Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Immunol ; 212(4): 645-662, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38180157

ABSTRACT

Collectin is a crucial component of the innate immune system and plays a vital role in the initial line of defense against pathogen infection. In mammals, collectin kidney 1 (CL-K1) is a soluble collectin that has recently been identified to have significant functions in host defense. However, the evolutionary origins of immune defense of CL-K1 and its mechanism in clearance of pathogenic microorganisms remain unclear, especially in early vertebrates. In this study, the Oreochromis niloticus CL-K1 (OnCL-K1) protein was purified and identified, which was capable of binding to two important pathogens of tilapia, Streptococcus agalactiae and Aeromonas hydrophila. Interestingly, OnCL-K1 exhibited direct bactericidal activity by binding to lipoteichoic acid or LPS on cell walls, disrupting the permeability and integrity of the bacterial membrane in vitro. Upon bacterial challenge, OnCL-K1 significantly inhibited the proliferation of pathogenic bacteria, reduced the inflammatory response, and improved the survival of tilapia. Further research revealed that OnCL-K1 could associate with OnMASPs to initiate and regulate the lectin complement pathway. Additionally, OnCD93 reduced the complement-mediated hemolysis by competing with OnMASPs for binding to OnCL-K1. More importantly, OnCL-K1 could facilitate phagocytosis by collaborating with cell surface CD93 in a lectin pathway-independent manner. Moreover, OnCL-K1 also promoted the formation of phagolysosomes, which degraded and killed ingested bacteria. Therefore, this study reveals the antibacterial response mechanism of CL-K1 in primitive vertebrates, including promoting complement activation, enhancing opsonophagocytosis, and killing of macrophages, as well as its internal links, all of which provide (to our knowledge) new insights into the understanding of the evolutionary origins and regulatory roles of the collectins in innate immunity.


Subject(s)
Macrophages , Opsonization , Animals , Macrophages/metabolism , Complement Activation , Kidney/metabolism , Vertebrates , Collectins/metabolism , Fish Proteins/metabolism , Mammals/metabolism
2.
Proc Natl Acad Sci U S A ; 120(15): e2217590120, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37011197

ABSTRACT

Antibodies play a central role in the immune defense against SARS-CoV-2. Emerging evidence has shown that nonneutralizing antibodies are important for immune defense through Fc-mediated effector functions. Antibody subclass is known to affect downstream Fc function. However, whether the antibody subclass plays a role in anti-SARS-CoV-2 immunity remains unclear. Here, we subclass-switched eight human IgG1 anti-spike monoclonal antibodies (mAbs) to the IgG3 subclass by exchanging their constant domains. The IgG3 mAbs exhibited altered avidities to the spike protein and more potent Fc-mediated phagocytosis and complement activation than their IgG1 counterparts. Moreover, combining mAbs into oligoclonal cocktails led to enhanced Fc- and complement receptor-mediated phagocytosis, superior to even the most potent single IgG3 mAb when compared at equivalent concentrations. Finally, in an in vivo model, we show that opsonic mAbs of both subclasses can be protective against a SARS-CoV-2 infection, despite the antibodies being nonneutralizing. Our results suggest that opsonic IgG3 oligoclonal cocktails are a promising idea to explore for therapy against SARS-CoV-2, its emerging variants, and potentially other viruses.


Subject(s)
COVID-19 , Immunoglobulin G , Humans , Opsonization , SARS-CoV-2 , Phagocytosis , Antibodies, Monoclonal/pharmacology
3.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35058363

ABSTRACT

Gram-positive organisms with their thick envelope cannot be lysed by complement alone. Nonetheless, antibody-binding on the surface can recruit complement and mark these invaders for uptake and killing by phagocytes, a process known as opsonophagocytosis. The crystallizable fragment of immunoglobulins (Fcγ) is key for complement recruitment. The cell surface of S. aureus is coated with Staphylococcal protein A (SpA). SpA captures the Fcγ domain of IgG and interferes with opsonization by anti-S. aureus antibodies. In principle, the Fcγ domain of therapeutic antibodies could be engineered to avoid the inhibitory activity of SpA. However, the SpA-binding site on Fcγ overlaps with that of the neonatal Fc receptor (FcRn), an interaction that is critical for prolonging the half-life of serum IgG. This evolutionary adaptation poses a challenge for the exploration of Fcγ mutants that can both weaken SpA-IgG interactions and retain stability. Here, we use both wild-type and transgenic human FcRn mice to identify antibodies with enhanced half-life and increased opsonophagocytic killing in models of S. aureus infection and demonstrate that antibody-based immunotherapy can be improved by modifying Fcγ. Our experiments also show that by competing for FcRn-binding, staphylococci effectively reduce the half-life of antibodies during infection. These observations may have profound impact in treating cancer, autoimmune, and asthma patients colonized or infected with S. aureus and undergoing monoclonal antibody treatment.


Subject(s)
Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Opsonization/immunology , Protein Engineering , Amino Acid Sequence , Antibody-Dependent Cell Cytotoxicity/immunology , Complement Activation , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Humans , Phagocytosis/immunology , Protein Binding , Protein Engineering/methods , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/immunology , Receptors, Fc/genetics , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology
4.
J Immunol ; 208(5): 1232-1247, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35110419

ABSTRACT

The ß protein from group B Streptococcus (GBS) is a ∼132-kDa, cell-surface exposed molecule that binds to multiple host-derived ligands, including complement factor H (FH). Many details regarding this interaction and its significance to immune evasion by GBS remain unclear. In this study, we identified a three-helix bundle domain within the C-terminal half of the B75KN region of ß as the major FH-binding determinant and determined its crystal structure at 2.5 Å resolution. Analysis of this structure suggested a role in FH binding for a loop region connecting helices α1 and α2, which we confirmed by mutagenesis and direct binding studies. Using a combination of protein cross-linking and mass spectrometry, we observed that B75KN bound to complement control protein (CCP)3 and CCP4 domains of FH. Although this binding site lies within a complement regulatory region of FH, we determined that FH bound by ß retained its decay acceleration and cofactor activities. Heterologous expression of ß by Lactococcus lactis resulted in recruitment of FH to the bacterial surface and a significant reduction of C3b deposition following exposure to human serum. Surprisingly, we found that FH binding by ß was not required for bacterial resistance to phagocytosis by neutrophils or killing of bacteria by whole human blood. However, loss of the B75KN region significantly diminished bacterial survival in both assays. Although our results show that FH recruited to the bacterial surface through a high-affinity interaction maintains key complement-regulatory functions, they raise questions about the importance of FH binding to immune evasion by GBS as a whole.


Subject(s)
Bacterial Proteins/metabolism , Immune Evasion/immunology , Membrane Proteins/metabolism , Streptococcus agalactiae/immunology , Binding Sites/physiology , Complement C3b/metabolism , Complement Factor H/metabolism , Humans , Neutrophils/immunology , Opsonization/immunology , Protein Binding/immunology , Protein Domains/genetics , Protein Domains/immunology , Streptococcal Infections/immunology , Streptococcal Infections/pathology
5.
Biochem Biophys Res Commun ; 656: 10-15, 2023 05 14.
Article in English | MEDLINE | ID: mdl-36940638

ABSTRACT

The "biological identity" of nanoparticles (NPs) is governed by a shell consisting of various biomolecules that is formed upon exposure to biological media, the so-called biomolecule corona. Consequently, supplementation of cell culture media with e.g. different sera is likely to affect interactions between cells and NPs ex-vivo, especially endocytosis. We aimed to investigate the differential impact of human and fetal-bovine serum on the endocytosis of poly (lactic-co-glycolic acid) NPs by human peripheral blood mononuclear cells via flow cytometry. Furthermore, we employed different methods to inhibit endocytosis, providing mechanistic insights. The resulting biomolecule corona was characterized via denaturing gel electrophoresis. We found profound differences between human and fetal bovine serum regarding the endocytosis of fluorescently labeled PLGA nanoparticles by different classes of human leukocytes. Uptake by B-lymphocytes was particularly sensitive. We further present evidence, that these effects are mediated by a biomolecule corona. We demonstrate to our knowledge for the first time that the complement is an important contributor to the endocytosis of non-surface-engineered PLGA-nanoparticles prepared via emulsion solvent evaporation by human immune cells. Our data demonstrates that results obtained with xenogeneic culture supplements such as fetal bovine serum may have to be interpreted with caution.


Subject(s)
Nanoparticles , Polyglycolic Acid , Humans , Polylactic Acid-Polyglycolic Acid Copolymer , Serum Albumin, Bovine , Lactic Acid , Leukocytes, Mononuclear , Opsonization , Endocytosis , Particle Size , Drug Carriers
6.
Cell Biochem Funct ; 41(8): 1031-1043, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37933222

ABSTRACT

Opsonization plays a pivotal role in hindering controlled drug release from nanoformulations due to macrophage-mediated nanoparticle destruction. While first and second-generation delivery systems, such as lipoplexes (50-150 nm) and quantum dots, hold immense potential in revolutionizing disease treatment through spatiotemporal controlled drug delivery, their therapeutic efficacy is restricted by the selective labeling of nanoparticles for uptake by reticuloendothelial system and mononuclear phagocyte system via various molecular forces, such as electrostatic, hydrophobic, and van der Waals bonds. This review article presents novel insights into surface-modification techniques utilizing macromolecule-mediated approaches, including PEGylation, di-block copolymerization, and multi-block polymerization. These techniques induce stealth properties by generating steric forces to repel micromolecular-opsonins, such as fibrinogen, thereby mitigating opsonization effects. Moreover, advanced biological methods, like cellular hitchhiking and dysopsonic protein adsorption, are highlighted for their potential to induce biological camouflage by adsorbing onto the nanoparticulate surface, leading to immune escape. These significant findings pave the way for the development of long-circulating next-generation nanoplatforms capable of delivering superior therapy to patients. Future integration of artificial intelligence-based algorithms, integrated with nanoparticle properties such as shape, size, and surface chemistry, can aid in elucidating nanoparticulate-surface morphology and predicting interactions with the immune system, providing valuable insights into the probable path of opsonization.


Subject(s)
Nanoparticles , Polyethylene Glycols , Humans , Polyethylene Glycols/chemistry , Opsonization , Artificial Intelligence , Drug Delivery Systems , Opsonin Proteins/chemistry , Opsonin Proteins/metabolism , Nanoparticles/chemistry
7.
J Nanobiotechnology ; 21(1): 376, 2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37838659

ABSTRACT

BACKGROUND: Nanoparticles represent one of the most important innovations in the medical field. Among nanocarriers, polymeric nanoparticles (PNPs) attracted much attention due to their biodegradability, biocompatibility, and capacity to increase efficacy and safety of encapsulated drugs. Another important improvement in the use of nanoparticles as delivery systems is the conjugation of a targeting agent that enables the nanoparticles to accumulate in a specific tissue. Despite these advantages, the clinical translation of therapeutic approaches based on nanoparticles is prevented by their interactions with blood proteins. In fact, the so-formed protein corona (PC) drastically alters the biological identity of the particles. Adsorbed activated proteins of the complement cascade play a pivotal role in the clearance of nanoparticles, making them more easily recognized by macrophages, leading to their rapid elimination from the bloodstream and limiting their efficacy. Since the mouse is the most used preclinical model for human disease, this work compared human and mouse PC formed on untargeted PNPs (uPNPs) and targeted PNPs (tPNPs), paying particular attention to complement activation. RESULTS: Mouse and human serum proteins adsorbed differently to PNPs. The differences in the binding of mouse complement proteins are minimal, whereas human complement components strongly distinguish the two particles. This is probably due to the human origin of the Fc portion of the antibody used as targeting agent on tPNPs. tPNPs and uPNPs mainly activate complement via the classical and alternative pathways, respectively, but this pattern did not affect their binding and internalization in macrophages and only a limited consumption of the activity of the human complement system was documented. CONCLUSIONS: The results clearly indicate the presence of complement proteins on PNPs surface but partially derived from an unspecific deposition rather than an effective complement activation. The presence of a targeting antibody favors the activation of the classical pathway, but its absence allows an increased activation of the alternative pathway. This results in similar opsonization of both PNPs and similar phagocytosis by macrophages, without an impairment of the activity of circulating complement system and, consequently, not enhancing the susceptibility to infection.


Subject(s)
Nanoparticles , Protein Corona , Humans , Mice , Animals , Opsonization , Complement System Proteins/metabolism , Antibodies , Polymers
8.
Nano Lett ; 22(16): 6516-6522, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35943299

ABSTRACT

Folic acid (FA) is one of the most widely utilized small-molecule ligands for cancer targeted drug delivery. Natural IgM was recently found to avidly absorb on the surface of FA-functionalized liposomes (FA-sLip), negatively regulating the in vivo performance by efficiently activating complement. Herein, FA-functionalized lipodiscs (FA-Disc) were constructed to successfully circumvent IgM-mediated opsonization and retained binding activity with folate receptors in vivo. The FA moiety along with the bound IgM was restricted to the highly curved rim of lipodiscs, leading to IgM incapability of presenting the membrane-bound conformation to trigger complement activation. The C1q docking, C3 binding, and C5a release were blocked and accelerated blood clearance phenomenon was mitigated of FA-Disc. FA-Disc retained folate binding activity and could effectively target folate receptor positive tumors in vivo. The present study provides a useful solution to avoid the negative regulation by IgM and achieve FA-enabled targeting by exploring disc-shaped nanocarriers.


Subject(s)
Nanoparticles , Neoplasms , Cell Line, Tumor , Drug Delivery Systems , Folic Acid/chemistry , Folic Acid/metabolism , Humans , Immunoglobulin M , Liposomes/chemistry , Opsonization
9.
J Bacteriol ; 204(1): e0018421, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34633872

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen that can cause life-threatening infections, particularly in immunocompromised individuals. The high-level virulence of S. aureus largely relies on its diverse and variable collection of virulence factors and immune evasion proteins, including the six serine protease-like proteins SplA to SplF. Spl proteins are expressed by most clinical isolates of S. aureus, but little is known about the molecular mechanisms by which these proteins modify the host's immune response for the benefit of the bacteria. Here, we identify SplB as a protease that inactivates central human complement proteins, i.e., C3, C4, and the activation fragments C3b and C4b, by preferentially cleaving their α-chains. SplB maintained its proteolytic activity in human serum, degrading C3 and C4. SplB further cleaved the components of the terminal complement pathway, C5, C6, C7, C8, and C9. In contrast, the important soluble human complement regulators factor H and C4b-binding protein (C4BP), as well as C1q, were left intact. Thereby, SplB reduced C3b-mediated opsonophagocytosis by human neutrophils as well as C5b-9 deposition on the bacterial surface. In conclusion, we identified the first physiological substrates of the S. aureus extracellular protease SplB. This enzyme inhibits all three complement pathways and blocks opsonophagocytosis. Thus, SplB can be considered a novel staphylococcal complement evasion protein. IMPORTANCE The success of bacterial pathogens in immunocompetent humans depends on the control and inactivation of host immunity. S. aureus, like many other pathogens, efficiently blocks host complement attack early in infection. Aiming to understand the role of the S. aureus-encoded orphan proteases of the Spl operon, we asked whether these proteins play a role in immune escape. We found that SplB inhibits all three complement activation pathways as well as the lytic terminal complement pathway. This blocks the opsonophagocytosis of the bacteria by neutrophils. We also clarified the molecular mechanisms: SplB cleaves the human complement proteins C3, C4, C5, C6, C7, C8, and C9 as well as factor B but not the complement inhibitors factor H and C4BP. Thus, we identify the first physiological substrates of the extracellular protease SplB of S. aureus and characterize SplB as a novel staphylococcal complement evasion protein.


Subject(s)
Bacterial Proteins/metabolism , Complement System Proteins/metabolism , Opsonization/physiology , Peptide Hydrolases/metabolism , Staphylococcus aureus/enzymology , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Humans , Peptide Hydrolases/genetics , Staphylococcus aureus/metabolism
10.
Small ; 18(14): e2106529, 2022 04.
Article in English | MEDLINE | ID: mdl-35187804

ABSTRACT

Coating nanoparticles with poly(ethylene glycol) (PEG) is widely used to achieve long-circulating properties after infusion. While PEG reduces binding of opsonins to the particle surface, immunogenic anti-PEG side-effects show that PEGylated nanoparticles are not truly "stealth" to surface active proteins. A major obstacle for understanding the complex interplay between opsonins and nanoparticles is the averaging effects of the bulk assays that are typically applied to study protein adsorption to nanoparticles. Here, a microscopy-based method for directly quantifying opsonization at the single nanoparticle level is presented. Various surface coatings are investigated on liposomes, including PEG, and show that opsonization by both antibodies and complement C3b is highly dependent on the surface chemistry. It is further demonstrated that this opsonization is heterogeneous, with opsonized and non-opsonized liposomes co-existing in the same ensemble. Surface coatings modify the percentage of opsonized liposomes and/or opsonin surface density on the liposomes, with strikingly different patterns for antibodies and complement. Thus, this assay provides mechanistic details about opsonization at the single nanoparticle level previously inaccessible to established bulk assays.


Subject(s)
Liposomes , Opsonin Proteins , Antibodies , Complement System Proteins/metabolism , Liposomes/chemistry , Opsonin Proteins/metabolism , Opsonization , Polyethylene Glycols/chemistry
11.
Fish Shellfish Immunol ; 125: 17-25, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35525410

ABSTRACT

C-type lectins (CTLs), as a member of the Ca2+-dependent carbohydrate recognition protein superfamily, play multiple roles in non-self recognition and the elimination of invading pathogens. In this study, a C-type lectin was identified and characterized from the Pacific abalone Haliotis discus hannai (designed as HdClec), and its open reading frame (ORF) encoded a polypeptide of 163 amino acids containing a typical signal peptide and only one carbohydrate-recognition domain (CRD). The deduced amino acid sequence of CRD in HdClec shared identities ranging from 22.4% to 39.8% with that of other identified CRDs of CTLs. A novel NPN motif was found in Ca2+-binding site 2 of HdClec. The mRNA transcripts of HdClec were detectable in all the examined tissues of non-stimulated abalones, with the highest expression in hepatopancreas (224.13-fold of that in gills). The expression of HdClec mRNA in hemocytes was significantly up-regulated after Vibrio harveyi challenge. Recombinant HdClec protein (rHdClec) could bind lipopolysaccharide (LPS) and peptidoglycan (PGN) in vitro in the presence of Ca2+. Coinciding with the PAMPs binding assay, rHdClec displayed broad agglutination activities towards Gram-negative bacteria V. splendidus, V. anguillarum, V. parahaemolyticus, V. harveyi, Escherichia coli, and Gram-positive bacteria Micrococcus luteus. Moreover, rHdClec could significantly elicit the chemotactic response of hemocytes in vitro. And the phagocytosis and encapsulation ability of hemocytes could be significantly enhanced by rHdClec. All these results showed that HdClec could function as pattern recognition receptors (PRRs) and further enhance the opsonization of hemocytes, which might play a crucial role in the innate immune responses of Pacific abalone.


Subject(s)
Hemocytes , Lectins, C-Type , Animals , Carbohydrates , Immunity, Innate/genetics , Opsonization , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/metabolism
12.
J Infect Dis ; 224(12): 2133-2147, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34036366

ABSTRACT

Monoclonal antibodies (mAbs) are gaining significant momentum as novel therapeutics for infections caused by antibiotic-resistant bacteria. We evaluated the mechanism by which antibacterial mAb therapy protects against Acinetobacter baumannii infections. Anticapsular mAb enhanced macrophage opsonophagocytosis and rescued mice from lethal infections by harnessing complement, macrophages, and neutrophils; however, the degree of bacterial burden did not correlate with survival. Furthermore, mAb therapy reduced proinflammatory (interleukin-1ß [IL-1ß], IL-6, tumor necrosis factor-α [TNF-α]) and anti-inflammatory (IL-10) cytokines, which correlated inversely with survival. Although disrupting IL-10 abrogated the survival advantage conferred by the mAb, IL-10-knockout mice treated with mAb could still survive if TNF-α production was suppressed directly (via anti-TNF-α neutralizing antibody) or indirectly (via macrophage depletion). Thus, even for a mAb that enhances microbial clearance via opsonophagocytosis, clinical efficacy required modulation of pro- and anti-inflammatory cytokines. These findings may inform future mAb development targeting bacteria that trigger the sepsis cascade.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter Infections/immunology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Immunomodulation , Acinetobacter Infections/microbiology , Animals , Anti-Bacterial Agents , Cytokines/blood , Cytokines/immunology , Interleukin-10 , Mice , Opsonization , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factor-alpha
13.
J Bacteriol ; 203(21): e0024421, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34460311

ABSTRACT

Melioidosis is a neglected tropical disease caused by the bacterium Burkholderia pseudomallei. The bacterium is intrinsically resistant to various antibiotics, and melioidosis is therefore difficult to treat successfully without a relapse in infection. B. pseudomallei is an intracellular pathogen and therefore, to eradicate the infection, antimicrobials must be able to access bacteria in an intracellular niche. This study assessed the ability of a panel of monoclonal antibodies (MAbs) to opsonize Burkholderia species and determine the effect that each antibody has on bacterial virulence in vitro. Murine macrophage infection assays demonstrated that monoclonal antibodies against the capsule of B. pseudomallei are opsonizing. Furthermore, one of these monoclonal antibodies reduced bacterial actin tail formation in our in vitro assays, indicating that antibodies could reduce the intracellular spread of Burkholderia thailandensis. The data presented in this paper demonstrate that monoclonal antibodies are opsonizing and can decrease bacterial actin tail formation, thus decreasing their intracellular spread. These data have informed selection of an antibody for development of an antibody-antibiotic conjugate (AAC) for melioidosis. IMPORTANCE Melioidosis is difficult to treat successfully due to the causal bacterium being resistant to many classes of antibiotics, therefore limiting available therapeutic options. New and improved therapies are urgently required to treat this disease. Here, we have investigated the potential of monoclonal antibodies to target this intracellular pathogen. We have demonstrated that monoclonal antibodies can target the bacterium, increase uptake into macrophages, and reduce actin tail formation required by the bacterium for spread between cells. Through targeting the bacterium with antibodies, we hope to disarm the pathogen, reducing the spread of infection. Ultimately, we aim to use an opsonizing antibody to deliver antibiotics intracellularly by developing an antibody-antibiotic conjugate therapeutic for melioidosis.


Subject(s)
Actins/metabolism , Antibodies, Monoclonal/immunology , Burkholderia pseudomallei/immunology , Macrophages/immunology , Macrophages/microbiology , Animals , Mice , Opsonization , RAW 264.7 Cells
14.
Glia ; 69(5): 1216-1240, 2021 05.
Article in English | MEDLINE | ID: mdl-33386754

ABSTRACT

Peripheral nerve section with subsequent disconnection of motor neuron (MN) cell bodies from their skeletal muscle targets leads to a rapid reactive response involving the recruitment and activation of microglia. In addition, the loss of afferent synapses on MNs occurs in concomitance with microglial reaction by a process described as synaptic stripping. However, the way in which postaxotomy-activated microglia adjacent to MNs are involved in synaptic removal is less defined. Here, we used confocal and electron microscopy to examine interactions between recruited microglial cells and presynaptic terminals in axotomized MNs between 1 and 15 days after sciatic nerve transection in mice. We did not observe any bulk engulfment of synaptic boutons by microglia. Instead, microglial cells internalized small membranous-vesicular fragments which originated from the acute disruption of synaptic terminals involving the activation of the necroptotic pathway. The presence of abundant extracellular vesicles in the perineuronal space after axotomy, together with the increased expression of phospho-mixed lineage kinase domain-like protein and, later, of extracellular vesicle markers, such as CD9, CD63, and flotillin, indicate that the vesicles mainly originated in synapses and were transferred to microglia. The upregulation of Rab7 and Rab10 in microglia interacting with injured MNs, indicated the activation of endocytosis. As activated microglia and synaptic boutons displayed positive C1q immunoreactivity, a complement-mediated opsonization may also contribute to microglial-mediated synaptic disruption. In addition to the relevance of our data in the context of neuroinflammation and MN disease, they should also be taken into account for understanding functional recovery after peripheral nerve injury.


Subject(s)
Peripheral Nerve Injuries , Presynaptic Terminals , Animals , Mice , Microglia , Motor Neurons , Neuroinflammatory Diseases , Opsonization , Spinal Cord
15.
Bioconjug Chem ; 32(10): 2154-2166, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34499487

ABSTRACT

Translation of intravenously administered nanomaterials to the clinic is limited due to adverse infusion reactions. While these reactions are infrequent, with up to 10% prone to experiencing infusion reactions, the reactions can be severe and life-threatening. One of the innate immune pathways, the complement activation pathway, plays a significant role in mediating this response. Nanoparticle surface properties are a relevant design feature, as they control the blood proteins the nanoparticles interact with and allow the nanoparticles to evade the immune reaction. PEGylation of nanosurfaces is critical in improving the blood circulation of nanoparticles and reducing opsonization. Our goal was to understand whether modifying the surface architecture by varying the PEG density and architecture can impact the complement response in vitro. We utilized block copolymers of poly(lactic acid)-b-poly(ethylene glycol) prepared with poly(ethylene glycol) macroinitiators of molecular weights 3400 and 5000 Da. Tracking the complement biomarker C5a, we monitored the impact of changing PEGylation of the nanoparticles. We also investigated how the changing PEG length on the nanoparticle surface impacts further strengthening the stealth properties. Lastly, we determined which cytokines change upon blood incubation with nanoparticles in vitro to understand the extent to which inflammation may occur and the crosstalk between the complement and immune responses. Increasing PEGylation reduced the generation of complement-mediated anaphylatoxin C5a in vitro, with 5000 Da PEG more effectively reducing levels of C5a generated compared to 3400 Da PEG. The insights gathered regarding the impact of PEG density and PEG chain length would be critical in developing stealth nanoparticles that do not lead to infusion reactions upon intravenous administration.


Subject(s)
Opsonization , Polyesters , Lactates , Nanoparticles , Polyethylene Glycols
16.
J Immunol Methods ; 528: 113668, 2024 May.
Article in English | MEDLINE | ID: mdl-38574804

ABSTRACT

Complement plays a critical role in the immune response toward nanomaterials. The complement attack on a foreign surface results in the deposition of C3, assembly of C3 convertases, the release of anaphylatoxins C3a and C5a, and finally, the formation of membrane attack complex C5b-9. Various technologies can measure complement activation markers in the fluid phase, but measurements of surface C3 deposition are less common. Previously, we developed an ultracentrifugation-based dot blot immunoassay (DBI) to measure the deposition of C3 and other protein corona components on nanoparticles. Here, we validate the repeatability of the DBI and its correlation with pathway-specific and common fluid phase markers. Moreover, we discuss the advantages of DBI, such as cost-effectiveness and versatility, while addressing potential limitations. This study provides insights into complement activation at the nanosurface level, offering a valuable tool for nanomedicine researchers in the field.


Subject(s)
Nanoparticles , Opsonization , Complement Activation , Complement Membrane Attack Complex/metabolism , Immunoassay , Complement C3a , Complement C5a , Complement C5
17.
J Biomol Struct Dyn ; 42(2): 687-695, 2024.
Article in English | MEDLINE | ID: mdl-36995305

ABSTRACT

Metal encapsulation delivers a straightforward strategy to improve miscellaneous nanoparticle properties and qualifies the resulting nanocomposite for exceptional application, including bioimaging, drug release, and theranostic development. Besides crucial applications, investigations associated with the nanocomposite impact on the biological media are highly relevant from a pharmacological viewpoint. Such studies can be conducted by exploring nanocomposite attributes and all aspects of their interaction with proteins existing in biofluids. Based on these aspects, the present work examines manganese-encapsulated carbonaceous nanocomposite (MnCQD) and their interaction with plasma proteins. On one side, the obtained nanocomposite has almost spherical shapes (≈12 nm in size), an appropriate composition and interesting optical properties for bioimaging applications. On another side, MnCQD quenches the fluorescence of two plasma proteins (BSA and HTF) following a static mechanism, confirming the formation of the MnCQD-BSA and MnCQD-HTF complexes. Although hydrophobic forces guide the stability of both formed complexes, MnCQD binds preferentially to BSA compared to HTF, with affinity constants differing by almost an order of magnitude. Furthermore, HTF and BSA underwent modifications in their secondary structure provoked due to contact with the nanocomposite, which also presented neglectable opsonization levels when exposed to appropriate biological media. These results highlight the MnCQD outstanding potential to be employed in diverse bioapplications.Communicated by Ramaswamy H. Sarma.


Subject(s)
Manganese , Nanocomposites , Opsonization , Fluorescence , Blood Proteins , Nanocomposites/chemistry , Serum Albumin, Bovine/chemistry , Protein Binding , Spectrometry, Fluorescence
18.
Front Cell Infect Microbiol ; 14: 1328185, 2024.
Article in English | MEDLINE | ID: mdl-38510967

ABSTRACT

Pseudomonas aeruginosa is an important human opportunistic pathogen responsible for a wide range of infections. The complement system is the main early host defense mechanism to control these infections. P. aeruginosa counteracts complement attack by binding Factor H (FH), a complement regulator that inactivates C3b, preventing the formation of the C3-convertase and complement amplification on the bacterial surface. Factor H-related proteins (FHRs) are a group of plasma proteins evolutionarily related to FH that have been postulated to interfere in this bacterial mechanism of resisting complement. Here, we show that FHR-1 binds to P. aeruginosa via the outer membrane protein OprG in a lipopolysaccharide (LPS) O antigen-dependent manner. Binding assays with purified components or with FHR-1-deficient serum supplemented with FHR-1 show that FHR-1 competes with FH for binding to P. aeruginosa. Blockage of FH binding to C3b deposited on the bacteria reduces FH-mediated cofactor activity of C3b degradation, increasing the opsonization of the bacteria and the formation of the potent chemoattractant C5a. Overall, our findings indicate that FHR-1 is a host factor that promotes complement activation, facilitating clearance of P. aeruginosa by opsonophagocytosis.


Subject(s)
Blood Proteins , Complement Factor H , Pseudomonas aeruginosa , Humans , Complement Factor H/metabolism , Pseudomonas aeruginosa/metabolism , Opsonization , Protein Binding , Complement System Proteins/metabolism , Bacteria/metabolism
19.
Vaccine ; 41(35): 5079-5084, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37455161

ABSTRACT

Low and very-low-birth-weight (V/LBW) neonates are highly susceptible to bacterial sepsis and meningitis. Bacterial infections caused by Staphylococcus aureus can be particularly dangerous for neonates and can result in high mortality and long-term disabilities.Antibody-based strategies have been attempted to protect V/LBW neonates against staphylococcal disease. However, these efforts have so far been unsuccessful. Failures were attributed to the immaturity of the neonatal immune system but did not account for the anti-opsonic activity of Staphylococcal protein A (SpA). Here we show that monoclonal antibody 3F6, which blocks SpA activity, promotes complement-dependent cell-mediated phagocytosis of S. aureus in human umbilical cord blood. A substitution in the crystallizable fragment (Fc) region of 3F6 that enhances recruitment of complement component C1q further increases the phagocytic activity of cord blood. Our data demonstrate that the neonatal immune system possesses bactericidal activity that can be harnessed by antibodies that circumvent a key innate immune strategy of S. aureus.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Infant, Newborn , Humans , Staphylococcal Protein A/metabolism , Fetal Blood , Opsonization , Antibodies, Bacterial , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal
20.
Microbiol Spectr ; 11(1): e0327922, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36651756

ABSTRACT

Nontuberculous mycobacteria (NTM), including Mycobacterium avium, are clinically important pathogens in cystic fibrosis (CF). The innate immune response to M. avium remains incompletely understood. We evaluated the role of complement opsonization in neutrophil-mediated killing of M. avium. Killing assays were performed using neutrophils from healthy donors (HDs) and persons with CF (pwCF). Clinical isolates of M. avium were opsonized with plasma from HDs or pwCF, which was intact or heat-treated to inactivate complement. HD neutrophils had killing activity against M. avium opsonized with intact HD plasma and killing was significantly reduced when M. avium was opsonized with heat-inactivated HD plasma. When opsonized with HD plasma, CF neutrophils had killing activity against M. avium that was not different than HD neutrophils. When opsonized with intact plasma from pwCF, HD neutrophil killing of M. avium was significantly reduced. Opsonization of M. avium with C3-depleted serum or IgM-depleted plasma resulted in significantly reduced killing. Plasma C3 levels were elevated in pwCF with NTM infection compared to pwCF without NTM infection. These studies demonstrate that human neutrophils efficiently kill M. avium when opsonized in the presence of plasma factors from HD that include C3 and IgM. Killing efficiency is significantly lower when the bacteria are opsonized with plasma from pwCF. This indicates a novel role for opsonization in neutrophil killing of M. avium and a deficiency in complement opsonization as a mechanism of impaired M. avium killing in CF. IMPORTANCE Mycobacterium avium is a member of a group of bacterial species termed nontuberculous mycobacteria (NTM) that cause lung disease in certain populations, including persons with cystic fibrosis (CF). NTM infections are challenging to diagnose and can be even more difficult to treat. This study investigated how the immune system responds to M. avium infection in CF. We found that neutrophils, the most abundant immune cell in the lungs in CF, can effectively kill M. avium in individuals both with and without CF. Another component of the immune response called the complement system is also required for this process. Levels of complement proteins are altered in persons with CF who have a history of NTM compared to those without a history of NTM infection. These results add to our understanding of how the immune system responds to M. avium, which can help pave the way toward better diagnostic and treatment strategies.


Subject(s)
Cystic Fibrosis , Mycobacterium Infections, Nontuberculous , Humans , Cystic Fibrosis/microbiology , Neutrophils , Mycobacterium avium , Opsonization , Mycobacterium Infections, Nontuberculous/microbiology , Nontuberculous Mycobacteria , Complement System Proteins , Immunoglobulin M
SELECTION OF CITATIONS
SEARCH DETAIL