Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Int J Cell Biol ; 2023: 7121512, 2023.
Article in English | MEDLINE | ID: mdl-37941807

ABSTRACT

Inducible gene regulation methods are indispensable in diverse biological applications, yet many of them have severe limitations in their applicability. These include inducer toxicity, a limited variety of organisms the given system can be used in, and side effects of the induction method. In this study, a novel inducible system, the RuX system, was created using a mutant ligand-binding domain of the glucocorticoid receptor (CS1/CD), used together with various genetic elements such as the Gal4 DNA-binding domain or Cre recombinase. The RuX system is shown to be capable of over 1000-fold inducibility, has flexible applications, and is offered for use in cell cultures.

2.
Front Immunol ; 13: 845953, 2022.
Article in English | MEDLINE | ID: mdl-35392081

ABSTRACT

Components of the extracellular matrix (ECM), when exposed to body fluids may promote local complement activation and inflammation. Pathologic complement activation at the glomerular basement membrane and at the Bruch's membrane is implicated in renal and eye diseases, respectively. Binding of soluble complement inhibitors to the ECM, including factor H (FH), is important to prevent excessive complement activation. Since the FH-related (FHR) proteins FHR1 and FHR5 are also implicated in these diseases, our aim was to study whether these FHRs can also bind to ECM components and affect local FH activity and complement activation. Both FH and the FHRs showed variable binding to ECM components. We identified laminin, fibromodulin, osteoadherin and PRELP as ligands of FHR1 and FHR5, and found that FHR1 bound to these ECM components through its C-terminal complement control protein (CCP) domains 4-5, whereas FHR5 bound via its middle region, CCPs 3-7. Aggrecan, biglycan and decorin did not bind FH, FHR1 and FHR5. FHR5 also bound to immobilized C3b, a model of surface-deposited C3b, via CCPs 3-7. By contrast, soluble C3, C3(H2O), and the C3 fragments C3b, iC3b and C3d bound to CCPs 8-9 of FHR5. Properdin, which was previously described to bind via CCPs 1-2 to FHR5, did not bind in its physiologically occurring serum forms in our assays. FHR1 and FHR5 inhibited the binding of FH to the identified ECM proteins in a dose-dependent manner, which resulted in reduced FH cofactor activity. Moreover, both FHR1 and FHR5 enhanced alternative complement pathway activation on immobilized ECM proteins when exposed to human serum, resulting in the increased deposition of C3-fragments, factor B and C5b-9. Thus, our results identify novel ECM ligands of FH family proteins and indicate that FHR1 and FHR5 are competitive inhibitors of FH on ECM and, when bound to these ligands, they may enhance local complement activation and promote inflammation under pathological conditions.


Subject(s)
Complement Activation , Complement C3b Inactivator Proteins , Complement Factor H , Complement System Proteins , Complement C3b Inactivator Proteins/metabolism , Complement System Proteins/metabolism , Extracellular Matrix , Humans , Inflammation , Ligands
3.
Front Immunol ; 12: 720183, 2021.
Article in English | MEDLINE | ID: mdl-34566977

ABSTRACT

Background: Factor H-related protein 5 (FHR-5) is a member of the complement Factor H protein family. Due to the homology to Factor H, the main complement regulator of the alternative pathway, it may also be implicated in the pathomechanism of kidney diseases where Factor H and alternative pathway dysregulation play a role. Here, we report the first observational study on CFHR5 variations along with serum FHR-5 levels in immune complex-mediated membranoproliferative glomerulonephritis (IC-MPGN) and C3 glomerulopathy (C3G) patients together with the clinical, genetic, complement, and follow-up data. Methods: A total of 120 patients with a histologically proven diagnosis of IC-MPGN/C3G were enrolled in the study. FHR-5 serum levels were measured in ELISA, the CFHR5 gene was analyzed by Sanger sequencing, and selected variants were studied as recombinant proteins in ELISA and surface plasmon resonance (SPR). Results: Eight exonic CFHR5 variations in 14 patients (12.6%) were observed. Serum FHR-5 levels were lower in patients compared to controls. Low serum FHR-5 concentration at presentation associated with better renal survival during the follow-up period; furthermore, it showed clear association with signs of complement overactivation and clinically meaningful clusters. Conclusions: Our observations raise the possibility that the FHR-5 protein plays a fine-tuning role in the pathogenesis of IC-MPGN/C3G.


Subject(s)
Antigen-Antibody Complex/immunology , Biomarkers , Complement C3/immunology , Complement System Proteins/genetics , Complement System Proteins/metabolism , Genetic Variation , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/etiology , Adolescent , Adult , Alleles , Case-Control Studies , Complement Activation , Disease Management , Disease Susceptibility , Enzyme-Linked Immunosorbent Assay , Female , Genetic Predisposition to Disease , Glomerulonephritis, Membranoproliferative/diagnosis , Glomerulonephritis, Membranoproliferative/mortality , Humans , Kidney Function Tests , Male , Polymorphism, Single Nucleotide , Prognosis , ROC Curve , Symptom Assessment , Young Adult
4.
Front Immunol ; 11: 1297, 2020.
Article in English | MEDLINE | ID: mdl-32765490

ABSTRACT

Complement plays an essential role in the opsonophagocytic clearance of apoptotic/necrotic cells. Dysregulation of this process may lead to inflammatory and autoimmune diseases. Factor H (FH), a major soluble complement inhibitor, binds to dead cells and inhibits excessive complement activation on their surface, preventing lysis, and the release of intracellular material, including DNA. The FH-related (FHR) proteins share common ligands with FH, due to their homology with this complement regulator, but they lack the domains that mediate the complement inhibitory activity of FH. Because their roles in complement regulation is controversial and incompletely understood, we studied the interaction of FHR-1 and FHR-5 with DNA and dead cells and investigated whether they influence the regulatory role of FH and the complement activation on DNA and dead cells. FH, FHR-1, and FHR-5 bound to both plasmid DNA and human genomic DNA, where both FHR proteins inhibited FH-DNA interaction. The FH cofactor activity was inhibited by FHR-1 and FHR-5 due to the reduced binding of FH to DNA in the presence of the FHRs. Both FHRs caused increased complement activation on DNA. FHR-1 and FHR-5 bound to late apoptotic and necrotic cells and recruited monomeric C-reactive protein and pentraxin 3, and vice versa. Interactions of the FHRs with pentraxins resulted in enhanced activation of both the classical and the alternative complement pathways on dead cells when exposed to human serum. Altogether, our results demonstrate that FHR-1 and FHR-5 are competitive inhibitors of FH on DNA; moreover, FHR-pentraxin interactions promote opsonization of dead cells.


Subject(s)
Complement C3b Inactivator Proteins/metabolism , Complement System Proteins/metabolism , DNA/metabolism , Apoptosis/immunology , Cell Death/genetics , Cell Death/immunology , Cell Line, Tumor , Complement Activation , Complement System Proteins/immunology , Endothelial Cells , Extracellular Traps/genetics , Extracellular Traps/immunology , Flow Cytometry , Humans , Necrosis/immunology , Protein Binding
5.
Semin Immunol ; 45: 101341, 2019 10.
Article in English | MEDLINE | ID: mdl-31757608

ABSTRACT

The complement system, while being an essential and very efficient effector component of innate immunity, may cause damage to the host and result in various inflammatory, autoimmune and infectious diseases or cancer, when it is improperly activated or regulated. Factor H is a serum glycoprotein and the main regulator of the activity of the alternative complement pathway. Factor H, together with its splice variant factor H-like protein 1 (FHL-1), inhibits complement activation at the level of the central complement component C3 and beyond. In humans, there are also five factor H-related (FHR) proteins, whose function is poorly characterized. While data indicate complement inhibiting activity for some of the FHRs, there is increasing evidence that FHRs have an opposite role compared with factor H and FHL-1, namely, they enhance complement activation directly and also by competing with the regulators FH and FHL-1. This review summarizes the current stand and recent data on the roles of factor H family proteins in health and disease, with focus on the function of FHR proteins.


Subject(s)
Blood Proteins/metabolism , Carrier Proteins/metabolism , Complement Activation/immunology , Complement Factor H/immunology , Complement Factor H/metabolism , Animals , Blood Proteins/chemistry , Blood Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Complement Factor H/chemistry , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Susceptibility , Humans , Immunomodulation , Ligands , Multigene Family , Protein Binding , Structure-Activity Relationship
6.
Front Immunol ; 8: 1145, 2017.
Article in English | MEDLINE | ID: mdl-28974948

ABSTRACT

Factor H-related (FHR) proteins consist of varying number of complement control protein domains that display various degrees of sequence identity to respective domains of the alternative pathway complement inhibitor factor H (FH). While such FHR proteins are described in several species, only human FHRs were functionally investigated. Their biological role is still poorly understood and in part controversial. Recent studies on some of the human FHRs strongly suggest a role for FHRs in enhancing complement activation via competing with FH for binding to certain ligands and surfaces. The aim of the current study was the functional characterization of a murine FHR, FHR-B. To this end, FHR-B was expressed in recombinant form. Recombinant FHR-B bound to human C3b and was able to compete with human FH for C3b binding. FHR-B supported the assembly of functionally active C3bBb alternative pathway C3 convertase via its interaction with C3b. This activity was confirmed by demonstrating C3 activation in murine serum. In addition, FHR-B bound to murine pentraxin 3 (PTX3), and this interaction resulted in murine C3 fragment deposition due to enhanced complement activation in mouse serum. FHR-B also induced C3 deposition on C-reactive protein, the extracellular matrix (ECM) extract Matrigel, and endothelial cell-derived ECM when exposed to mouse serum. Moreover, mouse C3 deposition was strongly enhanced on necrotic Jurkat T cells and the mouse B cell line A20 by FHR-B. FHR-B also induced lysis of sheep erythrocytes when incubated in mouse serum with FHR-B added in excess. Altogether, these data demonstrate that, similar to human FHR-1 and FHR-5, mouse FHR-B modulates complement activity by promoting complement activation via interaction with C3b and via competition with murine FH.

7.
J Immunol ; 199(1): 292-303, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28533443

ABSTRACT

Factor H-related protein (FHR) 1 is one of the five human FHRs that share sequence and structural homology with the alternative pathway complement inhibitor FH. Genetic studies on disease associations and functional analyses indicate that FHR-1 enhances complement activation by competitive inhibition of FH binding to some surfaces and immune proteins. We have recently shown that FHR-1 binds to pentraxin 3. In this study, our aim was to investigate whether FHR-1 binds to another pentraxin, C-reactive protein (CRP), analyze the functional relevance of this interaction, and study the role of FHR-1 in complement activation and regulation. FHR-1 did not bind to native, pentameric CRP, but it bound strongly to monomeric CRP via its C-terminal domains. FHR-1 at high concentration competed with FH for CRP binding, indicating possible complement deregulation also on this ligand. FHR-1 did not inhibit regulation of solid-phase C3 convertase by FH and did not inhibit terminal complement complex formation induced by zymosan. On the contrary, by binding C3b, FHR-1 allowed C3 convertase formation and thereby enhanced complement activation. FHR-1/CRP interactions increased complement activation via the classical and alternative pathways on surfaces such as the extracellular matrix and necrotic cells. Altogether, these results identify CRP as a ligand for FHR-1 and suggest that FHR-1 enhances, rather than inhibits, complement activation, which may explain the protective effect of FHR-1 deficiency in age-related macular degeneration.


Subject(s)
C-Reactive Protein/immunology , C-Reactive Protein/metabolism , Complement Activation , Complement C3b Inactivator Proteins/immunology , Complement C3b Inactivator Proteins/metabolism , Binding Sites , C-Reactive Protein/chemistry , C-Reactive Protein/pharmacology , Complement C3-C5 Convertases , Complement C3b/immunology , Complement C3b/pharmacology , Complement C3b Inactivator Proteins/pharmacology , Complement Factor H , Extracellular Matrix/drug effects , Extracellular Matrix/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Ligands , Macular Degeneration/immunology , Protein Binding , Serum Amyloid P-Component/immunology , Serum Amyloid P-Component/metabolism
8.
Front Immunol ; 8: 1800, 2017.
Article in English | MEDLINE | ID: mdl-29321782

ABSTRACT

Dysregulation of the complement alternative pathway is involved in the pathogenesis of several diseases, including the kidney diseases atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G). In a patient, initially diagnosed with chronic glomerulonephritis, possibly C3G, and who 6 years later had an episode of aHUS, a heterozygous missense mutation leading to a tryptophan to arginine exchange (W198R) in the factor H (FH) complement control protein (CCP) 3 domain has previously been identified. The aim of this study was to clarify the functional relevance of this mutation. To this end, wild-type (FH1-4WT) and mutant (FH1-4W198R) CCPs 1-4 of FH were expressed as recombinant proteins. The FH1-4W198R mutant showed decreased C3b binding compared with FH1-4WT. FH1-4W198R had reduced cofactor and decay accelerating activity compared with the wild-type protein. Hemolysis assays demonstrated impaired capacity of FH1-4W198R to protect rabbit erythrocytes from human complement-mediated lysis, and also to prevent lysis of sheep erythrocytes in human serum induced by a monoclonal antibody binding in FH CCP5 domain, compared with that of FH1-4WT. Thus, the FH W198R exchange results in impaired complement alternative pathway regulation. The heterozygous nature of this mutation in the index patient may explain the manifestation of two diseases, likely due to different triggers leading to complement dysregulation in plasma or on cell surfaces.

9.
MAbs ; 6(4): 991-9, 2014.
Article in English | MEDLINE | ID: mdl-24828435

ABSTRACT

The Fc receptor (FcγRIIb) inhibits B cell responses when coengaged with B cell receptor (BCR), and has become a target for new autoimmune disease therapeutics. For example, BCR and FcγRIIb coengagement via the Fc-engineered anti-CD19 XmAb5871 suppresses humoral immune responses. We now assess effects of XmAb5871 on other activation pathways, including the pathogen-associated molecular pattern receptor, TLR9. Since TLR9 signaling is implicated in autoimmune diseases, we asked if XmAb5871 could inhibit TLR9 costimulation. We show that XmAb5871 decreases ERK and AKT activation, cell proliferation, cytokine, and IgG production induced by BCR and/or TLR9 signals. XmAb5871 also inhibited differentiation of citrullinated peptide-specific plasma cells from rheumatoid arthritis patients. XmAb5871 may therefore have potential to suppress pathogenic B cells in autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD19 , B-Lymphocytes/immunology , Immunity, Innate/drug effects , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Receptors, IgG/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal/immunology , B-Lymphocytes/pathology , Cell Proliferation/drug effects , Cytokines/immunology , Female , Humans , Immunoglobulin G/immunology , Immunosuppressive Agents/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Male , Middle Aged , Receptors, Antigen, B-Cell/immunology , Receptors, IgG/immunology , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/immunology
SELECTION OF CITATIONS
SEARCH DETAIL