151
|
|
152
|
Ding JD, Kelly U, Landowski M, Toomey CB, Groelle M, Miller C, Smith SG, Klingeborn M, Singhapricha T, Jiang H, Frank MM, Bowes Rickman C. Expression of human complement factor H prevents age-related macular degeneration-like retina damage and kidney abnormalities in aged Cfh knockout mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:29-42. [PMID: 25447048 DOI: 10.1016/j.ajpath.2014.08.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/21/2014] [Accepted: 08/27/2014] [Indexed: 01/17/2023]
Abstract
Complement factor H (CFH) is an important regulatory protein in the alternative pathway of the complement system, and CFH polymorphisms increase the genetic risk of age-related macular degeneration dramatically. These same human CFH variants have also been associated with dense deposit disease. To mechanistically study the function of CFH in the pathogenesis of these diseases, we created transgenic mouse lines using human CFH bacterial artificial chromosomes expressing full-length human CFH variants and crossed these to Cfh knockout (Cfh(-/-)) mice. Human CFH protein inhibited cleavage of mouse complement component 3 and factor B in plasma and in retinal pigment epithelium/choroid/sclera, establishing that human CFH regulates activation of the mouse alternative pathway. One of the mouse lines, which express relatively higher levels of CFH, demonstrated functional and structural protection of the retina owing to the Cfh deletion. Impaired visual function, detected as a deficit in the scotopic electroretinographic response, was improved in this transgenic mouse line compared with Cfh(-/-) mice, and transgenics had a thicker outer nuclear layer and less sub-retinal pigment epithelium deposit accumulation. In addition, expression of human CFH also completely protected the mice from developing kidney abnormalities associated with loss of CFH. These humanized CFH mice present a valuable model for study of the molecular mechanisms of age-related macular degeneration and dense deposit disease and for testing therapeutic targets.
Collapse
Affiliation(s)
- Jin-Dong Ding
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Una Kelly
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Michael Landowski
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Christopher B Toomey
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - Marybeth Groelle
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Chelsey Miller
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Stephanie G Smith
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Mikael Klingeborn
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Terry Singhapricha
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Haixiang Jiang
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Michael M Frank
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
153
|
Shen L, Frazer-Abel A, Reynolds PR, Giclas PC, Chappell A, Pangburn MK, Younis H, Henry SP. Mechanistic understanding for the greater sensitivity of monkeys to antisense oligonucleotide-mediated complement activation compared with humans. J Pharmacol Exp Ther 2014; 351:709-17. [PMID: 25301170 DOI: 10.1124/jpet.114.219378] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Differences in sensitivity of monkeys and humans to antisense oligonucleotide (ASO)-induced complement alternative pathway (AP) activation were evaluated in monkeys, humans, and in serum using biochemical assays. Transient AP activation was evident in monkeys at higher doses of two 2'-O-methoxyethyl (2'-MOE) ASOs (ISIS 426115 and ISIS 183750). No evidence of AP activation was observed in humans for either ASO, even with plasma ASO concentrations that reached the threshold for activation in monkeys. The absence of complement activation in humans is consistent with a query of the Isis Clinical Safety Database containing 767 subjects. The in vivo difference in sensitivity was confirmed in vitro, as monkey and human serum exposed to increasing concentrations of ASO indicated that monkeys were more sensitive to AP activation with this class of compounds. The mechanistic basis for the greater sensitivity of monkeys to AP activation by 2'-MOE ASO was evaluated using purified human or monkey factor H protein. The binding affinities between a representative 2'-MOE ASO and either purified protein are similar. However, the IC50 of fluid-phase complement inhibition for monkey factor H is about 3-fold greater than that for human protein using either monkey serum or factor H-depleted human serum. Interestingly, there is a sequence variant in the monkey complement factor H gene similar to a single nucleotide polymorphism in humans that is correlated with decreased factor H protein function. These findings show that monkeys are more sensitive to 2'-MOE ASO-mediated complement activation than humans likely because of differences in factor H inhibitory capacity.
Collapse
Affiliation(s)
- Lijiang Shen
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| | - Ashley Frazer-Abel
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| | - Paul R Reynolds
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| | - Patricia C Giclas
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| | - Alfred Chappell
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| | - Michael K Pangburn
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| | - Husam Younis
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| | - Scott P Henry
- Isis Pharmaceuticals, Inc., Carlsbad, California (L.S., A.C., H.Y., S.P.H.); National Jewish Health, Denver, Colorado (A.F.-A., P.R.R., P.C.G.); and University of Texas Health Science Center, Tyler, Texas (M.K.P.)
| |
Collapse
|
154
|
Johnson SA, Wong EKS, Taylor CM. Making sense of the spectrum of glomerular disease associated with complement dysregulation. Pediatr Nephrol 2014; 29:1883-94. [PMID: 23852337 DOI: 10.1007/s00467-013-2559-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 05/14/2013] [Accepted: 05/29/2013] [Indexed: 01/15/2023]
Abstract
Over recent years, complement has emerged as a major player in the development of a number of glomerular diseases, including atypical haemolytic uraemic syndrome, membranoproliferative glomerulonephritis and the recently described C3 glomerulonephritis. Some patients and pedigrees show overlapping features of these conditions. Intriguingly, a few complement gene mutations are common to different disease phenotypes. In this review, we explore the evidence for complement dysregulation in these diseases and the clinical interface between them, and present a hypothesis to explain the variable phenotype associated with dysregulation of the alternative complement pathway.
Collapse
Affiliation(s)
- Sally Ann Johnson
- Paediatric Nephrology, Great North Children's Hospital, Newcastle Upon Tyne, UK,
| | | | | |
Collapse
|
155
|
Horie-Inoue K, Inoue S. Genomic aspects of age-related macular degeneration. Biochem Biophys Res Commun 2014; 452:263-75. [PMID: 25111812 DOI: 10.1016/j.bbrc.2014.08.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/04/2014] [Indexed: 11/29/2022]
Abstract
Age-related macular degeneration (AMD) is a major late-onset posterior eye disease that causes central vision to deteriorate among elderly populations. The predominant lesion of AMD is the macula, at the interface between the outer retina and the inner choroid. Recent advances in genetics have revealed that inflammatory and angiogenic pathways play critical roles in the pathophysiology of AMD. Genome-wide association studies have identified ARMS2/HTRA1 and CFH as major AMD susceptibility genes. Genetic studies for AMD will contribute to the prevention of central vision loss, the development of new treatment, and the maintenance of quality of vision for productive aging.
Collapse
Affiliation(s)
- Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan.
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Department of Anti-Aging Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
156
|
Andreeva K, Zhang M, Fan W, Li X, Chen Y, Rebolledo-Mendez JD, Cooper NG. Time-dependent Gene Profiling Indicates the Presence of Different Phases for Ischemia/Reperfusion Injury in Retina. OPHTHALMOLOGY AND EYE DISEASES 2014; 6:43-54. [PMID: 25210480 PMCID: PMC4149383 DOI: 10.4137/oed.s17671] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 02/05/2023]
Abstract
Ischemia/reperfusion (IR) injury has been associated with several retinal pathologies, and a few genes/gene products have been linked to IR injury. However, the big picture of temporal changes, regarding the affected gene networks, pathways, and processes remains to be determined. The purpose of the present study was to investigate initial, intermediate, and later stages to characterize the etiology of IR injury in terms of the pathways affected over time. Analyses indicated that at the initial stage, 0-hour reperfusion following the ischemic period, the ischemia-associated genes were related to changes in metabolism. In contrast, at the 24-hour time point, the signature events in reperfusion injury include enhanced inflammatory and immune responses as well as cell death indicating that this would be a critical period for the development of any interventional therapeutic strategies. Genes in the signal transduction pathways, particularly transmitter receptors, are downregulated at this time. Activation of the complement system pathway clearly plays an important role in the later stages of reperfusion injury. Together, these results demonstrate that the etiology of injury related to IR is characterized by the appearance of specific patterns of gene expression at any given time point during retinal IR injury. These results indicate that evaluation of treatment strategies with respect to time is very critical.
Collapse
Affiliation(s)
- Kalina Andreeva
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Meixia Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Fan
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaohong Li
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Yinlu Chen
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Jovan D Rebolledo-Mendez
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nigel G Cooper
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| |
Collapse
|
157
|
Mehta G, Ferreira VP, Skerka C, Zipfel PF, Banda NK. New insights into disease-specific absence of complement factor H related protein C in mouse models of spontaneous autoimmune diseases. Mol Immunol 2014; 62:235-48. [PMID: 25033230 DOI: 10.1016/j.molimm.2014.06.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 06/21/2014] [Indexed: 12/18/2022]
Abstract
Complement factor H (CFH) protein is an inhibitor of the alternative pathway of complement (AP) both in the fluid phase and on the surface of host cells. Mouse and human complement factor H-related (CFHR) proteins also belong to the fH family of plasma glycoproteins. The main goal of the current study was to compare the presence of mRNA for two mCFHR proteins in spontaneously developing autoimmune diseases in mice such as dense deposit disease (DDD), diabetes mellitus (DM), basal laminar deposits (BLD), collagen antibody-induced arthrits (CAIA) and systemic lupus erythematosus (SLE). Here we report for the first time that the CFHR-C mRNA was universally absent in the liver from three strains of lupus-prone mice and in a diabetic-prone mouse strain. The mRNA levels (pg/ng) for CFH and CFHR-B in MRL-lpr/lpr, at 9 wks and 23 wks were 707.2±44.4, 54.5±5.75 and 729±252.9, 74.04±22.76, respectively. The mRNA levels for CFH and CFHR-B in NZB/NZW mice, at 9 wks and 54 wks were 579.9±23.8, 58.8±1.41 and 890.3±135.2, 63.30±9.2, respectively. CFHR-C protein was absent in the circulation of MRL-lpr/lpr and NZB/NZW mice before and after the development of lupus. Similarly, mRNA and protein for CFHR-C was universally absent in liver and other organs and in the circulation of NOD mice before and after the development of DM. In contrast, the mRNAs for CFH, CFHR-B and CFHR-C were universally present in the liver from mice with and without DDD, BLD and CAIA. The levels of mRNA for CFHR-B in mice with and without BLD were ∼4 times higher than the mice with lupus. The complete absence of mRNA for CFHR-C in lupus and diabetic-prone strains indicates that polymorphic variation within the mouse CFHR family exists and raises the possibility that such variation contributes to lupus and diabetic phenotypes.
Collapse
Affiliation(s)
- Gaurav Mehta
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | | | | - Peter F Zipfel
- Hans Knoell Institute, Jena, Germany; Friedrich Schiller University, Jena, Germany
| | - Nirmal K Banda
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
158
|
Abstract
Complement activation and recruitment of inflammatory leukocytes is an important defense mechanism against bacterial infection. However, complement also can mediate cellular injury and contribute to the pathogenesis of various diseases. With the appreciation that the C5b-9 membrane attack complex can injure cells in the absence of leukocytes, a role for the terminal complement pathway in inducing cell injury and kidney disease was shown in several experimental models, including the rat passive Heymann nephritis model of human membranous nephropathy. In podocytes, sublytic C5b-9 activates a variety of downstream pathways including protein kinases, lipid metabolism, reactive oxygen species, growth factors/gene transcription, endoplasmic reticulum stress, and the ubiquitin-proteasome system, and it impacts the integrity of the cytoskeleton and slit diaphragm proteins. C5b-9 also injures other kidney cells, including mesangial, glomerular endothelial, and tubular epithelial cells, and it contributes to the pathogenesis of mesangial-proliferative glomerulonephritis, thrombotic microangiopathy, and acute kidney injury. Conversely, certain C5b-9 signals limit complement-induced injury, or promote recovery of cells. In addition to C5b-9, complement cleavage products, such as C5a and C1q, can injure kidney cells. Thus, the complement system contributes to various kidney pathologies by causing cellular damage in both an inflammation-dependent and inflammation-independent manner.
Collapse
Affiliation(s)
- Tomoko Takano
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
159
|
Abstract
Activation of the complement system is tightly regulated by plasma and cell-associated complement regulatory proteins (CRPs), such as factor H (fH), decay-accelerating factor, and membrane cofactor protein. Animal models of disease have provided considerable insights into the important roles for CRPs in the kidney. Mice deficient in fH have excessive fluid phase C3 activation and inactivation, leading to deposition of inactivated C3b in glomerular capillary walls (GCW), comparable with dense deposit disease. In contrast, when fH lacks C-terminal surface targeting regions, local activation on the GCW leads to a disease reminiscent of thrombotic microangiopathy. The uniquely rodent protein, CR1-related y (Crry), has features analogous to human membrane cofactor protein. Defective Crry leads to unrestricted alternative pathway activation in the tubulointerstitium, resulting in pathologic features ranging from thrombotic microangiopathy (TMA), acute kidney injury, and tubulointerstitium nephritis. In the presence of initiators of the classic or lectin pathways, commonly in the form of immune complexes in human glomerular diseases, complement regulation is stressed, with the potential for recruitment of the spontaneously active alternative pathway. The threshold for this activation is set by CRPs; pathology is more likely when complement regulation is defective. Within the endocapillary region of the GCW, fH is key, while decay-accelerating factor and Crry are protective on mesangial cells and podocytes. Arguably, acquired alterations in these CRPs is a more common event, extending from pathologic states of cellular injury or production of inhibitory antibodies, to physiological fine tuning of the adaptive immune response.
Collapse
|
160
|
Ruseva MM, Takahashi M, Fujita T, Pickering MC. C3 dysregulation due to factor H deficiency is mannan-binding lectin-associated serine proteases (MASP)-1 and MASP-3 independent in vivo. Clin Exp Immunol 2014; 176:84-92. [PMID: 24279761 PMCID: PMC3958157 DOI: 10.1111/cei.12244] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2013] [Indexed: 11/28/2022] Open
Abstract
Uncontrolled activation of the complement alternative pathway is associated with complement-mediated renal disease. Factor B and factor D are essential components of this pathway, while factor H (FH) is its major regulator. In complete FH deficiency, uncontrolled C3 activation through the alternative pathway results in plasma C3 depletion and complement-mediated renal disease. These are dependent on factor B. Mannan-binding lectin-associated serine proteases 1 and 3 (MASP-1, MASP-3) have been shown recently to contribute to alternative pathway activation by cleaving pro-factor D to its active form, factor D. We studied the contribution of MASP-1 and MASP-3 to uncontrolled alternative pathway activation in experimental complete FH deficiency. Co-deficiency of FH and MASP-1/MASP-3 did not ameliorate either the plasma C3 activation or glomerular C3 accumulation in FH-deficient mice. Our data indicate that MASP-1 and MASP-3 are not essential for alternative pathway activation in complete FH deficiency.
Collapse
Affiliation(s)
- M M Ruseva
- Centre for Complement & Inflammation Research, Imperial College London, London, UK
| | | | | | | |
Collapse
|
161
|
Touzot M, Obada EN, Beaudreuil S, François H, Durrbach A. Complement modulation in solid-organ transplantation. Transplant Rev (Orlando) 2014; 28:119-25. [PMID: 24996770 DOI: 10.1016/j.trre.2014.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 03/08/2014] [Indexed: 01/18/2023]
Abstract
The complement system is a major constituent of the innate immune system. It has a critical role in defense against pathogens but dysregulation of complement activation may lead to tissue injury and modulate the adaptive immune response. In organ transplantation, local complement activation is involved in hyper-acute rejection and antibody-mediated rejection. This last decade, interest in complement activation has increased due to new insights into the pathophysiology of antibody-mediated rejection, but also since the availability of news drugs that target terminal complement activation. In this review, we discuss our current understanding of how local complement activation induces acute and chronic graft injury, and review recent advances in clinical trials that block complement activation using the anti-C5 monoclonal antibody, eculizumab. Finally, we discuss how complement-targeted therapy may be integrated into our current immunosuppressive regimen and what type of patient will benefit most from this therapy.
Collapse
Affiliation(s)
- Maxime Touzot
- Nephrology Department, IFRNT, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale INSERM U1014, Villejuif, France
| | | | - Severine Beaudreuil
- Nephrology Department, IFRNT, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale INSERM U1014, Villejuif, France
| | - Hélène François
- Nephrology Department, IFRNT, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale INSERM U1014, Villejuif, France
| | - Antoine Durrbach
- Nephrology Department, IFRNT, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale INSERM U1014, Villejuif, France.
| |
Collapse
|
162
|
Frémeaux-Bacchi V. [Pathophysiology of atypical hemolytic uremic syndrome. Ten years of progress, from laboratory to patient]. Biol Aujourdhui 2014; 207:231-40. [PMID: 24594571 DOI: 10.1051/jbio/2013027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Indexed: 11/14/2022]
Abstract
Hemolytic Uremic Syndrome (HUS) is characterized by the triad of hemolytic anemia, thrombocytopenia and acute renal failure. The most frequent form in children is caused by Shiga-toxin producing Escherichia coli. In absence of Shiga-toxin infection, the HUS is called atypical (aHUS). Some HUS are secondary to Streptococcus pneumonia or human immunodeficiency virus infection, cancer, anti-cancer drugs, or cyclosporine. During the last decade, aHUS has been demonstrated to be a disorder of complement alternative pathway regulation. aHUS must be regarded as a complex polygenic disease which results from a combination of genetic risk factors. Approximately 60% of patients have mutations in the genes encoding complement factor H (20-30% of patients), MCP (10-15%), factor I (4-10%), factor B (1-2%) or C3 (5-10%), and 6% have anti-factor H antibodies. Prognosis is severe whereas the clinical features vary according to complement abnormality. aHUS touches both children and adults, but in children very early onset is characteristic of factor H and factor I-HUS, while MCP-HUS is not observed before the age of 1. Half of patients with adult onset have a rapid evolution to end-stage renal disease, but half recover. The best prognosis is in patients with MCP (Membrane Cofactor Protein) mutation and a pediatric onset of the disease, who have a relapsing course, but a risk of end-stage renal disease of only 15-30% at 5 years follow-up. Anti-factor H antibodies-HUS is mainly observed in (pre)adolescents and appears to have a favourable outcome if treated early. There is a high risk of post-transplant recurrence in all groups, except MCP-HUS. These findings have paved the way for innovative therapeutic strategies based on complement blockade, and eculizumab, a monoclonal antibody targeting the human complement component 5, is now widely used to treat aHUS. Mutations in the gene of thrombomodulin and diacylglycerol kinase epsilon (DGKe) have been reported, suggesting the possibility of an alternative or more complex disease-causing mechanism than previously thought.
Collapse
Affiliation(s)
- Véronique Frémeaux-Bacchi
- INSERM UMR_S1135, Centre de Recherche des Cordeliers, Équipe « Complément et Maladies », 15 rue de l'École de Médecine, 75006 Paris, France - Hôpital Européen Georges Pompidou, Laboratoire d'Immunologie, 20 rue Leblanc, 75015 Paris, France
| |
Collapse
|
163
|
Complement factor H and related proteins in age-related macular degeneration. C R Biol 2014; 337:178-84. [DOI: 10.1016/j.crvi.2013.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 12/17/2022]
|
164
|
Bonomo JA, Palmer ND, Hicks PJ, Lea JP, Okusa MD, Langefeld CD, Bowden DW, Freedman BI. Complement factor H gene associations with end-stage kidney disease in African Americans. Nephrol Dial Transplant 2014; 29:1409-14. [PMID: 24586071 DOI: 10.1093/ndt/gfu036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mutations in the complement factor H gene (CFH) region associate with renal-limited mesangial proliferative forms of glomerulonephritis including IgA nephropathy (IgAN), dense deposit disease (DDD) and C3 glomerulonephritis (C3GN). Lack of kidney biopsies could lead to under diagnosis of CFH-associated end-stage kidney disease (ESKD) in African Americans (AAs), with incorrect attribution to other causes. A prior genome-wide association study in AAs with non-diabetic ESKD implicated an intronic CFH single nucleotide polymorphism (SNP). METHODS Thirteen CFH SNPs (8 exonic, 2 synonymous, 2 3'UTR, and the previously associated intronic variant rs379489) were tested for association with common forms of non-diabetic and type 2 diabetes-associated (T2D) ESKD in 3770 AAs (1705 with non-diabetic ESKD, 1305 with T2D-ESKD, 760 controls). Most cases lacked kidney biopsies; those with known IgAN, DDD or C3GN were excluded. RESULTS Adjusting for age, gender, ancestry and apolipoprotein L1 gene risk variants, single SNP analyses detected 6 CFH SNPs (5 exonic and the intronic variant) as significantly associated with non-diabetic ESKD (P = 0.002-0.01), three of these SNPs were also associated with T2D-ESKD. Weighted CFH locus-wide Sequence Kernel Association Testing (SKAT) in non-diabetic ESKD (P = 0.00053) and T2D-ESKD (P = 0.047) confirmed significant evidence of association. CONCLUSIONS CFH was associated with commonly reported etiologies of ESKD in the AA population. These results suggest that a subset of cases with ESKD clinically ascribed to the effects of hypertension or glomerulosclerosis actually have CFH-related forms of mesangial proliferative glomerulonephritis. Genetic testing may prove useful to identify the causes of renal-limited kidney disease in patients with ESKD who lack renal biopsies.
Collapse
Affiliation(s)
- Jason A Bonomo
- Department of Molecular Medicine and Translational Science, Wake Forest School of Medicine, Winston-Salem, NC, USA Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nicholette D Palmer
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Pamela J Hicks
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Janice P Lea
- Division of Renal Medicine, Department of Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Mark D Okusa
- Division of Nephrology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Carl D Langefeld
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Donald W Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barry I Freedman
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
165
|
Chauvet S, Servais A, Frémeaux-Bacchi V. [C3 glomerulopathy]. Nephrol Ther 2014; 10:78-85. [PMID: 24508002 DOI: 10.1016/j.nephro.2013.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 08/13/2013] [Accepted: 09/10/2013] [Indexed: 10/25/2022]
Abstract
C3 glomerulopathy is an heterogeneous group of glomerular diseases associated with acquired or genetic abnormalities of complement alternative pathway (AP) components. It is characterized by predominant C3 deposits in the mesangium and along the glomerular basement membrane (GBM). Presenting features comprise proteinuria (sometimes with nephritic syndrome), haematuria, hypertension and renal failure. C3 glomerulopathy have a poor renal prognosis with progression to end stage renal disease (ESRD) in 50% of cases during the first decade after initial presentation. Moreover, C3 deposits recur in most of cases after renal transplantation. Patients frequently have low serum C3 level attributed to the activation of the alternative pathway of complement. Animal models have confirmed the role of excessive C3 activation in the pathogenesis of C3 glomerulopathy. To date, the optimal treatment remains unknown. It is currently based on the use of angiotensin-converting-enzyme inhibitors (ACEI) and angiotensin II-receptor blockers (ARB), sometimes associated with immunosuppressive therapy. Blockade of C5a release with eculizumab, a monoclonal anti-C5 antibody, may be of particular interest in the treatment of C3G.
Collapse
Affiliation(s)
- Sophie Chauvet
- Service de néphrologie, hôpital Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France.
| | - Aude Servais
- Service de néphrologie, hôpital Necker-Enfants-Malades, 149, rue de Sèvres, 75015 Paris, France
| | | |
Collapse
|
166
|
Rowan S, Weikel K, Chang ML, Nagel BA, Thinschmidt JS, Carey A, Grant MB, Fliesler SJ, Smith D, Taylor A. Cfh genotype interacts with dietary glycemic index to modulate age-related macular degeneration-like features in mice. Invest Ophthalmol Vis Sci 2014; 55:492-501. [PMID: 24370827 DOI: 10.1167/iovs.13-12413] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Age-related macular degeneration (AMD) is a leading cause of visual impairment worldwide. Genetics and diet contribute to the relative risk for developing AMD, but their interactions are poorly understood. Genetic variations in Complement Factor H (CFH), and dietary glycemic index (GI) are major risk factors for AMD. We explored the effects of GI on development of early AMD-like features and changes to central nervous system (CNS) inflammation in Cfh-null mice. METHODS Aged 11-week-old wild type (WT) C57Bl/6J or Cfh-null mice were group pair-fed high or low GI diets for 33 weeks. At 10 months of age, mice were evaluated for early AMD-like features in the neural retina and RPE by light and electron microscopy. Brains were analyzed for Iba1 macrophage/microglia immunostaining, an indicator of inflammation. RESULTS The 10-month-old WT mice showed no retinal abnormalities on either diet. The Cfh-null mice, however, showed distinct early AMD-like features in the RPE when fed a low GI diet, including vacuolation, disruption of basal infoldings, and increased basal laminar deposits. The Cfh-null mice also showed thinning of the RPE, hypopigmentation, and increased numbers of Iba1-expressing macrophages in the brain, irrespective of diet. CONCLUSIONS The presence of early AMD-like features by 10 months of age in Cfh-null mice fed a low GI diet is surprising, given the apparent protection from the development of such features in aged WT mice or humans consuming lower GI diets. Our findings highlight the need to consider gene-diet interactions when developing animal models and therapeutic approaches to treat AMD.
Collapse
Affiliation(s)
- Sheldon Rowan
- JM-USDA Human Nutrition Research Center on Aging (HNRCA), Tufts University, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Abstract
Membranoproliferative GN represents a pattern of injury seen on light microscopy. Historically, findings on electron microscopy have been used to further subclassify this pathologic entity. Recent advances in understanding of the underlying pathobiology have led to a proposed classification scheme based on immunofluorescence findings. Dysregulation of the complement system has been shown to be a major risk factor for the development of a membranoproliferative GN pattern of injury on kidney biopsy. Evaluation and treatment of this complex disorder rest on defining the underlying mechanisms.
Collapse
Affiliation(s)
- Naveed Masani
- Division of Nephrology, Winthrop University Hospital, Mineola, New York, †Division of Kidney Diseases and Hypertension, Hofstra North Shore-Long Island Jewish School of Medicine, Great Neck, New York
| | | | | |
Collapse
|
168
|
Ding JD, Kelly U, Groelle M, Christenbury JG, Zhang W, Bowes Rickman C. The role of complement dysregulation in AMD mouse models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:213-9. [PMID: 24664701 DOI: 10.1007/978-1-4614-3209-8_28] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Variations in several complement genes are now known to be significant risk factors for the development of age-related macular degeneration (AMD). Despite dramatic effects on disease susceptibility, the underlying mechanisms by which common polymorphisms in complement proteins alter disease risk have remained unclear. Genetically modified mice in which the activity of the complement has been altered are available and can be used to investigate the role of complement in the pathogenesis of AMD. In this mini review, we will discuss some existing complement models of AMD and our efforts to develop and characterize the ocular phenotype in a variety of mice in which complement is either chronically activated or inhibited. A spectrum of complement dysregulation was modeled on the APOE4 AMD mouse model by crossing these mice to complement factor H knockout (cfh-/-) mice to test the impact of excess complement activation, and by crossing them to soluble-complement-receptor-1-related protein y (sCrry) mice, in which sCrry acts as a potent inhibitor of mouse complement acting in a manner similar to CFH. In addition, we have also generated humanized CFH mice expressing normal and risk variants of CFH.
Collapse
Affiliation(s)
- Jin-Dong Ding
- Department of Ophthalmology, Duke University Medical Center, 2351 Erwin Rd, 27710, Durham, NC, USA,
| | | | | | | | | | | |
Collapse
|
169
|
Bowes Rickman C, Farsiu S, Toth CA, Klingeborn M. Dry age-related macular degeneration: mechanisms, therapeutic targets, and imaging. Invest Ophthalmol Vis Sci 2013; 54:ORSF68-80. [PMID: 24335072 DOI: 10.1167/iovs.13-12757] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration is the leading cause of irreversible visual dysfunction in individuals over 65 in Western Society. Patients with AMD are classified as having early stage disease (early AMD), in which visual function is affected, or late AMD (generally characterized as either "wet" neovascular AMD, "dry" atrophic AMD or both), in which central vision is severely compromised or lost. Until recently, there have been no therapies available to treat the disorder(s). Now, the most common wet form of late-stage AMD, choroidal neovascularization, generally responds to treatment with anti-vascular endothelial growth factor therapies. Nevertheless, there are no current therapies to restore lost vision in eyes with advanced atrophic AMD. Oral supplementation with the Age-Related Eye Disease Study (AREDS) or AREDS2 formulation (antioxidant vitamins C and E, lutein, zeaxanthin, and zinc) has been shown to reduce the risk of progression to advanced AMD, although the impact was in neovascular rather than atrophic AMD. Recent findings, however, have demonstrated several features of early AMD that are likely to be druggable targets for treatment. Studies have established that much of the genetic risk for AMD is associated with complement genes. Consequently, several complement-based therapeutic treatment approaches are being pursued. Potential treatment strategies against AMD deposit formation and protein and/or lipid deposition will be discussed, including anti-amyloid therapies. In addition, the role of autophagy in AMD and prevention of oxidative stress through modulation of the antioxidant system will be explored. Finally, the success of these new therapies in clinical trials and beyond relies on early detection, disease typing, and predicting disease progression, areas that are currently being rapidly transformed by improving imaging modalities and functional assays.
Collapse
|
170
|
Abstract
C3 glomerulopathy refers to those renal lesions characterized histologically by predominant C3 accumulation within the glomerulus, and pathogenetically by aberrant regulation of the alternative pathway of complement. Dense deposit disease is distinguished from other forms of C3 glomerulopathy by its characteristic appearance on electron microscopy. The extent to which dense deposit disease also differs from other forms of C3 glomerulopathy in terms of clinical features, natural history, and outcomes of treatment including renal transplantation is less clear. We discuss the pathophysiology of C3 glomerulopathy, with evidence for alternative pathway dysregulation obtained from affected individuals and complement factor H (Cfh)-deficient animal models. Recent linkage studies in familial C3 glomerulopathy have shown genomic rearrangements in the Cfh-related genes, for which the novel pathophysiologic concept of Cfh deregulation has been proposed.
Collapse
Affiliation(s)
- Thomas D Barbour
- Kidney Research UK, Centre for Complement and Inflammation Research, Imperial College London, London, United Kingdom.
| | | | | |
Collapse
|
171
|
Affiliation(s)
- David J Salant
- Department of Medicine, Boston University Medical Center, Boston, MA
| |
Collapse
|
172
|
Feng S, Liang X, Cruz MA, Vu H, Zhou Z, Pemmaraju N, Dong JF, Kroll MH, Afshar-Kharghan V. The interaction between factor H and Von Willebrand factor. PLoS One 2013; 8:e73715. [PMID: 23991205 PMCID: PMC3753316 DOI: 10.1371/journal.pone.0073715] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 07/30/2013] [Indexed: 01/22/2023] Open
Abstract
Complement factor H (fH) is a plasma protein that regulates activation of the alternative pathway, and mutations in fH are associated with a rare form of thrombotic microangiopathy (TMA), known as atypical hemolytic uremic syndrome (aHUS). A more common TMA is thrombotic thrombocytopenic purpura, which is caused by the lack of normal ADAMTS-13-mediated cleavage of von Willebrand factor (VWF). We investigated whether fH interacts with VWF and affects cleavage of VWF. We found that factor H binds to VWF in plasma, to plasma-purified VWF, and to recombinant A1 and A2 domains of VWF as detected by co-immunoprecipitation (co-IP) and surface plasmon resonance assays. Factor H enhanced ADAMTS-13-mediated cleavage of recombinant VWF-A2 as determined by quantifying the cleavage products using Western-blotting, enhanced cleavage of a commercially available fragment of VWF-A2 (FRETS-VWF73) as determined by fluorometric assay, and enhanced cleavage of ultralarge (UL) VWF under flow conditions as determined by cleavage of VWF-platelet strings attached to histamine stimulated endothelial cells. Using recombinant full-length and truncated fH molecules, we found that the presence of the C-terminal half of fH molecule is important for binding to VWF-A2 and for enhancing cleavage of the A2 domain by ADAMTS-13. We conclude that factor H binds to VWF and may modulate cleavage of VWF by ADAMTS-13.
Collapse
Affiliation(s)
- Shuju Feng
- Division of Internal Medicine, Benign Hematology, University of Texas, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiaowen Liang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, United States of America
| | - Miguel A. Cruz
- Cardiovascular Research Section, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hangoc Vu
- Division of Internal Medicine, Benign Hematology, University of Texas, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Zhou Zhou
- Puget Sound Blood Center, Seattle, Washington, United States of America
| | - Naresh Pemmaraju
- Division of Internal Medicine, Benign Hematology, University of Texas, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Jing-Fei Dong
- Puget Sound Blood Center, Seattle, Washington, United States of America
| | - Michael H. Kroll
- Division of Internal Medicine, Benign Hematology, University of Texas, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, University of Texas, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
173
|
Jacob A, Chaves L, Eadon MT, Chang A, Quigg RJ, Alexander JJ. Curcumin alleviates immune-complex-mediated glomerulonephritis in factor-H-deficient mice. Immunology 2013; 139:328-37. [PMID: 23347386 DOI: 10.1111/imm.12079] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 12/31/2022] Open
Abstract
Complement factor H (Cfh) is a key regulator of the complement cascade and protects C57BL/6 mice from immune complex-mediated complement-dependent glomerulonephritis. In chronic serum sickness (CSS) there are increased deposits of immune complexes in the glomeruli with inflammation and a scarring phenotype. As cucurmin is an effective anti-inflammatory agent and reduces complement activation, we hypothesized that it should alleviate renal disease in this setting. To determine the effectiveness of curcumin, an apoferritin-induced CSS model in Cfh-deficient (Cfh(-/-)) mice was used. Curcumin treatment (30 mg/kg) given every day in parallel with apoferritin reduced glomerulonephritis and enhanced kidney function (blood urea nitrogen, 45·4 ± 7·5 versus 35·6 ± 5·1; albuminuria, 50·1 ± 7·1 versus 15·7 ± 7·1; glomerulonephritis, 2·62 + 0·25 versus 2 + 0·3, P < 0·05). In line with reduced IgG deposits in mice with CSS given curcumin, C9 deposits were reduced indicating reduced complement activation. Mice treated with curcumin had a significant reduction in the number of splenic CD19(+) B cells and the ratio of CD19 : CD3 cells (P < 0·05) with no change in the T-cell population. Myeloperoxidase assay showed reduced macrophages in the kidney. However, a significant reduction in the M2 subset of splenic macrophages by apoferritin was prevented by curcumin, suggesting a protective function. Curcumin treatment reduced mRNA expression of inflammatory proteins monocyte chemoattractant protein-1 and transforming growth factor-β and matrix proteins, fibronectin, laminin and collagen. Our results clearly illustrate that curcumin reduces glomerulosclerosis, improves kidney function and could serve as a therapeutic agent during serum sickness.
Collapse
Affiliation(s)
- Alexander Jacob
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | | | | | | | | |
Collapse
|
174
|
Zhang Y, Nester CM, Holanda DG, Marsh HC, Hammond RA, Thomas LJ, Meyer NC, Hunsicker LG, Sethi S, Smith RJH. Soluble CR1 therapy improves complement regulation in C3 glomerulopathy. J Am Soc Nephrol 2013; 24:1820-9. [PMID: 23907509 DOI: 10.1681/asn.2013010045] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) are widely recognized subtypes of C3 glomerulopathy. These ultra-rare renal diseases are characterized by fluid-phase dysregulation of the alternative complement pathway that leads to deposition of complement proteins in the renal glomerulus. Disease triggers are unknown and because targeted treatments are lacking, progress to end stage renal failure is a common final outcome. We studied soluble CR1, a potent regulator of complement activity, to test whether it restores complement regulation in C3 glomerulopathy. In vitro studies using sera from patients with DDD showed that soluble CR1 prevents dysregulation of the alternative pathway C3 convertase, even in the presence of C3 nephritic factors. In mice deficient in complement factor H and transgenic for human CR1, soluble CR1 therapy stopped alternative pathway activation, resulting in normalization of serum C3 levels and clearance of iC3b from glomerular basement membranes. Short-term use of soluble CR1 in a pediatric patient with end stage renal failure demonstrated its safety and ability to normalize activity of the terminal complement pathway. Overall, these data indicate that soluble CR1 re-establishes regulation of the alternative complement pathway and provide support for a limited trial to evaluate soluble CR1 as a treatment for DDD and C3GN.
Collapse
Affiliation(s)
- Yuzhou Zhang
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss emerging nomenclature, review the salient clinicopathological features and describe the therapeutic options available for the treatment of C3 glomerulopathy (C3G). RECENT FINDINGS C3G is minimally responsive to traditional immune suppression and randomized controlled trials to support therapy are absent. The burgeoning understanding of the role of the alternative complement pathway in C3G combined with animal data supporting the use of terminal complement blockade and a few reports suggesting that the anticomplement drug eculizumab may offer a therapeutic advantage have triggered great interest in the field of complement-mediated renal disease. SUMMARY Anticellular immune suppression and plasma therapy have limited efficacy in C3G. Data suggest that eculizumab may ameliorate disease in some C3G patients. The limited, recently published cohort data highlight crucial aspects of this group of diseases and support the need for extensive genetic and biomarker research to validate the pathologic mechanisms, delineate the spectrum of disease and guide the design of the rigorous trials to identify effective therapies for the treatment of C3G.
Collapse
|
176
|
Williams JAE, Greenwood J, Moss SE. Retinal changes precede visual dysfunction in the complement factor H knockout mouse. PLoS One 2013; 8:e68616. [PMID: 23844226 PMCID: PMC3699560 DOI: 10.1371/journal.pone.0068616] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/31/2013] [Indexed: 12/21/2022] Open
Abstract
We previously reported that aged mice lacking complement factor H (CFH) exhibit visual defects and structural changes in the retina. However, it is not known whether this phenotype is age-related or is the consequence of disturbed development. To address this question we investigated the effect of Cfh gene deletion on the retinal phenotype of young and mid-age mice. Cfh(-/-) mouse eyes exhibited thickening of the retina and reduced nuclear density, but relatively normal scotopic and photopic electroretinograms. At 12 months there was evidence of subtle astroglial activation in the Cfh(-/-) eyes, and significant elevation of the complement regulator, decay-accelerating factor (DAF) in Müller cells. In the retinal pigment epithelium (RPE) of young control and Cfh(-/-) animals mitochondria and melanosomes were oriented basally and apically respectively, whereas the apical positioning of melanosomes was significantly perturbed in the mid-age Cfh(-/-) RPE. We conclude that deletion of Cfh in the mouse leads to defects in the retina that precede any marked loss of visual function, but which become progressively more marked as the animals age. These observations are consistent with a lifelong role for CFH in retinal homeostasis.
Collapse
Affiliation(s)
| | - John Greenwood
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Stephen E. Moss
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
- * E-mail:
| |
Collapse
|
177
|
Lesher AM, Nilsson B, Song WC. Properdin in complement activation and tissue injury. Mol Immunol 2013; 56:191-8. [PMID: 23816404 DOI: 10.1016/j.molimm.2013.06.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 05/22/2013] [Indexed: 12/22/2022]
Abstract
The plasma protein properdin is the only known positive regulator of complement activation. Although regarded as an initiator of the alternative pathway of complement activation at the time of its discovery more than a half century ago, the role and mechanism of action of properdin in the complement cascade has undergone significant conceptual evolution since then. Despite the long history of research on properdin, however, new insight and unexpected findings on the role of properdin in complement activation, pathogen infection and host tissue injury are still being revealed by ongoing investigations. In this article, we provide a brief review on recent studies that shed new light on properdin biology, focusing on the following three topics: (1) its role as a pattern recognition molecule to direct and trigger complement activation, (2) its context-dependent requirement in complement activation on foreign and host cell surfaces, and (3) its involvement in alternative pathway complement-mediated immune disorders and considerations of properdin as a potential therapeutic target in human diseases.
Collapse
Affiliation(s)
- Allison M Lesher
- Department of Pharmacology and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
178
|
Hoh Kam J, Lenassi E, Malik TH, Pickering MC, Jeffery G. Complement component C3 plays a critical role in protecting the aging retina in a murine model of age-related macular degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:480-92. [PMID: 23747511 DOI: 10.1016/j.ajpath.2013.04.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 03/04/2013] [Accepted: 04/05/2013] [Indexed: 01/17/2023]
Abstract
Complement component C3 is the central complement component and a key inflammatory protein activated in age-related macular degeneration (AMD). AMD is associated with genetic variation in complement proteins that results in enhanced activation of C3 through the complement alternative pathway. These include complement factor H (CFH), a negative regulator of C3 activation. Both C3 inhibition and/or CFH augmentation are potential therapeutic strategies in AMD. Herein, we examined retinal integrity in aged (12 months) mice deficient in both factors H and C3 (CFH(-/-).C3(-/-)), CFH alone (CFH(-/-)), or C3 alone (C3(-/-)), and wild-type mice (C57BL/6). Retinal function was assessed by electroretinography, and retinal morphological features were analyzed at light and electron microscope levels. Retinas were also stained for amyloid β (Aβ) deposition, inflammation, and macrophage accumulation. Contrary to expectation, electroretinograms of CFH(-/-).C3(-/-) mice displayed more severely reduced responses than those of other mice. All mutant strains showed significant photoreceptor loss and thickening of Bruch's membrane compared with wild-type C57BL/6, but these changes were greater in CFH(-/-).C3(-/-) mice. CFH(-/-).C3(-/-) mice had significantly more Aβ on Bruch's membrane, fewer macrophages, and high levels of retinal inflammation than the other groups. Our data show that both uncontrolled C3 activation (CFH(-/-)) and complete absence of C3 (CFH(-/-).C3(-/-) and C3(-/-)) negatively affect aged retinas. These findings suggest that strategies that inhibit C3 in AMD may be deleterious.
Collapse
Affiliation(s)
- Jaimie Hoh Kam
- Institute of Ophthalmology, University College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
179
|
Thurman JM, Kulik L, Orth H, Wong M, Renner B, Sargsyan SA, Mitchell LM, Hourcade DE, Hannan JP, Kovacs JM, Coughlin B, Woodell AS, Pickering MC, Rohrer B, Holers VM. Detection of complement activation using monoclonal antibodies against C3d. J Clin Invest 2013; 123:2218-30. [PMID: 23619360 DOI: 10.1172/jci65861] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 02/21/2013] [Indexed: 12/21/2022] Open
Abstract
During complement activation the C3 protein is cleaved, and C3 activation fragments are covalently fixed to tissues. Tissue-bound C3 fragments are a durable biomarker of tissue inflammation, and these fragments have been exploited as addressable binding ligands for targeted therapeutics and diagnostic agents. We have generated cross-reactive murine monoclonal antibodies against human and mouse C3d, the final C3 degradation fragment generated during complement activation. We developed 3 monoclonal antibodies (3d8b, 3d9a, and 3d29) that preferentially bind to the iC3b, C3dg, and C3d fragments in solution, but do not bind to intact C3 or C3b. The same 3 clones also bind to tissue-bound C3 activation fragments when injected systemically. Using mouse models of renal and ocular disease, we confirmed that, following systemic injection, the antibodies accumulated at sites of C3 fragment deposition within the glomerulus, the renal tubulointerstitium, and the posterior pole of the eye. To detect antibodies bound within the eye, we used optical imaging and observed accumulation of the antibodies within retinal lesions in a model of choroidal neovascularization (CNV). Our results demonstrate that imaging methods that use these antibodies may provide a sensitive means of detecting and monitoring complement activation-associated tissue inflammation.
Collapse
Affiliation(s)
- Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Kokkinopoulos I. 670 nm LED ameliorates inflammation in the CFH(-/-) mouse neural retina. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2013; 122:24-31. [PMID: 23584451 DOI: 10.1016/j.jphotobiol.2013.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 03/12/2013] [Accepted: 03/17/2013] [Indexed: 12/27/2022]
Abstract
Para-inflammation in the neural retina is thought to contribute to the onset of some age-related retinal diseases. Continuous innate immune system activation, manifests in progressive chronic inflammation, macrophage invasion and cell loss, resulting in visual loss. We have previously shown that mitochondrial function is augmented following 670 nm LED exposure, leading to reduced retinal inflammation. Here, it was asked whether 670 nm LED regulates para-inflammation in an aged-related macular degeneration mouse model. Mutant CFH(-/-) mice were exposed to four 90 s exposures over 2 days for 1 week and 8 weeks. These regimes significantly reduced activated macrophage number, TNF-alpha and MIF protein expression levels. Immuno-reactivity to C3, C3b and calcitonin, all markers of inflammatory status were also altered. Finally, innate immune proteins, TLR 2 and 4, showed a marked decrease in protein expression. These findings support the notion that 670 nm LED regulates innate immunity, alleviating inflammation in the neural retina of an age-related macular degeneration mouse model.
Collapse
Affiliation(s)
- Ioannis Kokkinopoulos
- School of Biomedical and Health Sciences, Wolfson Centre for Age-Related Diseases, King's College London, UK.
| |
Collapse
|
181
|
Complement dysregulation and disease: from genes and proteins to diagnostics and drugs. Immunobiology 2013; 217:1034-46. [PMID: 22964229 DOI: 10.1016/j.imbio.2012.07.021] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 07/20/2012] [Accepted: 07/21/2012] [Indexed: 12/21/2022]
Abstract
During the last decade, numerous studies have associated genetic variations in complement components and regulators with a number of chronic and infectious diseases. The functional characterization of these complement protein variants, in addition to recent structural advances in understanding of the assembly, activation and regulation of the AP C3 convertase, have provided important insights into the pathogenic mechanisms involved in some of these complement related disorders. This knowledge has identified potential targets for complement inhibitory therapies which are demonstrating efficacy and generating considerable expectation in changing the natural history of these diseases. Comprehensive understanding of the genetic and non-genetic risk factors contributing to these disorders will also result in targeting of the right patient groups in a stratified medicine approach through better diagnostics and individually tailored treatments, thereby improving management of patients.
Collapse
|
182
|
Abstract
‘C3 glomerulopathy’ is a recent disease classification comprising several rare types of glomerulonephritis (GN), including dense deposit disease (DDD), C3 glomerulonephritis (C3GN) and CFHR5 nephropathy. These disorders share the key histological feature of isolated complement C3 deposits in the glomerulus. A common aetiology involving dysregulation of the alternative pathway (AP) of complement has been elucidated in the past decade, with genetic defects and/or autoantibodies able to be identified in a proportion of patients. We review the clinical and histological features of C3 glomerulopathy, relating these to underlying molecular mechanisms. The role of uncontrolled C3 activation in pathogenesis is emphasized, with important lessons from animal models. Methods, advantages and limitations of gene testing in the assessment of individuals or families with C3 glomerulopathy are discussed. While no therapy has yet been shown consistently effective, clinical evaluation of agents targeting specific components of the complement system is ongoing. However, limits to current knowledge regarding the natural history and the appropriate timing and duration of proposed therapies need to be addressed.
Collapse
Affiliation(s)
- Thomas D Barbour
- Centre for Complement & Inflammation Research (CCIR), Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK
| | | | | |
Collapse
|
183
|
Thurman JM, Rohrer B. Noninvasive detection of complement activation through radiologic imaging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:271-82. [PMID: 23402034 DOI: 10.1007/978-1-4614-4118-2_19] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
A wealth of experimental and clinical data demonstrates that the complement system is involved in the pathogenesis of numerous inflammatory diseases. Complement activation contributes to injury in disorders that involve nearly every tissue in the body. Concerted effort has been expended in recent years to develop therapeutic complement inhibitors. Eculizumab, an inhibitory antibody to C5, was recently approved for the treatment of several diseases, and many other complement inhibitors are in clinical development. As these drugs are developed, the need for improved methods of detecting and monitoring complement activation within particular tissues will be increasingly important. We have developed a magnetic resonance imaging (MRI)-based method for noninvasive detection of complement activation. This method utilizes iron-oxide nanoparticles that are targeted to sites of complement activation with a recombinant protein that contains the C3d-binding region of complement receptor (CR) 2. Iron-oxide nanoparticles darken (negatively enhance) images obtained by T2-weighted MRI. We have demonstrated that the CR2-targeted nanoparticles bind within the kidneys of mice with lupus-like kidney disease (MRL/1pr mice), causing a decrease in the T2 signal within the kidneys. This method discriminates diseased kidneys from healthy controls, and the magnitude of the negative enhancement in the cortex of MRL/lpr mice correlates with their disease severity. This method may be useful for identifying those patients most likely to benefit from complement inhibitors and for monitoring the response of these patients to treatment. These results may open up new avenues to develop tools for the monitoring of disease progression in complement-dependent diseases.
Collapse
Affiliation(s)
- Joshua M Thurman
- Department of Medicine, University of Colorado Denver School of Medicine, Denver, CO 80045, USA.
| | | |
Collapse
|
184
|
Dimerization of complement factor H-related proteins modulates complement activation in vivo. Proc Natl Acad Sci U S A 2013; 110:4685-90. [PMID: 23487775 DOI: 10.1073/pnas.1219260110] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The complement system is a key component regulation influences susceptibility to age-related macular degeneration, meningitis, and kidney disease. Variation includes genomic rearrangements within the complement factor H-related (CFHR) locus. Elucidating the mechanism underlying these associations has been hindered by the lack of understanding of the biological role of CFHR proteins. Here we present unique structural data demonstrating that three of the CFHR proteins contain a shared dimerization motif and that this hitherto unrecognized structural property enables formation of both homodimers and heterodimers. Dimerization confers avidity for tissue-bound complement fragments and enables these proteins to efficiently compete with the physiological complement inhibitor, complement factor H (CFH), for ligand binding. Our data demonstrate that these CFHR proteins function as competitive antagonists of CFH to modulate complement activation in vivo and explain why variation in the CFHRs predisposes to disease.
Collapse
|
185
|
Banda NK, Mehta G, Ferreira VP, Cortes C, Pickering MC, Pangburn MK, Arend WP, Holers VM. Essential role of surface-bound complement factor H in controlling immune complex-induced arthritis. THE JOURNAL OF IMMUNOLOGY 2013; 190:3560-9. [PMID: 23436934 DOI: 10.4049/jimmunol.1203271] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Factor H (fH) is an endogenous negative regulator of the alternative pathway (AP) that binds polyanions as well as complement activation fragments C3b and C3d. The AP is both necessary and sufficient to develop collagen Ab-induced arthritis (CAIA) in mice; the mechanisms whereby normal control of the AP is overcome and injury develops are unknown. Although primarily a soluble circulating protein, fH can also bind to tissues in a manner dependent on the carboxyl-terminal domain containing short consensus repeats 19 and 20. We examined the role of fH in CAIA by blocking its binding to tissues through administration of a recombinant negative inhibitor containing short consensus repeats 19 and 20 (rfH19-20), which impairs fH function and amplifies surface AP activation in vitro. Administration of rfH19-20, but not control rfH3-5, significantly worsened clinical disease activity, histopathologic injury, and C3 deposition in the synovium and cartilage in wild-type and fH(+/-) mice. In vitro studies demonstrated that rfH19-20 increased complement activation on cartilage extracts and injured fibroblast-like synoviocytes, two major targets of complement deposition in the joint. We conclude that endogenous fH makes a significant contribution to inhibition of the AP in CAIA through binding to sites of immune complex formation and complement activation.
Collapse
Affiliation(s)
- Nirmal K Banda
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | | | | | | | | | | | | | | |
Collapse
|
186
|
Barata L, Miwa T, Sato S, Kim D, Mohammed I, Song WC. Deletion of Crry and DAF on murine platelets stimulates thrombopoiesis and increases factor H-dependent resistance of peripheral platelets to complement attack. THE JOURNAL OF IMMUNOLOGY 2013; 190:2886-95. [PMID: 23390291 DOI: 10.4049/jimmunol.1202536] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Complement receptor 1-related gene/protein y (Crry) and decay-accelerating factor (DAF) are two murine membrane C3 complement regulators with overlapping functions. Crry deletion is embryonically lethal whereas DAF-deficient mice are generally healthy. Crry(-/-)DAF(-/-) mice were viable on a C3(-/-) background, but platelets from such mice were rapidly destroyed when transfused into C3-sufficient mice. In this study, we used the cre-lox system to delete platelet Crry in DAF(-/-) mice and studied Crry/DAF-deficient platelet development in vivo. Rather than displaying thrombocytopenia, Pf4-Cre(+)-Crry(flox/flox) mice had normal platelet counts and their peripheral platelets were resistant to complement attack. However, chimera mice generated with Pf4-Cre(+)-Crry(flox/flox) bone marrows showed platelets from C3(-/-) but not C3(+/+) recipients to be sensitive to complement activation, suggesting that circulating platelets in Pf4-Cre(+)-Crry(flox/flox) mice were naturally selected in a complement-sufficient environment. Notably, Pf4-Cre(+)-Crry(flox/flox) mouse platelets became complement susceptible when factor H function was blocked. Examination of Pf4-Cre(+)-Crry(flox/flox) mouse bone marrows revealed exceedingly active thrombopoiesis. Thus, under in vivo conditions, Crry/DAF deficiency on platelets led to abnormal platelet turnover, but peripheral platelet count was compensated for by increased thrombopoiesis. Selective survival of Crry/DAF-deficient platelets aided by factor H protection and compensatory thrombopoiesis demonstrates the cooperation between membrane and fluid phase complement inhibitors and the body's ability to adaptively respond to complement regulator deficiencies.
Collapse
Affiliation(s)
- Lidia Barata
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19401, USA
| | | | | | | | | | | |
Collapse
|
187
|
Complement evasion by Borrelia burgdorferi: it takes three to tango. Trends Parasitol 2013; 29:119-28. [PMID: 23298533 DOI: 10.1016/j.pt.2012.12.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 11/30/2012] [Accepted: 12/04/2012] [Indexed: 01/03/2023]
Abstract
The complement system is one of the major innate defense mechanisms Borrelia burgdorferi sensu lato has to overcome to establish an infection of mammalian hosts and to cause Lyme borreliosis in humans. Borrelia prevents complement-mediated killing during host colonization through (i) recruitment of host complement regulators by Borrelia, (ii) evasion mechanisms by Borrelia itself, and (iii) exploitation of tick proteins by Borrelia. These interactions with complement can be host species-specific. This review provides an overview of interactions between Borrelia, tick, and host leading to evasion of complement-mediated killing.
Collapse
|
188
|
|
189
|
Lesher AM, Zhou L, Kimura Y, Sato S, Gullipalli D, Herbert AP, Barlow PN, Eberhardt HU, Skerka C, Zipfel PF, Hamano T, Miwa T, Tung KS, Song WC. Combination of factor H mutation and properdin deficiency causes severe C3 glomerulonephritis. J Am Soc Nephrol 2012. [PMID: 23204401 DOI: 10.1681/asn.2012060570] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Factor H (fH) and properdin both modulate complement; however, fH inhibits activation, and properdin promotes activation of the alternative pathway of complement. Mutations in fH associate with several human kidney diseases, but whether inhibiting properdin would be beneficial in these diseases is unknown. Here, we found that either genetic or pharmacological blockade of properdin, which we expected to be therapeutic, converted the mild C3 GN of an fH-mutant mouse to a lethal C3 GN with features of human dense deposit disease. We attributed this phenotypic change to a differential effect of properdin on the dynamics of alternative pathway complement activation in the fluid phase and the cell surface in the fH-mutant mice. Thus, in fH mutation-related C3 glomerulopathy, additional factors that impact the activation of the alternative pathway of complement critically determine the nature and severity of kidney pathology. These results show that therapeutic manipulation of the complement system requires rigorous disease-specific target validation.
Collapse
Affiliation(s)
- Allison M Lesher
- Department of Pharmacology and Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Ruseva MM, Vernon KA, Lesher AM, Schwaeble WJ, Ali YM, Botto M, Cook T, Song W, Stover CM, Pickering MC. Loss of properdin exacerbates C3 glomerulopathy resulting from factor H deficiency. J Am Soc Nephrol 2012. [PMID: 23184055 DOI: 10.1681/asn.2012060571] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Complement factor H (CFH) is a negative regulator of the alternative pathway of complement, and properdin is the sole positive regulator. CFH-deficient mice (CFH(-/-)) develop uncontrolled C3 activation and spontaneous renal disease characterized by accumulation of C3 along the glomerular basement membrane, but the role of properdin in the pathophysiology is unknown. Here, we studied mice deficient in both CFH and properdin (CFH(-/-).P(-/-)). Although CFH(-/-) mice had plasma depleted of both C3 and C5, CFH(-/-).P(-/-) animals exhibited depletion of C3 predominantly, recapitulating the plasma complement profile observed in humans with properdin-independent C3 nephritic factors. Glomerular inflammation, thickening of the capillary wall, and glomerular C3 staining were significantly increased in CFH(-/-).P(-/-) compared with CFH(-/-) mice. We previously reported that exogenous CFH ameliorates C3 staining of the glomerular basement membrane and triggers the appearance of mesangial C3 deposits in CFH(-/-) mice; here, we show that these effects require properdin. In summary, during uncontrolled activation of C3 driven by complete CFH deficiency, properdin influences the intraglomerular localization of C3, suggesting that therapeutic inhibition of properdin would be detrimental in this setting.
Collapse
Affiliation(s)
- Marieta M Ruseva
- Centre for Complement and Inflammation Research, Imperial College, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Abstract
In the past decade, a large body of evidence has accumulated in support of the critical role of dysregulation of the alternative complement pathway in atypical haemolytic uraemic syndrome (aHUS) and C3 glomerulopathies. These findings have paved the way for innovative therapeutic strategies based on complement blockade, and eculizumab, a monoclonal antibody targeting the human complement component 5, is now widely used to treat aHUS. In this article, we review 28 case reports and preliminary data from 37 patients enrolled in prospective trials of eculizumab treatment for episodes of aHUS involving either native or transplanted kidneys. Eculizumab may be considered as an optimal first-line therapy when the diagnosis of aHUS is unequivocal and this treatment has the potential to rescue renal function when administered early after onset of the disease. However, a number of important issues require further study, including the appropriate duration of treatment according to an individual's genetic background and medical history, the optimal strategy to prevent post-transplantation recurrence of aHUS and a cost-efficacy analysis. Data regarding the efficacy of eculizumab in the control of C3 glomerulopathies are more limited and less clear, but several observations suggest that eculizumab may act on the most inflammatory forms of this disorder.
Collapse
|
192
|
Faber C, Williams J, Juel HB, Greenwood J, Nissen MH, Moss SE. Complement factor H deficiency results in decreased neuroretinal expression of Cd59a in aged mice. Invest Ophthalmol Vis Sci 2012; 53:6324-30. [PMID: 22918646 DOI: 10.1167/iovs.12-10385] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
PURPOSE The complement system is closely linked to the pathogenesis of AMD. Several complement genes are expressed in RPE, and complement proteins accumulate in drusen. Further, a common variant of complement factor H (CFH) confers increased risk of developing AMD. Because the mechanisms by which changes in the function of CFH influence development of AMD are unclear, we examined ocular complement expression as a consequence of age in control and CFH null mutant mice. METHODS Gene expression in neuroretinas and RPE/choroid from young and aged WT and Cfh(-/-) C57BL/6J mice was analyzed by microarrays. Expression of a wide range of complement genes was compared with expression in liver. RESULTS An age-associated increased expression of complement, particularly C1q, C3, and factor B, in the RPE/choroid coincided with increased expression of the negative regulators Cfh and Cd59a in the neuroretina. Young mice deficient in CFH expressed Cd59a similar to WT, but failed to upregulate Cd59a expression with age. Hepatic expression of Cd59a increased with age regardless of Cfh genotype. CONCLUSIONS While the connection between CFH deficiency and failure to upregulate CD59a remains unknown, these results suggest that expression of CD59 is tissue-specific and that neuroretinal regulation depends on CFH. This could contribute to the visual functional deficits and morphological changes in the Cfh(-/-) mouse retina that occur with age.
Collapse
Affiliation(s)
- Carsten Faber
- University of Copenhagen, Faculty of Health Sciences, ISIM, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
193
|
The C5a receptor has a key role in immune complex glomerulonephritis in complement factor H-deficient mice. Kidney Int 2012; 82:961-8. [PMID: 22832515 PMCID: PMC3472160 DOI: 10.1038/ki.2012.249] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Chronic serum sickness leads to the formation of glomerular immune complexes; however, C57BL/6 mice do not develop glomerulonephritis unless complement factor H (CFH) is absent from the plasma. Here we studied the role for C5a receptor (R) in this setting. The exaggerated humoral immune response in CFH−/− mice was normalized in CFH−/−C5aR−/− double knockout mice, highlighting the C5aR dependence. The CFH knockout mice developed proliferative glomerulonephritis with endocapillary F4/80+ macrophage infiltration, a process reduced in the double knockout mice. There was no interstitial inflammation by histologic criteria or flow cytometry for F4/80+Ly6ChiCCR2hi inflammatory macrophages. There were, however, more interstitial CD3+CD4+ T lymphocytes in CFH knockout mice with chronic serum sickness, while double knockout mice had greater than 5-fold more Ly6CloCCR2lo anti-inflammatory macrophages compared to the CFH knockout mice. Mice lacking C5aR were significantly protected from functional renal disease as assessed by blood urea nitrogen levels. Thus, IgG- and iC3b-containing immune complexes are not inflammatory in C57BL/6 mice. Yet when these mice lack CFH, sufficient C3b persists in glomeruli to generate C5a and activate C5aR.
Collapse
|
194
|
Therapeutic regulation of complement in patients with renal disease - where is the promise? Clin Nephrol 2012; 77:413-23. [PMID: 22551888 PMCID: PMC4407337 DOI: 10.5414/cn107220] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Numerous renal diseases are characterized by complement activation within the kidney, and several lines of evidence implicate complement activation as an important part of the pathogenesis of these diseases. Investigators have long anticipated that complement inhibitors would be important and effective therapies for renal diseases. Eculizumab is a monoclonal antibody to the complement protein C5 that has now been administered to patients with several types of renal disease. The apparent efficacy of this agent may herald a new era in the treatment of renal disease, but many questions about the optimal use of therapeutic complement inhibitors remain. Herein we review the rationale for using complement inhibitors in patients with renal disease and discuss several drugs and approaches that are currently under development.
Collapse
|
195
|
Pennesi ME, Neuringer M, Courtney RJ. Animal models of age related macular degeneration. Mol Aspects Med 2012; 33:487-509. [PMID: 22705444 DOI: 10.1016/j.mam.2012.06.003] [Citation(s) in RCA: 275] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Age related macular degeneration (AMD) is the leading cause of vision loss of those over the age of 65 in the industrialized world. The prevalence and need to develop effective treatments for AMD has lead to the development of multiple animal models. AMD is a complex and heterogeneous disease that involves the interaction of both genetic and environmental factors with the unique anatomy of the human macula. Models in mice, rats, rabbits, pigs and non-human primates have recreated many of the histological features of AMD and provided much insight into the underlying pathological mechanisms of this disease. In spite of the large number of models developed, no one model yet recapitulates all of the features of human AMD. However, these models have helped reveal the roles of chronic oxidative damage, inflammation and immune dysregulation, and lipid metabolism in the development of AMD. Models for induced choroidal neovascularization have served as the backbone for testing new therapies. This article will review the diversity of animal models that exist for AMD as well as their strengths and limitations.
Collapse
Affiliation(s)
- Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | |
Collapse
|
196
|
Abstract
The complement system has vital protective functions as a humoral component of the innate immune system and also through interactions with the adaptive immune system; however, when inappropriately activated or regulated, complement can cause inflammation and organ damage, and such processes are involved in the pathogenesis of many inflammatory conditions, not least rheumatic diseases. Furthermore, states of complement deficiency can predispose not only to infections, but also to autoimmune disorders, including rheumatic diseases such as systemic lupus erythematosus. In this Review, the mechanisms behind the pathogenic activities of complement in rheumatic diseases are discussed. Potential approaches to therapeutic intervention that focus on regulating complement activities in these disorders are also considered.
Collapse
|
197
|
Soltys J, Wu X. Complement regulatory protein Crry deficiency contributes to the antigen specific recall response in experimental autoimmune myasthenia gravis. JOURNAL OF INFLAMMATION-LONDON 2012; 9:20. [PMID: 22642809 PMCID: PMC3524051 DOI: 10.1186/1476-9255-9-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 04/18/2012] [Indexed: 12/20/2022]
Abstract
UNLABELLED BACKGROUND Myasthenia gravis (MG) and animal model of experimental autoimmune myasthenia gravis (EAMG) is the most common autoimmune disorder of neuromuscular transmission. The disease is caused by the breakdown of the acetylcholine receptor (AChR) which is largely due to complement activation at the neuromuscular junction (NMJ). Limited knowledge exists to the extent that complement receptor 1-related gene/protein y deficiency (Crry -/-) modulates the adaptive immune response and EAMG outcome. METHODS Mouse EAMG was induced by s.c. administrations of purified acetylcholine receptor (AChR) to Crry -/- and age- matched WT (C57BL/6) mice. Disease severity was assessed by clinical score assessment and muscle grip strength measurements. Serum complement activity was determined by hemolytic assay. ELISA was used to detect the level of AChR specific antibodies. Splenic cells were analyzed for T and B cells subsets distribution, release of cytokines and AChR specific recall responses. Deposition of complement components at the NMJ was assessed by immunofluorescence staining. RESULTS In comparison to WT EAMG, Crry -/- EAMG mice showed signs of augmented muscle weakness but differences, except for one time point, were not statistically significant. Serum complement activity was reduced in Crry -/- EAMG mice and no substantial changes in deposition of C3, C3b/iC3b and C5b-9 (MAC) at the NMJ between WT EAMG and Crry -/- EAMG mice were detected. Lack of Crry affected adaptive immune response. Crry -/- EAMG mice showed increases in the number of AChR specific splenic T-cells secreting IFN-γ and IL-4. Production of complement fixing antibodies (IgG2b, IgG2c) was also augmented. More Th1, Th2 and Th17 cytokines were released into the bloodstream of Crry -/- EAMG mice. CONCLUSIONS Data suggest that Crry deficiency modulates the adaptive immune response in EAMG, but its effect on disease outcome is limited. This was due to the generally lower serum complement level caused by increased C3 turnover. Modulation of complement activity with soluble or membrane bound regulators of complement activity represents a potentially effective approach to modify autoimmune processes in MG and EAMG.
Collapse
Affiliation(s)
- Jindrich Soltys
- Department of Neurology & Psychiatry, 1438 South Grand Boulevard, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA.
| | | |
Collapse
|
198
|
Thurman JM, Tchepeleva SN, Haas M, Panzer S, Boackle SA, Glogowska MJ, Quigg RJ, Holers VM. Complement alternative pathway activation in the autologous phase of nephrotoxic serum nephritis. Am J Physiol Renal Physiol 2012; 302:F1529-36. [PMID: 22492944 DOI: 10.1152/ajprenal.00422.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The complement cascade is an important part of the innate immune system, but pathological activation of this system causes tissue injury in several autoimmune and inflammatory diseases, including immune complex glomerulonephritis. We examined whether mice with targeted deletion of the gene for factor B (fB(-/-) mice) and selective deficiency in the alternative pathway of complement are protected from injury in the nephrotoxic serum (NTS) nephritis model of antibody-mediated glomerulonephritis. When the acute affects of the anti-glomerular basement membrane antibody were assessed, fB(-/-) mice developed a degree of injury similar to wild-type controls. If the mice were presensitized with sheep IgG or if the mice were followed for 5 mo postinjection, however, the fB(-/-) mice developed milder injury than wild-type mice. The immune response of fB(-/-) mice exposed to sheep IgG was similar to that of wild-type mice, but the fB(-/-) mice had less glomerular C3 deposition and lower levels of albuminuria. These results demonstrate that fB(-/-) mice are not significantly protected from acute heterologous injury in NTS nephritis but are protected from autologous injury in response to a planted glomerular antigen. Thus, although the glomerulus is resistant to antibody-initiated, alternative pathway-mediated injury, inhibition of this complement pathway may be beneficial in chronic immune complex-mediated diseases.
Collapse
Affiliation(s)
- Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA.
| | | | | | | | | | | | | | | |
Collapse
|
199
|
Servais A, Noël LH, Roumenina LT, Le Quintrec M, Ngo S, Dragon-Durey MA, Macher MA, Zuber J, Karras A, Provot F, Moulin B, Grünfeld JP, Niaudet P, Lesavre P, Frémeaux-Bacchi V. Acquired and genetic complement abnormalities play a critical role in dense deposit disease and other C3 glomerulopathies. Kidney Int 2012; 82:454-64. [PMID: 22456601 DOI: 10.1038/ki.2012.63] [Citation(s) in RCA: 375] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dense deposit disease and glomerulonephritis with isolated C3 deposits are glomerulopathies characterized by deposits of C3 within or along the glomerular basement membrane. Previous studies found a link between dysregulation of the complement alternative pathway and the pathogenesis of these diseases. We analyzed the role of acquired and genetic complement abnormalities in a cohort of 134 patients, of whom 29 have dense deposit disease, 56 have glomerulonephritis with isolated C3 deposits, and 49 have primary membranoproliferative glomerulonephritis type I, with adult and pediatric onset. A total of 53 patients presented with a low C3 level, and 65 were positive for C3 nephritic factor that was significantly more frequently detected in patients with dense deposit disease than in other histological types. Mutations in CFH and CFI genes were identified in 24 patients associated with a C3 nephritic factor in half the cases. We found evidence for complement alternative pathway dysregulation in 26 patients with membranoproliferative glomerulonephritis type I. The complement factor H Y402H variant was significantly increased in dense deposit disease. We identified one at-risk membrane cofactor protein (MCP) haplotype for glomerulonephritis with isolated C3 deposits and membranoproliferative glomerulonephritis type I. Thus, our results suggest a critical role of fluid-phase alternative pathway dysregulation in the pathogenesis of C3 glomerulopathies as well as in immune complex-mediated glomerular diseases. The localization of the C3 deposits may be under the influence of MCP expression.
Collapse
Affiliation(s)
- Aude Servais
- Department of Nephrology, Hôpital Necker-Enfants Malades AP-HP, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Gale DP, Pickering MC. Regulating complement in the kidney: insights from CFHR5 nephropathy. Dis Model Mech 2012; 4:721-6. [PMID: 22065842 PMCID: PMC3209641 DOI: 10.1242/dmm.008052] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Complement factor H related protein 5 (CFHR5) nephropathy is a monogenic disorder of complement regulation that is endemic in Cyprus. The disease is characterised by haematuria, C3 glomerulonephritis and kidney failure. Its identification suggests a role for the CFHR5 protein in the regulation of complement in the kidney. In this review, we discuss how studying CFHR5 nephropathy can contribute to our understanding of the role of complement in kidney diseases such as dense deposit disease, C3 glomerulonephritis and atypical haemolytic uraemic syndrome.
Collapse
Affiliation(s)
- Daniel P Gale
- UCL Centre for Nephrology, Royal Free Hospital, London, NW3 2PF, UK.
| | | |
Collapse
|