Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Pediatrics ; 140(3)2017 Sep.
Article in English | MEDLINE | ID: mdl-28864711

ABSTRACT

We describe a case of invasive meningococcal disease due to a vaccine-preventable and penicillin-resistant strain in a fully immunized young adult on long-term complement inhibitor therapy and daily penicillin chemoprophylaxis. Eculizumab is a humanized monoclonal antibody that binds human complement C5 protein and inhibits the terminal complement pathway. It is currently recommended for the treatment of complement-mediated thrombotic microangiopathies. An unwanted complication of inhibiting complement, however, is an increased risk of invasive meningococcal disease. Here, we report the first case of meningococcal group B vaccine failure in a young adult receiving eculizumab for atypical hemolytic uremic syndrome. She developed invasive meningococcal disease due to a vaccine-preventable and penicillin-resistant meningococcal group B strain 4 months after receiving 2 doses of meningococcal group B vaccine while on oral penicillin prophylaxis against meningococcal infection.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Meningococcal Infections/prevention & control , Meningococcal Vaccines/adverse effects , Neisseria meningitidis, Serogroup B/immunology , Penicillin Resistance/immunology , Adult , Female , Humans , Penicillins/therapeutic use , Young Adult
2.
Clin Nephrol ; 86(10): 200-2, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27616760

ABSTRACT

Eculizumab, a terminal complement inhibitor, has recently been used successfully to both prevent and treat the recurrence of atypical hemolytic uremic syndrome (aHUS) post renal transplantation. We describe a case that highlights the need to monitor the effects of eculizumab on the complement system and in this case alter the dosage. Despite taking the standard recommended dose of eculizumab for an adult, this aHUS patient developed a low-grade thrombotic microangiopathy on biopsy within months of renal transplantation. Complement assays (trough CH50) showed small amounts of residual terminal pathway activity suggesting inadequate complement blockade on 1,200 mg eculizumab every 2 weeks. Following an increase in the dose of eculizumab to 1,500 mg every 2 weeks, lactate dehydrogenase (LDH), proteinuria, and creatinine decreased, and CH50 assay showed 0%. This case emphasizes the need to monitor clinical parameters and complement activity to ensure that adequate therapeutic blockade is achieved.
.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/prevention & control , Kidney Transplantation/adverse effects , Adult , Atypical Hemolytic Uremic Syndrome/etiology , Complement C3/antagonists & inhibitors , Complement C5/antagonists & inhibitors , Complement System Proteins , Drug Monitoring/methods , Humans , Male , Recurrence
3.
J Med Case Rep ; 9: 92, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25925370

ABSTRACT

INTRODUCTION: Thrombotic microangiopathies are a group of diseases presenting as microangiopathic hemolytic anemia, thrombocytopenia and end-organ dysfunction. As the role of the complement system was elucidated in atypical hemolytic uremic syndrome pathogenesis, eculizumab was successfully introduced into clinical practice. We present a large pedigree with multiple individuals carrying a functionally significant novel factor H mutation. We describe the proband's presentation following a presumed infectious trigger requiring plasma exchange and hemodialysis. CASE PRESENTATION: A 32-year-old Caucasian woman presented with pyrexia and headache lasting one week to our Emergency Department. She gave no history of diarrhea or other symptoms to account for her high temperature. She was not taking any medication. She was pyrexial (38°C), tachycardic (110 bpm) and hypertensive (160/110 mmHg). Her fundoscopy revealed grade IV hypertensive retinopathy. She had mild pretibial and periorbital edema, with oliguria (450 mL/day). She had a pregnancy one year previously, during which she had hypertension, proteinuria and edema, with successful delivery at term. Her mother had died in her early 30s with a clinical picture consistent with thrombotic microangiopathy. Her laboratory evaluation showed microangiopathic hemolytic anemia. After 22 sessions of plasma exchange, her lactate dehydrogenase levels started to climb. As a result, she was classified as plasma resistant and eculizumab therapy was instituted. Her lactate dehydrogenase level and platelet count normalized, and her renal function recovered after three months of dialysis. CONCLUSIONS: We demonstrate that, even in patients with atypical hemolytic uremic syndrome and prolonged dialysis dependence, recovery of renal function can be seen with eculizumab treatment. We suggest a treatment regime of at least three months prior to evaluation of efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Factor H/genetics , Complement Inactivating Agents/therapeutic use , Mutation , Adult , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/therapy , Female , Humans , Male , Pedigree , Plasma Exchange , Renal Dialysis , Thrombotic Microangiopathies/drug therapy
4.
Mol Immunol ; 67(1): 21-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25929733

ABSTRACT

C3 glomerulopathy describes a spectrum of disorders with glomerular pathology associated with C3 cleavage product deposition and with defective complement action and regulation (Fakhouri et al., 2010; Sethi et al., 2012b). Kidney biopsies from these patients show glomerular accumulation or deposition of C3 cleavage fragments, but no or minor deposition of immunoglobulins (Appel et al., 2005; D'Agati and Bomback, 2012; Servais et al., 2007; Sethi and Fervenza, 2011). At present the current situation asks for a better definition of the underlining disease mechanisms, for precise biomarkers, and for a treatment for this disease. The complement system is a self activating and propelling enzymatic cascade type system in which inactive, soluble plasma components are activated spontaneously and lead into an amplification loop (Zipfel and Skerka, 2009). Activation of the alternative pathway is spontaneous, occurs by default, and cascade progression leads to amplification by complement activators. The system however is self-controlled by multiple regulators and inhibitors, like Factor H that control cascade progression in fluid phase and on surfaces. The activated complement system generates a series of potent effector components and activation products, which damage foreign-, as well as modified self cells, recruit innate immune cells to the site of action, coordinate inflammation and the response of the adaptive immune system in form of B cells and T lymphocytes (Kohl, 2006; Medzhitov and Janeway, 2002; Ogden and Elkon, 2006; Carroll, 2004; Kemper and Atkinson, 2007; Morgan, 1999; Muller-Eberhard, 1986; Ricklin et al., 2010). Complement controls homeostasis and multiple reactions in the vertebrate organism including defense against microbial infections (Diaz-Guillen et al., 1999; Mastellos and Lambris, 2002; Nordahl et al., 2004; Ricklin et al., 2010). In consequence defective control of the spontaneous self amplifying cascade or regulation is associated with numerous human disorders (Ricklin and Lambris, 2007; Skerka and Zipfel, 2008; Zipfel et al., 2006). Understanding the exact action and regulation of this sophisticated homeotic cascade system is relevant to understand disease pathology of various complement associated human disorders. Furthermore this knowledge is relevant for a better diagnosis and appropriate therapy. At present diagnosis of C3 glomerulopathy is primarily based on the kidney biopsy, and histological, immmunohistological and electron microscopical evaluation (D'Agati and Bomback, 2012; Fakhouri et al., 2010; Medjeral-Thomas et al., 2014a,b; Sethi et al., 2012b). The challenge is to define the actual cause of the diverse glomerular changes or damages, to define how C3 deposition results in the reported glomerular changes, the location of the cell damage and the formation of deposits.


Subject(s)
Complement C3 Nephritic Factor/chemistry , Complement C3/chemistry , Complement C3b Inactivator Proteins/chemistry , Glomerulonephritis, Membranoproliferative/pathology , Kidney Glomerulus/pathology , Protein Aggregation, Pathological/pathology , Adaptive Immunity , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Biomarkers/blood , Complement Activation , Complement C3/genetics , Complement C3 Nephritic Factor/genetics , Complement C3b Inactivator Proteins/genetics , Gene Expression , Glomerulonephritis, Membranoproliferative/diagnosis , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/immunology , Humans , Kidney Glomerulus/chemistry , Kidney Glomerulus/immunology , Protein Aggregation, Pathological/diagnosis , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
Mol Immunol ; 67(1): 31-42, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25843230

ABSTRACT

Tremendous advances in our understanding of the thrombotic microangiopathies (TMAs) have revealed distinct disease mechanisms within this heterogeneous group of diseases. As a direct result of this knowledge, both children and adults with complement-mediated TMA now enjoy higher expectations for long-term health. In this update on atypical hemolytic uremic syndrome, we review the clinical characteristics; the genetic and acquired drivers of disease; the broad spectrum of environmental triggers; and current diagnosis and treatment options. Many questions remain to be addressed if additional improvements in patient care and outcome are to be achieved in the coming decade.


Subject(s)
Atypical Hemolytic Uremic Syndrome/pathology , Communicable Diseases/pathology , Gene Expression Regulation/immunology , Kidney Neoplasms/pathology , Kidney/pathology , Adult , Atypical Hemolytic Uremic Syndrome/etiology , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/biosynthesis , Child , Communicable Diseases/complications , Communicable Diseases/genetics , Communicable Diseases/immunology , Complement Activation , Complement C3b/genetics , Complement C3b/immunology , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Genetic Predisposition to Disease , Humans , Kidney/immunology , Kidney Neoplasms/complications , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Transplantation
6.
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
7.
Br J Haematol ; 164(6): 759-66, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24387053

ABSTRACT

Thrombotic thrombocytopenic purpura (TTP) and atypical haemolytic uraemic syndrome (aHUS) are acute, rare life-threatening thrombotic microangiopathies that require rapid diagnosis and treatment. They are defined by microangiopathic haemolytic anaemia and thrombocytopenia, with renal involvement primarily in aHUS and neurological and cardiological sequelae in TTP. Prompt treatment for most cases of both conditions is with plasma exchange initially and monoclonal therapy (rituximab in TTP and eculizumab in aHUS) as the mainstay of therapy. Here we discuss the diagnosis and therapy for both disorders.


Subject(s)
Hemolytic-Uremic Syndrome/therapy , Purpura, Thrombotic Thrombocytopenic/therapy , ADAM Proteins/metabolism , Atypical Hemolytic Uremic Syndrome , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/diagnosis , Humans , Purpura, Thrombotic Thrombocytopenic/blood , Purpura, Thrombotic Thrombocytopenic/diagnosis
8.
J Clin Invest ; 124(1): 145-55, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24334459

ABSTRACT

The renal disorder C3 glomerulopathy with dense deposit disease (C3G-DDD) pattern results from complement dysfunction and primarily affects children and young adults. There is no effective treatment, and patients often progress to end-stage renal failure. A small fraction of C3G-DDD cases linked to factor H or C3 gene mutations as well as autoantibodies have been reported. Here, we examined an index family with 2 patients with C3G-DDD and identified a chromosomal deletion in the complement factor H-related (CFHR) gene cluster. This deletion resulted in expression of a hybrid CFHR2-CFHR5 plasma protein. The recombinant hybrid protein stabilized the C3 convertase and reduced factor H-mediated convertase decay. One patient was refractory to plasma replacement and exchange therapy, as evidenced by the hybrid protein quickly returning to pretreatment plasma levels. Subsequently, complement inhibitors were tested on serum from the patient for their ability to block activity of CFHR2-CFHR5. Soluble CR1 restored defective C3 convertase regulation; however, neither eculizumab nor tagged compstatin had any effect. Our findings provide insight into the importance of CFHR proteins for C3 convertase regulation and identify a genetic variation in the CFHR gene cluster that promotes C3G-DDD. Monitoring copy number and sequence variations in the CFHR gene cluster in C3G-DDD and kidney patients with C3G-DDD variations will help guide treatment strategies.


Subject(s)
Complement C3-C5 Convertases/metabolism , Complement C3b Inactivator Proteins/genetics , Complement System Proteins/genetics , Glomerulonephritis, Membranoproliferative/blood , Kidney Failure, Chronic/blood , Adult , Base Sequence , Child, Preschool , Chromosome Deletion , Complement Activation , Complement C3b/metabolism , Complement C3b Inactivator Proteins/metabolism , Complement System Proteins/metabolism , DNA Mutational Analysis , Enzyme Stability , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/therapy , HEK293 Cells , Humans , Kidney/pathology , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/therapy , Male , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Plasmapheresis , Protein Binding , Sequence Deletion , Treatment Outcome , Young Adult
9.
Semin Nephrol ; 33(6): 508-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24161037

ABSTRACT

Hemolytic uremic syndrome (HUS) is a triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure. The atypical form of HUS is a disease characterized by complement overactivation. Inherited defects in complement genes and acquired autoantibodies against complement regulatory proteins have been described. Incomplete penetrance of mutations in all predisposing genes is reported, suggesting that a precipitating event or trigger is required to unmask the complement regulatory deficiency. The underlying genetic defect predicts the prognosis both in native kidneys and after renal transplantation. The successful trials of the complement inhibitor eculizumab in the treatment of atypical HUS will revolutionize disease management.


Subject(s)
Complement Activation/physiology , Complement System Proteins/genetics , Hemolytic-Uremic Syndrome , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome , Autoantibodies , Complement Factor H/genetics , Complement Factor H/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/physiopathology , Humans , Kidney Transplantation/adverse effects , Kidney Transplantation/mortality , Precipitating Factors
10.
Mol Immunol ; 56(3): 199-212, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23810412

ABSTRACT

Central to the pathogenesis of atypical haemolytic uraemic syndrome (aHUS) is over-activation of the alternative pathway of complement. Inherited defects in complement genes and autoantibodies against complement regulatory proteins have been described. The use of plasma exchange to replace non-functioning complement regulators and hyper-functional complement components in addition to the removal of CFH-autoantibodies made this the 'gold-standard' for management of aHUS. In the last 4 years the introduction of the complement inhibitor Eculizumab has revolutionised the management of aHUS. In this review we shall discuss the available literature on treatment strategies to date.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Hemolytic-Uremic Syndrome/therapy , Plasma Exchange , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Atypical Hemolytic Uremic Syndrome , Complement System Proteins/immunology , Diacylglycerol Kinase/genetics , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/immunology , Humans , Penetrance , Prognosis
11.
Pediatr Transplant ; 17(3): E93-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23461281

ABSTRACT

We present three cases of organ transplantation for atypical haemolytic uraemic syndrome secondary to complement factor H mutation: one isolated renal transplant; one previously reported isolated liver transplant; and one combined liver and kidney transplant. All three patients were treated prior to the licensing of eculizumab for this condition, and all have had favourable outcomes with maintenance of graft function for years following transplantation. We discuss the evolution of transplantation therapy for aHUS over the last two decades. Transplantation decision-making in aHUS has evolved over this time with expanding knowledge of pathophysiology and genetics, alongside refined plasma exchange and anticoagulation protocols and improved centre experience. Our cases demonstrate how individual patient factors within this heterogeneous condition also underlie transplantation decisions and outcomes. Whilst our cases demonstrate that transplantation in aHUS can be a successful long-term treatment providing good quality of life, worldwide experience has proven that most curative treatment for aHUS strategies represents significant risks. Whether new pharmacotherapies such as eculizumab will alter this risk is yet to be determined.


Subject(s)
Hemolytic-Uremic Syndrome/therapy , Kidney Transplantation/methods , Liver Transplantation/methods , Antibodies, Monoclonal, Humanized/therapeutic use , Anticoagulants/therapeutic use , Atypical Hemolytic Uremic Syndrome , Child , Child, Preschool , Complement Factor H/metabolism , Female , Humans , Male , Plasma Exchange , Quality of Life , Renal Insufficiency/therapy , Risk , Treatment Outcome
12.
J Immunol Methods ; 390(1-2): 63-73, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23376722

ABSTRACT

BACKGROUND: Research on complement factor H (fH) in human disease is hampered by lack of an assay suitable for use in large-scale epidemiological studies. We describe the development and validation of a high throughput nephelometric assay for fH. METHODS: Reagents from a commercial radial immunodiffusion (RID) assay (The Binding Site) were adapted for use on the Siemens BNII high throughput nephelometric instrument. The assay was calibrated with a highly purified human fH preparation with rigorously determined concentration, and assay performance was comprehensively evaluated using samples from healthy human volunteers, with the commercial RID assay as a comparator. The distribution and determinants of circulating fH concentration in humans were then investigated in a large representative population sample. RESULTS: The nephelometric assay had recovery close to 100%, was reproducible with intra- and inter-assay CV's of 11% and 5-15% respectively, and had a wider operating range than the RID assay. fH values were unaffected after multiple freeze-thaw cycles demonstrating that it is evidently a stable analyte for immunoassay. fH concentration was unaltered by an acute inflammatory stimulus. The population study showed that plasma fH concentration is associated with circulating lipids and indices of body fat. CONCLUSION: We present the first high throughput assay for circulating fH; the assay is accurate and reliable with reproducible measures from stored samples. It has established the distribution of fH values at a population level and demonstrated important associations with circulating lipids and indices of body fat, thus providing an important reference for future clinical and epidemiological investigations.


Subject(s)
Nephelometry and Turbidimetry/methods , Adult , Blood Specimen Collection/methods , C-Reactive Protein/analysis , Cholesterol/blood , Cholesterol, HDL/blood , Complement Factor H/analysis , Cryopreservation/methods , Humans , Immunoassay/methods , Immunodiffusion/methods , Middle Aged , Protein Stability , Reproducibility of Results , Triglycerides/blood
13.
Pediatr Nephrol ; 26(11): 2073-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21717289

ABSTRACT

Atypical haemolytic uraemic syndrome (aHUS) is frequently associated with mutations in the gene encoding complement factor H (CFH). The clinical response to plasma therapy in aHUS is variable. We present here our experience of plasma therapy in three aHUS patients with CFH mutations. Three children presented aged 4, 22 and 6 months (patients 1-3 respectively) in acute kidney injury requiring dialysis. Plasma therapy consisting of plasma filtration (patient 1) or plasma exchange (PEX; patients 2 and 3) was commenced early following presentation. This resulted in aHUS remission and cessation of dialysis after 2 weeks, 9 days and 2 weeks respectively. Relapses were common and associated with increasing the interval between PEX, but all responded to intensification of PEX therapy. Patient 1 recovered 50% of renal function after first presentation. She had four relapses and started peritoneal dialysis 41 months after presentation. Mutation screening of CFH showed a missense mutation (c.3546 G > T, p.Arg1182Ser) in exon 23. PEX in patient 2 was slowly tapered over 4 months to fortnightly sessions, but she relapsed when PEX was extended to every 4 weeks. Renal function remained normal 12 months post-presentation. Mutation screening of CFH showed a mutation in exon 23 (c.3590 T > C, p.Val1197Ala) and two additional sequence variants in exons 3 and 4. Patient 3 had two relapses associated with intercurrent illnesses concurrent with reducing PEX to weekly doses. Renal function was normal 5 months post-presentation. All three patients showed a good response to PEX with improved renal function both initially and following a relapse. Further research is necessary to determine the best maintenance strategy to delay or prevent end-stage kidney disease.


Subject(s)
Complement Factor H/genetics , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/therapy , Mutation , Plasma Exchange , Plasmapheresis , Atypical Hemolytic Uremic Syndrome , DNA Mutational Analysis , Humans , Infant
14.
Am J Kidney Dis ; 58(1): 109-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21601332

ABSTRACT

Atypical hemolytic uremic syndrome was diagnosed in a 62-year-old man. Sequencing of the CFH gene, which encodes complement factor H, revealed a heterozygous adenine to guanine mutation at nucleotide 3550 of the complementary DNA, leading to a predicted substitution of alanine for threonine at amino acid position 1184 in the protein (c.3550A>G, p.Thr1184Ala). Three years later, he received a simultaneous liver-kidney transplant with plasmapheresis and intratransplant plasma infusion. The postoperative course was complicated by an anastomotic biliary stricture that was treated successfully using endoscopic stenting. One year later, he has excellent function of both transplants, emphasizing that simultaneous liver-kidney transplant is a valuable treatment option in the management of adult patients with atypical hemolytic uremic syndrome.


Subject(s)
Complement Factor H/genetics , Kidney Transplantation , Liver Transplantation , Atypical Hemolytic Uremic Syndrome , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/surgery , Humans , Male , Middle Aged , Mutation , Treatment Outcome
15.
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
16.
Nephron Clin Pract ; 118(1): c37-42, 2011.
Article in English | MEDLINE | ID: mdl-21071971

ABSTRACT

Atypical haemolytic uraemic syndrome (aHUS) is a disease characterized by complement overactivation in which inherited defects in complement genes and acquired autoantibodies against complement regulatory proteins have been described. Identification of the underlying defect can both predict disease outcome and guide treatment. The ability to remove inhibitory autoantibodies and hyper-active complement components in addition to its ability to replace defective complement regulators means that plasma exchange is currently first-line therapy. In those with factor H and factor I mutations who do progress to end-stage renal failure, renal transplantation usually fails due to recurrent HUS. In this situation, combined liver-kidney transplantation has been suggested to correct the underlying genetic defect. Newer agents, such as the complement inhibitor eculizumab, may herald a new era in the treatment of aHUS.


Subject(s)
Autoantibodies/genetics , Complement System Proteins/genetics , Genetic Predisposition to Disease/genetics , Genetic Testing/methods , Hemolytic-Uremic Syndrome , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/therapy , Humans
17.
Semin Thromb Hemost ; 36(6): 653-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20865642

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a disease characterized by overactivation of complement. Recurrence following renal transplantation is determined by a genetic predisposition. Genetic screening of all individuals with aHUS should be performed prior to listing for transplantation. Individuals with isolated mutations in MCP have a low risk of recurrence and may be considered for kidney transplantation alone. In individuals with CFH and CFI mutations, the risk of recurrence following renal transplantation is high. Combined liver/kidney transplantation has been used successfully in individuals with CFH mutations following the introduction of perioperative plasma exchange; however, such a procedure is not without its risks. Liver/kidney transplantation has yet to be performed on individuals with CFI and C3 mutations but may be predicted to be successful. In individuals with CFH autoantibodies, a reduction in titer through plasma exchange and rituximab has been successful. Clinical trials of the complement C5 inhibitor eculizumab may improve prospects for isolated renal transplantation in individuals with complement protein mutations.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hemolytic-Uremic Syndrome/therapy , Kidney Transplantation , Liver Transplantation , Complement System Proteins/genetics , Complement System Proteins/immunology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/immunology , Humans , Mutation , Plasma Exchange
18.
Pediatr Nephrol ; 25(12): 2431-42, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20526633

ABSTRACT

Central to the pathogenesis of atypical hemolytic uremic syndrome (aHUS) is over-activation of the alternative pathway of complement. Following the initial discovery of mutations in the complement regulatory protein, factor H, mutations have been described in factor I, membrane cofactor protein and thrombomodulin, which also result in decreased complement regulation. Autoantibodies to factor H have also been reported to impair complement regulation in aHUS. More recently, gain of function mutations in the complement components C3 and Factor B have been seen. This review focuses on the genetic causes of aHUS, their functional consequences, and clinical effect.


Subject(s)
Complement Activation/genetics , Complement System Proteins/genetics , Mutation , Atypical Hemolytic Uremic Syndrome , Autoantibodies/blood , Complement System Proteins/immunology , Genetic Predisposition to Disease , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/immunology , Humans , Penetrance , Phenotype , Risk Assessment , Risk Factors
19.
Am J Kidney Dis ; 55(4): 708-11, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19854549

ABSTRACT

Kidney transplant in patients with atypical hemolytic uremic syndrome (aHUS) is associated with a poor outcome because of recurrent disease, especially in patients known to have a factor H mutation. Long-term prophylactic plasma exchange and combined liver-kidney transplant have prevented graft loss caused by recurrence. However, the mortality associated with liver transplant is not negligible, and prophylactic plasma exchange requires permanent vascular access and regular hospitalization and exposes the patient to potential allergic reactions to plasma. Eculizumab is a high-affinity humanized monoclonal antibody that binds to C5 and thus prevents generation of C5a and the membrane attack complex. We report the case of a 17-year-old girl with aHUS associated with a mutation in the gene for complement factor H (CFH; c.3572C>T, Ser1191Leu) who was highly dependent on plasma exchange. Because of severe allergic reactions to plasma after the third renal graft, eculizumab was introduced in place of plasma exchange without problems. This and other reports suggest that the promise of complement inhibitors in the management of aHUS is going to be fulfilled.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/surgery , Kidney Transplantation/physiology , Antibodies, Monoclonal, Humanized , Child, Preschool , Complement Factor H/genetics , Female , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/therapy , Humans , Mutation , Plasma Exchange , Reoperation
20.
Br J Haematol ; 148(1): 37-47, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19821824

ABSTRACT

Atypical haemolytic uraemic syndrome (aHUS) is associated with a poor prognosis with regard to survival at presentation, recovery of renal function and transplantation. It is now established that aHUS is a disease of complement dysregulation with mutations in the genes encoding both complement regulators and activators, and autoantibodies against the complement regulator factor H. Identification of the underlying molecular abnormality in an individual patient can now help to guide their future management. In these guidelines we make recommendations for the investigation and management of aHUS patients both at presentation and in the long-term. We particularly address the role of renal transplantation alone and combined liver-kidney transplantation.


Subject(s)
Hemolytic-Uremic Syndrome/therapy , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Clinical Trials as Topic , Hemolytic-Uremic Syndrome/classification , Hemolytic-Uremic Syndrome/diagnosis , Humans , Kidney Transplantation , Liver Transplantation , Plasma Exchange/methods , Terminology as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...