Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.626
Filter
1.
Front Cell Infect Microbiol ; 14: 1400068, 2024.
Article in English | MEDLINE | ID: mdl-39310788

ABSTRACT

Complement C3 (C3) is usually deposited spontaneously on the surfaces of invading bacteria prior to internalization, but the impact of C3 coating on cellular responses is largely unknown. Staphylococcus aureus (S. aureus) is a facultative intracellular pathogen that subverts autophagy and replicates in both phagocytic and nonphagocytic cells. In the present study, we deposited C3 components on the surface of S. aureus by complement opsonization before cell infection and confirmed that C3-coatings remained on the surface of the bacteria after they have invaded the cells, suggesting S. aureus cannot escape or degrade C3 labeling. We found that the C3 deposition on S. aureus notably enhanced cellular autophagic responses, and distinguished these responses as xenophagy, in contrast to LC3-associated phagocytosis (LAP). Furthermore, this upregulation was due to the recruitment of and direct interaction with autophagy-related 16-like 1 (ATG16L1), thereby resulting in autophagy-dependent resistance to bacterial growth within cells. Interestingly, this autophagic effect occurred only after C3 activation by enzymatic cleavage because full-length C3 without cleavage of the complement cascade reaction, although capable of binding to ATG16L1, failed to promote autophagy. These findings demonstrate the biological function of intracellular C3 upon bacterial infection in enhancing autophagy against internalized S. aureus.


Subject(s)
Autophagy , Complement C3 , Phagocytosis , Staphylococcal Infections , Staphylococcus aureus , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology , Complement C3/metabolism , Humans , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Animals , Host-Pathogen Interactions , Mice , Opsonization , Complement Activation
2.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273426

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare and potentially life-threatening hematologic disorder caused by a somatic mutation in a relevant portion of hematopoietic stem cells. Mutation of the phosphatidylinositol glycan biosynthesis class A (PIGA) gene prevents the expression of cell-surface proteins, including the complement regulatory proteins CD55 and CD59. With decreased or a lack of CD55 and CD59 expression on their membranes, PNH red blood cells become susceptible to complement-mediated hemolysis (symptoms of which include anemia, dysphagia, abdominal pain, and fatigue), leading to thrombosis. State-of-the-art PNH treatments act by inhibiting the dysregulated complement at distinct points in the activation pathway: late at the C5 level (C5 inhibitors, eculizumab, ravulizumab, and crovalimab), centrally at the C3 level (C3/C3b inhibitors and pegcetacoplan), and early at the initiation and amplification of the alternative pathway (factor B inhibitor, iptacopan; factor D inhibitor, danicopan). Through their differing mechanisms of action, these treatments elicit varying profiles of disease control and offer valuable insights into the molecular underpinnings of PNH. This narrative review provides an overview of the mechanisms of action of the six complement inhibitors currently approved for PNH, with a focus on the C3/C3b-targeted therapy, pegcetacoplan.


Subject(s)
Complement Inactivating Agents , Hemoglobinuria, Paroxysmal , Humans , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/metabolism , Complement Inactivating Agents/therapeutic use , CD59 Antigens/metabolism , CD59 Antigens/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Complement Activation/drug effects , CD55 Antigens/metabolism , CD55 Antigens/genetics
3.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273570

ABSTRACT

The complement system and neutrophils play crucial roles in innate immunity. Neutrophils release neutrophil extracellular traps (NETs), which are composed of decondensed DNA entangled with granular contents, as part of their innate immune function. Mechanisms governing complement-mediated NET formation remain unclear. In this study, we tested a two-step NETosis mechanism, as follows: classical complement-mediated neutrophil activation in serum and subsequent NET formation in serum-free conditions, using neutrophils from healthy donors, endothelial cells, and various assays (Fluo-4AM, DHR123, and SYTOX), along with flow cytometry and confocal microscopy. Our findings reveal that classical complement activation on neutrophils upregulated the membrane-anchored complement regulators CD46, CD55, and CD59. Additionally, complement activation increased CD11b on neutrophils, signifying activation and promoting their attachment to endothelial cells. Complement activation induced calcium influx and citrullination of histone 3 (CitH3) in neutrophils. However, CitH3 formation alone was insufficient for NET generation. Importantly, NET formation occurred only when neutrophils were in serum-free conditions. In such environments, neutrophils induced NADPH oxidase-dependent reactive oxygen species (ROS) production, leading to NET formation. Hence, we propose that complement-mediated NET formation involves a two-step process, as follows: complement deposition, neutrophil priming, calcium influx, CitH3 formation, and attachment to endothelial cells in serum. This is followed by NADPH-dependent ROS production and NET completion in serum-free conditions. Understanding this process may unveil treatment targets for pathologies involving complement activation and NET formation.


Subject(s)
Calcium , Complement Activation , Extracellular Traps , NADPH Oxidases , Neutrophil Activation , Neutrophils , Reactive Oxygen Species , Extracellular Traps/metabolism , Humans , Neutrophils/metabolism , Neutrophils/immunology , NADPH Oxidases/metabolism , Calcium/metabolism , Reactive Oxygen Species/metabolism , Complement System Proteins/metabolism , Endothelial Cells/metabolism , Culture Media, Serum-Free/pharmacology , Histones/metabolism
4.
Nephrology (Carlton) ; 29 Suppl 2: 44-46, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39327741

ABSTRACT

The role of complement in the pathogenesis of IgA nephropathy has been heavily explored over the past 50 years. This has led to the general acceptance that complement plays an important role in the clinical presentation and risk for progression of disease in patients with IgA nephropathy. Herein, we review the evidence for complement activation in IgA nephropathy, focusing on evidence that the lectin and alternate pathways are the main actors. We are entering an era of intense investigation of various inhibitors of complement, which should ultimately be the best indicator of contributions of the lectin, alternate and common complement pathways to disease burden. More importantly, we will see if these efforts result in the discovery of clinically relevant options in managing this important disease.


Subject(s)
Complement Activation , Complement Inactivating Agents , Glomerulonephritis, IGA , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/drug therapy , Humans , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology , Complement Activation/drug effects , Complement System Proteins/immunology , Complement System Proteins/metabolism , Animals , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/immunology
6.
Front Immunol ; 15: 1449003, 2024.
Article in English | MEDLINE | ID: mdl-39295874

ABSTRACT

Pseudomonas aeruginosa is a leading cause of nosocomial bloodstream infections. The outcome of these infections depends on the virulence of the microorganism as well as host-related conditions and factors. The complement system plays a crucial role in defense against bloodstream infections. P. aeruginosa counteracts complement attack by recruiting Factor H (FH) that inhibits 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 this bacterial evasion mechanism. In this study, we demonstrate that FHR-3 competes with purified FH for binding to P. aeruginosa and identify EF-Tu as a common bacterial target for both complement regulator factors. Importantly, elevated levels of FHR-3 in human serum promote complement activation, leading to increased opsonization and killing of P. aeruginosa. Conversely, physiological concentrations of FHR-3 have no significant effect. Our findings suggest that FHR-3 may serve as a protective host factor against P. aeruginosa infections.


Subject(s)
Complement Factor H , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/immunology , Humans , Pseudomonas Infections/immunology , Complement Factor H/metabolism , Complement Factor H/immunology , Bacteremia/immunology , Bacteremia/microbiology , Complement Activation/immunology , Host-Pathogen Interactions/immunology , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Protein Binding
7.
Immunity ; 57(9): 2140-2156.e10, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39226900

ABSTRACT

Venous thromboembolism (VTE) is a common, deadly disease with an increasing incidence despite preventive efforts. Clinical observations have associated elevated antibody concentrations or antibody-based therapies with thrombotic events. However, how antibodies contribute to thrombosis is unknown. Here, we show that reduced blood flow enabled immunoglobulin M (IgM) to bind to FcµR and the polymeric immunoglobulin receptor (pIgR), initiating endothelial activation and platelet recruitment. Subsequently, the procoagulant surface of activated platelets accommodated antigen- and FcγR-independent IgG deposition. This leads to classical complement activation, setting in motion a prothrombotic vicious circle. Key elements of this mechanism were present in humans in the setting of venous stasis as well as in the dysregulated immunothrombosis of COVID-19. This antibody-driven thrombosis can be prevented by pharmacologically targeting complement. Hence, our results uncover antibodies as previously unrecognized central regulators of thrombosis. These findings carry relevance for therapeutic application of antibodies and open innovative avenues to target thrombosis without compromising hemostasis.


Subject(s)
Blood Platelets , COVID-19 , Complement Activation , Immunoglobulin M , Thrombosis , Humans , Thrombosis/immunology , Animals , Immunoglobulin M/immunology , Complement Activation/immunology , Mice , Blood Platelets/immunology , Blood Platelets/metabolism , COVID-19/immunology , COVID-19/complications , SARS-CoV-2/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Platelet Activation/immunology , Immunoglobulin G/immunology , Male
8.
Allergy Asthma Proc ; 45(5): 305-309, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39294906

ABSTRACT

The complement system is an important component of innate and adaptive immunity that consists of three activation pathways. The classic complement pathway plays a role in humoral immunity, whereas the alternative and lectin pathways augment the innate response. Impairment, deficiency, or overactivation of any of the known 50 complement proteins may lead to increased susceptibility to infection with encapsulated organisms, autoimmunity, hereditary angioedema, or thrombosis, depending on the affected protein. Classic pathway defects result from deficiencies of complement proteins C1q, C1r, C1s, C2, and C4, and typically manifest with features of systemic lupus erythematosus and infections with encapsulated organisms. Alternative pathway defects due to deficiencies of factor B, factor D, and properdin may present with increased susceptibility to Neisseria infections. Lectin pathway defects, including Mannose-binding protein-associated serine protease 2 (MASP2) and ficolin 3, may be asymptomatic or lead to pyogenic infections and autoimmunity. Complement protein C3 is common to all pathways, deficiency of which predisposes patients to severe frequent infections and glomerulonephritis. Deficiencies in factor H and factor I, which regulate the alternative pathway, may lead to hemolytic uremic syndrome. Disseminated Neisseria infections result from terminal pathway defects (i.e., C5, C6, C7, C8, and C9). Diagnosis of complement deficiencies involves screening with functional assays (i.e., total complement activity [CH50], alternative complement pathway activity [AH50], enzyme-linked immunosorbent assay [ELISA]) followed by measurement of individual complement factors by immunoassay. Management of complement deficiencies requires a comprehensive and individualized approach with special attention to vaccination against encapsulated bacteria, consideration of prophylactic antibiotics, treatment of comorbid autoimmunity, and close surveillance.


Subject(s)
Complement System Proteins , Immunologic Deficiency Syndromes , Humans , Complement System Proteins/immunology , Complement System Proteins/metabolism , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/immunology , Complement Activation
9.
Proc Natl Acad Sci U S A ; 121(37): e2404542121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39240968

ABSTRACT

Human C-reactive protein (CRP) is a pentameric complex involved in immune defense and regulation of autoimmunity. CRP is also a therapeutic target, with both administration and depletion of serum CRP being pursued as a possible treatment for autoimmune and cardiovascular diseases, among others. CRP binds to phosphocholine (PC) moieties on membranes to activate the complement system via the C1 complex, but it is unknown how CRP, or any pentraxin, binds to C1. Here, we present a cryoelectron tomography (cryoET)-derived structure of CRP bound to PC ligands and the C1 complex. To gain control of CRP binding, a synthetic mimotope of PC was synthesized and used to decorate cell-mimetic liposome surfaces. Structure-guided mutagenesis of CRP yielded a fully active complex able to bind PC-coated liposomes that was ideal for cryoET and subtomogram averaging. In contrast to antibodies, which form Fc-mediated hexameric platforms to bind and activate the C1 complex, CRP formed rectangular platforms assembled from four laterally associated CRP pentamers that bind only four of the six available globular C1 head groups. Potential residues mediating lateral association of CRP were identified from interactions between unit cells in existing crystal structures, which rationalized previously unexplained mutagenesis data regarding CRP-mediated complement activation. The structure also enabled interpretation of existing biochemical data regarding interactions mediating C1 binding and identified additional residues for further mutagenesis studies. These structural data therefore provide a possible mechanism for regulation of complement by CRP, which limits complement progression and has consequences for how the innate immune system influences autoimmunity.


Subject(s)
C-Reactive Protein , Humans , C-Reactive Protein/chemistry , C-Reactive Protein/metabolism , C-Reactive Protein/immunology , Complement Activation , Complement C1/metabolism , Complement C1/chemistry , Complement Pathway, Classical/immunology , Cryoelectron Microscopy , Liposomes/metabolism , Liposomes/chemistry , Models, Molecular , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Protein Binding
10.
Cell Mol Life Sci ; 81(1): 405, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39284944

ABSTRACT

Emerging evidence indicates that activation of complement system leading to the formation of the membrane attack complex (MAC) plays a detrimental role in COVID-19. However, their pathogenic roles have never been experimentally investigated before. We used three knock out mice strains (1. C3-/-; 2. C7-/-; and 3. Cd59ab-/-) to evaluate the role of complement in severe COVID-19 pathogenesis. C3 deficient mice lack a key common component of all three complement activation pathways and are unable to generate C3 and C5 convertases. C7 deficient mice lack a complement protein needed for MAC formation. Cd59ab deficient mice lack an important inhibitor of MAC formation. We also used anti-C5 antibody to block and evaluate the therapeutic potential of inhibiting MAC formation. We demonstrate that inhibition of complement activation (in C3-/-) and MAC formation (in C3-/-. C7-/-, and anti-C5 antibody) attenuates severe COVID-19; whereas enhancement of MAC formation (Cd59ab-/-) accelerates severe COVID-19. The degree of MAC but not C3 deposits in the lungs of C3-/-, C7-/- mice, and Cd59ab-/- mice as compared to their control mice is associated with the attenuation or acceleration of SARS-CoV-2-induced disease. Further, the lack of terminal complement activation for the formation of MAC in C7 deficient mice protects endothelial dysfunction, which is associated with the attenuation of diseases and pathologic changes. Our results demonstrated the causative effect of MAC in severe COVID-19 and indicate a potential avenue for modulating the complement system and MAC formation in the treatment of severe COVID-19.


Subject(s)
CD59 Antigens , COVID-19 , Complement Activation , Complement Membrane Attack Complex , Mice, Knockout , SARS-CoV-2 , Animals , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Complement Activation/immunology , Complement Membrane Attack Complex/metabolism , Complement Membrane Attack Complex/immunology , Mice , SARS-CoV-2/immunology , CD59 Antigens/metabolism , CD59 Antigens/genetics , CD59 Antigens/immunology , Complement C3/immunology , Complement C3/metabolism , Complement C3/genetics , Mice, Inbred C57BL , Humans , Complement C5/immunology , Complement C5/metabolism , Complement C5/antagonists & inhibitors , Disease Models, Animal
11.
Nat Commun ; 15(1): 8100, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285158

ABSTRACT

Antibody-dependent complement activation plays a key role in the natural human immune response to infections. Currently, the understanding of which antibody-antigen combinations drive a potent complement response on bacteria is limited. Here, we develop an antigen-agnostic approach to stain and single-cell sort human IgG memory B cells recognizing intact bacterial cells, keeping surface antigens in their natural context. With this method we successfully identified 29 antibodies against K. pneumoniae, a dominant cause of hospital-acquired infections with increasing antibiotic resistance. Combining genetic tools and functional analyses, we reveal that the capacity of antibodies to activate complement on K. pneumoniae critically depends on their antigenic target. Furthermore, we find that antibody combinations can synergistically activate complement on K. pneumoniae by strengthening each other's binding in an Fc-independent manner. Understanding the molecular basis of effective complement activation by antibody combinations to mimic a polyclonal response could accelerate the development of antibody-based therapies against problematic infections.


Subject(s)
Antibodies, Bacterial , Complement Activation , Immunoglobulin G , Klebsiella pneumoniae , Humans , Complement Activation/immunology , Antibodies, Bacterial/immunology , Klebsiella pneumoniae/immunology , Immunoglobulin G/immunology , B-Lymphocytes/immunology , Memory B Cells/immunology
12.
Sci Rep ; 14(1): 19311, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164435

ABSTRACT

Autoimmune related kidney diseases (ARKDs), including minimal change nephropathy (MCN), membranous nephropathy (MN), IgA nephropathy (IgAN), and lupus nephritis (LN), significantly affect renal function. These diseases are characterized by the formation of local immune complexes and the subsequent activation of the complement system, leading to kidney damage and proteinuria. Despite the known patterns of glomerular injury, the specific molecular mechanisms that contribute to renal tubular damage across ARKDs remain underexplored. Laser capture microdissection and liquid chromatography-tandem mass spectrometry (LC-MS/MS) were used to conduct a comparative proteomic analysis of renal tubular tissues from formalin-fixed paraffin-embedded samples. The cohort comprised of 10 normal controls (NC), 5 MCN, 4 MN, 17 IgAN, and 21 LN patients. Clinical parameters and histopathological assessments were integrated with proteomic findings to comprehensively investigate underlying pathogenic processes. Clinical evaluation indicated significant glomerular damage, as reflected by elevated urinary protein levels and reduced plasma albumin levels in patients with ARKD. Histological analyses confirmed varying degrees of tubular damage and deposition of immune complexes. Proteomic analyses identified significant changes in protein expression, particularly in complement components (C3, C4A, C4B, C8G, CFB, and SERPINA1) and mitochondrial proteins (ATP5F1E and ATP5PD), highlighting the common alterations in the complement system and mitochondrial proteins across ARKDs. These alterations suggest a novel complement-mitochondrial-epithelial-mesenchymal transition (EMT) pathway axis that contributes to tubular damage in ARKDs. Notably, significant alterations in CFB in tubular ARKD patients were revealed, implicating it as a therapeutic target. This study underscores the importance of complement activation and mitochondrial dysfunction in the pathogenesis of ARKDs, and proposes CFB as a potential therapeutic target to inhibit complement activation and mitigate tubular damage. Future research should validate the complement-mitochondrial-EMT pathway axis and explore the effects and mechanisms of CFB inhibitors in alleviating ARKD progression.


Subject(s)
Complement Activation , Mitochondria , Proteomics , Humans , Proteomics/methods , Female , Male , Adult , Mitochondria/metabolism , Middle Aged , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Tandem Mass Spectrometry , Laser Capture Microdissection , Complement System Proteins/metabolism , Chromatography, Liquid
13.
ACS Nano ; 18(33): 22122-22138, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39119697

ABSTRACT

Binding of anti-PEG antibodies to poly(ethylene glycol) (PEG) on the surface of PEGylated liposomal doxorubicin (PLD) in vitro and in rats can activate complement and cause the rapid release of doxorubicin from the liposome interior. Here, we find that irinotecan liposomes (IL) and L-PLD, which have 16-fold lower levels of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE)-PEG2000 in their liposome membrane as compared to PLD, generate less complement activation but remain sensitive to destabilization and drug release by anti-PEG antibodies. Complement activation and liposome destabilization correlated with the theoretically estimated number of antibody molecules bound per liposome. Drug release from liposomes proceeded through the alternative complement pathway but was accelerated by the classical complement pathway. In contrast to PLD destabilization by anti-PEG immunoglobulin G (IgG), which proceeded by the insertion of membrane attack complexes in the lipid bilayer of otherwise intact PLD, anti-PEG IgG promoted the fusion of L-PLD, and IL to form unilamellar and oligo-vesicular liposomes. Anti-PEG immunoglobulin M (IgM) induced drug release from all liposomes (PLD, L-PLD, and IL) via the formation of unilamellar and oligo-vesicular liposomes. Anti-PEG IgG destabilized both PLD and L-PLD in rats, indicating that the reduction of PEG levels on liposomes is not an effective approach to prevent liposome destabilization by anti-PEG antibodies.


Subject(s)
Doxorubicin , Liposomes , Polyethylene Glycols , Polyethylene Glycols/chemistry , Liposomes/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/analogs & derivatives , Animals , Rats , Antibodies/chemistry , Antibodies/immunology , Complement Activation/drug effects , Phosphatidylethanolamines/chemistry , Drug Liberation
14.
Neurol Neuroimmunol Neuroinflamm ; 11(5): e200293, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39133885

ABSTRACT

OBJECTIVES: In myelin oligodendrocyte glycoprotein IgG-associated disease (MOGAD) and aquaporin-4 IgG+ neuromyelitis optica spectrum disorder (AQP4+NMOSD), the autoantibodies are mainly composed of IgG1, and complement-dependent cytotoxicity is a primary pathomechanism in AQP4+NMOSD. We aimed to evaluate the CSF complement activation in MOGAD. METHODS: CSF-C3a, CSF-C4a, CSF-C5a, and CSF-C5b-9 levels during the acute phase before treatment in patients with MOGAD (n = 12), AQP4+NMOSD (n = 11), multiple sclerosis (MS) (n = 5), and noninflammatory neurologic disease (n = 2) were measured. RESULTS: CSF-C3a and CSF-C5a levels were significantly higher in MOGAD (mean ± SD, 5,629 ± 1,079 pg/mL and 2,930 ± 435.8 pg/mL) and AQP4+NMOSD (6,017 ± 3,937 pg/mL and 2,544 ± 1,231 pg/mL) than in MS (1,507 ± 1,286 pg/mL and 193.8 ± 0.53 pg/mL). CSF-C3a, CSF-C4a, and CSF-C5a did not differ between MOGAD and AQP4+NMOSD while CSF-C5b-9 (membrane attack complex, MAC) levels were significantly lower in MOGAD (17.4 ± 27.9 ng/mL) than in AQP4+NMOSD (62.5 ± 45.1 ng/mL, p = 0.0019). Patients with MOGAD with severer attacks (Expanded Disability Status Scale [EDSS] ≥ 3.5) had higher C5b-9 levels (34.0 ± 38.4 ng/m) than those with milder attacks (EDSS ≤3.0, 0.9 ± 0.7 ng/mL, p = 0.044). DISCUSSION: The complement pathway is activated in both MOGAD and AQP4+NMOSD, but MAC formation is lower in MOGAD, particularly in those with mild attacks, than in AQP4+NMOSD. These findings may have pathogenetic and therapeutic implications in MOGAD.


Subject(s)
Aquaporin 4 , Complement Activation , Immunoglobulin G , Myelin-Oligodendrocyte Glycoprotein , Neuromyelitis Optica , Humans , Neuromyelitis Optica/cerebrospinal fluid , Neuromyelitis Optica/immunology , Neuromyelitis Optica/blood , Aquaporin 4/immunology , Male , Female , Middle Aged , Adult , Myelin-Oligodendrocyte Glycoprotein/immunology , Immunoglobulin G/cerebrospinal fluid , Immunoglobulin G/blood , Immunoglobulin G/immunology , Autoantibodies/cerebrospinal fluid , Autoantibodies/blood , Aged , Complement C5a/cerebrospinal fluid , Complement C5a/metabolism , Complement C5a/immunology , Young Adult , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Complement C3a/metabolism , Complement C3a/cerebrospinal fluid , Complement C3a/immunology , Complement Membrane Attack Complex/metabolism , Complement Membrane Attack Complex/cerebrospinal fluid , Complement Membrane Attack Complex/immunology
15.
J Innate Immun ; 16(1): 440-450, 2024.
Article in English | MEDLINE | ID: mdl-39159606

ABSTRACT

BACKGROUND: Respiratory diseases seriously threaten human health worldwide, and lung injury is an important component of respiratory disease. Complement activation is an important function of the innate immune system. Complement activation helps the body defend against invasion by external microorganisms, whereas excessive complement activation can exacerbate tissue damage or lead to unwanted side effects. Ficolins are a class of immune-related proteins in the lectin pathway that play important roles in the body's immune defense. Although individual ficolins are not well understood, current information suggests that ficolins may play an important regulatory role in lung injury. SUMMARY: Several studies have shown that ficolins are involved in the immune response in the lung, particularly in the response to infectious and inflammatory processes. KEY MESSAGES: This review summarizes the role of ficolins in lung injury. Ficolins may influence the development and repair of lung injury by recognizing and binding pathogenic microorganisms, modulating the inflammatory response, and promoting the clearance of immune cells. In addition, ficolins are associated with the development and progression of lung diseases (such as pneumonia and ARDS) and may have an important impact on the pathophysiological processes of inflammatory diseases.


Subject(s)
Ficolins , Immunity, Innate , Lectins , Lung Injury , Humans , Animals , Lung Injury/immunology , Lectins/metabolism , Lectins/immunology , Complement Activation/immunology , Lung/immunology , Inflammation/immunology
16.
Front Immunol ; 15: 1396800, 2024.
Article in English | MEDLINE | ID: mdl-39100680

ABSTRACT

Introduction: Bone marrow embolization may complicate orthopedic surgery, potentially causing fat embolism syndrome. The inflammatory potential of bone marrow emboli is unclear. We aimed to investigate the inflammatory response to femoral intramedullary nailing, specifically the systemic inflammatory effects in plasma, and local tissue responses. Additionally, the plasma response was compared to that following intravenous injection of autologous bone marrow. Methods: Twelve pigs underwent femoral nailing (previously shown to have fat emboli in lung and heart), four received intravenous bone marrow, and four served as sham controls. Blood samples were collected hourly and tissue samples postmortem. Additionally, we incubated bone marrow and blood, separately and in combination, from six pigs in vitro. Complement activation was detected by C3a and the terminal C5b-9 complement complex (TCC), and the cytokines TNF, IL-1ß, IL-6 and IL-10 as well as the thrombin-antithrombin complexes (TAT) were all measured using enzyme-immunoassays. Results: After nailing, plasma IL-6 rose 21-fold, compared to a 4-fold rise in sham (p=0.0004). No plasma differences in the rest of the inflammatory markers were noted across groups. However, nailing yielded 2-3-times higher C3a, TCC, TNF, IL-1ß and IL-10 in lung tissue compared to sham (p<0.0001-0.03). Similarly, heart tissue exhibited 2-times higher TCC and IL-1ß compared to sham (p<0.0001-0.03). Intravenous bone marrow yielded 8-times higher TAT than sham at 30 minutes (p<0.0001). In vitro, incubation of bone marrow for four hours resulted in 95-times higher IL-6 compared to whole blood (p=0.03). Discussion: A selective increase in plasma IL-6 was observed following femoral nailing, whereas lung and heart tissues revealed a broad local inflammatory response not reflected systemically. In vitro experiments may imply bone marrow to be the primary IL-6 source.


Subject(s)
Embolism, Fat , Interleukin-6 , Lung , Animals , Swine , Interleukin-6/blood , Embolism, Fat/etiology , Embolism, Fat/blood , Embolism, Fat/immunology , Lung/immunology , Lung/pathology , Lung/metabolism , Bone Marrow/metabolism , Fracture Fixation, Intramedullary/adverse effects , Myocardium/metabolism , Myocardium/pathology , Myocardium/immunology , Inflammation/blood , Inflammation/immunology , Female , Cytokines/blood , Cytokines/metabolism , Bone Nails , Complement Activation , Femur/metabolism , Disease Models, Animal
17.
Int J Mol Sci ; 25(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39125933

ABSTRACT

The complement (C) system is implicated in the etiopathogenesis of rheumatoid arthritis (RA). However, there is a lack of studies characterizing all three C pathways in RA patients. This study aimed to evaluate the association between an in-depth examination of the C system and RA patient characteristics, focusing on disease activity and the presence of rheumatoid factor and anti-citrullinated protein autoantibodies (ACPA). In a cohort of 430 RA patients, functional assays of the three C pathways (classical, alternative, and lectin) and serum levels of their components were assessed. Components included C1q (classical); factor D and properdin (alternative); lectin (lectin); C1-inhibitor; C2, C4, and C4b (classical and lectin); C3, C3a, and C4b (common); and C5, C5a, and C9 (terminal). A multivariable linear regression analysis showed significant positive correlations between C-reactive protein and C system proteins and functional assays, especially in the terminal and common pathways. Disease activity, measured by scores with or without acute phase reactants, positively correlated with the classical pathway functional test and terminal pathway products. Conversely, rheumatoid factor or ACPA presence was associated with lower classical pathway values and decreased C3a and C4b levels, suggesting complement depletion. In conclusion, RA disease activity increases C molecules and functional complement assays, while rheumatoid factor or ACPA positivity is linked to C consumption. Our study offers a detailed analysis of the complement system's role in RA, potentially guiding the development of more targeted and effective treatment strategies.


Subject(s)
Arthritis, Rheumatoid , Rheumatoid Factor , Humans , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Male , Female , Middle Aged , Aged , Rheumatoid Factor/blood , Adult , Complement System Proteins/metabolism , Complement System Proteins/immunology , Anti-Citrullinated Protein Antibodies/blood , Complement Pathway, Alternative , Complement Activation , C-Reactive Protein/metabolism , C-Reactive Protein/analysis , Complement Pathway, Classical
19.
Int J Biol Macromol ; 276(Pt 1): 134117, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084989

ABSTRACT

The degradation of Belamcanda chinensis (L.) DC. polysaccharides was carried out by five concentrations of trifluoroacetic acid (TFA) (1-5 mol/L), and their physicochemical properties, degradation kinetics and anticomplementary activity were investigated. The findings revealed a notable reduction in the molecular weight of BCP, from an initial value of 2.622 × 105 g/mol to a final value of 6.255 × 104 g/mol, and the water solubility index increased from 90.66 ± 0.42 % to 97.78 ± 0.43 %. The degraded polysaccharides of B. chinensis exhibited a comparable monosaccharide composition comprising Man, GalA, Glc, Gal, and Ara. As the concentration of TFA increased, the degradation rate constant increased from 1.468 × 10-3 to 5.943 × 10-3, and the process followed the first-order degradation kinetic model (R2 > 0.97) and the random fracture model (R2 > 0.96). Furthermore, the five degraded polysaccharides still exhibit good thermal stability. In vitro experiments showed that DBCP-3 exhibited more potent anticomplementary activity than the original polysaccharides and positive drugs, which was strongly correlated with its Mw (r = 0.6-0.8), inhibiting complement activation by blocking C2 and C4. These results indicated that TFA degradation has a positive effect on polysaccharides, of which DBCP-3 is expected to treat diseases involving hyperactivation of the complement system.


Subject(s)
Molecular Weight , Polysaccharides , Trifluoroacetic Acid , Polysaccharides/chemistry , Polysaccharides/pharmacology , Trifluoroacetic Acid/chemistry , Kinetics , Monosaccharides/analysis , Monosaccharides/chemistry , Animals , Solubility , Caryophyllaceae/chemistry , Complement Activation/drug effects
20.
Drug Res (Stuttg) ; 74(7): 325-334, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38991528

ABSTRACT

Complement cascade is a defence mechanism useful for eliminating pathogenic microorganisms and damaged cells. However, activation of alternative complement system can also cause inflammation and promote kidney and retinal disease progression. Inflammation causes tissue hypoxia, which induces hypoxia-inducible factor (HIF) and HIF helps the body to adapt to inflammation. In this study, we investigated the effect of HIF stabilizer desidustat in complement-mediated diseases. Oral administration of desidustat (15 mg/kg) was effective to reduce the kidney injury in mice that was induced by either lipopolysaccharide (LPS), doxorubicin or bovine serum albumin (BSA)-overload. Complement activation-induced membrane attack complex (MAC) formation and factor B activity were also reduced by desidustat treatment. In addition, desidustat was effective against membranous nephropathy caused by cationic BSA and retinal degeneration induced by sodium iodate in mice. C3-deposition, proteinuria, malondialdehyde, and interleukin-1ß were decreased and superoxide dismutase was increased by desidustat treatment in cBSA-induced membranous nephropathy. Desidustat specifically inhibited alternative complement system, without affecting the lectin-, or classical complement pathway. This effect appears to be mediated by inhibition of factor B. These data demonstrate the potential therapeutic value of HIF stabilization by desidustat in treatment of complement-mediated diseases.


Subject(s)
Complement Activation , Animals , Mice , Complement Activation/drug effects , Glomerulonephritis, Membranous/drug therapy , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/pathology , Male , Lipopolysaccharides , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Serum Albumin, Bovine , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Complement Membrane Attack Complex/metabolism , Disease Models, Animal , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Interleukin-1beta/metabolism , Protective Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL