Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunol Rev ; 313(1): 60-63, 2023 01.
Article in English | MEDLINE | ID: mdl-36089772

ABSTRACT

Primitive underpinnings of the alternative pathway (AP), namely, a C3-like protein, likely arose more than a billion years ago. The development of an AP amplification loop, while greatly enhancing speed and potency, also presents a double-edged sword. Although critical to combat an infectious disease, it is also potentially destructive, particularly in a chronic disease process involving vital organs where scarring and reduction of regulatory function can occur. Furthermore, new knowledge is pointing to genetic factors involved in an increasing number of complement-related diseases such as age-related macular degeneration. However, even a normal functioning repertoire of complement components can drive cellular damage as a result of low-level complement activation over time. Thus, the modern human AP now faces a new challenge: cumulatively-driven tissue damage from chronic inflammatory processes that mediate cellular injury. The impact of ongoing low-level AP-enhanced complement activation in disease processes is just beginning to be appreciated and studied. However, the sheer numbers of individuals affected by chronic diseases emphasize the need for novel therapeutic agents capable of modulating the AP. The more we learn about this ancient system, the greater is the likelihood of developing fresh perspectives that could contribute to improved human health.


Subject(s)
Complement Activation , Complement Pathway, Alternative , Humans , Complement Pathway, Alternative/genetics , Complement System Proteins/metabolism , Inflammation
2.
J Virol ; 96(3): e0082621, 2022 02 09.
Article in English | MEDLINE | ID: mdl-34787457

ABSTRACT

Human adenovirus serotype 26 (Ad26) is used as a gene-based vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and HIV-1. However, its primary receptor portfolio remains controversial, potentially including sialic acid, coxsackie and adenovirus receptor (CAR), integrins, and CD46. We and others have shown that Ad26 can use CD46, but these observations were questioned on the basis of the inability to cocrystallize Ad26 fiber with CD46. Recent work demonstrated that Ad26 binds CD46 with its hexon protein rather than its fiber. We examined the functional consequences of Ad26 for infection in vitro and in vivo. Ectopic expression of human CD46 on Chinese hamster ovary cells increased Ad26 infection significantly. Deletion of the complement control protein domain CCP1 or CCP2 or the serine-threonine-proline (STP) region of CD46 reduced infection. Comparing wild-type and sialic acid-deficient CHO cells, we show that the usage of CD46 is independent of its sialylation status. Ad26 transduction was increased in CD46 transgenic mice after intramuscular (i.m.) injection but not after intranasal (i.n.) administration. Ad26 transduction was 10-fold lower than Ad5 transduction after intratumoral (i.t.) injection of CD46-expressing tumors. Ad26 transduction of liver was 1,000-fold lower than that ofAd5 after intravenous (i.v.) injection. These data demonstrate the use of CD46 by Ad26 in certain situations but also show that the receptor has little consequence by other routes of administration. Finally, i.v. injection of high doses of Ad26 into CD46 mice induced release of liver enzymes into the bloodstream and reduced white blood cell counts but did not induce thrombocytopenia. This suggests that Ad26 virions do not induce direct clotting side effects seen during coronavirus disease 2019 (COVID-19) vaccination with this serotype of adenovirus. IMPORTANCE The human species D Ad26 is being investigated as a low-seroprevalence vector for oncolytic virotherapy and gene-based vaccination against HIV-1 and SARS-CoV-2. However, there is debate in the literature about its tropism and receptor utilization, which directly influence its efficiency for certain applications. This work was aimed at determining which receptor(s) this virus uses for infection and its role in virus biology, vaccine efficacy, and, importantly, vaccine safety.


Subject(s)
Adenovirus Infections, Human/metabolism , Adenovirus Infections, Human/virology , Adenoviruses, Human/classification , Adenoviruses, Human/physiology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Host-Pathogen Interactions , Membrane Cofactor Protein/metabolism , Adenoviruses, Human/ultrastructure , Animals , Biomarkers , Blood Cell Count , CHO Cells , Cell Line , Coxsackie and Adenovirus Receptor-Like Membrane Protein/chemistry , Cricetulus , Disease Models, Animal , Gene Expression , Humans , Membrane Cofactor Protein/chemistry , Membrane Cofactor Protein/genetics , Mice, Transgenic , Models, Biological , Models, Molecular , Mutagenesis , Protein Binding , Protein Conformation , Serogroup , Sialic Acids/metabolism , Sialic Acids/pharmacology , Structure-Activity Relationship
3.
Immunity ; 39(6): 1143-57, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24315997

ABSTRACT

Complement is viewed as a critical serum-operative component of innate immunity, with processing of its key component, C3, into activation fragments C3a and C3b confined to the extracellular space. We report here that C3 activation also occurred intracellularly. We found that the T cell-expressed protease cathepsin L (CTSL) processed C3 into biologically active C3a and C3b. Resting T cells contained stores of endosomal and lysosomal C3 and CTSL and substantial amounts of CTSL-generated C3a. While "tonic" intracellular C3a generation was required for homeostatic T cell survival, shuttling of this intracellular C3-activation-system to the cell surface upon T cell stimulation induced autocrine proinflammatory cytokine production. Furthermore, T cells from patients with autoimmune arthritis demonstrated hyperactive intracellular complement activation and interferon-γ production and CTSL inhibition corrected this deregulated phenotype. Importantly, intracellular C3a was observed in all examined cell populations, suggesting that intracellular complement activation might be of broad physiological significance.


Subject(s)
B-Lymphocyte Subsets/cytology , CD4-Positive T-Lymphocytes/immunology , Cathepsin L/metabolism , Cell Differentiation , Complement Activation/physiology , Complement C3/metabolism , Homeostasis/physiology , Adult , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cell Survival/immunology , Child , Complement C3/immunology , Complement C3a/metabolism , Complement C3b/metabolism , Gene Expression Regulation/immunology , Humans
4.
J Biol Chem ; 294(13): 4878-4888, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30670593

ABSTRACT

ER protein 57 (ERp57), a thiol isomerase secreted from vascular cells, is essential for complete thrombus formation in vivo, but other extracellular ERp57 functions remain unexplored. Here, we employed a kinetic substrate-trapping approach to identify extracellular protein substrates of ERp57 in platelet-rich plasma. MS-based identification with immunochemical confirmation combined with gene ontology enrichment analysis revealed that ERp57 targets, among other substrates, components of the lectin pathway of complement activation: mannose-binding lectin, ficolin-2, ficolin-3, collectin-10, collectin-11, mannose-binding lectin-associated serine protease-1, and mannose-binding lectin-associated serine protease-2. Ficolin-3, the most abundant lectin pathway initiator in humans, circulates as disulfide-linked multimers of a monomer. ERp57 attenuated ficolin-3 ligand recognition and complement activation by cleaving intermolecular disulfide bonds in large ficolin-3 multimers, thereby reducing multimer size and ligand-binding affinity. We used MS to identify the disulfide-bonding pattern in ficolin-3 multimers and the disulfide bonds targeted by ERp57 and found that Cys6 and Cys23 in the N-terminal region of ficolin-3 form the intermolecular disulfide bonds in ficolin-3 multimers that are reduced by ERp57. Our results not only demonstrate that ERp57 can negatively regulate complement activation, but also identify a control mechanism for lectin pathway initiation in the vasculature. We conclude that extensive multimerization in large ficolin-3 multimers leads to a high affinity for ligands and strong complement-activating potential and that ERp57 suppresses complement activation by cleaving disulfide bonds in ficolin-3 and reducing its multimer size.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Glycoproteins/metabolism , Lectins/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Multimerization , Proteolysis , Glycoproteins/genetics , Humans , Lectins/genetics , Protein Disulfide-Isomerases/genetics
5.
EMBO J ; 35(10): 1133-49, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27013439

ABSTRACT

Regulators of complement activation (RCA) inhibit complement-induced immune responses on healthy host tissues. We present crystal structures of human RCA (MCP, DAF, and CR1) and a smallpox virus homolog (SPICE) bound to complement component C3b. Our structural data reveal that up to four consecutive homologous CCP domains (i-iv), responsible for inhibition, bind in the same orientation and extended arrangement at a shared binding platform on C3b. Large sequence variations in CCP domains explain the diverse C3b-binding patterns, with limited or no contribution of some individual domains, while all regulators show extensive contacts with C3b for the domains at the third site. A variation of ~100° rotation around the longitudinal axis is observed for domains binding at the fourth site on C3b, without affecting the overall binding mode. The data suggest a common evolutionary origin for both inhibitory mechanisms, called decay acceleration and cofactor activity, with variable C3b binding through domains at sites ii, iii, and iv, and provide a framework for understanding RCA disease-related mutations and immune evasion.


Subject(s)
Complement C3b/chemistry , Complement C3b/metabolism , Binding Sites , CD55 Antigens/chemistry , CD55 Antigens/metabolism , Complement Activation , Humans , Membrane Cofactor Protein/chemistry , Membrane Cofactor Protein/metabolism , Protein Domains , Receptors, Complement 3b/chemistry , Receptors, Complement 3b/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism
6.
Immunol Rev ; 274(1): 9-15, 2016 11.
Article in English | MEDLINE | ID: mdl-27782327

ABSTRACT

The complement system is an evolutionarily ancient component of immunity that revolves around the central component C3. With the recent description of intracellular C3 stores in many types of human cells, our view of the complement system has expanded. In this article, we hypothesize that a primitive version of C3 comprised the first element of the original complement system and initially functioned intracellularly and on the membrane of single-celled organisms. With increasing specialization and multicellularity, C3 evolved a secretory capacity that allowed it to play a protective role in the interstitial space. Upon development of a pumped circulatory system, C3 was synthesized in large amounts and secreted by the liver to protect the intravascular space. Recent discoveries of intracellular C3 activation, a C3-based recycling pathway and C3 being a driver and programmer of cell metabolism suggest that the complement system utilizes C3 to guard not only extracellular but also the intracellular environment. We predict that the major functions of C3 in all four locations (i.e. intracellular, membrane, interstitium and circulation) are similar: opsonization, membrane perturbation, triggering inflammation, and metabolic reprogramming.


Subject(s)
Biological Evolution , Blood Vessels/physiology , Complement C3/immunology , Inflammation/immunology , Liver/physiology , Animals , Cell Membrane/metabolism , Complement Activation , Extracellular Space , Humans , Immunity , Intracellular Space , Models, Immunological
7.
Am J Respir Cell Mol Biol ; 60(2): 144-157, 2019 02.
Article in English | MEDLINE | ID: mdl-30156437

ABSTRACT

The complement system provides host defense against pathogens and environmental stress. C3, the central component of complement, is present in the blood and increases in BAL fluid after injury. We recently discovered that C3 is taken up by certain cell types and cleaved intracellularly to C3a and C3b. C3a is required for CD4+ T-cell survival. These observations made us question whether complement operates at environmental interfaces, particularly in the respiratory tract. We found that airway epithelial cells (AECs, represented by both primary human tracheobronchial cells and BEAS-2B [cell line]) cultured in C3-free media were unique from other cell types in that they contained large intracellular stores of de novo synthesized C3. A fraction of this protein reduced ("storage form") but the remainder did not, consistent with it being pro-C3 ("precursor form"). These two forms of intracellular C3 were absent in CRISPR knockout-induced C3-deficient AECs and decreased with the use of C3 siRNA, indicating endogenous generation. Proinflammatory cytokine exposure increased both stored and secreted forms of C3. Furthermore, AECs took up C3 from exogenous sources, which mitigated stress-associated cell death (e.g., from oxidative stress or starvation). C3 stores were notably increased within AECs in lung tissues from individuals with different end-stage lung diseases. Thus, at-risk cells furnish C3 through biosynthesis and/or uptake to increase locally available C3 during inflammation, while intracellularly, these stores protect against certain inducers of cell death. These results establish the relevance of intracellular C3 to airway epithelial biology and suggest novel pathways for complement-mediated host protection in the airway.


Subject(s)
Bronchi/cytology , Complement C3/metabolism , Epithelial Cells/physiology , Cell Death , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Complement C3/genetics , Epithelial Cells/cytology , Epithelial Cells/drug effects , Goblet Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , Lung Diseases/metabolism , Lung Diseases/pathology , Stress, Physiological
8.
Kidney Int ; 90(1): 123-34, 2016 07.
Article in English | MEDLINE | ID: mdl-27236750

ABSTRACT

Atypical hemolytic uremic syndrome and thrombotic thrombocytopenic purpura have traditionally been considered separate entities. Defects in the regulation of the complement alternative pathway occur in atypical hemolytic uremic syndrome, and defects in the cleavage of von Willebrand factor (VWF)-multimers arise in thrombotic thrombocytopenic purpura. However, recent studies suggest that both entities are related as defects in the disease-causing pathways overlap or show functional interactions. Here we investigate the possible functional link of VWF-multimers and the complement system on endothelial cells. Blood outgrowth endothelial cells (BOECs) were obtained from 3 healthy individuals and 2 patients with Type 3 von Willebrand disease lacking VWF. Cells were exposed to a standardized complement challenge via the combination of classical and alternative pathway activation and 50% normal human serum resulting in complement fixation to the endothelial surface. Under these conditions we found the expected release of VWF-multimers causing platelet adhesion onto BOECs from healthy individuals. Importantly, in BOECs derived from patients with von Willebrand disease complement C3c deposition and cytotoxicity were more pronounced than on BOECs derived from normal individuals. This is of particular importance as primary glomerular endothelial cells display a heterogeneous expression pattern of VWF with overall reduced VWF abundance. Thus, our results support a mechanistic link between VWF-multimers and the complement system. However, our findings also identify VWF as a new complement regulator on vascular endothelial cells and suggest that VWF has a protective effect on endothelial cells and complement-mediated injury.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement Pathway, Alternative/immunology , Endothelial Cells/immunology , Purpura, Thrombotic Thrombocytopenic/immunology , von Willebrand Factor/metabolism , Blood Platelets/immunology , Cell Adhesion/immunology , Complement C3c/metabolism , Humans , Kidney Glomerulus/cytology , Primary Cell Culture , von Willebrand Disease, Type 3/blood
9.
Hum Genomics ; 9: 7, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26054645

ABSTRACT

Membrane cofactor protein (MCP; CD46) is an ubiquitously expressed complement regulatory protein that protects host cells from injury by complement. This type-I membrane glycoprotein serves as a cofactor for the serine protease factor I to mediate inactivation of C3b and C4b deposited on host cells. More than 60 disease-associated mutations in MCP have now been identified. The majority of the mutations are linked to a rare thrombotic microangiopathic-based disease, atypical hemolytic uremic syndrome (aHUS), but new putative links to systemic lupus erythematosus, glomerulonephritis, and pregnancy-related disorders among others have also been identified. This review summarizes our current knowledge of disease-associated mutations in this complement inhibitor.


Subject(s)
Glomerulonephritis/genetics , Lupus Erythematosus, Systemic/genetics , Membrane Cofactor Protein/genetics , Complement C3b/genetics , Complement C3b/metabolism , Complement C4b/genetics , Complement C4b/metabolism , Female , Glomerulonephritis/pathology , Humans , Lupus Erythematosus, Systemic/pathology , Membrane Cofactor Protein/metabolism , Mutation , Pregnancy , Pregnancy Complications/genetics
11.
Nat Genet ; 39(9): 1068-70, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17660820

ABSTRACT

Autosomal dominant retinal vasculopathy with cerebral leukodystrophy is a microvascular endotheliopathy with middle-age onset. In nine families, we identified heterozygous C-terminal frameshift mutations in TREX1, which encodes a 3'-5' exonuclease. These truncated proteins retain exonuclease activity but lose normal perinuclear localization. These data have implications for the maintenance of vascular integrity in the degenerative cerebral microangiopathies leading to stroke and dementias.


Subject(s)
Brain Diseases/genetics , Exodeoxyribonucleases/genetics , Mutation , Phosphoproteins/genetics , Retinal Diseases/genetics , Amino Acid Sequence , Brain Diseases/enzymology , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Genes, Dominant , Genetic Predisposition to Disease , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Molecular Sequence Data , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Diseases/enzymology , Sequence Homology, Amino Acid , Transfection
12.
J Biol Chem ; 289(1): 450-63, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24214979

ABSTRACT

To survive and replicate within the human host, malaria parasites must invade erythrocytes. Invasion can be mediated by the P. falciparum reticulocyte-binding homologue protein 4 (PfRh4) on the merozoite surface interacting with complement receptor type 1 (CR1, CD35) on the erythrocyte membrane. The PfRh4 attachment site lies within the three N-terminal complement control protein modules (CCPs 1-3) of CR1, which intriguingly also accommodate binding and regulatory sites for the key complement activation-specific proteolytic products, C3b and C4b. One of these regulatory activities is decay-accelerating activity. Although PfRh4 does not impact C3b/C4b binding, it does inhibit this convertase disassociating capability. Here, we have employed ELISA, co-immunoprecipitation, and surface plasmon resonance to demonstrate that CCP 1 contains all the critical residues for PfRh4 interaction. We fine mapped by homologous substitution mutagenesis the PfRh4-binding site on CCP 1 and visualized it with a solution structure of CCPs 1-3 derived by NMR and small angle x-ray scattering. We cross-validated these results by creating an artificial PfRh4-binding site through substitution of putative PfRh4-interacting residues from CCP 1 into their homologous positions within CCP 8; strikingly, this engineered binding site had an ∼30-fold higher affinity for PfRh4 than the native one in CCP 1. These experiments define a candidate site on CR1 by which P. falciparum merozoites gain access to human erythrocytes in a non-sialic acid-dependent pathway of merozoite invasion.


Subject(s)
Membrane Proteins/metabolism , Merozoites/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Receptors, Complement 3b/metabolism , Binding Sites , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/metabolism , Complement C4b/chemistry , Complement C4b/genetics , Complement C4b/metabolism , Erythrocytes/chemistry , Erythrocytes/metabolism , Erythrocytes/parasitology , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Merozoites/chemistry , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Receptors, Complement 3b/chemistry , Receptors, Complement 3b/genetics , Scattering, Small Angle , Surface Plasmon Resonance , X-Ray Diffraction
13.
Eur J Immunol ; 43(6): 1617-29, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23508668

ABSTRACT

Since a tightly regulated complement system is needed for a successful pregnancy, we hypothesized that alterations in complement inhibitors may be associated with idiopathic, recurrent miscarriage. We sequenced all exons coding for three complement inhibitors: C4b-binding protein (C4BP), CD46, and CD55 in 384 childless women with at least two miscarriages that could not be explained by known risk factors. Several alterations were found in C4BPA, of which the R120H, I126T, and the G423T mutations affected the expression level and/or the ability of recombinant C4BP to serve as cofactor for factor I. The only variant in C4BPB was located in the C-terminal part, and did not impair the polymerization of the molecule. Our results identify for the first time alterations in C4BP in women experiencing recurrent miscarriages. We also found four CD46 alterations in individual patients that were not found in healthy controls. One of the rare variants, P324L, showed decreased expression, whereas N213I resulted in deficient protein processing as well as an impaired cofactor activity in the degradation of both C4b and C3b. The identified alterations may result in in vivo consequences and contribute to the disorder but the degree of association must be evaluated in larger cohorts.


Subject(s)
Abortion, Habitual/genetics , Abortion, Habitual/immunology , CD55 Antigens/genetics , Complement C4b-Binding Protein/genetics , Membrane Cofactor Protein/genetics , Adult , DNA Mutational Analysis , Female , Fibrinogen/metabolism , Humans , Mutation/genetics , Polymorphism, Genetic , Pregnancy
15.
Genet Med ; 15(4): 310-3, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23037936

ABSTRACT

PURPOSE: Autosomal recessive congenital short bowel syndrome is caused by mutations in CLMP. No mutations were found in the affected males of a family with presumed X-linked congenital short bowel syndrome or in an isolated male patient. Our aim was to identify the disease-causing mutation in these patients. METHODS: We performed mutation analysis of the second exon of FLNA in the two surviving affected males of the presumed X-linked family and in the isolated patient. RESULTS: We identified a novel 2-base-pair deletion in the second exon of FLNA in all these male patients. The deletion is located between two nearby methionines at the N-terminus of filamin A. Previous studies showed that translation of FLNA occurs from both methionines, resulting in two isoforms of the protein. We hypothesized that the longer isoform is no longer translated due to the mutation and that this mutation is therefore not lethal for males in utero. CONCLUSION: Our findings emphasize that congenital short bowel syndrome can be the presenting symptom in male patients with mutations in FLNA.


Subject(s)
Filamins/genetics , Mutation , Short Bowel Syndrome/diagnosis , Short Bowel Syndrome/genetics , Adolescent , Adult , Base Sequence , Exons , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Humans , Male , Pedigree , Phenotype , Sequence Deletion , Young Adult
16.
Hum Pathol ; 135: 22-34, 2023 05.
Article in English | MEDLINE | ID: mdl-36871865

ABSTRACT

Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) is a rare autosomal dominant disease resulting from a frame-shift mutation in TREX1, an intracellular 3'-5' exonuclease 1. Hepatic findings include an elevated alkaline phosphatase (ALP) and nodular regenerative hyperplasia (NRH). Affected individuals typically succumb to brain lesions before clinically apparent hepatic manifestations; thus, little else is known about the hepatic pathology. Autopsy reports and a liver section from each (n = 11) of three unrelated kindreds with the most common mutation in TREX1 (V235Gfs∗6) were studied with standard and immunohistochemical stains. Cases were compared with "normal liver" controls from similar autopsy years. Cases consisted of six men and five women who died at a median age of 50 yr (range, 41-60 yr.). Seven had elevated ALP. Two had liver atrophy. Foci of NRH were variably detected in all. Inhomogeneous distribution of other findings included patternless parenchymal fibrous bands, approximation of vascular structures, and commonly, architectural changes of vascular structures. Only bile duct epithelia were unaffected. In addition, small trichrome-positive nodules were found along vein walls or isolated in the parenchyma. Rare foci of non-NRH hepatocytic nodules were noted in 3. Increased CD34 and altered α-SMA IHC expression were variably noted. Periportal ductules and perivenular K7 IHC expression were increased to unpredictable degrees. The extensive but inhomogeneous histopathologic findings in livers of autopsied patients with RVCL-S appear to involve hepatic vascular structures. These findings validate inclusion of vascular liver involvement beyond NRH in this complex hereditary disorder.


Subject(s)
Leukoencephalopathies , Liver Diseases , Vascular Diseases , Male , Humans , Female , Hyperplasia/pathology , Liver/pathology , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Vascular Diseases/genetics , Vascular Diseases/pathology , Liver Diseases/genetics , Liver Diseases/pathology
17.
PLoS Med ; 8(3): e1001013, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21445332

ABSTRACT

BACKGROUND: Pregnancy in women with systemic lupus erythematosus (SLE) or antiphospholipid antibodies (APL Ab)--autoimmune conditions characterized by complement-mediated injury--is associated with increased risk of preeclampsia and miscarriage. Our previous studies in mice indicate that complement activation targeted to the placenta drives angiogenic imbalance and placental insufficiency. METHODS AND FINDINGS: We use PROMISSE, a prospective study of 250 pregnant patients with SLE and/or APL Ab, to test the hypothesis in humans that impaired capacity to limit complement activation predisposes to preeclampsia. We sequenced genes encoding three complement regulatory proteins--membrane cofactor protein (MCP), complement factor I (CFI), and complement factor H (CFH)--in 40 patients who had preeclampsia and found heterozygous mutations in seven (18%). Five of these patients had risk variants in MCP or CFI that were previously identified in atypical hemolytic uremic syndrome, a disease characterized by endothelial damage. One had a novel mutation in MCP that impairs regulation of C4b. These findings constitute, to our knowledge, the first genetic defects associated with preeclampsia in SLE and/or APL Ab. We confirmed the association of hypomorphic variants of MCP and CFI in a cohort of non-autoimmune preeclampsia patients in which five of 59 were heterozygous for mutations. CONCLUSION: The presence of risk variants in complement regulatory proteins in patients with SLE and/or APL Ab who develop preeclampsia, as well as in preeclampsia patients lacking autoimmune disease, links complement activation to disease pathogenesis and suggests new targets for treatment of this important public health problem. STUDY REGISTRATION: ClinicalTrials.gov NCT00198068.


Subject(s)
Complement System Proteins/genetics , Genetic Predisposition to Disease , Mutation/genetics , Pre-Eclampsia/genetics , Pre-Eclampsia/immunology , Adult , Animals , Autoimmune Diseases/complications , Autoimmune Diseases/immunology , Biomarkers/metabolism , Cohort Studies , Complement Activation/immunology , Complement C3b/metabolism , Complement C4b/metabolism , Complement Factor H/genetics , Complement Factor I/genetics , Complement System Proteins/immunology , Female , Humans , Membrane Cofactor Protein/genetics , Mice , Pregnancy , Pregnancy Outcome , Young Adult
18.
J Immunol ; 183(5): 3150-9, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19667083

ABSTRACT

Although smallpox was eradicated as a global illness more than 30 years ago, variola virus and other related pathogenic poxviruses, such as monkeypox, remain potential bioterrorist weapons or could re-emerge as natural infections. Poxviruses express virulence factors that down-modulate the host's immune system. We previously compared functional profiles of the poxviral complement inhibitors of smallpox, vaccinia, and monkeypox known as SPICE, VCP (or VICE), and MOPICE, respectively. SPICE was the most potent regulator of human complement and attached to cells via glycosaminoglycans. The major goals of the present study were to further characterize the complement regulatory and heparin binding sites of SPICE and to evaluate a mAb that abrogates its function. Using substitution mutagenesis, we established that (1) elimination of the three heparin binding sites severely decreases but does not eliminate glycosaminoglycan binding, (2) there is a hierarchy of activity for heparin binding among the three sites, and (3) complement regulatory sites overlap with each of the three heparin binding motifs. By creating chimeras with interchanges of SPICE and VCP residues, a combination of two SPICE amino acids (H77 plus K120) enhances VCP activity approximately 200-fold. Also, SPICE residue L131 is critical for both complement regulatory function and accounts for the electrophoretic differences between SPICE and VCP. An evolutionary history for these structure-function adaptations of SPICE is proposed. Finally, we identified and characterized a mAb that inhibits the complement regulatory activity of SPICE, MOPICE, and VCP and thus could be used as a therapeutic agent.


Subject(s)
Complement Activating Enzymes/antagonists & inhibitors , Complement Activating Enzymes/metabolism , Variola virus/immunology , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/physiology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Binding Sites/genetics , Binding Sites/immunology , Binding Sites, Antibody , CHO Cells , Complement Activating Enzymes/genetics , Complement C3b/metabolism , Cricetinae , Cricetulus , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/metabolism , Heparin/metabolism , Humans , Hybridomas , Mice , Molecular Sequence Data , Point Mutation , Variola virus/genetics , Variola virus/pathogenicity , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virulence Factors/antagonists & inhibitors , Virulence Factors/genetics , Virulence Factors/physiology
19.
Curr Opin Immunol ; 72: 126-134, 2021 10.
Article in English | MEDLINE | ID: mdl-34004375

ABSTRACT

Membrane cofactor protein (MCP; CD46), a ubiquitously expressed complement regulatory protein, serves as a cofactor for serine protease factor I to cleave and inactivate C3b and C4b deposited on host cells. However, CD46 also plays roles in human reproduction, autophagy, modulating T cell activation and effector functions and is a member of the newly identified intracellular complement system (complosome). CD46 also is a receptor for 11 pathogens ('pathogen magnet'). While CD46 deficiencies contribute to inflammatory disorders, its overexpression in cancers and role as a receptor for some adenoviruses has led to its targeting by oncolytic agents and adenoviral-based therapeutic vectors, including coronavirus disease of 2019 (COVID-19) vaccines. This review focuses on recent advances in identifying disease-causing CD46 variants and its pathogen connections.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Membrane Cofactor Protein/metabolism , SARS-CoV-2/physiology , T-Lymphocytes/immunology , Animals , Autophagy , Complement Activation , Host-Pathogen Interactions , Humans , Lymphocyte Activation , Membrane Cofactor Protein/genetics , Oncolytic Virotherapy , Polymorphism, Genetic , Reproduction
20.
Viruses ; 13(7)2021 06 24.
Article in English | MEDLINE | ID: mdl-34202570

ABSTRACT

Dengue is a mosquito-borne viral disease causing significant health and economic burdens globally. The dengue virus (DENV) comprises four serotypes (DENV1-4). Usually, the primary infection is asymptomatic or causes mild dengue fever (DF), while secondary infections with a different serotype increase the risk of severe dengue disease (dengue hemorrhagic fever, DHF). Complement system activation induces inflammation and tissue injury, contributing to disease pathogenesis. However, in asymptomatic or primary infections, protective immunity largely results from the complement system's lectin pathway (LP), which is activated through foreign glycan recognition. Differences in N-glycans displayed on the DENV envelope membrane influence the lectin pattern recognition receptor (PRR) binding efficiency. The important PRR, mannan binding lectin (MBL), mediates DENV neutralization through (1) a complement activation-independent mechanism via direct MBL glycan recognition, thereby inhibiting DENV attachment to host target cells, or (2) a complement activation-dependent mechanism following the attachment of complement opsonins C3b and C4b to virion surfaces. The serum concentrations of lectin PRRs and their polymorphisms influence these LP activities. Conversely, to escape the LP attack and enhance the infectivity, DENV utilizes the secreted form of nonstructural protein 1 (sNS1) to counteract the MBL effects, thereby increasing viral survival and dissemination.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Dengue Virus/immunology , Dengue Virus/pathogenicity , Dengue/immunology , Dengue/virology , Animals , Humans , Immune Evasion , Mannose-Binding Lectin/blood , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Mannose-Binding Lectin/metabolism , Polymorphism, Single Nucleotide , Polysaccharides/immunology , Polysaccharides/metabolism , Receptors, Pattern Recognition/blood , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Severe Dengue/immunology , Severe Dengue/virology , Viral Nonstructural Proteins/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL