion molecules. The expression of adhesion molecules and activation markers on basophils from healthy blood donors was analysed by flow cytometry. Cross‐linking of CD203c, CD62L, CD11b and CD49d induced a significant upregulation of CD63 and CD203c. To mimic in vivo conditions, valid also for miBAT, CD62L and CD49d were cross‐linked followed by IgE‐dependent activation (anti‐IgE), which caused a reduced CD63 expression compared with anti‐IgE activation only. IL‐3 and IL‐33 priming caused increased CD63 expression after IgE‐independent activation (fMLP). Together, our data suggest that mechanisms operational both in the microfluidic chip and in vivo during basophil adhesion may impact basophil anaphylactic and piecemeal degranulation procedures and hence their immune regulatory function.
Collapse
Affiliation(s)
- Frida Kalm
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden.,Division of Nanobiotechnology, Department of Protein Sciences, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Ladan Mansouri
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Aman Russom
- Division of Nanobiotechnology, Department of Protein Sciences, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Joachim Lundahl
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Anna Nopp
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
8
|
Mödinger Y, Teixeira GQ, Neidlinger-Wilke C, Ignatius A. Role of the Complement System in the Response to Orthopedic Biomaterials. Int J Mol Sci 2018; 19:ijms19113367. [PMID: 30373272 PMCID: PMC6274916 DOI: 10.3390/ijms19113367] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022] Open
Abstract
Various synthetic biomaterials are used to replace lost or damaged bone tissue that, more or less successfully, osseointegrate into the bone environment. Almost all biomaterials used in orthopedic medicine activate the host-immune system to a certain degree. The complement system, which is a crucial arm of innate immunity, is rapidly activated by an implanted foreign material into the human body, and it is intensely studied regarding blood-contacting medical devices. In contrast, much less is known regarding the role of the complement system in response to implanted bone biomaterials. However, given the increasing knowledge of the complement regulation of bone homeostasis, regeneration, and inflammation, complement involvement in the immune response following biomaterial implantation into bone appears very likely. Moreover, bone cells can produce complement factors and are target cells of activated complement. Therefore, new bone formation or bone resorption around the implant area might be greatly influenced by the complement system. This review aims to summarize the current knowledge on biomaterial-mediated complement activation, with a focus on materials primarily used in orthopedic medicine. In addition, methods to modify the interactions between the complement system and bone biomaterials are discussed, which might favor osseointegration and improve the functionality of the device.
Collapse
Affiliation(s)
- Yvonne Mödinger
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Graciosa Q Teixeira
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Cornelia Neidlinger-Wilke
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| |
Collapse
|
9
|
Hardersen R, Enebakk T, Christiansen D, Bergseth G, Brekke OL, Mollnes TE, Lappegård KT, Hovland A. Granulocyte and monocyte CD11b expression during plasma separation is dependent on complement factor 5 (C5) - an ex vivo study with blood from a C5-deficient individual. APMIS 2018; 126:342-352. [PMID: 29575196 DOI: 10.1111/apm.12821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 01/21/2018] [Indexed: 12/18/2022]
Abstract
The aim of the study was to investigate the role of complement factor 5 (C5) in reactions elicited by plasma separation using blood from a C5-deficient (C5D) individual, comparing it to C5-deficient blood reconstituted with C5 (C5DR) and blood from healthy donors. Blood was circulated through an ex vivo plasma separation model. Leukocyte CD11b expression and leukocyte-platelet conjugates were measured by flow cytometry during a 30-min period. Other markers were assessed during a 240-min period. Granulocyte and monocyte CD11b expression did not increase in C5D blood during plasma separation. In C5DR samples granulocytes CD11b expression, measured by mean fluorescence intensity (MFI), increased from 10481 ± 6022 (SD) to 62703 ± 4936, and monocytes CD11b expression changed from 13837 ± 7047 to 40063 ± 713. Granulocyte-platelet conjugates showed a 2.5-fold increase in the C5DR sample compared to the C5D sample. Monocyte-platelet conjugates increased independently of C5. In the C5D samples, platelet count decreased from 210 × 109 /L (201-219) (median and range) to 51 × 109 /L (50-51), and C3bc increased from 14 CAU/mL (21-7) to 198 CAU/mL (127-269), whereas TCC formation was blocked during plasma separation. In conclusion, up-regulation of granulocyte and monocyte CD11b during plasma separation was C5-dependent. The results also indicate C5 dependency in granulocyte-platelet conjugates formation.
Collapse
Affiliation(s)
- Randolf Hardersen
- Department of Nephrology, Division of Internal Medicine, Nordland Hospital, Bodø, Norway
| | - Terje Enebakk
- Department of Nephrology, Division of Internal Medicine, Nordland Hospital, Bodø, Norway
| | | | | | - Ole-Lars Brekke
- Institute of Clinical Medicine and K. G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Department of Laboratory Medicine, Nordland Hospital, Bodø, Norway
| | - Tom Eirik Mollnes
- Research Laboratory, Nordland Hospital, Bodø, Norway.,Institute of Clinical Medicine and K. G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway.,K.J. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Knut Tore Lappegård
- Institute of Clinical Medicine and K. G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Department of Cardiology, Division of Internal Medicine, Nordland Hospital, Bodø, Norway
| | - Anders Hovland
- Institute of Clinical Medicine and K. G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Department of Cardiology, Division of Internal Medicine, Nordland Hospital, Bodø, Norway
| |
Collapse
|
10
|
Abstract
Sulfonated cellulose beads were prepared by oxidation of Cladophora nanocellulose to 2,3-dialdehyde cellulose followed by sulfonation using bisulfite. The physicochemical properties of the sulfonated beads, i.e., high surface area, high degree of oxidation, spherical shape, and the possibility of tailoring the porosity, make them interesting candidates for the development of immunosorbent platforms, including their application in extracorporeal blood treatments. A desired property for materials used in such applications is blood compatibility; therefore in the present work, we investigate the hemocompatibility of the sulfonated cellulose beads using an in vitro whole blood model. Complement system activation (C3a and sC5b-9 levels), coagulation activation (thrombin-antithrombin (TAT) levels) and hemolysis were evaluated after whole blood contact with the sulfonated beads and the results were compared with the values obtained with the unmodified Cladophora nanocellulose. Results showed that neither of the cellulosic materials presented hemolytic activity. A marked decrease in TAT levels was observed after blood contact with the sulfonated beads, compared with Cladophora nanocellulose. However, the chemical modification did not promote an improvement in Cladophora nanocellulose hemocompatibility in terms of complement system activation. Even though the sulfonated beads presented a significant reduction in pro-coagulant activity compared with the unmodified material, further modification strategies need to be investigated to control the complement activation by the cellulosic materials.
Collapse
|
11
|
Blatt AZ, Pathan S, Ferreira VP. Properdin: a tightly regulated critical inflammatory modulator. Immunol Rev 2017; 274:172-190. [PMID: 27782331 PMCID: PMC5096056 DOI: 10.1111/imr.12466] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The complement alternative pathway is a powerful arm of the innate immune system that enhances diverse inflammatory responses in the human host. Key to the effects of the alternative pathway is properdin, a serum glycoprotein that can both initiate and positively regulate alternative pathway activity. Properdin is produced by many different leukocyte subsets and circulates as cyclic oligomers of monomeric subunits. While the formation of non‐physiological aggregates in purified properdin preparations and the presence of potential properdin inhibitors in serum have complicated studies of its function, properdin has, regardless, emerged as a key player in various inflammatory disease models. Here, we review basic properdin biology, emphasizing the major hurdles that have complicated the interpretation of results from properdin‐centered studies. In addition, we elaborate on an emerging role for properdin in thromboinflammation and discuss the potential utility of properdin inhibitors as long‐term therapeutic options to treat diseases marked by increased formation of platelet/granulocyte aggregates. Finally, we describe the interplay between properdin and the alternative pathway negative regulator, Factor H, and how aiming to understand these interactions can provide scientists with the most effective ways to manipulate alternative pathway activation in complex systems.
Collapse
Affiliation(s)
- Adam Z Blatt
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sabina Pathan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
12
|
Campelo CS, Chevallier P, Vaz JM, Vieira RS, Mantovani D. Sulfonated chitosan and dopamine based coatings for metallic implants in contact with blood. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 72:682-691. [PMID: 28024638 DOI: 10.1016/j.msec.2016.11.133] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/01/2016] [Accepted: 11/26/2016] [Indexed: 11/28/2022]
Abstract
Thrombosis and calcification constitute the main clinical problems when blood-interacting devices are implanted in the body. Coatings with thin polymer layers represent an acknowledged strategy to modulate interactions between the material surface and the blood environment. To ensure the implant success, at short-term the coating should limit platelets adhesion and delay the clot formation, and at long-term it should delay the calcification process. Sulfonated chitosan, if compared to native chitosan, shows the unique ability to reduce proteins adsorption, decrease thrombogenic properties and limit calcification. In this work, stainless steel surfaces, commonly used for cardiovascular applications, were coated with sulfonated chitosan, by using dopamine and PEG as anchors, and the effect of these grafted surfaces on platelet adhesion, clot formation as well as on calcification were investigated. Surface characterization techniques evidenced that the coating formation was successful, and the sulfonated chitosan grafted sample exhibited a higher roughness and hydrophilicity, if compared to native chitosan one. Moreover, sulfonated surface limited platelet activation and the process of clot formation, thus confirming its high biological performances in blood. Calcium deposits were also lower on the sulfonated chitosan sample compared to the chitosan one, thus showing that calcification was minimal in presence of sulfonate groups. In conclusion, this sulfonated-modified surface has potential to be as blood-interacting material.
Collapse
Affiliation(s)
- Clayton S Campelo
- Lab Biomaterials and Bioengineering, CRC-Tier I, Department of Min-Met-Materials Engineering, Laval University & CHU de Quebec Research Center, G1V 0A6 Quebec City, QC, Canada; Federal University of Ceará, Department of Chemical Engineering, Campus do Pici - Bloco 709, Fortaleza, Ceará 60455-760, Brazil
| | - Pascale Chevallier
- Lab Biomaterials and Bioengineering, CRC-Tier I, Department of Min-Met-Materials Engineering, Laval University & CHU de Quebec Research Center, G1V 0A6 Quebec City, QC, Canada
| | - Juliana M Vaz
- Lab Biomaterials and Bioengineering, CRC-Tier I, Department of Min-Met-Materials Engineering, Laval University & CHU de Quebec Research Center, G1V 0A6 Quebec City, QC, Canada
| | - Rodrigo S Vieira
- Federal University of Ceará, Department of Chemical Engineering, Campus do Pici - Bloco 709, Fortaleza, Ceará 60455-760, Brazil.
| | - Diego Mantovani
- Lab Biomaterials and Bioengineering, CRC-Tier I, Department of Min-Met-Materials Engineering, Laval University & CHU de Quebec Research Center, G1V 0A6 Quebec City, QC, Canada.
| |
Collapse
|
13
|
Bian Z, Shi L, Guo YL, Lv Z, Tang C, Niu S, Tremblay A, Venkataramani M, Culpepper C, Li L, Zhou Z, Mansour A, Zhang Y, Gewirtz A, Kidder K, Zen K, Liu Y. Cd47-Sirpα interaction and IL-10 constrain inflammation-induced macrophage phagocytosis of healthy self-cells. Proc Natl Acad Sci U S A 2016; 113:E5434-43. [PMID: 27578867 PMCID: PMC5027463 DOI: 10.1073/pnas.1521069113] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rapid clearance of adoptively transferred Cd47-null (Cd47(-/-)) cells in congeneic WT mice suggests a critical self-recognition mechanism, in which CD47 is the ubiquitous marker of self, and its interaction with macrophage signal regulatory protein α (SIRPα) triggers inhibitory signaling through SIRPα cytoplasmic immunoreceptor tyrosine-based inhibition motifs and tyrosine phosphatase SHP-1/2. However, instead of displaying self-destruction phenotypes, Cd47(-/-) mice manifest no, or only mild, macrophage phagocytosis toward self-cells except under the nonobese diabetic background. Studying our recently established Sirpα-KO (Sirpα(-/-)) mice, as well as Cd47(-/-) mice, we reveal additional activation and inhibitory mechanisms besides the CD47-SIRPα axis dominantly controlling macrophage behavior. Sirpα(-/-) mice and Cd47(-/-) mice, although being normally healthy, develop severe anemia and splenomegaly under chronic colitis, peritonitis, cytokine treatments, and CFA-/LPS-induced inflammation, owing to splenic macrophages phagocytizing self-red blood cells. Ex vivo phagocytosis assays confirmed general inactivity of macrophages from Sirpα(-/-) or Cd47(-/-) mice toward healthy self-cells, whereas they aggressively attack toward bacteria, zymosan, apoptotic, and immune complex-bound cells; however, treating these macrophages with IL-17, LPS, IL-6, IL-1β, and TNFα, but not IFNγ, dramatically initiates potent phagocytosis toward self-cells, for which only the Cd47-Sirpα interaction restrains. Even for macrophages from WT mice, phagocytosis toward Cd47(-/-) cells does not occur without phagocytic activation. Mechanistic studies suggest a PKC-Syk-mediated signaling pathway, to which IL-10 conversely inhibits, is required for activating macrophage self-targeting, followed by phagocytosis independent of calreticulin Moreover, we identified spleen red pulp to be one specific tissue that provides stimuli constantly activating macrophage phagocytosis albeit lacking in Cd47(-/-) or Sirpα(-/-) mice.
Collapse
Affiliation(s)
- Zhen Bian
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Lei Shi
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Ya-Lan Guo
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Zhiyuan Lv
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Cong Tang
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Shuo Niu
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Alexandra Tremblay
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Mahathi Venkataramani
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Courtney Culpepper
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Limin Li
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhen Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Ahmed Mansour
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Life Science Institute (LSI) Immunology Programme, National University of Singapore, Singapore 117456
| | - Andrew Gewirtz
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303
| | - Koby Kidder
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302; Department of Cell Biology, Rutgers University, New Brunswick, NJ 08901
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yuan Liu
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302; Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303;
| |
Collapse
|
14
|
Blatt AZ, Saggu G, Kulkarni KV, Cortes C, Thurman JM, Ricklin D, Lambris JD, Valenzuela JG, Ferreira VP. Properdin-Mediated C5a Production Enhances Stable Binding of Platelets to Granulocytes in Human Whole Blood. THE JOURNAL OF IMMUNOLOGY 2016; 196:4671-4680. [PMID: 27183616 DOI: 10.4049/jimmunol.1600040] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/28/2016] [Indexed: 12/11/2022]
Abstract
Enhanced levels of platelet/granulocyte aggregates (PGAs) are found in patients suffering from many different inflammatory vascular diseases, and their formation in animal models of vascular disease is associated with increased thromboinflammation and worsened outcomes. The complement system, a part of the innate immune system, influences PGA formation, but the mechanisms for its effects are unknown. In this study, we have defined complement-mediated mechanisms that enhance PGA formation in human whole blood stimulated with thrombin receptor-activating peptide (TRAP) using ex vivo flow cytometry assays. We demonstrate that physiological properdin, a positive regulator of complement alternative pathway activity, increases PGA formation when added to TRAP-stimulated blood. All physiological properdin forms increase PGA formation, but properdin tetramers are the most efficient at increasing complement activity and PGA formation. Inhibition of endogenous properdin, either circulating in the blood or produced locally by leukocytes, impairs TRAP-mediated PGA formation to the same level as specific inhibition of either the alternative or classical pathway. Additionally, blocking the interaction of C5a with its cellular receptor prevents properdin-mediated increases in PGA formation. Adding either properdin tetramers or C5a to whole blood increases CD11b expression on granulocytes, and this increase is prevented by blockade of the C5a-C5a receptor axis. Finally, we demonstrate that the effects of properdin on PGA formation are tightly regulated by Factor H. Cumulatively, our data indicate that properdin enhances PGA formation via increased production of C5a, and that inhibition of properdin function has therapeutic potential to limit thromboinflammation in diseases characterized by increased PGA formation.
Collapse
Affiliation(s)
- Adam Z Blatt
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Gurpanna Saggu
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Koustubh V Kulkarni
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Claudio Cortes
- Department of Biomedical Sciences, Oakland University William Beaumont School of Medicine, Rochester, Michigan
| | - Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel Ricklin
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John D Lambris
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, LMVR, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
15
|
Schöttler S, Klein K, Landfester K, Mailänder V. Protein source and choice of anticoagulant decisively affect nanoparticle protein corona and cellular uptake. NANOSCALE 2016; 8:5526-36. [PMID: 26804616 DOI: 10.1039/c5nr08196c] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Protein adsorption on nanoparticles has been a focus of the field of nanocarrier research in the past few years and more and more papers are dealing with increasingly detailed lists of proteins adsorbed to a plethora of nanocarriers. While there is an urgent need to understand the influence of this protein corona on nanocarriers' interactions with cells the strong impact of the protein source on corona formation and the consequence for interaction with different cell types are factors that are regularly neglected, but should be taken into account for a meaningful analysis. In this study, the importance of the choice of protein source used for in vitro protein corona analysis is concisely investigated. Major and decisive differences in cellular uptake of a polystyrene nanoparticle incubated in fetal bovine serum, human serum, human citrate and heparin plasma are reported. Furthermore, the protein compositions are determined for coronas formed in the respective incubation media. A strong influence of heparin, which is used as an anticoagulant for plasma generation, on cell interaction is demonstrated. While heparin enhances the uptake into macrophages, it prevents internalization into HeLa cells. Taken together we can give the recommendation that human plasma anticoagulated with citrate seems to give the most relevant results for in vitro studies of nanoparticle uptake.
Collapse
Affiliation(s)
- S Schöttler
- Max Planck Institute of Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katja Klein
- Max Planck Institute of Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - K Landfester
- Max Planck Institute of Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - V Mailänder
- Max Planck Institute of Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
16
|
Yu K, Mei Y, Hadjesfandiari N, Kizhakkedathu JN. Engineering biomaterials surfaces to modulate the host response. Colloids Surf B Biointerfaces 2014; 124:69-79. [PMID: 25193153 DOI: 10.1016/j.colsurfb.2014.08.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/29/2014] [Accepted: 08/09/2014] [Indexed: 12/16/2022]
Abstract
Undesirable host response is responsible for the surface induced thrombus generation, activation of the complement system and the inflammatory reactions by the blood-contacting biomaterials. The surface interaction of biomaterials with different blood components is thought to be the critical factor that dictates the host response to biomaterials. Surface engineering can be utilized as a method to enhance the biocompatibility and tailor the biological response to biomaterials. This review provides a brief account of various polymer brush based approaches used for biomaterials surface modification, both passive and bioactive, to make the material surfaces biocompatible and antibacterial. Initially we discuss the utilization of polymer brushes with different structure and chemistry as a novel strategy to design the surface non-fouling that passively prevent the subsequent biological responses. Further we explore the utility of different bioactive agents including peptides, carbohydrates and proteins which can be conjugated the polymer brush to make the surface actively interact with the body and modulate the host response. A number of such avenues have also been explored in this review.
Collapse
Affiliation(s)
- Kai Yu
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Yan Mei
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Narges Hadjesfandiari
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada; Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6 T 1Z3, Canada.
| |
Collapse
|
17
|
Gore S, Andersson J, Biran R, Underwood C, Riesenfeld J. Heparin surfaces: Impact of immobilization chemistry on hemocompatibility and protein adsorption. J Biomed Mater Res B Appl Biomater 2014; 102:1817-24. [DOI: 10.1002/jbm.b.33154] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 02/25/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Savannah Gore
- W.L. Gore & Associates; 4100 W Kiltie Lane Flagstaff Arizona 86005
| | | | - Roy Biran
- W.L. Gore & Associates; 4100 W Kiltie Lane Flagstaff Arizona 86005
| | | | | |
Collapse
|
18
|
Han S, Huang W, Liu Y, Pan S, Feng Z, Li S. Does leukocyte-depleted blood cardioplegia reduce myocardial reperfusion injury in cardiac surgery? A systematic review and meta-analysis. Perfusion 2013; 28:474-83. [PMID: 23784875 DOI: 10.1177/0267659113492837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Blood cardioplegia in cardiac surgery contains leukocytes, which causes the inflammatory reaction and promotes myocardial reperfusion injury. The removal of leukocytes from the cardioplegia line, using specialized filters, has been proposed as one of the effective methods in attenuating the inflammatory response. We performed a two-level search to identify randomized, controlled trials concerning the effects of leukocyte-depleted blood cardioplegia on myocardial reperfusion injury. Sixteen studies, comprising 738 patients, met the selection criteria. There are significant reductions in creatinine kinase isoenzyme MB (CK-MB) during 4-8h postoperatively (SMD - 0.577; 95% CI -0.795 to -0.358; p=0.000), CK-MB peak (SMD - 0.713; 95% CI -1.027 to -0.400; p=0.000), troponin in the period of 4-8h postoperatively (SMD - 0.502; 95% CI -0.935 to -0.069; p=0.023), troponin peak (SMD - 0.826; 95% CI -1.373 to -0.279; p=0.003) and inotropic support (RR, 0.500; 95% CI 0.269 to 0.931; p=0.029). Leukocyte-depleted blood cardioplegia may reduce myocardial reperfusion injury in the early postoperative period, but there has been no evidence to support the clinically significant difference. Larger and more precise randomized control trials are needed to further elucidate the cardioprotective effects of cardioplegia leukofiltration.
Collapse
Affiliation(s)
- S Han
- 1First College of Clinical Medicine, Guangxi Medical University, Nanning, China
| | | | | | | | | | | |
Collapse
|
19
|
Targeted complement inhibition as a promising strategy for preventing inflammatory complications in hemodialysis. Immunobiology 2013; 217:1097-105. [PMID: 22964235 DOI: 10.1016/j.imbio.2012.07.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 07/17/2012] [Indexed: 01/10/2023]
Abstract
Hemodialysis is the most common method used to remove waste and hazardous products of metabolism in patients suffering from renal failure. Hundreds of thousands of people with end-stage renal disease undergo hemodialysis treatment in the United States each year. Strikingly, the 5-year survival rate for all dialysis patients is only 35%. Most of the patients succumb to cardiovascular disease that is exacerbated by the chronic induction of inflammation caused by contact of the blood with the dialysis membrane. The complement system, a strong mediator of pro-inflammatory networks, is a key contributor to such biomaterial-induced inflammation. Though only evaluated in experimental ex vivo settings, specific targeting of complement activation during hemodialysis has uncovered valuable information that points toward the therapeutic use of complement inhibitors as a means to control the unwelcomed inflammatory responses and consequent pathologies in hemodialysis patients.
Collapse
|
20
|
Johnson G, Curry B, Cahalan L, Prater R, Biggerstaff J, Hussain A, Gartner M, Cahalan P. Effects of surface-bound and intravenously administered heparin on cell-surface interactions: inflammation and coagulation. Perfusion 2013; 28:263-71. [DOI: 10.1177/0267659113475834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Intravenous administration of heparin and heparin-bonded extracorporeal circuits are frequently used to mitigate the deleterious effects of blood contact with synthetic materials. The work described here utilized human blood in a micro-perfusion circuit to experimentally examine the effects of intravenous and surface-bound heparin on cellular activation. Activation markers of coagulation and of the inflammatory response were examined using flow cytometry; specifically, markers of platelet, monocyte, polymorphonuclear leukocyte (PMN), and lymphocyte activation were quantified. The results indicate that surface-bound heparin reduces the inflammatory response whereas systemically administered heparin does not. This finding has important implications for blood-contacting devices, particularly within the context of recently elucidated connections between inflammation pathways and coagulation disorders. Data presented indicate that surface-bound heparin and intravenously administered heparin play distinct, but vital roles in rendering biomaterial surfaces compatible with blood.
Collapse
Affiliation(s)
| | - B Curry
- University of Tennessee – Knoxville, Knoxville, TN, USA
| | | | - R Prater
- University of Tennessee – Knoxville, Knoxville, TN, USA
| | - J Biggerstaff
- University of Tennessee – Knoxville, Knoxville, TN, USA
| | | | | | | |
Collapse
|
21
|
Ekdahl KN, Hong J, Hamad OA, Larsson R, Nilsson B. Evaluation of the blood compatibility of materials, cells, and tissues: basic concepts, test models, and practical guidelines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:257-70. [PMID: 23402033 DOI: 10.1007/978-1-4614-4118-2_18] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Medicine today uses a wide range of biomaterials, most of which make contact with blood permanently or transiently upon implantation. Contact between blood and nonbiological materials or cells or tissue of nonhematologic origin initiates activation of the cascade systems (complement, contact activation/coagulation) of the blood, which induces platelet and leukocyte activation. Although substantial progress regarding biocompatibility has been made, many materials and medical treatment procedures are still associated with severe side effects. Therefore, there is a great need for adequate models and guidelines for evaluating the blood compatibility of biomaterials. Due to the substantial amount of cross talk between the different cascade systems and cell populations in the blood, it is advisable to use an intact system for evaluation. Here, we describe three such in vitro models for the evaluation of the biocompatibility of materials and therapeutic cells and tissues. The use of different anticoagulants and specific inhibitors in order to be able to dissect interactions between the different cascade systems and cells of the blood is discussed. In addition, we describe two clinically relevant medical treatment modalities, the integration of titanium implants and transplantation of islets of Langerhans to patients with type 1 diabetes, whose mechanisms of action we have addressed using these in vitro models.
Collapse
Affiliation(s)
- Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory C5:3, Uppsala University, SE-751 85 Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
22
|
Sokolov A, Hellerud BC, Tønnessen TI, Johannessen EA, Mollnes TE. Activation of coagulation and platelets by candidate membranes of implantable devices in a whole blood model without soluble anticoagulant. J Biomed Mater Res A 2012; 101:575-81. [PMID: 22949225 DOI: 10.1002/jbm.a.34348] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 06/19/2012] [Accepted: 06/20/2012] [Indexed: 02/01/2023]
Abstract
Implantable devices are challenged with thrombus formation at their biomaterial interface. Thus the importance of identifying compatible biomaterials that will help to improve the performance of these devices are becoming increasingly paramount. The aim of this study was to evaluate the activation of coagulation and platelets by candidate membranes considered for use in implantable devices on the basis of an adapted whole blood model without soluble anticoagulants. Evaluated materials were incubated with whole blood without soluble anticoagulant in wells coated with heparin. Prothrombin fragment 1+2 (PTF 1+2), thrombin-antithrombin complex (TAT), and β-thromboglobulin (BTG) were analyzed in plasma samples using enzyme immunoassays. The C5 inhibitor eculizumab was used to evaluate the role of complement. Incubation of two of the polyamide membranes PAR and PATF led to an increase in concentration of PTF 1+2 and TAT (p < 0.01 for PAR, ns for PATF). The BTG concentration was significantly increased for five materials [PAR, PATF, polycarbonate (PC), and two polyarylethersulphone membranes PAES-1 and PAES-2]. Complement inhibition had no effect on coagulation or platelet activation induced by PAR and PATF. In conclusion, PAR and PATF were not compatible with blood and should be avoided for use in implantable devices.
Collapse
Affiliation(s)
- A Sokolov
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, P.O. 4950, Nydalen 0424 Oslo, Norway.
| | | | | | | | | |
Collapse
|
23
|
Walensi M, de Groot H, Schulz R, Hartmann M, Petrat F. Mesenteric ischemia-reperfusion injury: clearly improved hemodynamics but only minor protection of the rat small intestine by (sub)therapeutic heparin sodium and enoxaparin doses. J Surg Res 2012; 179:e57-69. [PMID: 22494914 DOI: 10.1016/j.jss.2012.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 11/17/2011] [Accepted: 01/03/2012] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tissue protection against ischemia (I)/reperfusion (R) injury by heparins can be due to their anticoagulant and/or non-anticoagulant properties. Here we studied the protective potential of the anticoagulant and the non-anticoagulant features of heparin sodium (HepSo) and enoxaparin (Enox) against mesenteric I/R injury in a rat model. MATERIALS AND METHODS Mesenteric I/R was induced in rats (n = 6 per group) by superior mesenteric artery occlusion (SMAO; 90 min) and reopening (120 min). Therapeutic/clinical and subtherapeutic/non-anticoagulant doses of HepSo (0.25 mg/kg bolus + 0.25 mg/kg × h; 0.05 mg/kg bolus + 0.1 mg/kg × h) or Enox (0.5 mg/kg bolus + 0.5 mg/kg × h; 0.05 mg/kg bolus + 0.1 mg/kg × h) were administered intravenously starting 30 min before SMAO to the end of reperfusion. Systemic/vital and intestinal microcirculatory parameters were measured during the whole experimental procedure, those of small intestine injury at the end. RESULTS During intestinal reperfusion, mean arterial blood pressure and heart rates were significantly increased by HepSo and, less effectively, by Enox, in a dose-dependent manner. Intestinal microcirculation was only affected by the therapeutic HepSo dose, which decreased the microvascular flow and S(O2) during reperfusion. The subtherapeutic Enox treatment, as opposed to any HepSo dose, most effectively diminished I/R-induced intestinal hemorrhages, myeloperoxidase activity (as a measure of neutrophil invasion), and histopathological changes. CONCLUSION Therapeutic but, to a lesser extent, also the subtherapeutic doses of both HepSo and Enox clearly improve hemodynamics during mesenteric reperfusion, while intestinal protection is exclusively provided by Enox, especially at its subtherapeutic dose. Alterations in intestinal microcirculation are not responsible for these effects. Thus, non-anticoagulant Enox doses and, preferably, heparin(oid)s unable to affect coagulation, could diminish clinical risks of I/R-induced gastrointestinal complications.
Collapse
Affiliation(s)
- Mikolaj Walensi
- Institut für Physiologische Chemie, Universitätsklinikum, Universität Duisburg-Essen, Essen, Germany
| | | | | | | | | |
Collapse
|
24
|
Ekdahl KN, Lambris JD, Elwing H, Ricklin D, Nilsson PH, Teramura Y, Nicholls IA, Nilsson B. Innate immunity activation on biomaterial surfaces: a mechanistic model and coping strategies. Adv Drug Deliv Rev 2011; 63:1042-50. [PMID: 21771620 PMCID: PMC3166435 DOI: 10.1016/j.addr.2011.06.012] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 06/13/2011] [Accepted: 06/25/2011] [Indexed: 01/28/2023]
Abstract
When an artificial biomaterial (e.g., a stent or implantable pump) is exposed to blood, plasma proteins immediately adhere to the surface, creating a new interface between the biomaterial and the blood. The recognition proteins within the complement and contact activation/coagulation cascade systems of the blood will be bound to, or inserted into, this protein film and generate different mediators that will activate polymorphonuclear leukocytes and monocytes, as well as platelets. Under clinical conditions, the ultimate outcome of these processes may be thrombotic and inflammatory reactions, and consequently the composition and conformation of the proteins in the initial layer formed on the surface will to a large extent determine the outcome of a treatment involving the biomaterial, affecting both the functionality of the material and the patient's life quality. This review presents models of biomaterial-induced activation processes and describes various strategies to attenuate potential adverse reactions by conjugating bioactive molecules to surfaces or by introducing nanostructures.
Collapse
Affiliation(s)
- Kristina N Ekdahl
- Dept of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, SE-751 85 Uppsala, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Sokolov A, Hellerud BC, Pharo A, Johannessen EA, Mollnes TE. Complement activation by candidate biomaterials of an implantable microfabricated medical device. J Biomed Mater Res B Appl Biomater 2011; 98:323-9. [DOI: 10.1002/jbm.b.31855] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 01/05/2011] [Accepted: 02/24/2011] [Indexed: 11/06/2022]
|
26
|
Alginate microbeads are complement compatible, in contrast to polycation containing microcapsules, as revealed in a human whole blood model. Acta Biomater 2011; 7:2566-78. [PMID: 21402181 DOI: 10.1016/j.actbio.2011.03.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 02/17/2011] [Accepted: 03/09/2011] [Indexed: 11/23/2022]
Abstract
Alginate microbeads and microcapsules are presently under evaluation for future cell-based therapy. Defining their inflammatory properties with regard to humans is therefore essential. A lepirudine-based human whole blood model was used as an inflammation predictor by measuring complement and leukocyte stimulation. Alginate microbeads were complement-compatible since they did not activate complement as measured by the soluble terminal complement complex (sTCC), Bb or the anaphylatoxins C3a and C5a. In addition, alginate microbeads were free of surface adherent leukocytes. In contrast, microcapsules containing poly-L-lysine (PLL) induced elevated levels of sTCC, Bb, C3a and C5a, surface active C3 convertase and leukocyte adhesion. The soluble PLL induced elevated levels of sTCC and up-regulated leukocyte CD11b expression. PMCG microcapsules containing poly(methylene-co-guanidine) complexed with sodium alginate and cellulose sulfate triggered a fast sTCC response and C3 deposition. The PMCG microcapsules were still less activating than PLL-containing microcapsules as a function of time. The amounts of anaphylatoxins C3a and C5a were diminished by the PMCG microcapsules, whereas leukocyte adherence demonstrated surface activating properties. We propose the whole blood model as an important tool for measuring bioincompatibility of microcapsules and microbeads for future applications as well as determining the mechanisms leading to inflammatory reactions.
Collapse
|
27
|
Hussaini BE, Treanor PR, Healey NA, Lu XG, Khuri SF, Thatte HS. Multifactorial Comparison of Modified and Conventional Perfusion Strategies in A Porcine Model of Cardiopulmonary Bypass. J Surg Res 2011; 168:e7-15. [DOI: 10.1016/j.jss.2010.01.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Revised: 01/12/2010] [Accepted: 01/21/2010] [Indexed: 11/15/2022]
|
28
|
Wu YQ, Qu H, Sfyroera G, Tzekou A, Kay BK, Nilsson B, Ekdahl KN, Ricklin D, Lambris JD. Protection of nonself surfaces from complement attack by factor H-binding peptides: implications for therapeutic medicine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:4269-77. [PMID: 21339361 PMCID: PMC3123137 DOI: 10.4049/jimmunol.1003802] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5-18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.
Collapse
Affiliation(s)
- You-Qiang Wu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Hongchang Qu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Georgia Sfyroera
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Apostolia Tzekou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Brian K. Kay
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607
| | - Bo Nilsson
- Rudbeck Laboratory C5, Section of Clinical Immunology, Department of Oncology, Radiology and Clinical Immunology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden
| | - Kristina Nilsson Ekdahl
- Rudbeck Laboratory C5, Section of Clinical Immunology, Department of Oncology, Radiology and Clinical Immunology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden
- School of Natural Sciences, Linnæus University, SE-391 82 Kalmar, Sweden
| | - Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
29
|
Bergseth G, Lambris JD, Mollnes TE, Lappegård KT. Artificial surface-induced inflammation relies on complement factor 5: proof from a deficient person. Ann Thorac Surg 2011; 91:527-33. [PMID: 21256307 PMCID: PMC3123536 DOI: 10.1016/j.athoracsur.2010.10.084] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 10/27/2010] [Accepted: 10/28/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND Exposing blood to artificial surfaces results in an inflammatory response, including complement activation and cytokine release. The aim of this investigation was to study complement-dependency and independency in artificial surface-induced inflammation in human whole blood from a patient with a genetic deficiency of complement factor 5 (C5). METHODS Whole blood from a C5-deficient patient, C5 protein reconstituted blood, and blood from a control subject was used. The complement inhibitor compstatin (C3 inhibitor) and a C5a receptor antagonist were used to block complement. Blood was circulated in closed loops of polyvinyl chloride tubing. Leukocyte CD11b expression and release of granule enzymes (myeloperoxidase, elastase, lactoferrin), cytokines (interleukins, chemokines, and growth factors; n = 27) as well as complement activation were measured after incubation. RESULTS In C5-deficient blood, there was no formation of the terminal complement complex, as opposed to reconstituted or control blood. Release of granule enzymes was partly dependent on C3, revealed by a compstatin-dependent effect in C5-deficient blood, and partly C5a-dependent as evident from the reconstitution and control blood. The chemokines interleukin-8 and monocyte chemoattractant protein-1 were also highly complement dependent, the effect being C5a-mediated, whereas platelet-derived and vascular endothelial growth factors were partly complement dependent. Interferon-γ increased in a complement-independent manner, whereas the rest of the cytokines did not respond to the surface. Leukocyte expression of CD11b was only marginally increased in deficient blood exposed to the surface, whereas reconstitution induced a considerable, C5a-dependent increase, comparable with that of the control. CONCLUSIONS The polyvinyl chloride surface induced a defined inflammatory response, which largely depended on C5.
Collapse
Affiliation(s)
- Grethe Bergseth
- Research Laboratory and Division of Internal Medicine, Nordland Hospital, Bodø, Norway.
| | | | | | | |
Collapse
|
30
|
Fleming SD, Pope MR, Hoffman SM, Moses T, Bukovnik U, Tomich JM, Wagner LM, Woods KM. Domain V peptides inhibit beta2-glycoprotein I-mediated mesenteric ischemia/reperfusion-induced tissue damage and inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:6168-78. [PMID: 20956350 PMCID: PMC3001127 DOI: 10.4049/jimmunol.1002520] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Reperfusion of ischemic tissue induces significant tissue damage in multiple conditions, including myocardial infarctions, stroke, and transplantation. Although not as common, the mortality rate of mesenteric ischemia/reperfusion (IR) remains >70%. Although complement and naturally occurring Abs are known to mediate significant damage during IR, the target Ags are intracellular molecules. We investigated the role of the serum protein, β2-glycoprotein I as an initiating Ag for Ab recognition and β2-glycoprotein I (β2-GPI) peptides as a therapeutic for mesenteric IR. The time course of β2-GPI binding to the tissue indicated binding and complement activation within 15 min postreperfusion. Treatment of wild-type mice with peptides corresponding to the lipid binding domain V of β2-GPI blocked intestinal injury and inflammation, including cellular influx and cytokine and eicosanoid production. The optimal therapeutic peptide (peptide 296) contained the lysine-rich region of domain V. In addition, damage and most inflammation were also blocked by peptide 305, which overlaps with peptide 296 but does not contain the lysine-rich, phospholipid-binding region. Importantly, peptide 296 retained efficacy after replacement of cysteine residues with serine. In addition, infusion of wild-type serum containing reduced levels of anti-β2-GPI Abs into Rag-1(-/-) mice prevented IR-induced intestinal damage and inflammation. Taken together, these data suggest that the serum protein β2-GPI initiates the IR-induced intestinal damage and inflammatory response and as such is a critical therapeutic target for IR-induced damage and inflammation.
Collapse
Affiliation(s)
- Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
In all three complement pathways, the central molecule is C3, which, upon activation cleavage, forms the major opsonin C3b - the key component of complement. C3b is also essential for propagation of the complement cascade to the stage of the lytic terminal complement complexes. In order to prevent damage to self cells and tissues and restrict overconsumption of the complement components, C3b molecules need to be controlled by factor H. Defect in C3 functions leads to compromised microbial defence and increased susceptibility to certain autoimmune diseases. Deficiency of factor H, or a functional defect in its N terminus, often leads to membranoproliferative glomerulonephritis and complement depletion, owing to continuous overconsumption of C3. Defect in the factor H C terminus leads to a dramatically increased risk of atypical hemolytic uremic syndrome. In addition, recently, a polymorphism in the middle part of factor H (Y402H) has been shown to be the major risk factor for the most common cause of blindness in the industrialized world: age-related macular degeneration. In future, analysis of patient samples for defects in these key complement components may prove useful in diagnosis of these diseases and new therapeutic targets will certainly be the aim for use in the recently recognized factor H-related diseases.
Collapse
Affiliation(s)
- T Sakari Jokiranta
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
32
|
Bioengineering of improved biomaterials coatings for extracorporeal circulation requires extended observation of blood-biomaterial interaction under flow. J Biomed Biotechnol 2010; 2007:29464. [PMID: 18317517 PMCID: PMC2246072 DOI: 10.1155/2007/29464] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 07/04/2007] [Accepted: 12/03/2007] [Indexed: 12/04/2022] Open
Abstract
Extended use of cardiopulmonary bypass (CPB) systems is often
hampered by thrombus formation and infection. Part of these
problems relates to imperfect hemocompatibility of the CPB
circuitry. The engineering of biomaterial surfaces with genuine
long-term hemocompatibility is essentially virgin territory in
biomaterials science. For example, most experiments with the
well-known Chandler loop model, for evaluation of
blood-biomaterial interactions under flow, have been described for
a maximum duration of 2 hours only. This study reports a systematic
evaluation of two commercial CPB tubings, each with a
hemocompatible coating, and one uncoated control. The experiments
comprised (i) testing over 5 hours under flow, with human whole
blood from 4 different donors; (ii) measurement of essential blood
parameters of hemocompatibility; (iii) analysis of the luminal
surfaces by scanning electron microscopy and thrombin generation
time measurements. The dataset indicated differences in
hemocompatibility of the tubings. Furthermore, it appeared that
discrimination between biomaterial coatings can be made only after
several hours of blood-biomaterial contact. Platelet counting,
myeloperoxidase quantification, and scanning electron microscopy
proved to be the most useful methods. These findings are believed
to be relevant with respect to the bioengineering of
extracorporeal devices that should function in contact with blood
for extended time.
Collapse
|
33
|
Human genetic deficiencies reveal the roles of complement in the inflammatory network: lessons from nature. Proc Natl Acad Sci U S A 2009; 106:15861-6. [PMID: 19717455 DOI: 10.1073/pnas.0903613106] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Complement component C5 is crucial for experimental animal inflammatory tissue damage; however, its involvement in human inflammation is incompletely understood. The responses to gram-negative bacteria were here studied taking advantage of human genetic complement-deficiencies--nature's own knockouts--including a previously undescribed C5 defect. Such deficiencies provide a unique tool for investigating the biological role of proteins. The experimental conditions allowed cross-talk between the different inflammatory pathways using a whole blood model based on the anticoagulant lepirudin, which does not interfere with the complement system. Expression of tissue factor, cell adhesion molecules, and oxidative burst depended highly on C5, mediated through the activation product C5a, whereas granulocyte enzyme release relied mainly on C3 and was C5a-independent. Release of cytokines and chemokines was mediated to varying degrees by complement and CD14; for example, interleukin (IL)-1beta and IL-8 were more dependent on complement than IFN-gamma and IL-6, which were highly dependent on CD14. IL-1 receptor antagonist (IL-1ra) and IFN-gamma inducible protein 10 (IP-10) were fully dependent on CD14 and inversely regulated by complement, that is, complement deficiency and complement inhibition enhanced their release. Granulocyte responses were mainly complement-dependent, whereas monocyte responses were more dependent on CD14. Notably, all responses were abolished by combined neutralization of complement and CD14. The present study provides important insight into the comprehensive role of complement in human inflammatory responses to gram-negative bacteria.
Collapse
|
34
|
Socha M, Bartecki P, Passirani C, Sapin A, Damgé C, Lecompte T, Barré J, Ghazouani FE, Maincent P. Stealth nanoparticles coated with heparin as peptide or protein carriers. J Drug Target 2009; 17:575-85. [DOI: 10.1080/10611860903112909] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- M. Socha
- Nancy-Université, Faculty of Pharmacy, Laboratory of Pharmaceutical Technology, Nancy Cedex, France
| | - P. Bartecki
- Nancy-Université, Faculty of Pharmacy, Laboratory of Pharmaceutical Technology, Nancy Cedex, France
| | - C. Passirani
- INSERM U646, Faculty of Pharmacy, Angers, France
| | - A. Sapin
- Nancy-Université, Faculty of Pharmacy, Laboratory of Pharmaceutical Technology, Nancy Cedex, France
| | - C. Damgé
- Institute of Physiology, Faculty of Medicine, Strasbourg, France
| | - T. Lecompte
- CHU de Nancy, Nancy-Université, Inserm U 961, Fédération de Recherche, Bioingéniereie Moléculaire, Cellulaire et Thérapeutique, Nancy, France
| | - J. Barré
- Unité Fonctionnelle de Pharmacologie-Toxicologie, Centre Hospitalier intercommunal, Créteil cedex, France
| | - F. El Ghazouani
- Nancy-Université, Faculty of Pharmacy, Laboratory of Pharmaceutical Technology, Nancy Cedex, France
| | - P. Maincent
- Nancy-Université, Faculty of Pharmacy, Laboratory of Pharmaceutical Technology, Nancy Cedex, France
| |
Collapse
|
35
|
Hoel TN, Thiara AS, Videm V, Fiane AE, Mollnes TE, Castellheim A, Svennevig JL. In vitro evaluation of PHISIO-coated sets for pediatric cardiac surgery. SCAND CARDIOVASC J 2009; 43:129-35. [DOI: 10.1080/14017430802302316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
36
|
De Somer F, Van Landschoot A, Van Nooten G, Delanghe J. Interaction of plasma proteins with commercial protein repellent polyvinyl chloride (PVC): a word of caution. Perfusion 2009; 23:215-21. [PMID: 19181753 DOI: 10.1177/0267659108099814] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Protein adsorption onto polymers remains a problem. In recent years, several protein-repellent PVC tubings have been developed. Although several studies report the interaction between plasma coagulation proteins and PVC, few address the interaction with other plasma proteins. Two commercial brands of untreated medical grade PVC tubing, phosphorylcholine-coated PVC tubing, triblock-copolymer (polycaprolactone-polydimethylsiloxane-polycaprolactone)-treated PVC tubing and poly-2-methoxyethylacrylate (PMEA)-coated tubing were exposed for 60 minutes to human plasma. A broad spectrum of plasma proteins was found on all tubing. The adsorbed albumin to total protein ratio is lower than the similar ratio in plasma while alpha1 and alpha2 globulins are over-represented in the protein spectrum. On PMEA tubing, not only alpha globulins, but also beta and gamma globulins, are found in high concentrations in the adsorbed protein. PMEA tubing and uncoated PVC tubing of brand B had a higher amount of protein adsorbed compared against all other tubing (p < 0.05). There were no statistical differences in protein adsorption between the triblock-copolymer-treated tubing, the phosphorylcholine-coated tubing and the uncoated PVC tubing of brand A. The average thickness of the protein layer was 23 nm. Plasma protein adsorption still exists on uncoated and protein-repellent tubing and can initiate a systemic inflammatory reaction.
Collapse
Affiliation(s)
- F De Somer
- Heart Centre, University Hospital Ghent, Belgium.
| | | | | | | |
Collapse
|
37
|
Lappegård KT, Bergseth G, Riesenfeld J, Pharo A, Magotti P, Lambris JD, Mollnes TE. The artificial surface-induced whole blood inflammatory reaction revealed by increases in a series of chemokines and growth factors is largely complement dependent. J Biomed Mater Res A 2008; 87:129-35. [PMID: 18085644 PMCID: PMC2814839 DOI: 10.1002/jbm.a.31750] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exposing blood to an artificial surface results in a systemic inflammatory response, including cytokine release and complement activation. We studied the artificial surface-induced inflammation in human whole blood using an extensive panel of inflammatory mediators including proinflammatory cytokines, chemokines and growth-factors and investigated the role of the complement system in the induction of this response. Using multiplex technology, 27 different inflammatory mediators were measured after circulating blood for 4 hours in polyvinyl chloride tubing. The C3 inhibitor compstatin was used to block complement activation. A significant (p < 0.05) increase in 14 of the 27 mediators was induced by the surface, of which 7 were chemokines (IL-8, MCP-1, MIP-1alpha, MIP-1beta, RANTES, eotaxin and IP-10) and 5 were growth-factors (G-CSF, GM-CSF, VEGF, PDGF and FGF). The traditional proinflammatory cytokines like IL-1beta, TNFalpha and IL-6 were not induced, although IL-6, as well as IL-15 and IL-17 increased if the surface was coated with highly bioincompatible laminaran. Inhibition of complement activation with compstatin significantly (p < 0.05) reduced the formation of 12 of the 14 mediators. For 10 of the 12 mediators, the inhibition was by 2/3 or more, for the remaining two the inhibition was more moderate. A highly biocompatible heparin-coated PVC surface was used as negative control and completely abolished the whole inflammatory response. The artificial surface PVC markedly induced a broad spectrum of chemokines and growth-factors, which was largely dependent on activation of complement.
Collapse
Affiliation(s)
- K T Lappegård
- Department of Medicine, Nordland Hospital, Bodø, Norway.
| | | | | | | | | | | | | |
Collapse
|
38
|
Brekke OL, Christiansen D, Fure H, Pharo A, Fung M, Riesenfeld J, Mollnes TE. Combined inhibition of complement and CD14 abolish E. coli-induced cytokine-, chemokine- and growth factor-synthesis in human whole blood. Mol Immunol 2008; 45:3804-13. [PMID: 18606453 DOI: 10.1016/j.molimm.2008.05.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 05/24/2008] [Accepted: 05/25/2008] [Indexed: 10/21/2022]
Abstract
The relative role of complement and CD14 in E. coli-induced cytokine synthesis in an in vitro human whole blood model of sepsis was examined. Fresh lepirudin-anticoagulated whole blood was incubated with E. coli for 2h. Monoclonal antibodies or a C5a receptor antagonist were used to block complement. Inflammatory mediators (n=27) were measured by multiplex technology, selected cytokine mRNA by real time PCR, and CD11b, oxidative burst and phagocytosis by flow cytometry. E. coli significantly increased 18 of the 27 inflammatory mediators, including proinflammatory cytokines (TNF-alpha, IL-6, INF-gamma and IL-1beta), chemokines (IL-8, MCP-1, MIP-1alpha, MIP-1beta, eotaxin and IP-10), growth factors (VEGF, FGF-basic, G-CSF and GM-CSF) and other interleukins (IL-9, IL-15 and IL-17). Notably, the increases in all mediators were abolished by a combined inhibition of CD14 and complement using anti-C2 and anti-factor D in combination, whereas the relative effect of the inhibition of complement and CD14 varied. In comparison, a C5a receptor antagonist and anti-CD14 in combination reduced cytokine synthesis less efficiently. Real time PCR analysis confirmed that the cytokine synthesis was blocked at the mRNA level. Similarly, E. coli-induced CD11b up-regulation, oxidative burst and phagocytosis was totally inhibited by CD14, anti-C2 and anti-factor D in combination after 2h incubation. In conclusion, the combined inhibition of complement using anti-C2, anti-factor D and CD14 almost completely inhibits the E. coli-induced inflammatory response. The combined approach may therefore be a new treatment regimen in Gram-negative sepsis.
Collapse
Affiliation(s)
- Ole-Lars Brekke
- Department of Laboratory Medicine, Nordland Hospital, Bodø, Norway.
| | | | | | | | | | | | | |
Collapse
|
39
|
Maruvada R, Blom AM, Prasadarao NV. Effects of complement regulators bound to Escherichia coli K1 and Group B Streptococcus on the interaction with host cells. Immunology 2008; 124:265-76. [PMID: 18028369 PMCID: PMC2556302 DOI: 10.1111/j.1365-2567.2007.02764.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2007] [Revised: 09/27/2007] [Accepted: 10/23/2007] [Indexed: 11/29/2022] Open
Abstract
Escherichia coli K1 and Group B Streptococcus (GBS) are the most common bacteria that cause meningitis during the neonatal period. Complement, the first line of defence in the host, acts on these bacteria to opsonize with various components of complement for subsequent presentation to phagocytes. To counteract these opsonization effects, E. coli and GBS bind to the complement regulators C4 binding protein and Factor H, respectively. Nonetheless, the deposition of complement components on these two bacteria from neonatal serum and their effect on the host cell interaction is unclear. Here we demonstrated that the deposition of complement proteins from adult serum prevented the invasion of E. coli into human brain microvascular endothelial cells, whereas the invasion of GBS was enhanced. In contrast, treatment with cord serum had no effect on the invasion of both these bacteria. We also examined the effect of the deposited complement proteins on phagocytosis using THP-1 cells and THP-1 cells differentiated into macrophages. Escherichia coli treated with adult serum neither attached nor entered these cells, whereas GBS was phagocytosed and survived efficiently. We further demonstrate that the inhibitory effect of complement proteins is the result of the bound complement inhibitors C4b-binding protein, in the case of E. coli, and Factor H, in the case of GBS. Taken together, these results suggest that E. coli and GBS utilize contrasting mechanisms of complement-mediated interactions with their target cells for successful establishment of disease.
Collapse
Affiliation(s)
- Ravi Maruvada
- Division of Infectious Diseases, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | | | | |
Collapse
|
40
|
Management of the Patient after Cardiac Surgery. Crit Care Med 2008. [DOI: 10.1016/b978-032304841-5.50039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Ricklin D, Lambris JD. Compstatin: a complement inhibitor on its way to clinical application. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 632:273-92. [PMID: 19025129 PMCID: PMC2700864 DOI: 10.1007/978-0-387-78952-1_20] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutic modulation of the human complement system is considered a promising approach for treating a number of pathological conditions. Owing to its central position in the cascade, component C3 is a particularly attractive target for complement-specific drugs. Compstatin, a cyclic tridecapeptide, which was originally discovered from phage-display libraries, is a highly potent and selective C3 inhibitor that demonstrated clinical potential in a series of experimental models. A combination of chemical, biophysical, and computational approaches allowed a remarkable optimization of its binding affinity towards C3 and its inhibitory potency. With the recent announcement of clinical trials with a compstatin analog for the treatment of age-related macular degeneration, another important milestone has been reached on its way to a drug. Furthermore, the release of a co-crystal structure of compstatin with C3c allows a detailed insight into the binding mode and paves the way to the rational design of peptides and mimetics with improved activity. Considering the new incentives and the promising pre-clinical results, compstatin seems to be well equipped for the challenges on its way to a clinical therapeutic.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA,
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA,
| |
Collapse
|
42
|
Lim HK, Anderson J, Leong JY, Pepe S, Salamonsen RF, Rosenfeldt FL. What is the Role of Leukocyte Depletion in Cardiac Surgery? Heart Lung Circ 2007; 16:243-53. [PMID: 17360235 DOI: 10.1016/j.hlc.2007.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Revised: 12/07/2006] [Accepted: 01/07/2007] [Indexed: 11/30/2022]
Abstract
Leukocytes play an important pathogenic role in ischaemia-reperfusion injury. During cardiopulmonary bypass, leukocyte filters have the potential to remove leukocytes, thereby reducing contact of activated leukocytes with the endothelium of target organs. Improvement in the safety and efficacy of commercially available leukocyte filters in recent years has led to their increasing use in cardiac surgery. However, the benefits have been inconsistent. Current evidence suggests that leukocyte depletion may not have a significant impact in low risk elective coronary artery bypass grafting but may be beneficial in valve surgery and high-risk cardiac surgery. High-risk surgical groups that may benefit from leukocyte filtration are those with left ventricular hypertrophy (LV mass>300 g), poor ejection fraction (EF<40%), chronic obstructive airways disease (predicted FEV1<75%), prolonged ischaemia (cross clamp time>120 min or cardiac transplantation), paediatric cardiac surgery and patients in cardiogenic shock requiring emergency coronary artery bypass grafting. Future trials should be powered to detect important clinical end points and be designed to avoid premature exhaustion of the filter.
Collapse
Affiliation(s)
- Hou-Kiat Lim
- Cardiac Surgical Research Unit, Alfred Hospital, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
43
|
Rinder CS, Smith MJ, Rinder HM, Cortright DN, Brodbeck RM, Krause JE, Smith BR. Leukocyte effects of C5a-receptor blockade during simulated extracorporeal circulation. Ann Thorac Surg 2007; 83:146-52. [PMID: 17184649 DOI: 10.1016/j.athoracsur.2006.08.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2006] [Revised: 08/03/2006] [Accepted: 08/04/2006] [Indexed: 12/22/2022]
Abstract
BACKGROUND Distinct pathways of leukocyte activation during simulated cardiopulmonary bypass are mediated by the complement C5a anaphylatoxin. We hypothesized that a human C5a receptor antagonist would specifically inhibit the inflammatory response of neutrophils to simulated extracorporeal circulation, while preserving the C5b-9 pathway for innate immunity. METHODS An in vitro extracorporeal circuit recirculated fresh heparinized whole blood through a membrane oxygenator with and without addition of a small molecule human C5a receptor antagonist. Samples were periodically drawn over 90 minutes for complement and leukocyte activation studies. RESULTS Addition of the C5a receptor antagonist to simulated extracorporeal circulation abrogated both neutrophil CD11b upregulation and interleukin 8 release (p < 0.01 for both), despite full generation of C3a and C5b-9; however, elastase release from neutrophils was unaffected. Although C5a receptor blockade only trended toward inhibiting monocyte CD11b upregulation (p = 0.09), circuit clearance of both monocytes (p = 0.04) and neutrophils (p = 0.01) was significantly decreased. In addition, the C5a receptor antagonist completely blocked both neutrophil-platelet and monocyte-platelet conjugate formation (p < 0.001 for both), without affecting platelet P-selectin expression. CONCLUSIONS C5a receptor blockade during simulated extracorporeal circulation completely blocked neutrophil beta2 integrin upregulation and induction of plasma interleukin 8, suggesting an acute downregulatory effect on neutrophil chemotaxis-related pathways, while preserving terminal complement generation and neutrophil elastase release. Inhibition of leukocyte-platelet conjugate formation suggests a novel function for leukocyte adhesive receptors, possibly related to preservation of elastase generation.
Collapse
Affiliation(s)
- Christine S Rinder
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut 06520-8051, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Nilsson B, Ekdahl KN, Mollnes TE, Lambris JD. The role of complement in biomaterial-induced inflammation. Mol Immunol 2006; 44:82-94. [PMID: 16905192 DOI: 10.1016/j.molimm.2006.06.020] [Citation(s) in RCA: 296] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 06/25/2006] [Accepted: 06/27/2006] [Indexed: 12/11/2022]
Abstract
Biomaterials are regularly used in various types of artificial tissues and organs, such as oxygenators, plasmapheresis equipment, hemodialysers, catheters, prostheses, stents, vascular grafts, miniature pumps, sensors and heart aids. Although progress has been made regarding bioincompatibility, many materials and procedures are associated with side effects, in particular bioincompatibility-induced inflammation, infections and subsequent loss of function. After cardiopulmonary bypass, coagulopathies can occur and lead to cognitive disturbances, stroke and extended hospitalization. Hemodialysis is associated with anaphylatoid reactions that cause whole-body inflammation and may contribute to accelerated arteriosclerosis. Stents cause restenosis and, in severe cases, thrombotic reactions. This situation indicates that there is still a need to try to understand the mechanisms involved in these incompatibility reactions in order to be able to improve the biomaterials and to develop treatments that attenuate the reactions and thereby reduce patients' discomfort, treatment time and cost. This overview deals with the role of complement in the incompatibility reactions that occur when biomaterials come in contact with blood and other body fluids.
Collapse
Affiliation(s)
- Bo Nilsson
- Department of Radiology, Oncology and Clinical Immunology, Division of Clinical Immunology, The Rudbeck Laboratory, University Hospital, Uppsala, Sweden
| | | | | | | |
Collapse
|
46
|
Sperling C, Houska M, Brynda E, Streller U, Werner C. In vitro hemocompatibility of albumin-heparin multilayer coatings on polyethersulfone prepared by the layer-by-layer technique. J Biomed Mater Res A 2006; 76:681-9. [PMID: 16302224 DOI: 10.1002/jbm.a.30519] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Polyethersulfone foils (PES)--a unique material for blood purification membranes--were coated with a multilayer assembly of heparin (unfractionated or high anticoagulant activity fraction heparin) and albumin (albumin-heparin coatings), or with a multilayer of albumin (albumin coating), using the layer-by-layer technique. The coatings combine advantages of albumin (reduction of nonspecific interactions) and heparin (specific interactions with blood coagulation proteins). The differences between the two heparins, while significant for their biological activity, had only a minor effect on the multilayer assembly with albumin monitored in situ by reflection infrared spectroscopy (FTIR MIRS). Uncoated as well as modified PES surfaces were evaluated using an in vitro assay with freshly drawn, slightly heparinized (1.5 IU heparin/mL) human whole blood. The blood was circulated with a roller pump over the sample surfaces in shear flow across rectangular slit channels ( app. 6 mL/min and 120 s(-1)) for 1.5 h at 37 degrees C. All coatings effectively reduced platelet adhesion and activation according to the PF4 release. The activation of coagulation evaluated as TAT generation was significantly lowered for the coating composed of albumin and high activity heparin. A further beneficial effect of the heparin containing coatings was reduced complement activation as determined by different complement fragments.
Collapse
Affiliation(s)
- Claudia Sperling
- Department of Biocompatible Materials, Leibniz Institute of Polymer Research Dresden and The Max Bergmann Center of Biomaterials Dresden, Hohe Str. 6, 01069 Dresden, Germany.
| | | | | | | | | |
Collapse
|
47
|
Baufreton C, Corbeau JJ, Pinaud F. [Inflammatory response and haematological disorders in cardiac surgery: toward a more physiological cardiopulmonary bypass]. ACTA ACUST UNITED AC 2006; 25:510-20. [PMID: 16488106 DOI: 10.1016/j.annfar.2005.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Accepted: 12/23/2005] [Indexed: 10/25/2022]
Abstract
The systemic inflammatory response in cardiac surgery is closely related to the haemostasis disturbances. It is responsible of a significant morbidity and mortality that was previously suspected to be caused by cardiopulmonary bypass alone. However, it is time now to clearly identify the factors that are material-dependent from that material-independent. From this point of view, off-pump surgery allowed for better comprehension of the multiple sources of the inflammatory response. Numerous pathways are activated, involving complement, platelets, neutrophiles and monocytes. The tissue pathway of the coagulation system, through tissue factor, is of major importance and has to be surgically considered in order to reduce the whole body inflammatory response postoperatively. The quality of the extracorporeal perfusion through its consequences on organ perfusion, particularly in the splanchnic area, also participates to this pathophysiological process. Beyond the progress of technology provided by the industry, particularly the minimally extracorporeal circulation derived from off-pump surgery evolution, the surgical approach is of major importance in the control of the systemic inflammatory response and must not be ignored yet.
Collapse
Affiliation(s)
- C Baufreton
- Service de Chirurgie Cardiaque et Département d'Anesthésie-Réanimation, CHU d'Angers, 4, rue Larrey, 49933 Angers cedex 09, France.
| | | | | |
Collapse
|
48
|
Asberg AE, Videm V. Activation of Neutrophil Granulocytes in an In Vitro Model of a Cardiopulmonary Bypass. Artif Organs 2005; 29:927-36. [PMID: 16305648 DOI: 10.1111/j.1525-1594.2005.00162.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Activated neutrophils play a central role in the pathogenesis of postoperative organ dysfunction after surgery with cardiopulmonary bypass. The researchers used an in vitro roller pump model to investigate the relative importance of the biomaterial, platelets, plasma proteins including activated complement, and flow mode on neutrophil activation as shown by the adhesion, degranulation, and increased the surface expression of CD11b. Neutrophil adhesion to the biomaterial increased with platelet addition, but not with plasma. Biomaterial contact activated neutrophils in a serum-free buffer, but was significantly increased by activated complement. Platelets increased neutrophil degranulation in a serum-free buffer but tended to reduce it in plasma. CD11b expression increased in both media. Complement activation was higher with neutrophils alone than with neutrophils and platelets combined. The roller pump reduced neutrophil adhesion and increased degranulation compared to passive rotation. Neutrophil interaction with platelets and complement were more important for activation than biomaterial contact and use of the roller pump. Improvement of biocompatibility is dependent on modifying complement activation and platelet interaction with neutrophils.
Collapse
Affiliation(s)
- Ann Elisabeth Asberg
- Department of Immunology and Transfusion Medicine, Trondheim University Hospital, Trondheim, Norway.
| | | |
Collapse
|
49
|
Lappegård KT, Fung M, Bergseth G, Riesenfeld J, Mollnes TE. Artificial surface-induced cytokine synthesis: effect of heparin coating and complement inhibition. Ann Thorac Surg 2005; 78:38-44; discussion 44-5. [PMID: 15223398 DOI: 10.1016/j.athoracsur.2004.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2004] [Indexed: 12/30/2022]
Abstract
BACKGROUND Contact between blood and artificial surfaces induces an inflammatory response including activation of leukocytes and platelets, as well as complement and other plasma cascade systems. In the present study we investigated the roles of complement and surface modification in polyvinylchloride-induced cytokine production. METHODS Human whole blood was incubated in rotating loops of polyvinylchloride or heparin-coated polyvinylchloride tubing for 4 hours. Plasma concentrations of the cytokines tumor necrosis factor alpha, interleukin (IL) 1 beta, IL-6, IL-8, IL-10, and monocyte chemoattractant protein 1 (MCP-1) were quantified. RESULTS Polyvinylchloride induced a substantial increase in IL-8 and MCP-1, which was abolished by cycloheximide, indicating that they were synthesized during incubation. Interleukin 8 synthesis was completely complement-dependent since it was abolished by neutralizing antibodies to factor D and complement factor 5, as well as by a complement factor 5a receptor antagonist. Monocyte chemoattractant protein 1 synthesis was reduced by approximately half the amount by the complement inhibitors. Heparin-coated polyvinylchloride efficiently prevented synthesis of both IL-8 and MCP-1. Addition of recombinant human complement factor 5a to blood incubated in heparin-coated polyvinylchloride restored IL-8 and MCP-1 production completely and partly, respectively. In contrast to IL-8 and MCP-1, tumor necrosis factor alpha, IL-1 beta, interleukin 6 and IL-10 increased only marginally. A minor but significant increase in IL-1 beta was complement-dependent, whereas a similar increase in IL-10 was completely prevented by heparin-coated polyvinylchloride. No significant changes were observed for tumor necrosis factor alpha and IL-6. CONCLUSIONS Polyvinylchloride induced a marked increase in IL-8 and MCP-1, in contrast to a marginal increase in tumor necrosis factor alpha, IL-1 beta, IL-6, and IL-10. The increase in IL-8 and MCP-1 was prevented by heparin-coated polyvinylchloride. Interleukin 8 production was totally complement-dependent and mediated by complement factor 5a.
Collapse
|
50
|
Lappegård KT, Riesenfeld J, Brekke OL, Bergseth G, Lambris JD, Mollnes TE. Differential Effect of Heparin Coating and Complement Inhibition on Artificial Surface-Induced Eicosanoid Production. Ann Thorac Surg 2005; 79:917-23. [PMID: 15734405 DOI: 10.1016/j.athoracsur.2004.08.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2004] [Indexed: 10/25/2022]
Abstract
BACKGROUND Contact between blood and artificial surfaces induces an inflammatory response including activation of leukocytes and platelets, as well as complement and other plasma cascade systems. In the present study we investigated the roles of complement and surface modification in polyvinyl chloride-induced synthesis of eicosanoids (arachidonic acid metabolites). METHODS Human whole blood was incubated in rotating loops of polyvinyl chloride or heparin-coated polyvinyl chloride tubing for 4 hours. Plasma concentrations of the eicosanoids leukotriene B4, prostaglandin E2 and thromboxane B2 were quantified. RESULTS Polyvinyl chloride induced a substantial increase in leukotriene B4, prostaglandin E2, and thromboxane B2. Inhibition of complement activation by the complement factor 3 binding peptide compstatin or blockade of the complement factor 5a receptor with a specific antagonist significantly and specifically inhibited the synthesis of leukotriene B4, whereas thromboxane B2 and prostaglandin E2 synthesis were apparently complement independent. The increase in all three mediators was significantly reduced by the heparin coating. Indomethacin abolished the increase of the cyclooxygenase products prostaglandin E2 and thromboxane B2, but had no effect on the increase of the lipoxygenase product leukotriene B4, consistent with the specificity of indomethacin for the cyclooxygenase and confirming the specificity of complement inhibition. CONCLUSIONS Polyvinyl chloride-induced increase in all three eicosanoids was attenuated by heparin coating, whereas complement inhibition selectively reduced the synthesis of leukotriene B4.
Collapse
Affiliation(s)
- Knut Tore Lappegård
- Department of Medicine, Nordland Hospital, Bodø and University of Tromsø, Tromsø, Norway.
| | | | | | | | | | | |
Collapse
|