Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32571987

ABSTRACT

Even though both cellular and humoral immunities contribute to host defense, the role played by humoral immunity against the airborne opportunistic fungal pathogen Aspergillus fumigatus has been underexplored. In this study, we aimed at deciphering the role of the complement system, the major humoral immune component, against A. fumigatus Mass spectrometry analysis of the proteins extracted from A. fumigatus conidial (asexual spores and infective propagules) surfaces opsonized with human serum indicated that C3 is the major complement protein involved. Flow cytometry and immunolabeling assays further confirmed C3b (activated C3) deposition on the conidial surfaces. Assays using cell wall components of conidia indicated that the hydrophobin RodAp, ß-(1,3)-glucan (BG) and galactomannan (GM) could efficiently activate C3. Using complement component-depleted sera, we showed that while RodAp activates C3 by the alternative pathway, BG and GM partially follow the classical and lectin pathways, respectively. Opsonization facilitated conidial aggregation and phagocytosis, and complement receptor (CR3 and CR4) blockage on phagocytes significantly inhibited phagocytosis, indicating that the complement system exerts a protective role against conidia by opsonizing them and facilitating their phagocytosis mainly through complement receptors. Conidial opsonization with human bronchoalveolar lavage fluid (BALF) confirmed C3 to be the major complement protein interacting with conidia. Nevertheless, complement C2 and mannose-binding lectin (MBL), the classical and lectin pathway components, respectively, were not identified, indicating that BALF activates the alternative pathway on the conidial surface. Moreover, the cytokine profiles were different upon stimulation of phagocytes with serum- and BALF-opsonized conidia, highlighting the importance of studying interaction of conidia with complement proteins in their biological niche.


Subject(s)
Aspergillus fumigatus/immunology , Bronchoalveolar Lavage Fluid/immunology , Complement C3/immunology , Fungal Polysaccharides/pharmacology , Macrophages/drug effects , Serum/immunology , Spores, Fungal/immunology , Aspergillosis/genetics , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillus fumigatus/chemistry , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/microbiology , Cell Wall/chemistry , Cell Wall/immunology , Complement Activation/drug effects , Complement C3/genetics , Cytokines/biosynthesis , Cytokines/immunology , Fungal Polysaccharides/immunology , Fungal Polysaccharides/isolation & purification , Galactose/analogs & derivatives , Host Microbial Interactions/immunology , Humans , Immunity, Cellular , Immunity, Humoral , Integrin alphaXbeta2/genetics , Integrin alphaXbeta2/immunology , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/immunology , Macrophages/immunology , Macrophages/microbiology , Mannans/immunology , Mannans/isolation & purification , Mannans/pharmacology , Opsonin Proteins/pharmacology , Phagocytosis/drug effects , Primary Cell Culture , Protein Binding , Reactive Oxygen Species , Serum/chemistry , Serum/microbiology , Spores, Fungal/chemistry , beta-Glucans/immunology , beta-Glucans/isolation & purification , beta-Glucans/pharmacology
2.
J Biol Chem ; 293(40): 15538-15555, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30139746

ABSTRACT

Innate immunity in animals including humans encompasses the complement system, which is considered an important host defense mechanism against Aspergillus fumigatus, one of the most ubiquitous opportunistic human fungal pathogens. Previously, it has been shown that the alkaline protease Alp1p secreted from A. fumigatus mycelia degrades the complement components C3, C4, and C5. However, it remains unclear how the fungal spores (i.e. conidia) defend themselves against the activities of the complement system immediately after inhalation into the lung. Here, we show that A. fumigatus conidia contain a metalloprotease Mep1p, which is released upon conidial contact with collagen and inactivates all three complement pathways. In particular, Mep1p efficiently inactivated the major complement components C3, C4, and C5 and their activation products (C3a, C4a, and C5a) as well as the pattern-recognition molecules MBL and ficolin-1, either by directly cleaving them or by cleaving them to a form that is further broken down by other proteases of the complement system. Moreover, incubation of Mep1p with human serum significantly inhibited the complement hemolytic activity and conidial opsonization by C3b and their subsequent phagocytosis by macrophages. Together, these results indicate that Mep1p associated with and released from A. fumigatus conidia likely facilitates early immune evasion by disarming the complement defense in the human host.


Subject(s)
Aspergillus fumigatus/immunology , Complement C3/genetics , Complement C4/genetics , Complement C5/genetics , Invasive Pulmonary Aspergillosis/immunology , Metalloendopeptidases/immunology , Animals , Aspergillus fumigatus/growth & development , Aspergillus fumigatus/pathogenicity , Collagen/genetics , Collagen/immunology , Complement C3/metabolism , Complement C4/metabolism , Complement C5/metabolism , Disease Models, Animal , Fungal Proteins/genetics , Fungal Proteins/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Invasive Pulmonary Aspergillosis/genetics , Invasive Pulmonary Aspergillosis/microbiology , Invasive Pulmonary Aspergillosis/pathology , Lectins/genetics , Lectins/immunology , Lung/immunology , Lung/pathology , Macrophages/immunology , Macrophages/microbiology , Male , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Phagocytosis , Spores, Fungal/growth & development , Spores, Fungal/immunology , Spores, Fungal/pathogenicity , Ficolins
3.
Front Immunol ; 13: 978152, 2022.
Article in English | MEDLINE | ID: mdl-36211424

ABSTRACT

Invasive aspergillosis (IA) is a life-threatening fungal infection for immunocompromised hosts. It is, therefore, necessary to understand the immune pathways that control this infection. Although the primary infection site is the lungs, aspergillosis can disseminate to other organs through unknown mechanisms. Herein we have examined the in vivo role of various complement pathways as well as the complement receptors C3aR and C5aR1 during experimental systemic infection by Aspergillus fumigatus, the main species responsible for IA. We show that C3 knockout (C3-/-) mice are highly susceptible to systemic infection of A. fumigatus. Intriguingly, C4-/- and factor B (FB)-/- mice showed susceptibility similar to the wild-type mice, suggesting that either the complement pathways display functional redundancy during infection (i.e., one pathway compensates for the loss of the other), or complement is activated non-canonically by A. fumigatus protease. Our in vitro study substantiates the presence of C3 and C5 cleaving proteases in A. fumigatus. Examination of the importance of the terminal complement pathway employing C5-/- and C5aR1-/- mice reveals that it plays a vital role in the conidial clearance. This, in part, is due to the increased conidial uptake by phagocytes. Together, our data suggest that the complement deficiency enhances the susceptibility to systemic infection by A. fumigatus.


Subject(s)
Aspergillosis , Aspergillus fumigatus , Animals , Complement C5/genetics , Complement C5/metabolism , Complement Factor B/genetics , Lung , Mice , Spores, Fungal
4.
Commun Biol ; 2: 290, 2019.
Article in English | MEDLINE | ID: mdl-31396570

ABSTRACT

Regulation of complement activation in the host cells is mediated primarily by the regulators of complement activation (RCA) family proteins that are formed by tandemly repeating complement control protein (CCP) domains. Functional annotation of these proteins, however, is challenging as contiguous CCP domains are found in proteins with varied functions. Here, by employing an in silico approach, we identify five motifs which are conserved spatially in a specific order in the regulatory CCP domains of known RCA proteins. We report that the presence of these motifs in a specific pattern is sufficient to annotate regulatory domains in RCA proteins. We show that incorporation of the lost motif in the fourth long-homologous repeat (LHR-D) in complement receptor 1 regains its regulatory activity. Additionally, the motif pattern also helped annotate human polydom as a complement regulator. Thus, we propose that the motifs identified here are the determinants of functionality in RCA proteins.


Subject(s)
Cell Adhesion Molecules/metabolism , Complement Activation , Complement System Proteins/metabolism , Receptors, Complement 3b/metabolism , Amino Acid Motifs , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cnidaria/chemistry , Cnidaria/metabolism , Complement System Proteins/chemistry , Complement System Proteins/genetics , Conserved Sequence , Humans , Phylogeny , Protein Conformation , Protein Domains , Receptors, Complement 3b/chemistry , Receptors, Complement 3b/genetics , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/metabolism
5.
Front Immunol ; 10: 1282, 2019.
Article in English | MEDLINE | ID: mdl-31231391

ABSTRACT

Background: Atypical hemolytic uremic syndrome (aHUS), an important cause of acute kidney injury (AKI), is characterized by dysregulation of the alternative complement pathway. Autoantibodies to factor H (FH), a chief regulator of this pathway, account for a distinct subgroup. While high anti-FH titers predict relapse, they do not correlate well with disease activity and their functional characterization is required. Methods: Of 781 patients <18-year-old of aHUS in the nationwide database from 2007 to 2018, 436 (55.8%) had anti-FH antibodies. Clinical features and outcome of patients managed in the last 6-year (n = 317) were compared to before (n = 119). In plasma samples of 44 patients, levels of serial circulating FH immune complexes (CIC), free FH, soluble terminal complement complex (sC5b-9), sheep red blood cell (SRBC) lysis and epitope specificity (n = 8) were examined. Functional renal reserve, ambulatory hypertension, left ventricular hypertrophy (LVH), and proteinuria were evaluated in a subset. Results: Patients presented with markedly elevated anti-FH titers (10,633.2 ± 998.5 AU/ml). Management varied by center, comprising plasma exchange (PEX; 77.5%) and immunosuppression (73.9%). Patients managed in the last 6-year showed better renal survival at mean 28.5 ± 27.3 months (log rank P = 0.022). Mean anti-FH titers stayed 700-1,164 AU/ml during prolonged follow-up, correlating with CIC. Patients with relapse had lower free-FH during remission [Generalized estimating equations (GEE), P = 0.001]; anti-FH levels ≥1,330 AU/ml and free FH ≤440 mg/l predicted relapse (hazards ratio, HR 6.3; P = 0.018). Epitope specificity was similar during onset, remission and relapse. Antibody titer ≥8,000 AU/ml (HR 2.23; P = 0.024), time to PEX ≥14 days (HR 2.09; P = 0.071) and PEX for <14 days (HR 2.60; P = 0.017) predicted adverse renal outcomes. Combined PEX and immunosuppression improved long-term outcomes (HR 0.37; P = 0.026); maintenance therapy reduced risk of relapses (HR 0.11; P < 0.001). At 4.4±2.5 year, median renal reserve was 15.9%; severe ambulatory, masked and pre-hypertension were found in 38, 30, and 18%, respectively. Proteinuria and LVH occurred in 58 and 28% patients, respectively. Conclusion: Prompt recognition and therapy with PEX and immunosuppression, is associated with satisfactory outcomes. Free-FH predicts early relapses in patients with high anti-FH titers. A significant proportion of impaired functional reserve, ambulatory hypertension, proteinuria and LVH highlight the need for vigilant long-term follow-up.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Adolescent , Atypical Hemolytic Uremic Syndrome/pathology , Atypical Hemolytic Uremic Syndrome/therapy , Autoantibodies/immunology , Autoantigens/immunology , Child , Child, Preschool , Complement Factor H/immunology , Databases, Factual , Female , Humans , Immunosuppressive Agents/therapeutic use , Infant , Male
SELECTION OF CITATIONS
SEARCH DETAIL