Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 9(8): 909-925, 2021 08.
Article in English | MEDLINE | ID: mdl-34039652

ABSTRACT

The complement system is a powerful and druggable innate immune component of the tumor microenvironment. Nevertheless, it is challenging to elucidate the exact mechanisms by which complement affects tumor growth. In this study, we examined the processes by which the master complement regulator factor H (FH) affects clear cell renal cell carcinoma (ccRCC) and lung cancer, two cancers in which complement overactivation predicts poor prognosis. FH was present in two distinct cellular compartments: the membranous (mb-FH) and intracellular (int-FH) compartments. Int-FH resided in lysosomes and colocalized with C3. In ccRCC and lung adenocarcinoma, FH exerted protumoral action through an intracellular, noncanonical mechanism. FH silencing in ccRCC cell lines resulted in decreased proliferation, due to cell-cycle arrest and increased mortality, and this was associated with increased p53 phosphorylation and NFκB translocation to the nucleus. Moreover, the migration of the FH-silenced cells was reduced, likely due to altered morphology. These effects were cell type-specific because no modifications occurred upon CFH silencing in other FH-expressing cells tested: tubular cells (from which ccRCC originates), endothelial cells (human umbilical vein endothelial cells), and squamous cell lung cancer cells. Consistent with this, in ccRCC and lung adenocarcinoma, but not in lung squamous cell carcinoma, int-FH conferred poor prognosis in patient cohorts. Mb-FH performed its canonical function of complement regulation but had no impact on tumor cell phenotype or patient survival. The discovery of intracellular functions for FH redefines the role of the protein in tumor progression and its use as a prognostic biomarker or potential therapeutic target.See article by Daugan et al., p. 891 (36).


Subject(s)
Complement Activation/genetics , Complement Factor H/genetics , Animals , Cell Line , Disease Progression , Humans , Mice
2.
Cancer Immunol Res ; 9(8): 891-908, 2021 08.
Article in English | MEDLINE | ID: mdl-34039653

ABSTRACT

The complement system plays a complex role in cancer. In clear cell renal cell carcinoma (ccRCC), local production of complement proteins drives tumor progression, but the mechanisms by which they do this are poorly understood. We found that complement activation, as reflected by high plasma C4d or as C4d deposits at the tumor site, was associated with poor prognosis in two cohorts of patients with ccRCC. High expression of the C4-activating enzyme C1s by tumor cells was associated with poor prognosis in three cohorts. Multivariate Cox analysis revealed that the prognostic value of C1s was independent from complement deposits, suggesting the possibility of complement cascade-unrelated, protumoral functions for C1s. Silencing of C1s in cancer cell lines resulted in decreased proliferation and viability of the cells and in increased activation of T cells in in vitro cocultures. Tumors expressing high levels of C1s showed high infiltration of macrophages and T cells. Modification of the tumor cell phenotype and T-cell activation were independent of extracellular C1s levels, suggesting that C1s was acting in an intracellular, noncanonical manner. In conclusion, our data point to C1s playing a dual role in promoting ccRCC progression by triggering complement activation and by modulating the tumor cell phenotype and tumor microenvironment in a complement cascade-independent, noncanonical manner. Overexpression of C1s by tumor cells could be a new escape mechanism to promote tumor progression.See related Spotlight by Magrini and Garlanda, p. 855. See article by Daugan et al., p. 909 (40).


Subject(s)
Biomarkers, Tumor/metabolism , Complement C1s/metabolism , Complement C4/metabolism , Kidney Neoplasms/genetics , Animals , Case-Control Studies , Humans , Mice , Prognosis , Prospective Studies , Transfection
3.
Front Immunol ; 11: 1772, 2020.
Article in English | MEDLINE | ID: mdl-32849636

ABSTRACT

Intravascular hemolysis of any cause can induce acute kidney injury (AKI). Hemolysis-derived product heme activates the innate immune complement system and contributes to renal damage. Therefore, we explored the role of the master complement regulator Factor H (FH) in the kidney's resistance to hemolysis-mediated AKI. Acute systemic hemolysis was induced in mice lacking liver expression of FH (hepatoFH-/-, ~20% residual FH) and in WT controls, by phenylhydrazine injection. The impaired complement regulation in hepatoFH-/- mice resulted in a delayed but aggravated phenotype of hemolysis-related kidney injuries. Plasma urea as well as markers for tubular (NGAL, Kim-1) and vascular aggression peaked at day 1 in WT mice and normalized at day 2, while they increased more in hepatoFH-/- compared to the WT and still persisted at day 4. These were accompanied by exacerbated tubular dilatation and the appearance of tubular casts in the kidneys of hemolytic hepatoFH-/- mice. Complement activation in hemolytic mice occurred in the circulation and C3b/iC3b was deposited in glomeruli in both strains. Both genotypes presented with positive staining of FH in the glomeruli, but hepatoFH-/- mice had reduced staining in the tubular compartment. Despite the clear phenotype of tubular injury, no complement activation was detected in the tubulointerstitium of the phenylhydrazin-injected mice irrespective of the genotype. Nevertheless, phenylhydrazin triggered overexpression of C5aR1 in tubules, predominantly in hepatoFH-/- mice. Moreover, C5b-9 was deposited only in the glomeruli of the hemolytic hepatoFH-/- mice. Therefore, we hypothesize that C5a, generated in the glomeruli, could be filtered into the tubulointerstitium to activate C5aR1 expressed by tubular cells injured by hemolysis-derived products and will aggravate the tissue injury. Plasma-derived FH is critical for the tubular protection, since pre-treatment of the hemolytic hepatoFH-/- mice with purified FH attenuated the tubular injury. Worsening of acute tubular necrosis in the hepatoFH-/- mice was trigger-dependent, as it was also observed in LPS-induced septic AKI model but not in chemotherapy-induced AKI upon cisplatin injection. In conclusion, plasma FH plays a key role in protecting the kidneys, especially the tubules, against hemolysis-mediated injury. Thus, FH-based molecules might be explored as promising therapeutic agents in a context of AKI.


Subject(s)
Complement Activation , Complement Factor H/metabolism , Hemolysis , Hepatocytes/metabolism , Kidney Glomerulus/metabolism , Kidney Tubular Necrosis, Acute/prevention & control , Kidney Tubules/metabolism , Animals , Complement C5a/genetics , Complement C5a/metabolism , Complement Factor H/genetics , Disease Models, Animal , Gene Expression Regulation , Kidney Glomerulus/pathology , Kidney Tubular Necrosis, Acute/blood , Kidney Tubular Necrosis, Acute/chemically induced , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubules/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenylhydrazines , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction
4.
J Am Soc Nephrol ; 31(4): 829-840, 2020 04.
Article in English | MEDLINE | ID: mdl-32034108

ABSTRACT

BACKGROUND: The pathophysiology of the leading cause of pediatric acute nephritis, acute postinfectious GN, including mechanisms of the pathognomonic transient complement activation, remains uncertain. It shares clinicopathologic features with C3 glomerulopathy, a complement-mediated glomerulopathy that, unlike acute postinfectious GN, has a poor prognosis. METHODS: This retrospective study investigated mechanisms of complement activation in 34 children with acute postinfectious GN and low C3 level at onset. We screened a panel of anticomplement protein autoantibodies, carried out related functional characterization, and compared results with those of 60 children from the National French Registry who had C3 glomerulopathy and persistent hypocomplementemia. RESULTS: All children with acute postinfectious GN had activation of the alternative pathway of the complement system. At onset, autoantibodies targeting factor B (a component of the alternative pathway C3 convertase) were found in a significantly higher proportion of children with the disorder versus children with hypocomplementemic C3 glomerulopathy (31 of 34 [91%] versus 4 of 28 [14%], respectively). In acute postinfectious GN, anti-factor B autoantibodies were transient and correlated with plasma C3 and soluble C5b-9 levels. We demonstrated that anti-factor B antibodies enhance alternative pathway convertase activity in vitro, confirming their pathogenic effect. We also identified crucial antibody binding sites on factor B, including one correlated to disease severity. CONCLUSIONS: These findings elucidate the pathophysiologic mechanisms underlying acute postinfectious GN by identifying anti-factor B autoantibodies as contributing factors in alternative complement pathway activation. At onset of a nephritic syndrome with low C3 level, screening for anti-factor B antibodies might help guide indications for kidney biopsy to avoid misdiagnosed chronic glomerulopathy, such as C3 glomerulopathy, and to help determine therapy.


Subject(s)
Autoantibodies/blood , Complement Activation/physiology , Complement C3/metabolism , Complement Factor B/immunology , Glomerulonephritis/blood , Glomerulonephritis/diagnosis , Child , Child, Preschool , Complement C3 Nephritic Factor/metabolism , Female , France , Humans , Male , Retrospective Studies
5.
Proc Natl Acad Sci U S A ; 116(13): 6280-6285, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30850533

ABSTRACT

Hemolytic diseases are frequently linked to multiorgan failure subsequent to vascular damage. Deciphering the mechanisms leading to organ injury upon hemolytic event could bring out therapeutic approaches. Complement system activation occurs in hemolytic disorders, such as sickle cell disease, but the pathological relevance and the acquisition of a complement-activating phenotype during hemolysis remain unclear. Here we found that intravascular hemolysis, induced by injection of phenylhydrazine, resulted in increased alanine aminotransferase plasma levels and NGAL expression. This liver damage was at least in part complement-dependent, since it was attenuated in complement C3-/- mice and by injection of C5-blocking antibody. We evidenced C3 activation fragments' deposits on liver endothelium in mice with intravascular hemolysis or injected with heme as well as on cultured human endothelial cells (EC) exposed to heme. This process was mediated by TLR4 signaling, as revealed by pharmacological blockade and TLR4 deficiency in mice. Mechanistically, TLR4-dependent surface expression of P-selectin triggered an unconventional mechanism of complement activation by noncovalent anchoring of C3 activation fragments, including the typical fluid-phase C3(H2O), measured by surface plasmon resonance and flow cytometry. P-selectin blockade by an antibody prevented complement deposits and attenuated the liver stress response, measured by NGAL expression, in the hemolytic mice. In conclusion, these results revealed the critical impact of the triad TLR4/P-selectin/complement in the liver damage and its relevance for hemolytic diseases. We anticipate that blockade of TLR4, P-selectin, or the complement system could prevent liver injury in hemolytic diseases like sickle cell disease.


Subject(s)
Endothelium, Vascular/metabolism , Heme/metabolism , Hemolysis , P-Selectin/metabolism , Toll-Like Receptor 4/metabolism , Alanine Transaminase/blood , Anemia, Sickle Cell , Animals , Complement Activation , Complement C3/metabolism , Disease Models, Animal , Gene Silencing , Hemolysis/drug effects , Humans , Lipocalin-2/metabolism , Liver/injuries , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylhydrazines/antagonists & inhibitors , Signal Transduction , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics
6.
JCI Insight ; 3(12)2018 06 21.
Article in English | MEDLINE | ID: mdl-29925688

ABSTRACT

In hemolytic diseases, such as sickle cell disease (SCD), intravascular hemolysis results in the release of hemoglobin, heme, and heme-loaded membrane microvesicles in the bloodstream. Intravascular hemolysis is thus associated with inflammation and organ injury. Complement system can be activated by heme in vitro. We investigated the mechanisms by which hemolysis and red blood cell (RBC) degradation products trigger complement activation in vivo. In kidney biopsies of SCD nephropathy patients and a mouse model with SCD, we detected tissue deposits of complement C3 and C5b-9. Moreover, drug-induced intravascular hemolysis or injection of heme or hemoglobin in mice triggered C3 deposition, primarily in kidneys. Renal injury markers (Kim-1, NGAL) were attenuated in C3-/- hemolytic mice. RBC degradation products, such as heme-loaded microvesicles and heme, induced alternative and terminal complement pathway activation in sera and on endothelial surfaces, in contrast to hemoglobin. Heme triggered rapid P selectin, C3aR, and C5aR expression and downregulated CD46 on endothelial cells. Importantly, complement deposition was attenuated in vivo and in vitro by heme scavenger hemopexin. In conclusion, we demonstrate that intravascular hemolysis triggers complement activation in vivo, encouraging further studies on its role in SCD nephropathy. Conversely, heme inhibition using hemopexin may provide a novel therapeutic opportunity to limit complement activation in hemolytic diseases.


Subject(s)
Cell-Free System , Heme/metabolism , Hemolysis/physiology , Acute Kidney Injury , Anemia, Sickle Cell , Animals , Complement C3/metabolism , Complement Membrane Attack Complex/metabolism , Disease Models, Animal , Endothelial Cells , Erythrocytes , Female , Hemopexin/pharmacology , Hepatitis A Virus Cellular Receptor 1 , Kidney , Mice , Mice, Inbred C57BL , P-Selectin , Receptor, Anaphylatoxin C5a/metabolism , Receptors, G-Protein-Coupled/metabolism
7.
Front Immunol ; 9: 179, 2018.
Article in English | MEDLINE | ID: mdl-29545789

ABSTRACT

Intravascular erythrocyte destruction, accompanied by the release of pro-oxidative and pro-inflammatory components hemoglobin and heme, is a common event in the pathogenesis of numerous diseases with heterogeneous etiology and clinical features. A frequent adverse effect related to massive hemolysis is the renal injury and inflammation. Nevertheless, it is still unclear whether heme--a danger-associated molecular pattern--and ligand for TLR4 or upstream hemolysis-derived products are responsible for these effects. Well-characterized animal models of hemolysis with kidney impairment are needed to investigate how hemolysis drives kidney injury and to test novel therapeutic strategies. Here, we characterized the pathological processes leading to acute kidney injury and inflammation during massive intravascular hemolysis, using a mouse model of phenylhydrazine (PHZ)-triggered erythrocyte destruction. We observed profound changes in mRNA levels for markers of tubular damage (Kim-1, NGAL) and regeneration (indirect marker of tubular injury, Ki-67), and tissue and vascular inflammation (IL-6, E-selectin, P-selectin, ICAM-1) in kidneys of PHZ-treated mice, associated with ultrastructural signs of tubular injury. Moreover, mass spectrometry revealed presence of markers of tubular damage in urine, including meprin-α, cytoskeletal keratins, α-1-antitrypsin, and α-1-microglobulin. Signs of renal injury and inflammation rapidly resolved and the renal function was preserved, despite major changes in metabolic parameters of PHZ-injected animals. Mechanistically, renal alterations were largely heme-independent, since injection of free heme could not reproduce them, and scavenging heme with hemopexin in PHZ-administered mice could not prevent them. Reduced overall health status of the mice suggested multiorgan involvement. We detected amylasemia and amylasuria, two markers of acute pancreatitis. We also provide detailed characterization of renal manifestations associated with acute intravascular hemolysis, which may be mediated by hemolysis-derived products upstream of heme release. This analysis provides a platform for further investigations of hemolytic diseases and associated renal injury and the evaluation of novel therapeutic strategies that target intravascular hemolysis.


Subject(s)
Acute Kidney Injury/genetics , Acute Kidney Injury/immunology , Heme/metabolism , Hemolysis , Inflammation , Vascular Diseases/immunology , Acute Kidney Injury/chemically induced , Animals , Biomarkers/urine , Cells, Cultured , Disease Models, Animal , E-Selectin/genetics , Erythrocytes/drug effects , Female , Hepatitis A Virus Cellular Receptor 1/genetics , Human Umbilical Vein Endothelial Cells , Humans , Ki-67 Antigen/genetics , Kidney/pathology , Lipocalin-2/genetics , Mice , Mice, Inbred C57BL , Phenylhydrazines , Vascular Diseases/complications
8.
Front Immunol ; 9: 3008, 2018.
Article in English | MEDLINE | ID: mdl-30619356

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a severe disease characterized by microvascular endothelial cell (EC) lesions leading to thrombi formation, mechanical hemolysis and organ failure, predominantly renal. Complement system overactivation is a hallmark of aHUS. To investigate this selective susceptibility of the microvascular renal endothelium to complement attack and thrombotic microangiopathic lesions, we compared complement and cyto-protection markers on EC, from different vascular beds, in in vitro and in vivo models as well as in patients. No difference was observed for complement deposits or expression of complement and coagulation regulators between macrovascular and microvascular EC, either at resting state or after inflammatory challenge. After prolonged exposure to hemolysis-derived heme, higher C3 deposits were found on glomerular EC, in vitro and in vivo, compared with other EC in culture and in mice organs (liver, skin, brain, lungs and heart). This could be explained by a reduced complement regulation capacity due to weaker binding of Factor H and inefficient upregulation of thrombomodulin (TM). Microvascular EC also failed to upregulate the cytoprotective heme-degrading enzyme heme-oxygenase 1 (HO-1), normally induced by hemolysis products. Only HUVEC (Human Umbilical Vein EC) developed adaptation to heme, which was lost after inhibition of HO-1 activity. Interestingly, the expression of KLF2 and KLF4-known transcription factors of TM, also described as possible transcription modulators of HO-1- was weaker in micro than macrovascular EC under hemolytic conditions. Our results show that the microvascular EC, and especially glomerular EC, fail to adapt to the stress imposed by hemolysis and acquire a pro-coagulant and complement-activating phenotype. Together, these findings indicate that the vulnerability of glomerular EC to hemolysis is a key factor in aHUS, amplifying complement overactivation and thrombotic microangiopathic lesions.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement C3/immunology , Heme Oxygenase-1/metabolism , Heme/immunology , Kidney Glomerulus/immunology , Animals , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/pathology , Biopsy , Complement Activation , Disease Models, Animal , Disease Susceptibility/immunology , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Female , Heme/metabolism , Hemolysis/immunology , Human Umbilical Vein Endothelial Cells , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Kruppel-Like Factor 4 , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microvessels/cytology , Microvessels/immunology , Primary Cell Culture , Thrombomodulin/metabolism , Up-Regulation
9.
EMBO J ; 36(8): 1084-1099, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28264884

ABSTRACT

Properdin (FP) is an essential positive regulator of the complement alternative pathway (AP) providing stabilization of the C3 and C5 convertases, but its oligomeric nature challenges structural analysis. We describe here a novel FP deficiency (E244K) caused by a single point mutation which results in a very low level of AP activity. Recombinant FP E244K is monomeric, fails to support bacteriolysis, and binds weakly to C3 products. We compare this to a monomeric unit excised from oligomeric FP, which is also dysfunctional in bacteriolysis but binds the AP proconvertase, C3 convertase, C3 products and partially stabilizes the convertase. The crystal structure of such a FP-convertase complex suggests that the major contact between FP and the AP convertase is mediated by a single FP thrombospondin repeat and a small region in C3b. Small angle X-ray scattering indicates that FP E244K is trapped in a compact conformation preventing its oligomerization. Our studies demonstrate an essential role of FP oligomerization in vivo while our monomers enable detailed structural insight paving the way for novel modulators of complement.


Subject(s)
Complement C3-C5 Convertases/chemistry , Complement Pathway, Alternative , Properdin/chemistry , Protein Multimerization , Amino Acid Substitution , Complement C3-C5 Convertases/genetics , Complement C3-C5 Convertases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Mutation, Missense , Properdin/deficiency , Properdin/genetics , Properdin/metabolism , Protein Domains
10.
J Am Soc Nephrol ; 27(6): 1665-77, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26471127

ABSTRACT

C3 glomerulopathy is a recently described form of CKD. C3GN is a subtype of C3 glomerulopathy characterized by predominant C3 deposits in the glomeruli and is commonly the result of acquired or genetic abnormalities in the alternative pathway (AP) of the complement system. We identified and characterized the first mutation of the C3 gene (p. I734T) in two related individuals diagnosed with C3GN. Immunofluorescence and electron microscopy studies showed C3 deposits in the subendothelial space, associated with unusual deposits located near the complement receptor 1 (CR1)-expressing podocytes. In vitro, this C3 mutation exhibited decreased binding to CR1, resulting in less CR1-dependent cleavage of C3b by factor 1. Both patients had normal plasma C3 levels, and the mutant C3 interacted with factor B comparably to wild-type (WT) C3 to form a C3 convertase. Binding of mutant C3 to factor H was normal, but mutant C3 was less efficiently cleaved by factor I in the presence of factor H, leading to enhanced C3 fragment deposition on glomerular cells. In conclusion, our results reveal that a CR1 functional deficiency is a mechanism of intraglomerular AP dysregulation and could influence the localization of the glomerular C3 deposits.


Subject(s)
Complement C3/genetics , Glomerulonephritis, Membranoproliferative/genetics , Receptors, Complement 3b/genetics , Adult , Complement Factor H/genetics , Gene Expression Regulation , Humans , Male , Middle Aged , Pedigree
11.
Blood ; 125(15): 2359-69, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25608561

ABSTRACT

The pathogenesis of atypical hemolytic uremic syndrome (aHUS) is strongly linked to dysregulation of the alternative pathway of the complement system. Mutations in complement genes have been identified in about two-thirds of cases, with 5% to 15% being in C3. In this study, 23 aHUS-associated genetic changes in C3 were characterized relative to their interaction with the control proteins factor H (FH), membrane cofactor protein (MCP; CD46), and complement receptor 1 (CR1; CD35). In surface plasmon resonance experiments, 17 mutant recombinant proteins demonstrated a defect in binding to FH and/or MCP, whereas 2 demonstrated reduced binding to CR1. In the majority of cases, decreased binding affinity translated to a decrease in proteolytic inactivation (known as cofactor activity) of C3b via FH and MCP. These results were used to map the putative binding regions of C3b involved in the interaction with MCP and CR1 and interrogated relative to known FH binding sites. Seventy-six percent of patients with C3 mutations had low C3 levels that correlated with disease severity. This study expands our knowledge of the functional consequences of aHUS-associated C3 mutations relative to the interaction of C3 with complement regulatory proteins mediating cofactor activity.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/metabolism , Complement C3/genetics , Complement C3/metabolism , Protein Interaction Maps , Atypical Hemolytic Uremic Syndrome/pathology , Binding Sites , Cohort Studies , Complement C3/analysis , Female , Humans , Male , Models, Molecular , Mutation
12.
J Am Soc Nephrol ; 25(9): 2053-65, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24652797

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a genetic ultrarare renal disease associated with overactivation of the alternative pathway of complement. Four gain-of-function mutations that form a hyperactive or deregulated C3 convertase have been identified in Factor B (FB) ligand binding sites. Here, we studied the functional consequences of 10 FB genetic changes recently identified from different aHUS cohorts. Using several tests for alternative C3 and C5 convertase formation and regulation, we identified two gain-of-function and potentially disease-relevant mutations that formed either an overactive convertase (M433I) or a convertase resistant to decay by FH (K298Q). One mutation (R178Q) produced a partially cleaved protein with no ligand binding or functional activity. Seven genetic changes led to near-normal or only slightly reduced ligand binding and functional activity compared with the most common polymorphism at position 7, R7. Notably, none of the algorithms used to predict the disease relevance of FB mutations agreed completely with the experimental data, suggesting that in silico approaches should be undertaken with caution. These data, combined with previously published results, suggest that 9 of 15 FB genetic changes identified in patients with aHUS are unrelated to disease pathogenesis. This study highlights that functional assessment of identified nucleotide changes in FB is mandatory to confirm disease association.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Complement Factor B/genetics , Mutation , Amino Acid Substitution , Binding Sites/genetics , Complement C3 Convertase, Alternative Pathway/chemistry , Complement C3 Convertase, Alternative Pathway/genetics , Complement C3 Convertase, Alternative Pathway/metabolism , Complement C3b/metabolism , Complement C5 Convertase, Alternative Pathway/chemistry , Complement C5 Convertase, Alternative Pathway/genetics , Complement C5 Convertase, Alternative Pathway/metabolism , Complement Factor B/chemistry , Complement Factor B/metabolism , Complement Pathway, Alternative/genetics , Computer Simulation , Gene Frequency , Human Umbilical Vein Endothelial Cells , Humans , Ligands , Models, Molecular , Multiprotein Complexes/chemistry , Polymorphism, Genetic , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Nephrol Dial Transplant ; 28(11): 2899-907, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24009284

ABSTRACT

BACKGROUND: Atypical haemolytic uraemic syndrome (aHUS) is associated with dysfunction of the alternative pathway of complement. Disease activity subsides as renal failure progresses but recurs upon renal transplantation, indicating that viable renal tissue contributes to disease activity. We present evidence of cerebrovascular occlusive disease indicating that vascular injury may occur in the absence of kidneys. METHODS: A currently 12-year-old girl developed renal failure at the age of 20 months. She underwent bilateral nephrectomy and renal transplantation but lost the transplant due to recurrences. She was on haemodialysis for 7 years. At 10 years of age she developed a transient ischaemic attack. Imaging, genetic investigation and mutation characterization were performed. RESULTS: Imaging demonstrated occlusion and stenosis of the carotid arteries. Two complement mutations, a novel mutation in factor B and a previously described mutation in factor I, and the H3-factor H haplotype, were identified. The factor B mutation, L433S, did not induce excessive complement activation in vitro. Measurement of C3 degradation products indicated ongoing complement activation. In spite of the patient being anephric, treatment was initiated with eculizumab, a humanized anti-C5 antibody that blocks terminal complement activation. She underwent a successful kidney transplant 9 months later and has not developed a recurrence or progression of vascular stenosis 1 year later. CONCLUSIONS: The course of disease in this patient with aHUS suggests that complement-mediated vascular injury may occur in the total absence of renal tissue and overt recurrences. To our knowledge, this is the first description of eculizumab treatment in an anephric aHUS patient.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Cerebrovascular Disorders/drug therapy , Hemolytic-Uremic Syndrome/drug therapy , Kidney Transplantation , Nephrectomy/adverse effects , Renal Insufficiency/complications , Atypical Hemolytic Uremic Syndrome , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/pathology , Child , Complement C5/metabolism , Complement Factor B/genetics , Enzyme-Linked Immunosorbent Assay , Female , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/pathology , Humans , Magnetic Resonance Imaging , Mutation/genetics , Polymerase Chain Reaction , Prognosis , Renal Insufficiency/surgery , Surface Plasmon Resonance
14.
Mol Microbiol ; 56(4): 888-902, 2005 May.
Article in English | MEDLINE | ID: mdl-15853878

ABSTRACT

The presence of housekeeping gene promoters with a unique consensus sequence in Bacteroides fragilis, previously described by Bayley et al. (2000, FEMS Microbiol Lett 193: 149-154), suggested the existence of a particular primary sigma factor. The single rpoD-like gene observed in the B. fragilis genome, and similarly in those of other members of the Bacteroidetes phylum, was found to be essential. It encodes a protein, sigma(ABfr), of only 32.7 kDa that is produced with equal abundance during all phases of growth and was concluded to be the primary sigma factor. sigma(ABfr) and its orthologues in the Bacteroidetes are unusual primary sigma factors in that they lack region 1.1, have a unique signature made up of 29 strictly identical amino acids and are the only RpoD factors that cluster with the RpoS factors. Although binding to the Escherichia coli core RNA polymerase, sigma(ABfr) does not support transcription initiation from any promoter when it is part of the heterologous holoenzyme, while in the reconstituted homologous holoenzyme it does so only from typical B. fragilis, including rrs, promoters but not from the lacUV5 or RNA I promoters.


Subject(s)
Bacteroidetes/genetics , Gene Expression Regulation, Bacterial , Sigma Factor/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteroidetes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/classification , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic , Protein Conformation , Sequence Alignment , Sigma Factor/chemistry , Sigma Factor/classification , Sigma Factor/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...