Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Immunol Rev ; 313(1): 91-103, 2023 01.
Article in English | MEDLINE | ID: mdl-36258635

ABSTRACT

A number of clinical treatment modalities involve contact between blood and biomaterials: these include extracorporeal circuits such as hemodialysis, cardiopulmonary bypass, plasmapheresis, and intravascular treatments. Common side effects arising from these treatments are caused by activation of the cascade systems of the blood. Many of these side effects are mediated via the complement system, including thromboinflammatory reactions and rejection of implants. Depending on the composition of the materials, complement activation is triggered via all the activation pathways but is by far mostly driven by the alternative pathway amplification loop. On biomaterial surfaces the alternative pathway amplification is totally unregulated and leads under optimal conditions to deposition of complement fragments, mostly C3b, on the surface leading to a total masking of the underlying surface. In this review, we discuss the mechanism of the complement activation, clinical consequences of the activation, and potential strategies for therapeutic regulation of the activation, using hemodialysis as demonstrator.


Subject(s)
Complement Activation , Complement System Proteins , Humans , Complement Pathway, Alternative
2.
Semin Immunol ; 60: 101640, 2022 03.
Article in English | MEDLINE | ID: mdl-35853795

ABSTRACT

Patients in the intensive care unit (ICU) often straddle the divide between life and death. Understanding the complex underlying pathomechanisms relevant to such situations may help intensivists select broadly acting treatment options that can improve the outcome for these patients. As one of the most important defense mechanisms of the innate immune system, the complement system plays a crucial role in a diverse spectrum of diseases that can necessitate ICU admission. Among others, myocardial infarction, acute lung injury/acute respiratory distress syndrome (ARDS), organ failure, and sepsis are characterized by an inadequate complement response, which can potentially be addressed via promising intervention options. Often, ICU monitoring and existing treatment options rely on massive intervention strategies to maintain the function of vital organs, and these approaches can further contribute to an unbalanced complement response. Artificial surfaces of extracorporeal organ support devices, transfusion of blood products, and the application of anticoagulants can all trigger or amplify undesired complement activation. It is, therefore, worth pursuing the evaluation of complement inhibition strategies in the setting of ICU treatment. Recently, clinical studies in COVID-19-related ARDS have shown promising effects of central inhibition at the level of C3 and paved the way for prospective investigation of this approach. In this review, we highlight the fundamental and often neglected role of complement in the ICU, with a special focus on targeted complement inhibition. We will also consider complement substitution therapies to temporarily counteract a disease/treatment-related complement consumption.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Humans , Complement C3 , Prospective Studies , COVID-19/therapy , Intensive Care Units , Respiratory Distress Syndrome/therapy , Complement Activation
3.
J Immunol ; 204(10): 2779-2790, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32253242

ABSTRACT

We identified apolipoprotein E (ApoE) as one of the proteins that are found in complex with complement component C4d in pooled synovial fluid of rheumatoid arthritis (RA) patients. Immobilized human ApoE activated both the classical and the alternative complement pathways. In contrast, ApoE in solution demonstrated an isoform-dependent inhibition of hemolysis and complement deposition at the level of sC5b-9. Using electron microscopy imaging, we confirmed that ApoE interacts differently with C1q depending on its context; surface-bound ApoE predominantly bound C1q globular heads, whereas ApoE in a solution favored the hinge/stalk region of C1q. As a model for the lipidated state of ApoE in lipoprotein particles, we incorporated ApoE into phosphatidylcholine/phosphatidylethanolamine liposomes and found that the presence of ApoE on liposomes increased deposition of C1q and C4b from serum when analyzed using flow cytometry. In addition, posttranslational modifications associated with RA, such as citrullination and oxidation, reduced C4b deposition, whereas carbamylation enhanced C4b deposition on immobilized ApoE. Posttranslational modification of ApoE did not alter C1q interaction but affected binding of complement inhibitors factor H and C4b-binding protein. This suggests that changed ability of C4b to deposit on modified ApoE may play an important role. Our data show that posttranslational modifications of ApoE alter its interactions with complement. Moreover, ApoE may play different roles in the body depending on its solubility, and in diseased states such as RA, deposited ApoE may induce local complement activation rather than exert its typical role of inhibition.


Subject(s)
Apolipoproteins E/metabolism , Arthritis, Rheumatoid/immunology , Complement C1q/metabolism , Joints/immunology , Synovial Fluid/immunology , Complement Activation , Complement C4b-Binding Protein/metabolism , Complement Factor H/metabolism , Humans , Protein Binding , Protein Processing, Post-Translational , Protein-Arginine Deiminase Type 4/genetics , Protein-Arginine Deiminase Type 4/metabolism
4.
Sci Technol Adv Mater ; 23(1): 845-857, 2022.
Article in English | MEDLINE | ID: mdl-36518982

ABSTRACT

Liposome surface coating has been studied to avoid the immunological responses caused by the complement system, and alternative materials to poly(ethylene glycol) (PEG) have been explored recently since the production of anti-PEG IgM antibodies has been found in humans. We previously reported a liposome coating with poly(2-methacryloyloxyethyl phosphorylcholine) (poly(MPC))-conjugated lipids (PMPC-lipids) and demonstrated its protective effect on blood protein interactions. Here, we attempted to modify the liposome surface by exogenously adding PMPC-lipids, which were spontaneously incorporated into the outer membrane via hydrophobic interactions. The polymerization degree of the PMPC segment was regulated from 10 to 100. The incorporated ratio of PMPC-lipid increased with a decrease in the degree of PMPC polymerization. Due to surface modification with PMPC-lipids, increase in the length of the PMPC-chain increased the size of the liposomes. The modified liposomes were kept stable for 14 d in terms of their size, polydispersity, and surface properties, where approximately 70% of PMPC-lipids were incorporated into the liposome surface. We demonstrated that liposome surface modification with PMPC-lipids can inhibit protein adsorption when exposed to serum, regardless of the degree of polymerization of PMPC. In addition, the PMPC-lipid modified surface was not recognized by the anti-PEG IgM antibody, whereas PEG-lipid was recognized by the antibody. Thus, we successfully fabricated an inert liposome surface via spontaneous modification with PMPC-lipids, where only the outer bilayer surface was modified. This technique can be available for full loading of water-soluble active pharmaceutical ingredient inside the modified liposome.

5.
Langmuir ; 37(32): 9711-9723, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34342462

ABSTRACT

The use of amphiphilic molecules such as poly(ethylene glycol)-conjugated phospholipid (PEG-lipid) enables incorporation into liposome surfaces by exogenous addition as a result of the self-assembly with lipids. This technique can be applicable for manipulation of both liposomes and cells. In this study, we aimed to characterize Tat peptide (YGRKKRRQRRR)-conjugated PEG-lipids when used to exogenously surface modify liposomes (size: ca. 100 nm). We earlier reported that cells, which were surface modified with Tat peptides conjugated to PEG-lipids could attach spontaneously to material surfaces without any chemical modification. Here, we synthesized different types of Tat-PEG-lipids by combining PEG of different molecular weights (5 and 40 kDa) with different lipids with three acyl chains (myristoyl, palmitoyl, and stearoyl, respectively) and then studied the spontaneous adsorption of modified liposomes onto a substrate surface induced by the different Tat-PEG-lipids. The amount of adsorbed liposomes strongly depended on the number of incorporated Tat-PEG-lipid moieties: a decrease in both the PEG and the acyl chain lengths led to adsorption of higher amounts of liposomes. Furthermore, when a collagenase-cleavable amino acid sequence was inserted between the Tat sequence and the PEG segment, adsorbed liposomes could be harvested from the substrate by collagenase treatment with no difference in desorption efficiency between the different Tat-PEG-lipids. Thus, Tat-PEG-lipid can be a suitable tool for the manipulation of liposomes and cells.


Subject(s)
Cell-Penetrating Peptides , Liposomes , Adsorption , Humans , Phospholipids , Polyethylene Glycols
6.
Biomacromolecules ; 22(5): 1980-1989, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33813822

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) evoke great excitement for treating different human diseases due to their ability to home inflamed tissues, suppress inflammation, and promote tissue regeneration. Despite great promises, clinical trial results are disappointing as allotransplantation of MSCs trigger thrombotic activity and are damaged by the complement system, compromising their survival and function. To overcome this, a new strategy is presented by the silencing of tissue factor (TF), a transmembrane protein that mediates procoagulant activity. Novel Pluronic-based micelles are designed with the pendant pyridyl disulfide group, which are used to conjugate TF-targeting siRNA by the thiol-exchange reaction. This nanocarrier design effectively delivered the payload to MSCs resulting in ∼72% TF knockdown (KD) without significant cytotoxicity. Hematological evaluation of MSCs and TF-KD MSCs in an ex vivo human whole blood model revealed a significant reduction in an instant-blood-mediated-inflammatory reaction as evidenced by reduced platelet aggregation (93% of free platelets in the TF-KD group, compared to 22% in untreated bone marrow-derived MSCs) and thrombin-antithrombin complex formation. Effective TF silencing induced higher MSC differentiation in osteogenic and adipogenic media and showed stronger paracrine suppression of proinflammatory cytokines in macrophages and higher stimulation in the presence of endotoxins. Thus, TF silencing can produce functional cells with higher fidelity, efficacy, and functions.


Subject(s)
Mesenchymal Stem Cells , Cell Differentiation , Cells, Cultured , Humans , Micelles , Paracrine Communication , Poloxamer , Thromboplastin/genetics
7.
Rheumatology (Oxford) ; 59(11): 3264-3274, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32259250

ABSTRACT

OBJECTIVE: Complement components, including C4d, can be found on activated platelets, a process associated with vascular disease in SLE. We investigated whether platelet C4d (PC4d) adds additional value to traditional and known lupus-associated risk factors when identifying SLE patients with vascular disease. METHODS: This cross-sectional study included 308 well-characterized SLE patients and 308 matched general population controls. PC4d deposition was analysed using flow cytometry. Values >95% of controls were considered as PC4d positive (+). aPL were determined by Luminex, and the LA test was performed by DRVVT. History of vascular disease (composite and as separate outcomes) was defined at inclusion. RESULTS: SLE patients had increased PC4d deposition as compared with population controls (50 vs 5%, P < 0.0001). PC4d+ positively associated with any vascular events, and separately with venous and cerebrovascular events, and also with all investigated aPL profiles. The association for any vascular event remained statistically significant after adjustment for traditional and SLE-associated risk factors (odds ratio: 2.3, 95% CI: 1.3, 4.3, P = 0.008). Compared with patients negative for both PC4d and LA, patients with double positivity were more likely to have vascular disease (odds ratio: 12.3, 95% CI: 5.4, 29.3; attributable proportion due to interaction 0.8, 95% CI: 0.4, 1.1). CONCLUSION: PC4d+ is associated with vascular events in SLE, independently of traditional and SLE-associated risk factors. Concurrent presence of PC4d and LA seem to interact to further increase the odds for vascular events. Prospective studies should examine whether the aPL/PC4d combination can improve prediction of vascular events in SLE and/or APS.


Subject(s)
Blood Platelets/immunology , Complement C4b/analysis , Lupus Erythematosus, Systemic/immunology , Peptide Fragments/analysis , Vascular Diseases/immunology , Adult , Autoantigens/analysis , Biomarkers/analysis , Carotid Artery Diseases/diagnostic imaging , Case-Control Studies , Complement Activation , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Lupus Erythematosus, Systemic/complications , Male , Middle Aged , Odds Ratio , Ribonucleoproteins/analysis , Risk Factors , Vascular Diseases/etiology , SS-B Antigen
8.
Semin Immunol ; 28(3): 268-77, 2016 06.
Article in English | MEDLINE | ID: mdl-27211838

ABSTRACT

Therapeutic medicine today includes a vast number of procedures involving the use of biomaterials, transplantation of therapeutic cells or cell clusters, as well as of solid organs. These treatment modalities are obviously of great benefit to the patient, but also present a great challenge to the innate immune system, since they involve direct exposure of non-biological materials, cells of non-hematological origin as well as endothelial cells, damaged by ischemia-perfusion in solid organs to proteins and cells in the blood. The result of such an exposure may be an inappropriate activation of the complement and contact/kallikrein systems, which produce mediators capable of triggering the platelets and PMNs and monocytes, which can ultimately result in thrombotic and inflammatory (i.e., a thrombo-inflammatory) response to the treatment modality. In this concept review, we give an overview of the mechanisms of recognition within the innate immunity system, with the aim to identify suitable points for intervention. Finally, we discuss emerging and promising techniques for surface modification of biomaterials and cells with specific inhibitors in order to diminish thromboinflammation and improve clinical outcome.


Subject(s)
Biocompatible Materials/therapeutic use , Blood Platelets/immunology , Complement System Proteins/metabolism , Immunotherapy/methods , Inflammation/therapy , Thrombocytosis/therapy , Biocompatible Materials/adverse effects , Complement Activation , Humans , Immunity, Innate , Inflammation/immunology , Molecular Targeted Therapy , Thrombocytosis/immunology
9.
Immunol Rev ; 274(1): 245-269, 2016 11.
Article in English | MEDLINE | ID: mdl-27782319

ABSTRACT

Innate immunity is fundamental to our defense against microorganisms. Physiologically, the intravascular innate immune system acts as a purging system that identifies and removes foreign substances leading to thromboinflammatory responses, tissue remodeling, and repair. It is also a key contributor to the adverse effects observed in many diseases and therapies involving biomaterials and therapeutic cells/organs. The intravascular innate immune system consists of the cascade systems of the blood (the complement, contact, coagulation, and fibrinolytic systems), the blood cells (polymorphonuclear cells, monocytes, platelets), and the endothelial cell lining of the vessels. Activation of the intravascular innate immune system in vivo leads to thromboinflammation that can be activated by several of the system's pathways and that initiates repair after tissue damage and leads to adverse reactions in several disorders and treatment modalities. In this review, we summarize the current knowledge in the field and discuss the obstacles that exist in order to study the cross-talk between the components of the intravascular innate immune system. These include the use of purified in vitro systems, animal models and various types of anticoagulants. In order to avoid some of these obstacles we have developed specialized human whole blood models that allow investigation of the cross-talk between the various cascade systems and the blood cells. We in particular stress that platelets are involved in these interactions and that the lectin pathway of the complement system is an emerging part of innate immunity that interacts with the contact/coagulation system. Understanding the resulting thromboinflammation will allow development of new therapeutic modalities.


Subject(s)
Blood Platelets/immunology , Complement System Proteins/metabolism , Endothelial Cells/physiology , Inflammation/immunology , Thrombosis/immunology , Animals , Blood Coagulation , Homeostasis , Humans , Immunity, Innate , Kallikreins/metabolism , Kinins/metabolism
10.
Sci Technol Adv Mater ; 20(1): 688-698, 2019.
Article in English | MEDLINE | ID: mdl-31275460

ABSTRACT

In this review article, we focus on activation of the soluble components of the innate immune system triggered by nonbiological compounds and stress variances in activation due to the difference in size between nanoparticles (NPs) and larger particles or bulk material of the same chemical and physical composition. We then discuss the impact of the so-called protein corona which is formed on the surface of NPs when they come in contact with blood or other body fluids. For example, NPs which bind inert proteins, proteins which are prone to activate the contact system (e.g., factor XII), which may lead to clotting and fibrin formation or the complement system (e.g., IgG or C3), which may result in inflammation and vascular damage. Furthermore, we describe a whole blood model which we have developed to monitor activation and interaction between different components of innate immunity: blood protein cascade systems, platelets, leukocytes, cytokine generation, which are induced by NPs. Finally, we describe our own studies on innate immunity system activation induced by three fundamentally different species of NPs (two types of engineered NPs and diesel NPs) as demonstrator of the utility of an initial determination of the composition of the protein corona formed on NPs exposed to ethylenediaminetetraacetic acid (EDTA) plasma and subsequent analysis in our whole blood model.

11.
Nanomedicine ; 14(3): 735-744, 2018 04.
Article in English | MEDLINE | ID: mdl-29277639

ABSTRACT

Iron-oxide nanoparticles (NPs) generated by environmental events are likely to represent health problems. α-Fe2O3 NPs were synthesized, characterized and tested in a model for toxicity utilizing human whole blood without added anticoagulant. MALDI-TOF of the corona was performed and activation markers for plasma cascade systems (complement, contact and coagulation systems), platelet consumption and release of growth factors, MPO, and chemokine/cytokines from blood cells were analyzed. The coronas formed on the pristine α-Fe2O3 NPs contained contact system proteins and they induced massive activation of the contact (kinin/kallikrein) system, as well as thrombin generation, platelet activation, and release of two pro-angiogeneic growth factors: platelet-derived growth factor and vascular endothelial growth factor, whereas complement activation was unaffected. The α-Fe2O3 NPs exhibited a noticeable toxicity, with kinin/kallikrein activation, which may be associated with hypotension and long-term angiogenesis in vivo, with implications for cancer, arteriosclerosis and pulmonary disease.


Subject(s)
Blood Coagulation , Ferric Compounds/chemistry , Immunity, Innate/drug effects , Kallikrein-Kinin System , Metal Nanoparticles/administration & dosage , Humans , Metal Nanoparticles/chemistry , Platelet-Derived Growth Factor/metabolism , Protein Corona/metabolism , Vascular Endothelial Growth Factor A/metabolism
12.
J Immunol ; 194(7): 3305-16, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712219

ABSTRACT

The plasma protein C3 is a central element in the activation and effector functions of the complement system. A hereditary dysfunction of C3 that prevents complement activation via the alternative pathway (AP) was described previously in a Swedish family, but its genetic cause and molecular consequences have remained elusive. In this study, we provide these missing links by pinpointing the dysfunction to a point mutation in the ß-chain of C3 (c.1180T > C; p.Met(373)Thr). In the patient's plasma, AP activity was completely abolished and could only be reconstituted with the addition of normal C3. The M373T mutation was localized to the macroglobulin domain 4 of C3, which contains a binding site for the complement inhibitor compstatin and is considered critical for the interaction of C3 with the AP C3 convertase. Structural analyses suggested that the mutation disturbs the integrity of macroglobulin domain 4 and induces conformational changes that propagate into adjacent regions. Indeed, C3 M373T showed an altered binding pattern for compstatin and surface-bound C3b, and the presence of Thr(373) in either the C3 substrate or convertase-affiliated C3b impaired C3 activation and opsonization. In contrast to known gain-of-function mutations in C3, patients affected by this loss-of-function mutation did not develop familial disease, but rather showed diverse and mostly episodic symptoms. Our study therefore reveals the molecular mechanism of a relevant loss-of-function mutation in C3 and provides insight into the function of the C3 convertase, the differential involvement of C3 activity in clinical conditions, and some potential implications of therapeutic complement inhibition.


Subject(s)
Complement C3/genetics , Complement C3/immunology , Complement Pathway, Alternative , Mutation , Adult , Amino Acid Substitution , Complement Activation/genetics , Complement Activation/immunology , Complement C3/chemistry , Complement C3-C5 Convertases/metabolism , DNA Mutational Analysis , Exons , Female , Humans , Models, Molecular , Mutation, Missense , Pedigree , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs
13.
Clin Immunol ; 166-167: 89-95, 2016 05.
Article in English | MEDLINE | ID: mdl-27085202

ABSTRACT

Besides its vital role in immunity, the complement system also contributes to the shaping of the synaptic circuitry of the brain. We recently described that soluble Complement Receptor 2 (sCR2) is part of the nerve injury response in rodents. We here study CR2 in context of multiple sclerosis (MS) and explore the molecular effects of CR2 on C3 activation. Significant increases in sCR2 levels were evident in cerebrospinal fluid (CSF) from both patients with relapsing-remitting MS (n=33; 6.2ng/mL) and secondary-progressive MS (n=9; 7.0ng/mL) as compared to controls (n=18; 4.1ng/mL). Furthermore, CSF sCR2 levels correlated significantly both with CSF C3 and C1q as well as to a disease severity measure. In vitro, sCR2 inhibited the cleavage and down regulation of C3b to iC3b, suggesting that it exerts a modulatory role in complement activation downstream of C3. These results propose a novel function for CR2/sCR2 in human neuroinflammatory conditions.


Subject(s)
Complement C3/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis/immunology , Receptors, Complement 3d/immunology , Adult , Complement Activation/immunology , Complement C1q/cerebrospinal fluid , Complement C1q/immunology , Complement C3/cerebrospinal fluid , Electrophoresis, Polyacrylamide Gel , Female , Humans , Linear Models , Male , Middle Aged , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/pathology , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/pathology , Severity of Illness Index , Young Adult
15.
Adv Exp Med Biol ; 865: 211-33, 2015.
Article in English | MEDLINE | ID: mdl-26306452

ABSTRACT

The humoral barrier in transplant biology is the result of preformed donor-specific antibodies (DSAs), directed either against human leukocyte antigens (HLA) or non-HLA antigens such as blood group (ABO) molecules. The term "sensitization" applies to patients carrying these antibodies. Transplantation is widely accepted as a life-saving opportunity for patients with terminal end-organ disease. However, in sensitized patients, transplant outcome is hampered by antibody-mediated rejection (AMR) as a consequence of DSA exposure. Furthermore, sensitized patients have limited access to "matched" organs from the both living and deceased donor pool.Considering the crucial role of the complement system in the pathophysiology of AMR and the availability of complement intervention therapeutics, there is a growing interest in complement-targeting strategies. This review highlights the emerging importance of monitoring and modulation of the complement system in the context of enabling transplantation across humoral incompatibility in sensitized recipients with preformed anti-HLA or natural anti-ABO antibodies. It also discusses the significance of the complement system in the induction of accommodation and further emphasizes current and future perspectives of novel complement therapeutics.


Subject(s)
Complement Activation/drug effects , Complement Inactivating Agents/therapeutic use , Graft Rejection/prevention & control , Immunity, Humoral/drug effects , Kidney Transplantation , ABO Blood-Group System/genetics , ABO Blood-Group System/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibody Formation/drug effects , Blood Coagulation Factors/antagonists & inhibitors , Blood Coagulation Factors/genetics , Blood Coagulation Factors/immunology , Complement C1 Inhibitor Protein/therapeutic use , Complement System Proteins/drug effects , Complement System Proteins/genetics , Complement System Proteins/immunology , Desensitization, Immunologic/methods , Gene Expression , Graft Rejection/immunology , Graft Rejection/pathology , HLA Antigens/genetics , HLA Antigens/immunology , Histocompatibility Testing , Humans , Unrelated Donors
16.
Adv Exp Med Biol ; 865: 189-209, 2015.
Article in English | MEDLINE | ID: mdl-26306451

ABSTRACT

Transplantation of the pancreatic islets of Langerhans (islets) is a promising cell therapy for treating insulin-dependent type 1 diabetes mellitus. Islet transplantation is a minimally-invasive technique involving relatively simple surgery. However, after intraportal transplantation, the transplanted islets are attacked by the recipient's immune system, because they activate a number of systems, including coagulation, complement response, inflammation, immune rejection, and recurrence of autoimmune disease. We have developed a surface modification and microencapsulation technique that protects cells and islets with biomaterials and bioactive substances, which may be useful in clinical settings. This approach employs amphiphilic polymers, which can interact with lipid bilayer membranes, without increasing cell volume. Molecules attached to these polymers can protect transplanted cells and islets from attack by the host immune system. We expect that this surface modification technique will improve graft survival in clinical islet transplantation.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Graft Rejection/prevention & control , Islets of Langerhans Transplantation/methods , Islets of Langerhans/chemistry , Lipid Bilayers/chemistry , Polyethylene Glycols/chemistry , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival , Immobilized Proteins/chemistry , Immobilized Proteins/immunology , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Isogeneic , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/immunology
17.
Adv Exp Med Biol ; 865: 3-17, 2015.
Article in English | MEDLINE | ID: mdl-26306440

ABSTRACT

Thromboinflammation is primarily triggered by the humoral innate immune system, which mainly consists of the cascade systems of the blood, i.e., the complement, contact/coagulation and fibrinolytic systems. Activation of these systems subsequently induces activation of endothelial cells, leukocytes and platelets, finally resulting in thrombotic and inflammatory reactions. Such reactions are triggered by a number of medical procedures, e.g., treatment with biomaterials or drug delivery devices as well as in transplantation with cells, cell clusters or whole vascularized organs. Here, we (1) describe basic mechanisms for thromboinflammation; (2) review thromboinflammatory reactions in therapeutic medicine; and (3) discuss emerging strategies to dampen thromboinflammation.


Subject(s)
Anticoagulants/therapeutic use , Graft Rejection/prevention & control , Immunologic Factors/therapeutic use , Thrombosis/prevention & control , Tissue Transplantation , Biocompatible Materials/adverse effects , Blood Coagulation/drug effects , Blood Coagulation Factors/immunology , Blood Coagulation Factors/metabolism , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Graft Rejection/immunology , Graft Rejection/pathology , Humans , Immunity, Humoral/drug effects , Immunity, Innate/drug effects , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Leukocytes/cytology , Leukocytes/drug effects , Leukocytes/immunology , Thrombosis/immunology , Thrombosis/pathology
18.
Eur J Clin Invest ; 44(6): 587-96, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24754458

ABSTRACT

BACKGROUND: In several reports, C3 and C4 have been linked to diabetes and cardiovascular disease (CVD). Here, we investigate this link and the degree of C3 activation in elderly individuals. METHODS: In this study, C3 and C4 and the activation fragment C3a-desArg were analysed in 1016 subjects aged 70, in which blood pressure, lipid variables and fasting blood glucose were assessed. RESULTS: C3 levels were related to all the investigated classical cardiovascular risk factors and the metabolic syndrome (BMI, waist circumference, fat distribution, blood pressure, blood glucose levels, TG) except total cholesterol and LDL cholesterol in a highly significant fashion (Spearman up to 0,5; P < 0·0001). C4 and C3a-desArg were associated in the same fashion but less significantly, while the ratios C4/C3 or C3a-desArg/C3 were not, indicating that the association was not directly related to complement activation. The levels C3 and to a lesser degree C4 and C3a-desArg were associated particularly with CRP, but also with E-selectin and ICAM-1. In addition, C3 and C4 levels were shown to decline significantly in 15 female subjects enrolled in a weight-reduction programme over 4 months. CONCLUSION: A strong relation between C3, C4 and C3a-desArg levels, adipose tissue and risk factors of CVD was established. The data support that the adipose tissue produces complement components and generates initiators of inflammation, such as C3a and C5a, able to trigger a cyto/chemokine response, in proportion to the amount of adipose tissue. This corroborates the concept that complement contributes to the low-grade inflammation associated with obesity.


Subject(s)
Abdominal Fat/metabolism , Cardiovascular Diseases/etiology , Complement C3/metabolism , Complement C4/metabolism , Aged , Biomarkers/metabolism , Blood Glucose/metabolism , Cardiovascular Diseases/metabolism , Cytokines/metabolism , Female , Humans , Magnetic Resonance Imaging , Male , Metabolic Syndrome/metabolism , Obesity/complications , Obesity/metabolism , Risk Factors , Waist-Hip Ratio , Weight Loss/physiology
19.
Sci Rep ; 14(1): 15178, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987553

ABSTRACT

The evolution of endovascular therapies, particularly in the field of intracranial aneurysm treatment, has been truly remarkable and is characterized by the development of various stents. However, ischemic complications related to thrombosis or downstream emboli pose a challenge for the broader clinical application of such stents. Despite advancements in surface modification technologies, an ideal coating that fulfills all the desired requirements, including anti-thrombogenicity and swift endothelialization, has not been available. To address these issues, we investigated a new coating comprising 3-aminopropyltriethoxysilane (APTES) with both anti-thrombogenic and cell-adhesion properties. We assessed the anti-thrombogenic property of the coating using an in vitro blood loop model by evaluating the platelet count and the level of the thrombin-antithrombin (TAT) complex, and investigating thrombus formation on the surface using scanning electron microscopy (SEM). We then assessed endothelial cell adhesion on the metal surfaces. In vitro blood tests revealed that, compared to a bare stent, the coating significantly inhibited platelet reduction and thrombus formation; more human serum albumin spontaneously adhered to the coated surface to block thrombogenic activation in the blood. Cell adhesion tests also indicated a significant increase in the number of cells adhering to the APTES-coated surfaces compared to the numbers adhering to either the bare stent or the stent coated with an anti-fouling phospholipid polymer. Finally, we performed an in vivo safety test by implanting coated stents into the internal thoracic arteries and ascending pharyngeal arteries of minipigs, and subsequently assessing the health status and vessel patency of the arteries by angiography over the course of 1 week. We found that there were no adverse effects on the pigs and the vascular lumens of their vessels were well maintained in the group with APTES-coated stents. Therefore, our new coating exhibited both high anti-thrombogenicity and cell-adhesion properties, which fulfill the requirements of an implantable stent.


Subject(s)
Cell Adhesion , Coated Materials, Biocompatible , Propylamines , Silanes , Stents , Thrombosis , Silanes/chemistry , Silanes/pharmacology , Animals , Cell Adhesion/drug effects , Humans , Stents/adverse effects , Swine , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Propylamines/pharmacology , Propylamines/chemistry , Adsorption , Thrombosis/prevention & control , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL