1
|
Complement factor H in AMD: Bridging genetic associations and pathobiology. Prog Retin Eye Res 2017; 62:38-57. [PMID: 28928087 DOI: 10.1016/j.preteyeres.2017.09.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/08/2017] [Accepted: 09/13/2017] [Indexed: 01/28/2023]
Abstract
Age-Related Macular Degeneration (AMD) is a complex multifactorial disease characterized in its early stages by lipoprotein accumulations in Bruch's Membrane (BrM), seen on fundoscopic exam as drusen, and in its late forms by neovascularization ("wet") or geographic atrophy of the Retinal Pigmented Epithelial (RPE) cell layer ("dry"). Genetic studies have strongly supported a relationship between the alternative complement cascade, in particular the common H402 variant in Complement Factor H (CFH) and development of AMD. However, the functional significance of the CFH Y402H polymorphism remains elusive. In this article, we critically review the literature surrounding the functional significance of this polymorphism. Furthermore, based on our group's studies we propose a model in which CFH H402 affects CFH binding to heparan sulfate proteoglycans leading to accelerated lipoprotein accumulation in BrM and drusen progression. We also review the literature on the role of other complement components in AMD pathobiologies, including C3a, C5a and the membrane attack complex (MAC), and on transgenic mouse models developed to interrogate in vivo the effects of the CFH Y402H polymorphism.
Collapse
|
2
|
Abou-El-Hassan H, Zaraket H. Viral-derived complement inhibitors: current status and potential role in immunomodulation. Exp Biol Med (Maywood) 2016; 242:397-410. [PMID: 27798122 DOI: 10.1177/1535370216675772] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The complement system is one of the body's major innate immune defense mechanisms in vertebrates. Its function is to detect foreign bodies and promote their elimination through opsonisation or lysis. Complement proteins play an important role in the immunopathogenesis of several disorders. However, excessive complement activation does not confer more protection but instead leads to several autoimmune and inflammatory diseases. With inappropriate activation of the complement system, activated complement proteins and glycoproteins may damage both healthy and diseased tissues. Development of complement inhibitors represents an effective approach in controlling dysregulated complement activity and reducing disease severity, yet few studies have investigated the nature and role of novel complement inhibitory proteins of viral origin. Viral complement inhibitors have important implications in understanding the importance of complement inhibition and their role as a promising novel therapeutic approach in diseases caused by dysregulated complement function. In this review, we discuss the role and importance of complement inhibitors derived from several viruses in the scope of human inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- 1 Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon.,2 Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Zaraket
- 2 Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,3 Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
3
|
Abstract
Besides mediating primary hemostasis and thrombosis, platelets play a critical role in tissue repair and regeneration. They regulate fundamental mechanisms involved in the healing process including cellular migration, proliferation, and angiogenesis. Control of apoptosis/cell survival and interaction with progenitor cells, which are clinically relevant but poorly understood aspects of platelets in tissue repair, will be highlighted in this review. Gaining deeper insight into the less well-characterized molecular mechanisms is necessary to develop new therapeutic platelet-based options.
Collapse
|
4
|
Lee SW, Kim IJ, Jeong BY, Choi MH, Kim JY, Kwon KH, Lee JW, Yu A, Shin MG. Use of MDLC-DIGE and LC-MS/MS to identify serum biomarkers for complete remission in patients with acute myeloid leukemia. Electrophoresis 2012; 33:1863-72. [DOI: 10.1002/elps.201200047] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Seung-Won Lee
- Department of Anatomy, Chonnam National University Medical School; Gwangju; Korea
| | - In Jae Kim
- Department of Anatomy, Chonnam National University Medical School; Gwangju; Korea
| | - Bo Yoon Jeong
- National Cancer Control Institute; National Cancer Center; Goyang-si; Korea
| | - Mun-Ho Choi
- Department of Computer Science; College of Engineering; Chonnam National University; Gwangju; Korea
| | - Jin Young Kim
- Division of Mass Spectrometry Research; Korea Basic Science Institute; Ochang; Korea
| | - Kyung-Hoon Kwon
- Division of Mass Spectrometry Research; Korea Basic Science Institute; Ochang; Korea
| | - Jae Won Lee
- Department of Statistics; Korea University; Seoul; Korea
| | - Ami Yu
- Department of Statistics; Korea University; Seoul; Korea
| | - Myung-Geun Shin
- Laboratory Medicine; Chonnam National University Medical School; Gwangju; Korea
| |
Collapse
|
5
|
Samson M, Audia S, Leguy V, Berthier S, Janikashvili N, Martin L, Bonnotte B, Lorcerie B. Haemolytic-uraemic syndrome during severe lupus nephritis: efficacy of plasma exchange. Intern Med J 2012; 42:95-8. [PMID: 22276560 DOI: 10.1111/j.1445-5994.2011.02591.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Systemic lupus erythematosus (SLE) has been described as a cause of thrombotic microangiopathy, especially thrombotic thrombocytopenic purpura (TTP). Haemolytic-uraemic syndrome (HUS) is less frequent in SLE. We report a case of such an association during an episode of severe lupus nephritis in a young woman, who was successfully treated with steroids, cyclophosphamide and especially plasma exchange with plasma replacement. This report highlights the importance of recognising atypical HUS in SLE patients by looking for schistocytes in case of haemolytic anemia with a negative antiglobulin test, in order to begin plasma exchange.
Collapse
Affiliation(s)
- M Samson
- Department of Internal Medicine and Clinical Immunology, Dijon University Hospital, Dijon, France.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Gandhi J, Cashman SM, Kumar-Singh R. Soluble CD59 expressed from an adenovirus in vivo is a potent inhibitor of complement deposition on murine liver vascular endothelium. PLoS One 2011; 6:e21621. [PMID: 21720565 PMCID: PMC3123367 DOI: 10.1371/journal.pone.0021621] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 06/03/2011] [Indexed: 11/19/2022] Open
Abstract
Inappropriate activation of complement on the vascular endothelium of specific organs, or systemically, underlies the etiology of a number of diseases. These disorders include atypical hemolytic uremic syndrome, membranoproliferative glomerulonephritis, atherosclerosis, age-related macular degeneration, diabetic retinopathy, and transplant rejection. Inhibition of the terminal step of complement activation, i.e. formation of the membrane attack complex, using CD59 has the advantage of retaining the upstream processes of the complement cascade necessary for fighting pathogens and retaining complement's crucial role in tissue homeostasis. Previous studies have shown the necessity of membrane targeting of soluble CD59 in order for it to prove an effective inhibitor of complement deposition both in vitro and in vivo. In this study we have generated an in vivo model of human complement activation on murine liver vascular endothelium. This model should prove useful for the development of anti-complement therapies for complement-induced pathologies of vascular endothelium. Using this model, we have demonstrated the viability of a non membrane-targeted soluble CD59 to significantly inhibit complement deposition on the endothelium of murine liver vasculature when expressed in vivo from an adenovirus. This result, unanticipated based on prior studies, suggests that the use of non membrane-targeted sCD59 as an anti-complement therapy be re-visited.
Collapse
Affiliation(s)
- Jarel Gandhi
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Siobhan M. Cashman
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Rajendra Kumar-Singh
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
7
|
Abstract
Hemolytic uremic syndrome (HUS) is related to a renal thrombotic microangiopathy, inducing hypertension and acute renal failure (ARF). Its pathogenesis involves an activation/lesion of microvascular endothelial cells, mainly in the renal vasculature, secondary to bacterial toxins, drugs, or autoantibodies. An overactivation of the complement alternate pathway secondary to a heterozygote deficiency of regulatory proteins (factor H, factor I or MCP) or to an activating mutation of factor B or C3 can also result in HUS. Less frequently, renal microthrombi are due to an acquired or a constitutional deficiency in ADAMTS-13, the protease cleaving von Wilebrand factor. Hemolytic anemia with schistocytes, thrombocytopenia without evidence of disseminated intravascular coagulation, and renal failure are consistently found. In typical HUS, a prodromal diarrhea, with blood in the stools, is observed, related to pathogenic enterobacteria, most frequently E. Coli O157:H7. HUS may also occur in the post partum period, and is then related to a factor H or factor I deficiency. HUS may also occur after various treatments such as mitomycin C, gemcitabine, ciclosporin A, or tacrolimus, and as reported more recently bevacizumab, an anti VEGF antibody. Atypical HUS are not associated with diarrhea, may be sporadic or familial, and can be related to an overactivation of the complement alternate pathway. More recently, some of them have been related to a mutation of thrombomodulin, which also regulates the alternate pathway of complement. In adults, several HUS are encountered in the course of chronic nephropathies: nephroangiosclerosis, chronic glomerulonephritis, post irradiation nephropathy, scleroderma, disseminated lupus erythematosus, antiphospholipid syndrome. Overall the prognosis of HUS has improved, with a patient survival greater than 85% at 1 year. Chronic renal failure is observed as a sequella in 20 to 65% of the cases. Plasma infusions and plasma exchanges are effective in most of the cases to treat hemolysis and thrombocytopenia. Steroid therapy is debated, as well as immunosuppressive drugs, including rituximab, in autoimmune forms. A new monoclonal anti-C5 antibody is tested, and seems to be effective in atypical HUS with abnormal complement alternate pathway activation. If terminal renal failure occurs, renal transplantation can be performed but the risk of recurrence, which very low in post infectious forms of HUS, is about 70 to 80% in genetic forms of complement regulatory protein deficiency.
Collapse
Affiliation(s)
- Alexandre Hertig
- Service des urgences néphrologiques et transplantation rénale, hôpital Tenon, 4, rue de la Chine, 75020 Paris, France
| | | | | |
Collapse
|
8
|
Clinical practice. Today's understanding of the haemolytic uraemic syndrome. Eur J Pediatr 2010; 169:7-13. [PMID: 19707787 DOI: 10.1007/s00431-009-1039-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 07/30/2009] [Indexed: 12/19/2022]
Abstract
The haemolytic uraemic syndrome (HUS) includes the triad of haemolytic anaemia, thrombocytopenia, and acute renal failure. The classical form [D(+) HUS] is caused by infectious agents, and it is a common cause of acute renal failure in children. The enterohaemorrhagic Escherichia coli-producing Shiga toxin (Stx) is the most common infectious agent causing HUS. Other infectious agents are Shigella and Streptococcus pneumoniae. Infections by S. pneumoniae can be particularly severe and has a higher acute mortality and a higher long-term morbidity compared to HUS by Stx. Atypical HUS [D(-)Stx(-)HUS] are often used by paediatricians to indicate a presentation of HUS without preceding diarrhoea. Almost all patients with D(-)Stx(-)HUS have a defect in the alternative pathway, for example, mutations in the genes for complement factor H, factor I, and membrane co-factor protein. Mutations in the factor H gene are described more often. The majority of children with D(+) HUS develop some degree of renal insufficiency, and approximately two thirds of children with HUS will require dialysis therapy, while about one third will have milder renal involvement without the need for dialysis therapy. General management of acute renal failure includes appropriate fluid and electrolyte management, antihypertensive therapy, and the initiation of renal replacement therapy when appropriate. Specific management issues in HUS include management of the haematological complications of HUS, monitoring for extra-renal involvement, avoiding antidiarrhoeal drugs, and possibly avoiding of antibiotic therapy. In addition to the obligatory supportive treatment and tight control of hypertension, there is anecdotal evidence that plasma therapy may induce remission and, in some cases, maintain it. Fresh frozen plasma contains factor H at physiological concentrations. A new therapy for D(-)Stx(-)HUS is a humanised monoclonal antibody (Eculizumab) that blocks complement activity by cleavage of the complement protein C5. It prevents the generation of the inflammatory peptide C5a and the cytotoxic membrane-attack complex C5b-9. We have first positive results, but it is still not approved for HUS.
Collapse
|
9
|
Kamioka I, Nozu K, Fujita T, Kaito H, Tanaka R, Yoshiya K, Iijima K, Nakanishi K, Yoshikawa N, Matsuo M. Prognosis and pathological characteristics of five children with non-Shiga toxin-mediated hemolytic uremic syndrome. Pediatr Int 2007; 49:196-201. [PMID: 17445038 DOI: 10.1111/j.1442-200x.2007.02352.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The three major signs of hemolytic uremic syndrome (HUS) are hemolytic anemia, thrombopenia and acute renal failure. HUS is classified into Shiga toxin-mediated HUS (Stx-HUS) and non-Shiga toxin-mediated HUS (nStx-HUS). The prognosis of nStx-HUS is reported to be less favorable than that of Stx-HUS. Although the association between the prognosis and pathological characteristics of HUS have been reported such that the prognosis was considered to be poor for thrombotic microangiopathy (TMA) with predominant arterial involvement (arterial TMA), good for TMA with predominant glomerular involvement (glomerular TMA) and dependent on the extent of necrosis in cases of renal cortical necrosis, it is not yet clear whether pathological findings are also related to the renal prognosis of nStx-HUS cases. Therefore the purpose of the present paper was to analyze renal biopsy findings and prognosis for five children with nStx-HUS. METHODS Clinical records of five cases of nStx-HUS among 74 cases of diagnosed HUS were reviewed, and information and data were summarized. RESULTS Histological examination of the kidney led to the diagnosis of arterial TMA in three cases, and glomerular TMA and severe renal cortical necrosis in one case each. Analysis of the relationship between renal histological findings and the prognosis found that three patients with arterial TMA and one patient with severe renal cortical necrosis later developed end-stage renal failure while one patient with glomerular TMA has continued to show normal renal function. CONCLUSIONS These findings indicate that pathological findings are closely related to the prognosis in cases of nStx-HUS.
Collapse
Affiliation(s)
- Ichiro Kamioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Thrombotic microangiopathies (TMA) are microvascular occlusive disorders characterized by hemolytic anemia caused by fragmentation of erythrocytes and thrombocytopenia due to increased platelet aggregation and thrombus formation, eventually leading to disturbed microcirculation with reduced organ perfusion. Although several disease states may manifest as TMA, the two most relevant conditions associated with TMA are thrombotic thrombocytopenic purpura (TTP) and hemolytic uremic syndrome (HUS), characterized by prominent brain or renal lesions, respectively. However, occasionally the clinical distinction between these two conditions can be difficult. In this review, we focus on the epidemiologic and diagnostic criteria as well as on the most recent insights into the pathophysiology and treatment of these two conditions.
Collapse
Affiliation(s)
- Massimo Franchini
- Servizio di Immunoematologia e Trasfusione, Azienda Ospedaliera di Verona, Verona, Italy.
| |
Collapse
|
11
|
Moake JL. Thrombotic Thrombocytopenic Purpura and the Hemolytic-Uremic Syndrome. Platelets 2007. [DOI: 10.1016/b978-012369367-9/50812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Oppermann M, Manuelian T, Józsi M, Brandt E, Jokiranta TS, Heinen S, Meri S, Skerka C, Götze O, Zipfel PF. The C-terminus of complement regulator Factor H mediates target recognition: evidence for a compact conformation of the native protein. Clin Exp Immunol 2006; 144:342-52. [PMID: 16634809 PMCID: PMC1809651 DOI: 10.1111/j.1365-2249.2006.03071.x] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The complement inhibitor Factor H has three distinct binding sites for C3b and for heparin, but in solution uses specifically the most C-terminal domain, i.e. short consensus repeats (SCR) 20 for ligand interaction. Two novel monoclonal antibodies (mABs C14 and C18) that bind to the most C-terminal domain SCR 20 completely blocked interaction of Factor H with the ligands C3b, C3d, heparin and binding to endothelial cells. In contrast, several mAbs that bind to the N-terminus and to the middle regions of the molecule showed no or minor inhibitory effects when assayed by enzyme-linked immunosorbent assay (ELISA) and ligand interaction assays. This paradox between a single functional binding site identified for native Factor H versus multiple interaction sites reported for deletion constructs is explained by a compact conformation of the fluid phase protein with one accessible binding site. On zymosan particles mAbs C14 and C18 blocked alternative pathway activation completely. Thus demonstrating that native Factor H makes the first and initial contact with the C terminus, which is followed by N terminally mediated complement regulation. These results are explained by a conformational hypothetical model: the native Factor H protein has a compact structure and only one binding site accessible. Upon the first contact the protein unfolds and exposes the additional binding sites. This model does explain how Factor H mediates recognition functions during complement control and the clustering of disease associated mutations in patients with haemolytic uraemic syndrome that have been reported in the C-terminal recognition domain of Factor H.
Collapse
Affiliation(s)
- M Oppermann
- Department of Immunology, University of Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Vaziri-Sani F, Holmberg L, Sjöholm AG, Kristoffersson AC, Manea M, Frémeaux-Bacchi V, Fehrman-Ekholm I, Raafat R, Karpman D. Phenotypic expression of factor H mutations in patients with atypical hemolytic uremic syndrome. Kidney Int 2006; 69:981-8. [PMID: 16528247 DOI: 10.1038/sj.ki.5000155] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We investigated the phenotypic expression of factor H mutations in two patients with atypical hemolytic uremic syndrome (HUS). Factor H in serum was assayed by rocket immunoelectrophoresis, immunoblotting, and double immunodiffusion and in tissue by immunohistochemistry. Functional activity was analyzed by hemolysis of sheep erythrocytes and binding to endothelial cells. A homozygous mutation in complement control protein (CCP) domain 10 of factor H was identified in an adult man who first developed membranoproliferative glomerulonephritis and later HUS. C3 levels were very low. The patient had undetectable factor H levels in serum and a weak factor H 150 kDa band. Double immunodiffusion showed partial antigenic identity with factor H in normal serum owing to the presence of factor H-like protein 1. Strong specific labeling for factor H was detected in glomerular endothelium, mesangium and in glomerular and tubular epithelium as well as in bone marrow cells. A heterozygous mutation in CCP 20 of factor H was found in a girl with HUS. C3 levels were moderately decreased at onset. Factor H levels were normal and a normal 150 kDa band was present. Double immunodiffusion showed antigenic identity with normal factor H. Factor H labeling was minimal in the renal cortex. Factor H dysfunction was demonstrated by increased sheep erythrocyte hemolysis and decreased binding to endothelial cells. In summary, two different factor H mutations associated with HUS were examined: in one, factor H accumulated in cells, and in the other, membrane binding was reduced.
Collapse
Affiliation(s)
- F Vaziri-Sani
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Zipfel PF, Heinen S, Józsi M, Skerka C. Complement and diseases: Defective alternative pathway control results in kidney and eye diseases. Mol Immunol 2006; 43:97-106. [PMID: 16026839 DOI: 10.1016/j.molimm.2005.06.015] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The complement system is a central part of innate immunity and in its normal setting aimed to recognize and eliminate microbes. For elimination toxic activation products are generated locally and are reported directly of the surface of the invading microbe. A deregulation of the alternative pathway results in defective recognition and toxic activation products can be formed on the surface of host tissues and structures. Recent studies have shown that mutated or defective regulators of the alternative pathway of complement are associated with auto immune diseases of the kidney, including the atypical form of hemolytic uremic syndrome (HUS), membranoproliferative glomerulonephritis (MPGN) and also of the eye, such as age-related macular degeneration (ARMD). Current research provides clues how mutations occurring in genes coding for single complement components or the inactivation of single regulators lead to defective alternative pathway amplification, via the convertase C3bBb. These scenarios explain how defects of a single regulator lead to local, organ specific damage.
Collapse
Affiliation(s)
- Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Products Research and Infection Biology, Hans Knoell Institute, Beutenbergstr. 11a, 07745 Jena, Germany.
| | | | | | | |
Collapse
|
15
|
Franchini M, Zaffanello M, Veneri D. Advances in the pathogenesis, diagnosis and treatment of thrombotic thrombocytopenic purpura and hemolytic uremic syndrome. Thromb Res 2006; 118:177-84. [PMID: 16126255 DOI: 10.1016/j.thromres.2005.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 06/30/2005] [Accepted: 07/21/2005] [Indexed: 11/27/2022]
Abstract
The thrombotic microangiopathies are microvascular occlusive disorders characterized by hemolytic anemia caused by fragmentation of erythrocytes and thrombocytopenia due to increased platelet aggregation and thrombus formation, eventually leading to disturbed microcirculation with reduced organ perfusion. Depending on whether brain or renal lesions prevail, two different entities have been described: thrombotic thrombocytopenic purpura (TTP) and hemolytic uremic syndrome (HUS). However, not rarely the clinical distinctions between these two conditions remain questionable. Recent studies have contributed greatly to our current understanding of the molecular mechanisms leading to TTP and HUS. In this review, we briefly focus on the most important advances in the pathophysiology, diagnosis and treatment of these two thrombotic microangiopathies.
Collapse
Affiliation(s)
- Massimo Franchini
- Servizio di Immunoematologia e Trasfusione, Ospedale Policlinico, Piazzale L. Scuro Azienda Ospedaliera di Verona, Italy.
| | | | | |
Collapse
|
16
|
Licht C, Weyersberg A, Heinen S, Stapenhorst L, Devenge J, Beck B, Waldherr R, Kirschfink M, Zipfel PF, Hoppe B. Successful plasma therapy for atypical hemolytic uremic syndrome caused by factor H deficiency owing to a novel mutation in the complement cofactor protein domain 15. Am J Kidney Dis 2005; 45:415-21. [PMID: 15685522 DOI: 10.1053/j.ajkd.2004.10.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Quantitative or functional deficiency of complement factor H results in uncontrolled complement activation. This leads to thrombotic microangiopathy and finally causes renal failure (atypical hemolytic uremic syndrome [aHUS]). By regular analysis of factor H in patients with aHUS, the authors found a complete factor H deficiency in an infant in whom aHUS developed at 8 months of age. Factor H was quantified by enzyme-linked immunosorbent assay and further analyzed by Western blot using a factor H-specific antibody. Complement activation was determined by measuring total hemolytic activity of the classical (CH50) and alternative (APH50) pathways, C3 and C3d. The sequence of factor H gene was determined. Serial factor H measurements after fresh frozen plasma infusion allowed calculation of a factor H half-life. Factor H was absent in plasma (<1 mug/mL), and the complement system was highly activated (CH50, APH50, C3 decreased; C3d increased). Genetic analysis identified a novel homozygous factor H mutation (T2770A; Y899Stop) in CCP domain 15, most likely causing defective protein secretion. Time course measurements of factor H after plasma infusion established a factor H half-life of about 6 days. By repetitive plasma infusions (20 mL/kg over about 2 to 3 hours) the authors were able to interrupt the vicious circle of thrombotic microangiopathy in a factor H-deficient patient with aHUS. Based on the measured factor H half-life of about 6 days, regular plasma infusions in 2-week intervals were given, which prevented further aHUS episodes and stopped the decline of kidney function.
Collapse
Affiliation(s)
- Christoph Licht
- Division of Pediatric Nephrology, Children's Hospital of the University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Cheng ZZ, Hellwage J, Seeberger H, Zipfel PF, Meri S, Jokiranta TS. Comparison of surface recognition and C3b binding properties of mouse and human complement factor H. Mol Immunol 2005; 43:972-9. [PMID: 16023208 DOI: 10.1016/j.molimm.2005.05.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Indexed: 12/25/2022]
Abstract
Factor H (FH) is a central complement regulator both in plasma and on certain cellular and acellular surfaces that are in contact with plasma. Although FH deficiency has been shown to lead to similar diseases in man and mice (membranoproliferative glomerulonephritis or dense deposit disease) little is known about the similarity between the human and murine FH functions. We here characterize the interactions of murine FH (mFH) with C3b, glycosaminoglycans, and endothelial cells and compare these interactions with those of human FH (hFH). To achieve this we purified mFH and murine C3 from plasma, prepared murine C3b, and expressed recombinant mFH constructs containing domains 1-5 and 18-20 (mFH1-5 and mFH18-20). For comparisons, hFH, human C3b, and recombinant hFH1-5 and hFH18-20 were used. We demonstrate that mFH and mFH1-5 do act as cofactors for factor I-mediated cleavage of human C3b. Surface plasmon resonance analysis showed binding of mFH18-20 to murine C3b and weak binding to human C3b. The mFH18-20 construct bound to heparin in a manner comparable to hFH18-20. It was demonstrated by flow cytometry that mFH and mFH18-20 bind to human endothelial cells in a similar manner to hFH and hFH18-20. Taken together, locations of the key functions of mFH, i.e. complement regulation and surface recognition, are comparable to hFH. Recently, mutations in the carboxy-terminal end of hFH have been found to be associated with atypical hemolytic uremic syndrome (aHUS). Based on the results in this report it is conceptually attractive to establish a murine model for aHUS.
Collapse
Affiliation(s)
- Zhu-Zhu Cheng
- Department of Bacteriology and Immunology, Haartman Institute and HUSLAB, University and University Hospital of Helsinki, Haartmaninkatu 3, FIN-00290 Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
18
|
Anderson AJ, Najbauer J, Huang W, Young W, Robert S. Upregulation of complement inhibitors in association with vulnerable cells following contusion-induced spinal cord injury. J Neurotrauma 2005; 22:382-97. [PMID: 15785233 DOI: 10.1089/neu.2005.22.382] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We have previously described the activation of the classical, alternative, and terminal complement cascade pathways after acute contusion spinal cord injury using the New York University (NYU) weight-drop impactor. In the present study, we examined the induction of protein regulators of the complement cascade, factor H (FH), and clusterin, in the same experimental paradigm. The spinal cord of laminectomized adult rats was subjected to mild or severe injury using impactor weight-drop heights of 12.5 and 50 mm, respectively. The spinal cords of control and injured animals were evaluated at 1, 7, and 42 days after injury. Immunocytochemistry revealed a robust increase in the numbers and intensity of staining of FH, and clusterin-positive cells in the injured cord at all three time points, with the highest increases observed at 1 and 42 days after injury. FH and clusterin-positive cells were observed among neurons as well as oligodendrocytes. The increased expression was detected both rostrally and caudally from the injury site, in the latter case at distances up to 20 mm. The precise biological significance of injury-induced upregulation of these proteins remains to be determined. However, FH and clusterin are potent regulators of complement activity targeting upstream (FH) and downstream (clusterin) molecules of the pro-inflammatory cascade, which could be of vital importance in preventing a "runaway" inflammatory reaction in the injured spinal cord.
Collapse
Affiliation(s)
- Aileen J Anderson
- Department of Physical Medicine and Rehabilitation, and the Reeve-Irvine Center, University of California, Irvine, California, USA.
| | | | | | | | | |
Collapse
|
19
|
Yassa SK, Blessios G, Marinides G, Venuto RC. Anti-CD20 monoclonal antibody (Rituximab) for life-threatening hemolytic-uremic syndrome. Clin Transplant 2005; 19:423-6. [PMID: 15877809 DOI: 10.1111/j.1399-0012.2005.00334.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Rituximab is a chimaeric monoclonal antibody directed against the CD20 antigen. It has been successfully used in B-cell malignancy and its efficacy in the treatment of in autoimmune hemolytic anemia and other autoimmune diseases is being investigated. There are also few case reports of its success in thrombotic thrombocytopenic purpura, but no reports of its use in hemolytic-uremic syndrome (HUS). We report a 36-year-old patient who had lost the function of her native kidneys secondary to HUS. After more than 1 year in clinical remission, she received a living unrelated kidney transplant. This immediately precipitated a severe relapse of HUS. The process was abrogated but not completely inactivated, despite over 40 plasma exchange treatments. Consequently, she was given Rituximab in courses of two to three doses, each dose 375 mg/m(2), at weekly intervals with remarkable stabilization of her disease for approximately 6 months.
Collapse
Affiliation(s)
- S K Yassa
- Department of Medicine, Division of Nephrology, University at Buffalo School of Medicine, Buffalo, NY, USA
| | | | | | | |
Collapse
|
20
|
Hageman GS, Anderson DH, Johnson LV, Hancox LS, Taiber AJ, Hardisty LI, Hageman JL, Stockman HA, Borchardt JD, Gehrs KM, Smith RJH, Silvestri G, Russell SR, Klaver CCW, Barbazetto I, Chang S, Yannuzzi LA, Barile GR, Merriam JC, Smith RT, Olsh AK, Bergeron J, Zernant J, Merriam JE, Gold B, Dean M, Allikmets R. A common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc Natl Acad Sci U S A 2005; 102:7227-32. [PMID: 15870199 PMCID: PMC1088171 DOI: 10.1073/pnas.0501536102] [Citation(s) in RCA: 1482] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2005] [Indexed: 12/21/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most frequent cause of irreversible blindness in the elderly in developed countries. Our previous studies implicated activation of complement in the formation of drusen, the hallmark lesion of AMD. Here, we show that factor H (HF1), the major inhibitor of the alternative complement pathway, accumulates within drusen and is synthesized by the retinal pigmented epithelium. Because previous linkage analyses identified chromosome 1q25-32, which harbors the factor H gene (HF1/CFH), as an AMD susceptibility locus, we analyzed HF1 for genetic variation in two independent cohorts comprised of approximately 900 AMD cases and 400 matched controls. We found association of eight common HF1 SNPs with AMD; two common missense variants exhibit highly significant associations (I62V, chi2 = 26.1 and P = 3.2 x 10(-7) and Y402H, chi2 = 54.4 and P = 1.6 x 10(-13)). Haplotype analysis reveals that multiple HF1 variants confer elevated or reduced risk of AMD. One common at-risk haplotype is present at a frequency of 50% in AMD cases and 29% in controls [odds ratio (OR) = 2.46, 95% confidence interval (1.95-3.11)]. Homozygotes for this haplotype account for 24% of cases and 8% of controls [OR = 3.51, 95% confidence interval (2.13-5.78)]. Several protective haplotypes are also identified (OR = 0.44-0.55), further implicating HF1 function in the pathogenetic mechanisms underlying AMD. We propose that genetic variation in a regulator of the alternative complement pathway, when combined with a triggering event, such as infection, underlie a major proportion of AMD in the human population.
Collapse
Affiliation(s)
- Gregory S Hageman
- Department of Ophthalmology and Visual Sciences, Cell Biology and Functional Genomics Laboratory, University of Iowa, Iowa City, IA 52240, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Vaziri-Sani F, Hellwage J, Zipfel PF, Sjöholm AG, Iancu R, Karpman D. Factor H binds to washed human platelets. J Thromb Haemost 2005; 3:154-62. [PMID: 15634279 DOI: 10.1111/j.1538-7836.2004.01010.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Factor H regulates the alternative pathway of complement. The protein has three heparin-binding sites, is synthesized primarily in the liver and copurifies from platelets with thrombospondin-1. Factor H mutations at the C-terminus are associated with atypical hemolytic uremic syndrome, a condition in which platelets are consumed. Objectives The aim of this study was to investigate if factor H interacts with platelets. METHODS Binding of factor H, recombinant C- or N-terminus constructs and a C-terminus mutant to washed (plasma and complement-free) platelets was analyzed by flow cytometry. Binding of factor H and constructs to thrombospondin-1 was measured by surface plasmon resonance. RESULTS Factor H bound to platelets in a dose-dependent manner. The major binding site was localized to the C-terminus. The interaction was partially blocked by heparin. Inhibition with anti-GPIIb/IIIa, or with fibrinogen, suggested that the platelet GPIIb/IIIa receptor is involved in factor H binding. Factor H binds to thrombospondin-1. Addition of thrombospondin-1 increased factor H binding to platelets. Factor H mutated at the C-terminus also bound to platelets, albeit to a significantly lesser degree. CONCLUSIONS This study reports a novel property of factor H, i.e. binding to platelets, either directly via the GPIIb/IIIa receptor or indirectly via thrombospondin-1, in the absence of complement. Binding to platelets was mostly mediated by the C-terminal region of factor H and factor H mutated at the C-terminus exhibited reduced binding.
Collapse
Affiliation(s)
- F Vaziri-Sani
- Department of Pediatrics, Lund University, Lund, Sweden
| | | | | | | | | | | |
Collapse
|
22
|
Kirschfink M, Mollnes TE. Modern complement analysis. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2004; 10:982-9. [PMID: 14607856 PMCID: PMC262430 DOI: 10.1128/cdli.10.6.982-989.2003] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Fortin MC, Schürch W, Cardinal H, Hébert MJ. Complement factor H deficiency in acute allograft glomerulopathy and post-transplant hemolytic uremic syndrome. Am J Transplant 2004; 4:270-3. [PMID: 14974950 DOI: 10.1046/j.1600-6143.2003.00286.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Acute allograft glomerulopathy (AAG) is a distinct form of allograft rejection characterized by cytotoxic T-cell-mediated injury to the renal glomerular and arteriolar endothelium. Acute allograft glomerulopathy is characterized by mononuclear cell infiltration of glomerular capillary tufts in association with endothelial cell hypertrophy and injury. Intra-glomerular thrombi have been described in AAG, suggesting that overlapping features of AAG and post-transplant thrombotic microangiopathy (TMA) may coexist. We present a case suggesting that complement factor H deficiency, a known hereditary risk factor for TMA, may also favor development of AAG. We discuss the potential implications of factor H deficiency in the pathophysiology of renal allograft microvascular injury, leukocyte infiltration and formation of intraglomerular platelet thrombi. We propose that unopposed complement activation is a risk factor for both immune and nonimmune forms of microvascular injuries in renal allografts.
Collapse
Affiliation(s)
- Marie-Chantal Fortin
- Transplant Division, Centre Hospitalier de l'Université de Montréal, University of Montreal, Montreal, QC, Canada
| | | | | | | |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Haemolytic uraemic syndrome (HUS) is a disease of diverse origin. The last year has witnessed the identification of a novel genetic marker of this disease, the description of the frequency of the factor H associated form of HUS in a registry of over 100 patients and a better understanding of the pathophysiology of the disease. RECENT FINDINGS In patients with atypical HUS, heterozygous mutations in the gene coding for the soluble complement regulator factor H are reported and most of the mutations cluster in the C-terminal recognition domain of the protein. A novel genetic marker for HUS has also been identified. Mutations occurring in the gene of the von Willebrand factor cleaving protease, ADAMTS13, which were previously linked to thrombotic thrombocytopenic purpura have now been identified in HUS patients. The frequency of factor H-associated HUS was established as 14% in a registry of German speaking countries and also 16 novel disease associated mutations were reported. The pathophysiology of factor H-associated HUS was analysed. Three analysed mutant proteins show normal complement regulatory activities but display defective recognition functions: reduced binding to surface attached C3b, to heparin/polyanions and to endothelial cells. SUMMARY The identification of effector molecules of the complement as well as the coagulation cascade as disease associated molecules indicate a regulatory protein network, which maintains integrity of endothelial cells during stress or infection. Defining the individual components and how their functional interaction causes microangiopathies will identify additional disease markers and will allow the design of proper diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Peter F Zipfel
- Department of Infection Biology, Hans Knoell Institute for Natural Products Research, Jena, Germany.
| | | | | |
Collapse
|
25
|
Ren G, Doshi M, Hack BK, Alexander JJ, Quigg RJ. Rat glomerular epithelial cells produce and bear factor H on their surface that is up-regulated under complement attack. Kidney Int 2003; 64:914-22. [PMID: 12911541 DOI: 10.1046/j.1523-1755.2003.00188.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Factor H is a potent complement inhibitory molecule that is primarily produced by the liver and appears in plasma as a soluble protein. Yet there is evidence that other cells, including those in the kidney, can produce factor H, and that it can be cell-associated as well as present as a plasma protein. Here we studied factor H in rat glomerular epithelial cells (GEC). METHODS A polyclonal antibody to factor H was used to identify factor H protein. A polymerase chain reaction (PCR)-based strategy was utilized to clone the full-length cDNA of GEC factor H. The relative quantity of factor H mRNA was measured by quantitative reverse transcription (RT)-PCR in cultured GEC exposed to complement activation and in the passive Heymann nephritis (PHN) model of membranous nephropathy. RESULTS By immunofluorescence microscopy, factor H protein was present on the plasma membranes of cultured GEC. Based upon Western blot studies, this appeared to be the full-length 150 kD factor H protein. Factor H cDNA cloned from GEC was identical to the newly deposited sequence for rat liver factor H cDNA. In cultured GEC in which complement was activated, factor H mRNA increased over time. Similarly, in the PHN model in which complement was activated on GEC in vivo, factor H mRNA and protein also increased over time. CONCLUSION Cultured GEC and glomeruli express factor H mRNA and protein. As modeled both in vitro and in vivo in the rat, factor H is up-regulated in membranous nephropathy. This is likely to be a direct response of GEC to complement attack and may represent a protective response of this cell.
Collapse
Affiliation(s)
- Guohui Ren
- Section of Nephrology, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | | |
Collapse
|
26
|
Gerber A, Kirchhoff-Moradpour AH, Obieglo S, Brandis M, Kirschfink M, Zipfel PF, Goodship JA, Zimmerhackl LB. Successful (?) therapy of hemolytic-uremic syndrome with factor H abnormality. Pediatr Nephrol 2003; 18:952-5. [PMID: 12836093 DOI: 10.1007/s00467-003-1192-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2002] [Revised: 04/07/2003] [Accepted: 04/07/2003] [Indexed: 10/26/2022]
Abstract
We report a patient with continuously recurring hemolytic-uremic syndrome due to factor H deficiency. First at the age of 3 months he showed signs of hemolytic anemia, thrombocytopenia and renal insufficiency, often recurring concomitantly with respiratory tract infections, despite weekly to twice weekly plasma substitution (20 ml/kg body weight). Now at the age of 3.5 years glomerular filtration rate is approximately 50 ml/min/1.73 m(2) and psychomotoric development is normal. Since factor H is mainly synthesized in the liver, hepatic transplantation has been proposed as curative treatment. Before justification of liver transplantation as the ultimate treatment for these patients, an international registry should be developed to optimize and standardize therapeutic alternatives.
Collapse
Affiliation(s)
- Angela Gerber
- Zentrum für Kinderheilkunde und Jugendmedizin, Universität Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Friese MA, Manuelian T, Junnikkala S, Hellwage J, Meri S, Peter HH, Gordon DL, Eibel H, Zipfel PF. Release of endogenous anti-inflammatory complement regulators FHL-1 and factor H protects synovial fibroblasts during rheumatoid arthritis. Clin Exp Immunol 2003; 132:485-95. [PMID: 12780697 PMCID: PMC1808733 DOI: 10.1046/j.1365-2249.2003.02173.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Rheumatoid arthritis is a chronic inflammatory disease of unknown aetiology predominantly affecting cells and tissues of synovial joints. Here we show that the two important complement regulators FHL-1 and factor H play a protective anti-inflammatory role in rheumatoid arthritis. Expression analyses at the mRNA- and protein level show in vitro expression and secretion of both regulators by synovial fibroblasts derived from patients with rheumatoid arthritis. Similarly the two regulators are synthesized in vivo in diseased synovial tissue, and in particular synovial lining cells express high levels of FHL-1. The anti-inflammatory role of these regulators in rheumatoid arthritis is highlighted by their induction with IFN-gamma and dexamethasone, whilst the pro-inflammatory cytokine TNF-alpha had no effect. Transient transfection experiments with various FHL-1/factor H promoter-luciferase reporter constructs into cells of distinct origin show independent cell and tissue specific promoter regulated transcription of these two regulators. The inducible expression, specifically of FHL-1 has physiological consequences. By binding directly to surfaces the released proteins protect cells from inflammatory damage and complement-mediated cell lysis. This study shows a novel protective and anti-inflammatory role of the two important complement regulators FHL-1 and factor H in rheumatoid arthritis and suggests a disease controlling role of the two proteins.
Collapse
Affiliation(s)
- M A Friese
- Research Group of Biomolecular Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Manuelian T, Hellwage J, Meri S, Caprioli J, Noris M, Heinen S, Jozsi M, Neumann HPH, Remuzzi G, Zipfel PF. Mutations in factor H reduce binding affinity to C3b and heparin and surface attachment to endothelial cells in hemolytic uremic syndrome. J Clin Invest 2003; 111:1181-90. [PMID: 12697737 PMCID: PMC152934 DOI: 10.1172/jci16651] [Citation(s) in RCA: 257] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hemolytic uremic syndrome (HUS) is a disease characterized by microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure. Recent studies have identified a factor H-associated form of HUS, caused by gene mutations that cluster in the C-terminal region of the complement regulator factor H. Here we report how three mutations (E1172Stop, R1210C, and R1215G; each of the latter two identified in three independent cases from different, unrelated families) affect protein function. All three mutations cause reduced binding to the central complement component C3b/C3d to heparin, as well as to endothelial cells. These defective features of the mutant factor H proteins explain progression of endothelial cell and microvascular damage in factor H-associated genetic HUS and indicate a protective role of factor H for tissue integrity during thrombus formation.
Collapse
Affiliation(s)
- Tamara Manuelian
- Hans Knoell Institute for Natural Products Research, Beutenbergstrasse 11a, D-07745 Jena, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hegasy GA, Manuelian T, Hogasen K, Jansen JH, Zipfel PF. The molecular basis for hereditary porcine membranoproliferative glomerulonephritis type II: point mutations in the factor H coding sequence block protein secretion. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:2027-34. [PMID: 12466119 PMCID: PMC1850924 DOI: 10.1016/s0002-9440(10)64481-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Porcine membranoproliferative glomerulonephritis type II in piglets of the Norwegian Yorkshire breed is considered the first animal model of human dense deposit disease. Porcine dense deposit disease is caused by the absence of the complement regulator factor H in plasma. Here we report the molecular basis for this absence. Single nucleotide exchanges at position C1590G and T3610G in the coding region of the factor H gene result in amino acid exchanges at nonframework residues L493V and I1166R that are located within SCR 9 and SCR 20, respectively. Apparently the L493V mutation represents a polymorphism whereas the I1166R causes the physiological consequences a block in protein secretion. Expression analysis shows comparable mRNA levels for factor H in liver tissue derived from both affected and healthy animals. In affected piglets, factor H protein is detected in increased amounts in liver cells. Factor H accumulates inside the hepatocytes and is not released as shown by Western blot analysis and immunohistochemistry. These data demonstrate that single amino acid exchanges of two nonframework amino acids either alone or in combination block protein secretion of factor H. This observation is also of interest for other human diseases in which factor H is involved, such as human factor H-associated form of hemolytic uremic syndrome.
Collapse
Affiliation(s)
- Guido A Hegasy
- Research Group for Biomolecular Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | | | | |
Collapse
|
30
|
|
31
|
Sánchez-Corral P, Pérez-Caballero D, Huarte O, Simckes AM, Goicoechea E, López-Trascasa M, de Córdoba SR. Structural and functional characterization of factor H mutations associated with atypical hemolytic uremic syndrome. Am J Hum Genet 2002; 71:1285-95. [PMID: 12424708 PMCID: PMC378565 DOI: 10.1086/344515] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2002] [Accepted: 08/28/2002] [Indexed: 01/15/2023] Open
Abstract
Genetic studies have demonstrated the involvement of the complement regulator factor H in nondiarrheal, nonverocytotoxin (i.e., atypical) cases of hemolytic uremic syndrome. Different factor H mutations have been identified in 10%-30% of patients with atypical hemolytic uremic syndrome (aHUS), and most of these mutations alter single amino acids in the C-terminal region of factor H. Although these mutations are considered to be responsible for the disease, the precise role that factor H plays in the pathogenesis of aHUS is unknown. We report here the structural and functional characterization of three different factor H proteins purified from the plasma of patients with aHUS who carry the factor H mutations W1183L, V1197A, or R1210C. Structural anomalies in factor H were found only in R1210C carriers; these individuals show, in their plasma, a characteristic high-molecular-weight factor H protein that results from the covalent interaction between factor H and human serum albumin. Most important, all three aHUS-associated factor H proteins have a normal cofactor activity in the proteolysis of fluid-phase C3b by factor I but show very low binding to surface-bound C3b. This functional impairment was also demonstrated in recombinant mutant factor H proteins expressed in COS7 cells. These data support the hypothesis that patients with aHUS carry a specific dysfunction in the protection of cellular surfaces from complement activation, offering new possibilities to improve diagnosis and develop appropriate therapies.
Collapse
Affiliation(s)
- Pilar Sánchez-Corral
- Departamento de Inmunología, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Remuzzi G, Ruggenenti P, Codazzi D, Noris M, Caprioli J, Locatelli G, Gridelli B. Combined kidney and liver transplantation for familial haemolytic uraemic syndrome. Lancet 2002; 359:1671-2. [PMID: 12020532 DOI: 10.1016/s0140-6736(02)08560-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Recurrent haemolytic uraemic syndrome (HUS) is a genetic form of thrombotic microangiopathy that is mostly associated with low activity of complement factor H. The disorder usually develops in families, leads to end stage renal disease, and invariably recurs after kidney transplantation. We did a simultaneous kidney and liver transplantation in a 2-year-old child with HUS and a mutation in complement factor H to restore the defective factor H, with no recurrence of the disease. The operation was successful, and at discharge, the child had healthy kidney and liver function, with no sign of haemolysis.
Collapse
|
34
|
Perkins SJ, Goodship THJ. Molecular modelling of the C-terminal domains of factor H of human complement: a correlation between haemolytic uraemic syndrome and a predicted heparin binding site. J Mol Biol 2002; 316:217-24. [PMID: 11851332 DOI: 10.1006/jmbi.2001.5337] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Factor H (FH) of the complement system acts as a regulatory cofactor for the factor I-mediated cleavage of C3b and binds to polyanionic substrates. FH is composed of 20 short consensus/complement repeat (SCR) domains. A set of 12 missense mutations in the C-terminal domains between SCR-16 to SCR-20 is associated with haemolytic uraemic syndrome. Recent structural models for intact FH permit the molecular interpretation of these amino acid substitutions. As all nine SCR-20 substitutions correspond to normal amounts of FH in plasma, and were localised in mostly surface-exposed positions, these are inferred to lead to a functional defect in FH. The nine substitutions occur in the same spatial region of SCR-20. As this surface coincides with conserved basic residues in the C-terminal SCR-20 domain, the substitutions provide direct evidence for a polyanionic binding surface. The positions of these conserved basic residues coincide with those of heparin-binding residues in the crystal structure of the acidic fibroblast growth factor-heparin complex. A tenth substitution and another conserved basic residue in SCR-19 are proximate to this binding site. As the remaining FH substitutions could also be correlated with their proximity to conserved basic residues, haemolytic uraemic syndrome may result from a failure of FH to interact with polyanions at cell surfaces in the kidney.
Collapse
Affiliation(s)
- Stephen J Perkins
- Department of Biochemistry and Molecular Biology, Royal Free and University College Medical School, University College London, Rowland Hill Street, London, NW3 2PF, UK.
| | | |
Collapse
|