32 results on '"Pickering, Matthew C"'
Search Results
2. Cell-autonomous regulation of complement C3 by factor H limits macrophage efferocytosis and exacerbates atherosclerosis.
- Author
-
Kiss MG, Papac-Miličević N, Porsch F, Tsiantoulas D, Hendrikx T, Takaoka M, Dinh HQ, Narzt MS, Göderle L, Ozsvár-Kozma M, Schuster M, Fortelny N, Hladik A, Knapp S, Gruber F, Pickering MC, Bock C, Swirski FK, Ley K, Zernecke A, Cochain C, Kemper C, Mallat Z, and Binder CJ
- Subjects
- Animals, Humans, Mice, Complement Factor H genetics, Complement Factor H metabolism, Inflammation, Macrophages metabolism, Atherosclerosis metabolism, Complement C3 genetics, Complement C3 metabolism
- Abstract
Complement factor H (CFH) negatively regulates consumption of complement component 3 (C3), thereby restricting complement activation. Genetic variants in CFH predispose to chronic inflammatory disease. Here, we examined the impact of CFH on atherosclerosis development. In a mouse model of atherosclerosis, CFH deficiency limited plaque necrosis in a C3-dependent manner. Deletion of CFH in monocyte-derived inflammatory macrophages propagated uncontrolled cell-autonomous C3 consumption without downstream C5 activation and heightened efferocytotic capacity. Among leukocytes, Cfh expression was restricted to monocytes and macrophages, increased during inflammation, and coincided with the accumulation of intracellular C3. Macrophage-derived CFH was sufficient to dampen resolution of inflammation, and hematopoietic deletion of CFH in atherosclerosis-prone mice promoted lesional efferocytosis and reduced plaque size. Furthermore, we identified monocyte-derived inflammatory macrophages expressing C3 and CFH in human atherosclerotic plaques. Our findings reveal a regulatory axis wherein CFH controls intracellular C3 levels of macrophages in a cell-autonomous manner, evidencing the importance of on-site complement regulation in the pathogenesis of inflammatory diseases., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
3. Homodimeric Minimal Factor H: In Vivo Tracking and Extended Dosing Studies in Factor H Deficient Mice.
- Author
-
Kamala O, Malik TH, Hallam TM, Cox TE, Yang Y, Vyas F, Luli S, Connelly C, Gibson B, Smith-Jackson K, Denton H, Pappworth IY, Huang L, Kavanagh D, Pickering MC, and Marchbank KJ
- Subjects
- Animals, Complement Factor H deficiency, Kidney immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Complement C3 immunology, Complement Factor H immunology
- Abstract
C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway (AP) of complement and treatment options remain inadequate. Factor H (FH) is a potent regulator of the AP. An in-depth analysis of FH-related protein dimerised minimal (mini)-FH constructs has recently been published. This analysis showed that addition of a dimerisation module to mini-FH not only increased serum half-life but also improved complement regulatory function, thus providing a potential treatment option for C3G. Herein, we describe the production of a murine version of homodimeric mini-FH [mHDM-FH (mFH
1-5^18-20^R1-2 )], developed to reduce the risk of anti-drug antibody formation during long-term experiments in murine models of C3G and other complement-driven pathologies. Our analysis of mHDM-FH indicates that it binds with higher affinity and avidity to WT mC3b when compared to mouse (m)FH (mHDM-FH KD =505 nM; mFH KD =1370 nM) analogous to what we observed with the respective human proteins. The improved binding avidity resulted in enhanced complement regulatory function in haemolytic assays. Extended interval dosing studies in CFH-/- mice (5mg/kg every 72hrs) were partially effective and bio-distribution analysis in CFH-/- mice, through in vivo imaging technologies, demonstrates that mHDM-FH is preferentially deposited and remains fixed in the kidneys (and liver) for up to 4 days. Extended dosing using an AAV- human HDM-FH (hHDM-FH) construct achieved complete normalisation of C3 levels in CFH-/- mice for 3 months and was associated with a significant reduction in glomerular C3 staining. Our data demonstrate the ability of gene therapy delivery of mini-FH constructs to enhance complement regulation in vivo and support the application of this approach as a novel treatment strategy in diseases such as C3G., Competing Interests: KM has received research funding from Gemini Therapeutics, Idorsia Pharmaceuticals Ltd and Catalyst Biosciences as well as consultancy income from Freeline Therapeutics, Bath ASU & MPM Capital. MP provides consultancy for Alexion, Apellis, Gemini and Gyroscope Pharma. DK has received consultancy income from Gyroscope Therapeutics, Alexion Pharmaceuticals, Novartis, Apellis; Sarepta. TH is an employee of Gyroscope Therapeutics. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Kamala, Malik, Hallam, Cox, Yang, Vyas, Luli, Connelly, Gibson, Smith-Jackson, Denton, Pappworth, Huang, Kavanagh, Pickering and Marchbank.)- Published
- 2021
- Full Text
- View/download PDF
4. C3 Glomerulopathy and Related Disorders in Children: Etiology-Phenotype Correlation and Outcomes.
- Author
-
Wong EKS, Marchbank KJ, Lomax-Browne H, Pappworth IY, Denton H, Cooke K, Ward S, McLoughlin AC, Richardson G, Wilson V, Harris CL, Morgan BP, Hakobyan S, McAlinden P, Gale DP, Maxwell H, Christian M, Malcomson R, Goodship THJ, Marks SD, Pickering MC, Kavanagh D, Cook HT, and Johnson SA
- Subjects
- Adolescent, Child, Child, Preschool, Complement C3 genetics, Complement C3b immunology, Complement C4 metabolism, Complement Factor B immunology, Complement Factor H immunology, Disease Progression, Female, Follow-Up Studies, Glomerular Filtration Rate, Glomerulonephritis, Membranoproliferative pathology, Glomerulonephritis, Membranoproliferative therapy, Graft Survival, Humans, Kaplan-Meier Estimate, Kidney Failure, Chronic etiology, Kidney Failure, Chronic surgery, Kidney Transplantation, Male, Prognosis, Proportional Hazards Models, Prospective Studies, Recurrence, Registries, Risk Factors, Autoantibodies blood, Complement C3 metabolism, Glomerulonephritis, Membranoproliferative blood, Glomerulonephritis, Membranoproliferative etiology, Phenotype
- Abstract
Background and Objectives: Membranoproliferative GN and C3 glomerulopathy are rare and overlapping disorders associated with dysregulation of the alternative complement pathway. Specific etiologic data for pediatric membranoproliferative GN/C3 glomerulopathy are lacking, and outcome data are based on retrospective studies without etiologic data., Design, Setting, Participants, & Measurements: A total of 80 prevalent pediatric patients with membranoproliferative GN/C3 glomerulopathy underwent detailed phenotyping and long-term follow-up within the National Registry of Rare Kidney Diseases (RaDaR). Risk factors for kidney survival were determined using a Cox proportional hazards model. Kidney and transplant graft survival was determined using the Kaplan-Meier method., Results: Central histology review determined 39 patients with C3 glomerulopathy, 31 with immune-complex membranoproliferative GN, and ten with immune-complex GN. Patients were aged 2-15 (median, 9; interquartile range, 7-11) years. Median complement C3 and C4 levels were 0.31 g/L and 0.14 g/L, respectively; acquired (anticomplement autoantibodies) or genetic alternative pathway abnormalities were detected in 46% and 9% of patients, respectively, across all groups, including those with immune-complex GN. Median follow-up was 5.18 (interquartile range, 2.13-8.08) years. Eleven patients (14%) progressed to kidney failure, with nine transplants performed in eight patients, two of which failed due to recurrent disease. Presence of >50% crescents on the initial biopsy specimen was the sole variable associated with kidney failure in multivariable analysis (hazard ratio, 6.2; 95% confidence interval, 1.05 to 36.6; P< 0.05). Three distinct C3 glomerulopathy prognostic groups were identified according to presenting eGFR and >50% crescents on the initial biopsy specimen., Conclusions: Crescentic disease was a key risk factor associated with kidney failure in a national cohort of pediatric patients with membranoproliferative GN/C3 glomerulopathy and immune-complex GN. Presenting eGFR and crescentic disease help define prognostic groups in pediatric C3 glomerulopathy. Acquired abnormalities of the alternative pathway were commonly identified but not a risk factor for kidney failure., (Copyright © 2021 by the American Society of Nephrology.)
- Published
- 2021
- Full Text
- View/download PDF
5. Murine Factor H Co-Produced in Yeast With Protein Disulfide Isomerase Ameliorated C3 Dysregulation in Factor H-Deficient Mice.
- Author
-
Kerr H, Herbert AP, Makou E, Abramczyk D, Malik TH, Lomax-Browne H, Yang Y, Pappworth IY, Denton H, Richards A, Marchbank KJ, Pickering MC, and Barlow PN
- Subjects
- Animals, Gene Expression, Immunomodulation, Mice, Mice, Knockout, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, Yeasts genetics, Yeasts metabolism, Complement C3 immunology, Complement C3 metabolism, Complement Factor H biosynthesis, Complement Factor H deficiency, Protein Disulfide-Isomerases metabolism, Recombinant Proteins metabolism
- Abstract
Recombinant human factor H (hFH) has potential for treating diseases linked to aberrant complement regulation including C3 glomerulopathy (C3G) and dry age-related macular degeneration. Murine FH (mFH), produced in the same host, is useful for pre-clinical investigations in mouse models of disease. An abundance of FH in plasma suggests high doses, and hence microbial production, will be needed. Previously, Pichia pastoris produced useful but modest quantities of hFH. Herein, a similar strategy yielded miniscule quantities of mFH. Since FH has 40 disulfide bonds, we created a P. pastoris strain containing a methanol-inducible codon-modified gene for protein-disulfide isomerase (PDI) and transformed this with codon-modified DNA encoding mFH under the same promoter. What had been barely detectable yields of mFH became multiple 10s of mg/L. Our PDI-overexpressing strain also boosted hFH overproduction, by about tenfold. These enhancements exceeded PDI-related production gains reported for other proteins, all of which contain fewer disulfide-stabilized domains. We optimized fermentation conditions, purified recombinant mFH, enzymatically trimmed down its (non-human) N-glycans, characterised its functions in vitro and administered it to mice. In FH-knockout mice, our de-glycosylated recombinant mFH had a shorter half-life and induced more anti-mFH antibodies than mouse serum-derived, natively glycosylated, mFH. Even sequential daily injections of recombinant mFH failed to restore wild-type levels of FH and C3 in mouse plasma beyond 24 hours after the first injection. Nevertheless, mFH functionality appeared to persist in the glomerular basement membrane because C3-fragment deposition here, a hallmark of C3G, remained significantly reduced throughout and beyond the ten-day dosing regimen., Competing Interests: KM has received consultancy or research income from Gemini Therapeutics Inc, Freeline Therapeutics, MPM Capital, Idorsia Pharmaceuticals Ltd and Catalyst Biosciences. PB is a scientific co-founder of, and has received consultancy and research income from, Gemini Therapeutics Inc. AH is founder and Chief Scientific Officer of Invizius. EM is currently an employee of Invizius. AR has been employed at GlaxoSmithKline since October 2014. Results reported in this work were undertaken during her Wellcome Trust Intermediate Clinical Fellowship. Her spouse, David Kavanagh, is head of the National Renal Complement Therapeutics Centre, UK, and a board member and scientific advisor to Gyroscope Therapeutics Ltd. PNB and AR are co-authors on a recombinant CFH patent application (EP10803266A). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Kerr, Herbert, Makou, Abramczyk, Malik, Lomax-Browne, Yang, Pappworth, Denton, Richards, Marchbank, Pickering and Barlow.)
- Published
- 2021
- Full Text
- View/download PDF
6. Gain-of-function factor H-related 5 protein impairs glomerular complement regulation resulting in kidney damage.
- Author
-
Malik TH, Gitterman DP, Lavin DP, Lomax-Browne HJ, Hiemeyer EC, Moran LB, Boroviak K, Cook HT, Gilmore AC, Mandwie M, Ahmad A, Alexander IE, Logan GJ, Marchbank KJ, Bradley A, and Pickering MC
- Subjects
- Animals, Disease Models, Animal, Female, Humans, Kidney Glomerulus metabolism, Male, Mice, Mice, Transgenic, Sex Factors, Complement C3 metabolism, Complement System Proteins genetics, Gain of Function Mutation, Glomerulonephritis genetics, Glomerulonephritis metabolism, Kidney Glomerulus pathology
- Abstract
Genetic variation within the factor H-related (FHR) genes is associated with the complement-mediated kidney disease, C3 glomerulopathy (C3G). There is no definitive treatment for C3G, and a significant proportion of patients develop end-stage renal disease. The prototypical example is CFHR5 nephropathy, through which an internal duplication within a single CFHR5 gene generates a mutant FHR5 protein (FHR5mut) that leads to accumulation of complement C3 within glomeruli. To elucidate how abnormal FHR proteins cause C3G, we modeled CFHR5 nephropathy in mice. Animals lacking the murine factor H (FH) and FHR proteins, but coexpressing human FH and FHR5mut (hFH-FHR5mut), developed glomerular C3 deposition, whereas mice coexpressing human FH with the normal FHR5 protein (hFH-FHR5) did not. Like in patients, the FHR5mut had a dominant gain-of-function effect, and when administered in hFH-FHR5 mice, it triggered C3 deposition. Importantly, adeno-associated virus vector-delivered homodimeric mini-FH, a molecule with superior surface C3 binding compared to FH, reduced glomerular C3 deposition in the presence of the FHR5mut. Our data demonstrate that FHR5mut causes C3G by disrupting the homeostatic regulation of complement within the kidney and is directly pathogenic in C3G. These results support the use of FH-derived molecules with enhanced C3 binding for treating C3G associated with abnormal FHR proteins. They also suggest that targeting FHR5 represents a way to treat complement-mediated kidney injury., Competing Interests: Competing interest statement: M.C.P. has received consultancy fees for providing scientific advice for work for Alexion, ChemoCentryx, Achillion, Apellis, Gemini, Gyroscope, Ra, Silence Therapeutics, and Sobi Pharma. M.C.P. is a scientific advisory board member for Gemini and Gyroscope Pharma., (Copyright © 2021 the Author(s). Published by PNAS.)
- Published
- 2021
- Full Text
- View/download PDF
7. Complement activity is regulated in C3 glomerulopathy by IgG-factor H fusion proteins with and without properdin targeting domains.
- Author
-
Gilmore AC, Zhang Y, Cook HT, Lavin DP, Katti S, Wang Y, Johnson KK, Kim S, and Pickering MC
- Subjects
- Animals, Complement C3-C5 Convertases, Complement C5, Complement Factor H genetics, Complement Pathway, Alternative, Immunoglobulin G, Mice, Complement C3 genetics, Properdin genetics
- Abstract
C3 glomerulopathy is characterized by accumulation of complement C3 within glomeruli. Causes include, but are not limited to, abnormalities in factor H, the major negative regulator of the complement alternative pathway. Factor H-deficient (Cfh
-/- ) mice develop C3 glomerulopathy together with a reduction in plasma C3 levels. Using this model, we assessed the efficacy of two fusion proteins containing the factor H alternative pathway regulatory domains (FH1-5 ) linked to either a non-targeting mouse immunoglobulin (IgG-FH1-5 ) or to an anti-mouse properdin antibody (Anti-P-FH1-5 ). Both proteins increased plasma C3 and reduced glomerular C3 deposition to an equivalent extent, suggesting that properdin-targeting was not required for FH1-5 to alter C3 activation in either plasma or glomeruli. Following IgG-FH1-5 administration, plasma C3 levels temporally correlated with changes in factor B levels whereas plasma C5 levels correlated with changes in plasma properdin levels. Notably, the increases in plasma C5 and properdin levels persisted for longer than the increases in C3 and factor B. In Cfh-/- mice IgG-FH1-5 reduced kidney injury during accelerated serum nephrotoxic nephritis. Thus, our data demonstrate that IgG-FH1-5 restored circulating alternative pathway activity and reduced glomerular C3 deposition in Cfh-/- mice and that plasma properdin levels are a sensitive marker of C5 convertase activity in factor H deficiency. The immunoglobulin conjugated FH1-5 protein, through its comparatively long plasma half-life, may be a potential therapy for C3 glomerulopathy., (Copyright © 2020 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
8. Hyperfunctional complement C3 promotes C5-dependent atypical hemolytic uremic syndrome in mice.
- Author
-
Smith-Jackson K, Yang Y, Denton H, Pappworth IY, Cooke K, Barlow PN, Atkinson JP, Liszewski MK, Pickering MC, Kavanagh D, Cook HT, and Marchbank KJ
- Subjects
- Amino Acid Substitution, Animals, Complement C9 genetics, Complement C9 immunology, Disease Models, Animal, Glomerulonephritis, Membranous genetics, Glomerulonephritis, Membranous immunology, Glomerulonephritis, Membranous pathology, Mice, Mice, Transgenic, Atypical Hemolytic Uremic Syndrome genetics, Atypical Hemolytic Uremic Syndrome immunology, Atypical Hemolytic Uremic Syndrome pathology, Complement Activation, Complement C3 genetics, Complement C3 immunology, Complement C5 genetics, Complement C5 immunology, Kidney immunology, Kidney pathology, Mutation, Missense
- Abstract
Atypical hemolytic uremic syndrome (aHUS) is frequently associated in humans with loss-of-function mutations in complement-regulating proteins or gain-of-function mutations in complement-activating proteins. Thus, aHUS provides an archetypal complement-mediated disease with which to model new therapeutic strategies and treatments. Herein, we show that, when transferred to mice, an aHUS-associated gain-of-function change (D1115N) to the complement-activation protein C3 results in aHUS. Homozygous C3 p.D1115N (C3KI) mice developed spontaneous chronic thrombotic microangiopathy together with hematuria, thrombocytopenia, elevated creatinine, and evidence of hemolysis. Mice with active disease had reduced plasma C3 with C3 fragment and C9 deposition within the kidney. Therapeutic blockade or genetic deletion of C5, a protein downstream of C3 in the complement cascade, protected homozygous C3KI mice from thrombotic microangiopathy and aHUS. Thus, our data provide in vivo modeling evidence that gain-of-function changes in complement C3 drive aHUS. They also show that long-term C5 deficiency is not accompanied by development of other renal complications (such as C3 glomerulopathy) despite sustained dysregulation of C3. Our results suggest that this preclinical model will allow testing of novel complement inhibitors with the aim of developing precisely targeted therapeutics that could have application in many complement-mediated diseases.
- Published
- 2019
- Full Text
- View/download PDF
9. C3 glomerulopathy - understanding a rare complement-driven renal disease.
- Author
-
Smith RJH, Appel GB, Blom AM, Cook HT, D'Agati VD, Fakhouri F, Fremeaux-Bacchi V, Józsi M, Kavanagh D, Lambris JD, Noris M, Pickering MC, Remuzzi G, de Córdoba SR, Sethi S, Van der Vlag J, Zipfel PF, and Nester CM
- Subjects
- Autoantibodies immunology, Biopsy, Complement C3 metabolism, Humans, Kidney Diseases metabolism, Kidney Glomerulus metabolism, Rare Diseases, Autoimmunity, Complement C3 immunology, Kidney Diseases immunology, Kidney Glomerulus pathology
- Abstract
The C3 glomerulopathies are a group of rare kidney diseases characterized by complement dysregulation occurring in the fluid phase and in the glomerular microenvironment, which results in prominent complement C3 deposition in kidney biopsy samples. The two major subgroups of C3 glomerulopathy - dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) - have overlapping clinical and pathological features suggestive of a disease continuum. Dysregulation of the complement alternative pathway is fundamental to the manifestations of C3 glomerulopathy, although terminal pathway dysregulation is also common. Disease is driven by acquired factors in most patients - namely, autoantibodies that target the C3 or C5 convertases. These autoantibodies drive complement dysregulation by increasing the half-life of these vital but normally short-lived enzymes. Genetic variation in complement-related genes is a less frequent cause. No disease-specific treatments are available, although immunosuppressive agents and terminal complement pathway blockers are helpful in some patients. Unfortunately, no treatment is universally effective or curative. In aggregate, the limited data on renal transplantation point to a high risk of disease recurrence (both DDD and C3GN) in allograft recipients. Clinical trials are underway to test the efficacy of several first-generation drugs that target the alternative complement pathway.
- Published
- 2019
- Full Text
- View/download PDF
10. An Engineered Complement Factor H Construct for Treatment of C3 Glomerulopathy.
- Author
-
Yang Y, Denton H, Davies OR, Smith-Jackson K, Kerr H, Herbert AP, Barlow PN, Pickering MC, and Marchbank KJ
- Subjects
- Animals, Complement Factor H chemical synthesis, Complement Factor H genetics, Complement Pathway, Alternative, Cricetinae, Glomerular Basement Membrane metabolism, Glomerulonephritis, Membranoproliferative drug therapy, Half-Life, Mice, Protein Binding, Protein Engineering, Complement C3 metabolism, Complement C3b metabolism, Complement Factor H metabolism, Complement Factor H pharmacokinetics, Glomerulonephritis, Membranoproliferative metabolism
- Abstract
Background C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway of complement activation, and treatment options for C3G remain limited. Complement factor H (FH) is a potent regulator of the alternative pathway and might offer a solution, but the mass and complexity of FH makes generation of full-length FH far from trivial. We previously generated a mini-FH construct, with FH short consensus repeats 1-5 linked to repeats 18-20 (FH
1-5^18-20 ), that was effective in experimental C3G. However, the serum t1/2 of FH1-5^18-20 was significantly shorter than that of serum-purified FH. Methods We introduced the oligomerization domain of human FH-related protein 1 (denoted by R1-2) at the carboxy or amino terminus of human FH1-5^18-20 to generate two homodimeric mini-FH constructs (FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2 , respectively) in Chinese hamster ovary cells and tested these constructs using binding, fluid-phase, and erythrocyte lysis assays, followed by experiments in FH-deficient Cfh-/- mice. Results FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2 homodimerized in solution and displayed avid binding profiles on clustered C3b surfaces, particularly FHR1-2^1-5^18-20 Each construct was >10-fold more effective than FH at inhibiting cell surface complement activity in vitro and restricted glomerular basement membrane C3 deposition in vivo significantly better than FH or FH1-5^18-20 FH1-5^18-20^R1-2 had a C3 breakdown fragment binding profile similar to that of FH, a >5-fold increase in serum t1/2 compared with that of FH1-5^18-20 , and significantly better retention in the kidney than FH or FH1-5^18-20 Conclusions FH1-5^18-20^R1-2 may have utility as a treatment option for C3G or other complement-mediated diseases., (Copyright © 2018 by the American Society of Nephrology.)- Published
- 2018
- Full Text
- View/download PDF
11. Atypical hemolytic uremic syndrome and C3 glomerulopathy: conclusions from a "Kidney Disease: Improving Global Outcomes" (KDIGO) Controversies Conference.
- Author
-
Goodship TH, Cook HT, Fakhouri F, Fervenza FC, Frémeaux-Bacchi V, Kavanagh D, Nester CM, Noris M, Pickering MC, Rodríguez de Córdoba S, Roumenina LT, Sethi S, and Smith RJ
- Subjects
- Animals, Atypical Hemolytic Uremic Syndrome drug therapy, Atypical Hemolytic Uremic Syndrome genetics, Atypical Hemolytic Uremic Syndrome pathology, Complement C3 genetics, Complement Inactivating Agents therapeutic use, Genetic Predisposition to Disease, Glomerulonephritis drug therapy, Glomerulonephritis genetics, Glomerulonephritis pathology, Humans, Kidney drug effects, Kidney pathology, Phenotype, Risk Factors, Treatment Outcome, Atypical Hemolytic Uremic Syndrome immunology, Complement Activation drug effects, Complement C3 immunology, Glomerulonephritis immunology, Kidney immunology
- Abstract
In both atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G) complement plays a primary role in disease pathogenesis. Herein we report the outcome of a 2015 Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference where key issues in the management of these 2 diseases were considered by a global panel of experts. Areas addressed included renal pathology, clinical phenotype and assessment, genetic drivers of disease, acquired drivers of disease, and treatment strategies. In order to help guide clinicians who are caring for such patients, recommendations for best treatment strategies were discussed at length, providing the evidence base underpinning current treatment options. Knowledge gaps were identified and a prioritized research agenda was proposed to resolve outstanding controversial issues., (Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
12. The complement factor H-related proteins.
- Author
-
Medjeral-Thomas N and Pickering MC
- Subjects
- Animals, Autoimmunity, Blood Proteins immunology, Complement Factor H immunology, Homeostasis, Humans, Blood Proteins metabolism, Complement Activation, Complement C3 metabolism, Complement Factor H metabolism, Immunity, Innate
- Abstract
The role of the complement factor H-related (FHR) proteins in homeostasis, pathogen defense, and autoimmune disease has recently attracted considerable interest. We highlight the exciting research that has contributed to our understanding of the FHR protein family. Unlike factor H, a potent negative regulator of complement C3 activation, the FHR proteins appear to promote C3 activation. These data have important implications for understanding complement-mediated diseases because, depending on the context, the balance between the actions of factor H and the FHR proteins determines the degree of complement activation., (© 2016 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
13. Distinct roles for the complement regulators factor H and Crry in protection of the kidney from injury.
- Author
-
Laskowski J, Renner B, Le Quintrec M, Panzer S, Hannan JP, Ljubanovic D, Ruseva MM, Borza DB, Antonioli AH, Pickering MC, Holers VM, and Thurman JM
- Subjects
- Animals, Complement Factor H genetics, Glomerulonephritis genetics, Glomerulonephritis pathology, Kidney Glomerulus pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutation, Receptors, Complement genetics, Receptors, Complement 3b, Complement C3 metabolism, Complement Factor H metabolism, Complement Pathway, Alternative immunology, Glomerulonephritis immunology, Kidney Glomerulus immunology, Receptors, Complement metabolism
- Abstract
Mutations in the complement regulatory proteins are associated with several different diseases. Although these mutations cause dysregulated alternative pathway activation throughout the body, the kidneys are the most common site of injury. The susceptibility of the kidney to alternative pathway-mediated injury may be due to limited expression of complement regulatory proteins on several tissue surfaces within the kidney. To examine the roles of the complement regulatory proteins factor H and Crry in protecting distinct renal surfaces from alternative pathway mediated injury, we generated mice with targeted deletions of the genes for both proteins. Surprisingly, mice with combined genetic deletions of factor H and Crry developed significantly milder renal injury than mice deficient in only factor H. Deficiency of both factor H and Crry was associated with C3 deposition at multiple locations within the kidney, but glomerular C3 deposition was lower than that in factor H alone deficient mice. Thus, factor H and Crry are critical for regulating complement activation at distinct anatomic sites within the kidney. However, widespread activation of the alternative pathway reduces injury by depleting the pool of C3 available at any 1 location., (Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
14. Partial Complement Factor H Deficiency Associates with C3 Glomerulopathy and Thrombotic Microangiopathy.
- Author
-
Vernon KA, Ruseva MM, Cook HT, Botto M, Malik TH, and Pickering MC
- Subjects
- Animals, Complement Activation, Female, Hereditary Complement Deficiency Diseases, Kidney blood supply, Kidney Diseases complications, Male, Mice, Atypical Hemolytic Uremic Syndrome etiology, Complement C3, Complement Factor H deficiency, Kidney Diseases etiology, Kidney Glomerulus, Thrombotic Microangiopathies etiology
- Abstract
The complement-mediated renal diseases C3 glomerulopathy (C3G) and atypical hemolytic uremic syndrome (aHUS) strongly associate with inherited and acquired abnormalities in the regulation of the complement alternative pathway (AP). The major negative regulator of the AP is the plasma protein complement factor H (FH). Abnormalities in FH result in uncontrolled activation of C3 through the AP and associate with susceptibility to both C3G and aHUS. Although previously developed FH-deficient animal models have provided important insights into the mechanisms underlying susceptibility to these unique phenotypes, these models do not entirely reproduce the clinical observations. FH is predominantly synthesized in the liver. We generated mice with hepatocyte-specific FH deficiency and showed that these animals have reduced plasma FH levels with secondary reduction in plasma C3. Unlike mice with complete FH deficiency, hepatocyte-specific FH-deficient animals developed neither plasma C5 depletion nor accumulation of C3 along the glomerular basement membrane. In contrast, subtotal FH deficiency associated with mesangial C3 accumulation consistent with C3G. Although there was no evidence of spontaneous thrombotic microangiopathy, the hepatocyte-specific FH-deficient animals developed severe C5-dependent thrombotic microangiopathy after induction of complement activation within the kidney by accelerated serum nephrotoxic nephritis. Taken together, our data indicate that subtotal FH deficiency can give rise to either spontaneous C3G or aHUS after a complement-activating trigger within the kidney and that the latter is C5 dependent., (Copyright © 2016 by the American Society of Nephrology.)
- Published
- 2016
- Full Text
- View/download PDF
15. Update on C3 glomerulopathy.
- Author
-
Barbour TD, Ruseva MM, and Pickering MC
- Subjects
- Animals, Complement Activation, Humans, Kidney Glomerulus metabolism, Complement C3 metabolism, Complement Factor H metabolism, Kidney Diseases physiopathology, Kidney Glomerulus pathology
- Abstract
C3 glomerulopathy refers to a disease process in which abnormal control of complement activation, degradation or deposition results in predominant C3 fragment deposition within the glomerulus and glomerular damage. Recent studies have improved our understanding of its pathogenesis. The key abnormality is uncontrolled C3b amplification in the circulation and/or along the glomerular basement membrane. Family studies in which disease segregates with structurally abnormal complement factor H-related (CFHR) proteins demonstrate that abnormal CFHR proteins are important in some types of C3 glomerulopathy. This is currently thought to be due to the ability of these proteins to antagonize the major negative regulator of C3 activation, complement factor H (CFH), a process termed 'CFH de-regulation'. Recent clinicopathological cohort studies have led to further refinements in case definition, culminating in a 2013 consensus report, which provides recommendations regarding investigation and treatment. Early clinical experience with complement-targeted therapeutics, notably C5 inhibitors, has also now been published. Here, we summarize the latest developments in C3 glomerulopathy., (© The Author 2014. Published by Oxford University Press on behalf of ERA-EDTA.)
- Published
- 2016
- Full Text
- View/download PDF
16. Complement receptor 3 mediates renal protection in experimental C3 glomerulopathy.
- Author
-
Barbour TD, Ling GS, Ruseva MM, Fossati-Jimack L, Cook HT, Botto M, and Pickering MC
- Subjects
- Animals, CD11b Antigen genetics, Complement Factor H metabolism, Cytokines metabolism, Disease Models, Animal, Female, Hereditary Complement Deficiency Diseases, Mice, Inbred C57BL, Myeloid Cells metabolism, Complement C3 metabolism, Complement Factor H deficiency, Glomerulonephritis metabolism, Kidney Diseases metabolism, Macrophage-1 Antigen metabolism
- Abstract
C3 glomerulopathy is a complement-mediated renal disease that is frequently associated with abnormalities in regulation of the complement alternative pathway. Mice with deficiency of factor H (Cfh(-/-)), a negative alternative pathway regulator, are an established experimental model of C3 glomerulopathy in which complement C3 fragments including iC3b accumulate along the glomerular basement membrane. Here we show that deficiency of complement receptor 3 (CR3), the main receptor for iC3b, enhances the severity of spontaneous renal disease in Cfh(-/-) mice. This effect was found to be dependent on CR3 expression on bone marrow-derived cells. CR3 also mediated renal protection outside the setting of factor H deficiency, as shown by the development of enhanced renal injury in CR3-deficient mice during accelerated nephrotoxic nephritis. The iC3b-CR3 interaction downregulated the proinflammatory cytokine response of both murine and human macrophages to lipopolysaccharide stimulation in vitro, suggesting that the protective effect of CR3 on glomerular injury was mediated via modulation of macrophage-derived proinflammatory cytokines. Thus, CR3 has a protective role in glomerulonephritis and suggests that pharmacologic potentiation of the macrophage CR3 interaction with iC3b could be therapeutically beneficial., (Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
17. Efficacy of Targeted Complement Inhibition in Experimental C3 Glomerulopathy.
- Author
-
Ruseva MM, Peng T, Lasaro MA, Bouchard K, Liu-Chen S, Sun F, Yu ZX, Marozsan A, Wang Y, and Pickering MC
- Subjects
- Animals, Complement C3 metabolism, Disease Models, Animal, Kidney Diseases metabolism, Mice, Recombinant Fusion Proteins pharmacology, Complement C3 antagonists & inhibitors, Glomerular Basement Membrane metabolism, Kidney Diseases drug therapy, Recombinant Fusion Proteins therapeutic use
- Abstract
C3 glomerulopathy refers to renal disorders characterized by abnormal accumulation of C3 within the kidney, commonly along the glomerular basement membrane (GBM). C3 glomerulopathy is associated with complement alternative pathway dysregulation, which includes functional defects in complement regulator factor H (FH). There is no effective treatment for C3 glomerulopathy. We investigated the efficacy of a recombinant mouse protein composed of domains from complement receptor 2 (CR2) and FH (CR2-FH) in two models of C3 glomerulopathy with either preexisting or triggered C3 deposition along the GBM. FH-deficient mice spontaneously develop renal pathology associated with abnormal C3 accumulation along the GBM and secondary plasma C3 deficiency. CR2-FH partially restored plasma C3 levels in FH-deficient mice 2 hours after intravenous injection. CR2-FH specifically targeted glomerular C3 deposits, reduced the linear C3 reactivity assessed with anti-C3 and anti-C3b/iC3b/C3c antibodies, and prevented further spontaneous accumulation of C3 fragments along the GBM. Reduction in glomerular C3d and C9/C5b-9 reactivity was observed after daily administration of CR2-FH for 1 week. In a second mouse model with combined deficiency of FH and complement factor I, CR2-FH prevented de novo C3 deposition along the GBM. These data show that CR2-FH protects the GBM from both spontaneous and triggered C3 deposition in vivo and indicate that this approach should be tested in C3 glomerulopathy., (Copyright © 2016 by the American Society of Nephrology.)
- Published
- 2016
- Full Text
- View/download PDF
18. C3 glomerulopathy: the genetic and clinical findings in dense deposit disease and C3 glomerulonephritis.
- Author
-
Xiao X, Pickering MC, and Smith RJ
- Subjects
- Alleles, Biomarkers metabolism, Complement Pathway, Alternative, Genetic Variation, Humans, Immunity, Innate, Kidney Glomerulus immunology, Kidney Glomerulus pathology, Phenotype, Complement C3 immunology, Glomerulonephritis, Membranoproliferative genetics, Glomerulonephritis, Membranoproliferative immunology, Kidney Diseases genetics, Kidney Diseases immunology
- Abstract
C3 glomerulopathy (C3G) defines a group of very rare renal diseases in which dysregulation of the alternative and terminal complement pathways plays a pivotal pathogenic role. Dysregulation is driven by genetic and/or acquired defects, with interindividual variability giving rise to two broad subtypes of C3G-dense deposit disease (DDD) and C3 glomerulonephritis (C3GN). Patient evaluation should include genetic testing and biomarker profiling of complement activity. There is currently no effective targeted treatment option for C3G and, as a consequence, a variety of supportive measures are used. C3G remains an ideal disease in which new complement therapies can be tested as they become available. Trials must include a comprehensive evaluation of each patient at the genetic and biomarker level so that individual responses to therapy can be predicted and understood in light of the degree of complement dysregulation and underlying pathology., (Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.)
- Published
- 2014
- Full Text
- View/download PDF
19. C3 glomerulopathy: clinicopathologic features and predictors of outcome.
- Author
-
Medjeral-Thomas NR, O'Shaughnessy MM, O'Regan JA, Traynor C, Flanagan M, Wong L, Teoh CW, Awan A, Waldron M, Cairns T, O'Kelly P, Dorman AM, Pickering MC, Conlon PJ, and Cook HT
- Subjects
- Adolescent, Adult, Biomarkers analysis, Biopsy, Child, Disease Progression, Female, Glomerulonephritis complications, Glomerulonephritis immunology, Glomerulonephritis mortality, Glomerulonephritis pathology, Glomerulonephritis, Membranoproliferative complications, Glomerulonephritis, Membranoproliferative immunology, Glomerulonephritis, Membranoproliferative mortality, Glomerulonephritis, Membranoproliferative pathology, Humans, Ireland, Kaplan-Meier Estimate, Kidney Failure, Chronic etiology, London, Male, Middle Aged, Prognosis, Proportional Hazards Models, Retrospective Studies, Risk Factors, Time Factors, Young Adult, Complement C3 analysis, Glomerulonephritis diagnosis, Glomerulonephritis, Membranoproliferative diagnosis, Kidney immunology, Kidney pathology
- Abstract
Background and Objectives: The term C3 glomerulopathy describes renal disorders characterized by the presence of glomerular deposits composed of C3 in the absence of significant amounts of Ig. On the basis of electron microscopy appearance, subsets of C3 glomerulopathy include dense deposit disease (DDD) and C3 glomerulonephritis (C3GN). The full spectrum of histologic change observed in C3 glomerulopathy has yet to be defined and pathologic predictors of renal outcome within this patient population remain largely unknown. This study thus characterized a large C3 glomerulopathy cohort and identified clinicopathologic predictors of renal outcome., Design, Setting, Participants, & Measurements: All patients with kidney biopsies fulfilling criteria for C3 glomerulopathy from two quaternary renal centers within the United Kingdom and Ireland between 1992 and 2012 were retrospectively reviewed. We recorded histologic, demographic, and clinical data and determined predictors of ESRD using the Cox proportional hazards model., Results: Eighty patients with C3 glomerulopathy were identified: 21 with DDD and 59 with C3GN. Patients with DDD were younger, more likely to have low serum C3 levels, and more likely to have crescentic GN than patients with C3GN. Patients with C3GN were older and had more severe arteriolar sclerosis, glomerular sclerosis, and interstitial scarring than patients with DDD. Of 70 patients with available follow-up data, 20 (29%) progressed to ESRD after a median of 28 months. Age >16 years, DDD subtype, and crescentic GN were independent predictors of ESRD within the entire cohort. Renal impairment at presentation predicted ESRD only among patients with DDD., Conclusions: Although detailed serologic and genetic data are lacking, this study nevertheless identifies important clinicopathologic distinctions between patients with DDD and C3GN. These include independent predictors of renal outcome. If replicated in other cohorts, these predictors could be used to stratify patients, enabling application of emerging mechanism-based therapies to patients at high risk for poor renal outcome.
- Published
- 2014
- Full Text
- View/download PDF
20. Intracellular complement activation sustains T cell homeostasis and mediates effector differentiation.
- Author
-
Liszewski MK, Kolev M, Le Friec G, Leung M, Bertram PG, Fara AF, Subias M, Pickering MC, Drouet C, Meri S, Arstila TP, Pekkarinen PT, Ma M, Cope A, Reinheckel T, Rodriguez de Cordoba S, Afzali B, Atkinson JP, and Kemper C
- Subjects
- Adult, Arthritis, Rheumatoid immunology, CD4-Positive T-Lymphocytes metabolism, Cell Line, Cell Survival immunology, Child, Complement C3 immunology, Complement C3a metabolism, Complement C3b metabolism, Gene Expression Regulation immunology, Humans, B-Lymphocyte Subsets cytology, CD4-Positive T-Lymphocytes immunology, Cathepsin L metabolism, Cell Differentiation, Complement Activation physiology, Complement C3 metabolism, Homeostasis physiology
- Abstract
Complement is viewed as a critical serum-operative component of innate immunity, with processing of its key component, C3, into activation fragments C3a and C3b confined to the extracellular space. We report here that C3 activation also occurred intracellularly. We found that the T cell-expressed protease cathepsin L (CTSL) processed C3 into biologically active C3a and C3b. Resting T cells contained stores of endosomal and lysosomal C3 and CTSL and substantial amounts of CTSL-generated C3a. While "tonic" intracellular C3a generation was required for homeostatic T cell survival, shuttling of this intracellular C3-activation-system to the cell surface upon T cell stimulation induced autocrine proinflammatory cytokine production. Furthermore, T cells from patients with autoimmune arthritis demonstrated hyperactive intracellular complement activation and interferon-γ production and CTSL inhibition corrected this deregulated phenotype. Importantly, intracellular C3a was observed in all examined cell populations, suggesting that intracellular complement activation might be of broad physiological significance., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
21. C3 glomerulopathy: consensus report.
- Author
-
Pickering MC, D'Agati VD, Nester CM, Smith RJ, Haas M, Appel GB, Alpers CE, Bajema IM, Bedrosian C, Braun M, Doyle M, Fakhouri F, Fervenza FC, Fogo AB, Frémeaux-Bacchi V, Gale DP, Goicoechea de Jorge E, Griffin G, Harris CL, Holers VM, Johnson S, Lavin PJ, Medjeral-Thomas N, Paul Morgan B, Nast CC, Noel LH, Peters DK, Rodríguez de Córdoba S, Servais A, Sethi S, Song WC, Tamburini P, Thurman JM, Zavros M, and Cook HT
- Subjects
- Biomedical Research, Biopsy, Cooperative Behavior, Glomerulonephritis diagnosis, Glomerulonephritis therapy, Humans, International Cooperation, Kidney Glomerulus pathology, Predictive Value of Tests, Prognosis, Complement C3 analysis, Glomerulonephritis immunology, Kidney Glomerulus immunology
- Abstract
C3 glomerulopathy is a recently introduced pathological entity whose original definition was glomerular pathology characterized by C3 accumulation with absent or scanty immunoglobulin deposition. In August 2012, an invited group of experts (comprising the authors of this document) in renal pathology, nephrology, complement biology, and complement therapeutics met to discuss C3 glomerulopathy in the first C3 Glomerulopathy Meeting. The objectives were to reach a consensus on: the definition of C3 glomerulopathy, appropriate complement investigations that should be performed in these patients, and how complement therapeutics should be explored in the condition. This meeting report represents the current consensus view of the group.
- Published
- 2013
- Full Text
- View/download PDF
22. Dense deposit disease and C3 glomerulopathy.
- Author
-
Barbour TD, Pickering MC, and Terence Cook H
- Subjects
- Animals, Complement Factor H genetics, Disease Models, Animal, Glomerulonephritis, Membranoproliferative genetics, Humans, Kidney Glomerulus metabolism, Complement C3 metabolism, Complement Factor H deficiency, Complement Pathway, Alternative genetics, Glomerulonephritis, Membranoproliferative physiopathology, Kidney Glomerulus pathology
- 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., (© 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
23. Complement component C3 plays a critical role in protecting the aging retina in a murine model of age-related macular degeneration.
- Author
-
Hoh Kam J, Lenassi E, Malik TH, Pickering MC, and Jeffery G
- Subjects
- Amyloid beta-Peptides metabolism, Animals, Bruch Membrane ultrastructure, Complement C3 deficiency, Complement Factor H deficiency, Disease Models, Animal, Electroretinography, Macular Degeneration physiopathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Electron, Scanning, Photoreceptor Cells, Vertebrate metabolism, Photoreceptor Cells, Vertebrate physiology, Retina metabolism, Retinal Pigment Epithelium metabolism, Complement C3 physiology, Macular Degeneration etiology, Retina physiology
- 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., (Copyright © 2013 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
24. Recent insights into C3 glomerulopathy.
- Author
-
Barbour TD, Pickering MC, and Cook HT
- Subjects
- Animals, Humans, Kidney Diseases metabolism, Kidney Glomerulus metabolism, Complement C3 metabolism, Kidney Diseases pathology, Kidney Glomerulus pathology
- 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.
- Published
- 2013
- Full Text
- View/download PDF
25. C3 glomerulopathy-associated CFHR1 mutation alters FHR oligomerization and complement regulation.
- Author
-
Tortajada A, Yébenes H, Abarrategui-Garrido C, Anter J, García-Fernández JM, Martínez-Barricarte R, Alba-Domínguez M, Malik TH, Bedoya R, Cabrera Pérez R, López Trascasa M, Pickering MC, Harris CL, Sánchez-Corral P, Llorca O, and Rodríguez de Córdoba S
- Subjects
- Child, Complement C3 chemistry, Complement C3b Inactivator Proteins chemistry, Complement C3b Inactivator Proteins metabolism, Complement System Proteins isolation & purification, Complement System Proteins metabolism, Female, Gene Duplication, Hemolysis, Humans, Immobilized Proteins chemistry, Immobilized Proteins metabolism, Kidney pathology, Male, Middle Aged, Mutagenesis, Insertional, Pedigree, Protein Binding, Protein Multimerization, Protein Structure, Quaternary, Sequence Analysis, DNA, Complement C3 metabolism, Complement C3b Inactivator Proteins genetics, Kidney Diseases genetics
- Abstract
C3 glomerulopathies (C3G) are a group of severe renal diseases with distinct patterns of glomerular inflammation and C3 deposition caused by complement dysregulation. Here we report the identification of a familial C3G-associated genomic mutation in the gene complement factor H–related 1 (CFHR1), which encodes FHR1. The mutation resulted in the duplication of the N-terminal short consensus repeats (SCRs) that are conserved in FHR2 and FHR5. We determined that native FHR1, FHR2, and FHR5 circulate in plasma as homo- and hetero-oligomeric complexes, the formation of which is likely mediated by the conserved N-terminal domain. In mutant FHR1, duplication of the N-terminal domain resulted in the formation of unusually large multimeric FHR complexes that exhibited increased avidity for the FHR1 ligands C3b, iC3b, and C3dg and enhanced competition with complement factor H (FH) in surface plasmon resonance (SPR) studies and hemolytic assays. These data revealed that FHR1, FHR2, and FHR5 organize a combinatorial repertoire of oligomeric complexes and demonstrated that changes in FHR oligomerization influence the regulation of complement activation. In summary, our identification and characterization of a unique CFHR1 mutation provides insights into the biology of the FHRs and contributes to our understanding of the pathogenic mechanisms underlying C3G.
- Published
- 2013
- Full Text
- View/download PDF
26. A hybrid CFHR3-1 gene causes familial C3 glomerulopathy.
- Author
-
Malik TH, Lavin PJ, Goicoechea de Jorge E, Vernon KA, Rose KL, Patel MP, de Leeuw M, Neary JJ, Conlon PJ, Winn MP, and Pickering MC
- Subjects
- Adolescent, Adult, Biopsy, Child, Preschool, Female, Genetic Predisposition to Disease genetics, Genotype, Humans, Kidney metabolism, Kidney pathology, Male, Middle Aged, Pedigree, Blood Proteins genetics, Chimera genetics, Complement C3 metabolism, Complement C3b Inactivator Proteins genetics, Glomerulonephritis, IGA genetics, Glomerulonephritis, IGA metabolism
- Abstract
Controlled activation of the complement system, a key component of innate immunity, enables destruction of pathogens with minimal damage to host tissue. Complement factor H (CFH), which inhibits complement activation, and five CFH-related proteins (CFHR1-5) compose a family of structurally related molecules. Combined deletion of CFHR3 and CFHR1 is common and confers a protective effect in IgA nephropathy. Here, we report an autosomal dominant complement-mediated GN associated with abnormal increases in copy number across the CFHR3 and CFHR1 loci. In addition to normal copies of these genes, affected individuals carry a unique hybrid CFHR3-1 gene. In addition to identifying an association between these genetic observations and complement-mediated kidney disease, these results provide insight into the protective role of the combined deletion of CFHR3 and CFHR1 in IgA nephropathy.
- Published
- 2012
- Full Text
- View/download PDF
27. C3 glomerulopathy: a new classification.
- Author
-
Fakhouri F, Frémeaux-Bacchi V, Noël LH, Cook HT, and Pickering MC
- Subjects
- Glomerulonephritis etiology, Humans, Kidney Glomerulus metabolism, Kidney Glomerulus pathology, Risk Factors, Complement C3 metabolism, Glomerulonephritis classification
- Abstract
Several distinct pathological patterns of glomerular inflammation are associated with abnormal regulation of the complement system, specifically, with dysregulation of the alternative pathway of the complement system. However, these conditions share the pathological finding of complement C3 (C3) deposited within the glomerulus in the absence of substantial immunoglobulin. This finding has alerted us and others to the possible presence of genetic and acquired complement dysregulation in individual patients. This article summarizes our current understanding of the relationship between dysregulation of the complement system and glomerular inflammation. Here, we suggest that glomerular pathologies that are characterized by the isolated deposition of C3 could usefully be classified by the term C3 glomerulopathy. In our view, this classification would alert the pathologist and nephrologist to the importance of screening for acquired and genetic abnormalities in complement regulation. In the future, it could help to identify individuals who might benefit from therapeutic inhibition of the complement system.
- Published
- 2010
- Full Text
- View/download PDF
28. Decay-accelerating factor suppresses complement C3 activation and retards atherosclerosis in low-density lipoprotein receptor-deficient mice.
- Author
-
Leung VW, Yun S, Botto M, Mason JC, Malik TH, Song W, Paixao-Cavalcante D, Pickering MC, Boyle JJ, and Haskard DO
- Subjects
- Actins metabolism, Animals, Antibodies pharmacology, Aorta, Abdominal drug effects, Aorta, Abdominal immunology, Aorta, Abdominal pathology, Aorta, Thoracic drug effects, Aorta, Thoracic immunology, Aorta, Thoracic pathology, Atherosclerosis blood, Body Weight drug effects, Complement Activation drug effects, Complement C3d immunology, Complement Membrane Attack Complex immunology, Lipids blood, Mice, Microscopy, Confocal, Receptors, LDL metabolism, Reproducibility of Results, Atherosclerosis immunology, Atherosclerosis prevention & control, CD55 Antigens metabolism, Complement Activation immunology, Complement C3 immunology, Receptors, LDL deficiency
- Abstract
Decay-accelerating factor (DAF; CD55) is a membrane protein that regulates complement pathway activity at the level of C3. To test the hypothesis that DAF plays an essential role in limiting complement activation in the arterial wall and protecting from atherosclerosis, we crossed DAF gene targeted mice (daf-1(-/-)) with low-density lipoprotein-receptor deficient mice (Ldlr(-/-)). Daf-1(-/-)Ldlr(-/-) mice had more extensive en face Sudan IV staining of the thoracoabdominal aorta than Ldlr(-/-) mice, both following a 12-week period of low-fat diet or a high-fat diet. Aortic root lesions in daf-1(-/-)Ldlr(-/-) mice on a low-fat diet showed increased size and complexity. DAF deficiency increased deposition of C3d and C5b-9, indicating the importance of DAF for downstream complement regulation in the arterial wall. The acceleration of lesion development in the absence of DAF provides confirmation of the proinflammatory and proatherosclerotic potential of complement activation in the Ldlr(-/-) mouse model. Because upstream complement activation is potentially protective, this study underlines the importance of DAF in shielding the arterial wall from the atherogenic effects of complement.
- Published
- 2009
- Full Text
- View/download PDF
29. Crry deficiency in complement sufficient mice: C3 consumption occurs without associated renal injury.
- Author
-
Ruseva MM, Hughes TR, Donev RM, Sivasankar B, Pickering MC, Wu X, Harris CL, and Morgan BP
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Complement C3 genetics, Complement C5 antagonists & inhibitors, Complement C5 genetics, Complement C5 immunology, Complement C6 genetics, Complement C6 immunology, Complement Factor H genetics, Complement Factor H immunology, Crosses, Genetic, Embryo Loss genetics, Embryo Loss immunology, Female, Homeostasis genetics, Kidney injuries, Male, Mice, Mice, Knockout, Pregnancy, Receptors, Complement genetics, Receptors, Complement 3b, Complement C3 immunology, Homeostasis immunology, Kidney immunology, Receptors, Complement immunology
- Abstract
The rodent-specific complement regulator complement receptor 1-related gene/protein-y (Crry) is critical for complement homeostasis. Gene deletion is 100% embryonically lethal; Crry-deficient (Crry(-/-)) mice were rescued by back-crossing onto C3 deficiency, confirming that embryo loss was complement mediated. In order to rescue viable Crry(-/-) mice without deleting C3, we have tested inhibition of C5 during gestation. Crry(+/-) females were given neutralizing anti-C5 mAb immediately prior to mating with Crry(+/-) males and C5 inhibition maintained through pregnancy. A single, healthy Crry(-/-) female was obtained and mating with Crry(+/-) males yielded healthy litters containing equal numbers of Crry(+/-) and Crry(-/-) pups. Inter-crossing Crry(-/-) mice yielded healthy litters of expected size. Although the mice were not anemic, exposure of Crry(-/-) erythrocytes to normal mouse serum caused C3 deposition and lysis, while transfusion into normal or C6(-/-) mice resulted in rapid clearance. Complement activity and C3 levels in Crry(-/-) mice were markedly reduced. Comparison with factor H deficient (CfH(-/-)) mice revealed similar levels of residual C3; however, unlike the CfH(-/-) mice, Crry(-/-) mice showed no evidence of renal injury, demonstrating distinct roles for these regulators in protecting the kidney.
- Published
- 2009
- Full Text
- View/download PDF
30. Factor I is required for the development of membranoproliferative glomerulonephritis in factor H-deficient mice.
- Author
-
Rose KL, Paixao-Cavalcante D, Fish J, Manderson AP, Malik TH, Bygrave AE, Lin T, Sacks SH, Walport MJ, Cook HT, Botto M, and Pickering MC
- Subjects
- Animals, Complement C3 analysis, Complement Factor H administration & dosage, Complement Factor H genetics, Complement Factor I administration & dosage, Complement Factor I genetics, Glomerulonephritis, Membranoproliferative blood, Glomerulonephritis, Membranoproliferative immunology, Mice, Mice, Mutant Strains, Complement C3 metabolism, Complement Factor H deficiency, Complement Factor I physiology, Glomerulonephritis, Membranoproliferative genetics
- Abstract
The inflammatory kidney disease membranoproliferative glomerulonephritis type II (MPGN2) is associated with dysregulation of the alternative pathway of complement activation. MPGN2 is characterized by the presence of complement C3 along the glomerular basement membrane (GBM). Spontaneous activation of C3 through the alternative pathway is regulated by 2 plasma proteins, factor H and factor I. Deficiency of either of these regulators results in uncontrolled C3 activation, although the breakdown of activated C3 is dependent on factor I. Deficiency of factor H, but not factor I, is associated with MPGN2 in humans, pigs, and mice. To explain this discordance, mice with single or combined deficiencies of these factors were studied. MPGN2 did not develop in mice with combined factor H and I deficiency or in mice deficient in factor I alone. However, administration of a source of factor I to mice with combined factor H and factor I deficiency triggered both activated C3 fragments in plasma and GBM C3 deposition. Mouse renal transplant studies demonstrated that C3 deposited along the GBM was derived from plasma. Together, these findings provide what we believe to be the first evidence that factor I-mediated generation of activated C3 fragments in the circulation is a critical determinant for the development of MPGN2 associated with factor H deficiency.
- Published
- 2008
- Full Text
- View/download PDF
31. Uncontrolled C3 activation causes membranoproliferative glomerulonephritis in mice deficient in complement factor H.
- Author
-
Pickering MC, Cook HT, Warren J, Bygrave AE, Moss J, Walport MJ, and Botto M
- Subjects
- Animals, Complement C3 metabolism, Complement C9 metabolism, Complement Factor H metabolism, Disease Models, Animal, Female, Kidney pathology, Kidney ultrastructure, Male, Mice, Mice, Inbred Strains, Mice, Mutant Strains, Mutation, Complement Activation genetics, Complement C3 immunology, Complement Factor H genetics, Glomerulonephritis genetics, Glomerulonephritis physiopathology
- Abstract
The alternative pathway of complement is activated continuously in vivo through the C3 'tick-over' pathway. This pathway is triggered by the hydrolysis of C3, resulting in the formation of C3 convertase. This, in turn, generates C3b, which mediates many of the biological functions of complement. Factor H, the main regulator of this activation, prevents formation and promotes dissociation of the C3 convertase enzyme, and, together with factor I, mediates the proteolytic inactivation of C3b. Factor H deficiency, described in 29 individuals from 12 families and in pigs, allows unhindered activation of fluid-phase C3 and severe depletion of plasma C3 (ref. 11). Membranoproliferative glomerulonephritis (MPGN) occurs in factor H-deficient humans and pigs. Although MPGN has been reported in other conditions in which uncontrolled activation of C3 occurs, the role of C3 dysregulation in the pathogenesis of MPGN is not understood. Here we show that mice deficient in factor H (Cfh(-/-) mice) develop MPGN spontaneously and are hypersensitive to developing renal injury caused by immune complexes. Introducing a second mutation in the gene encoding complement factor B, which prevents C3 turnover in vivo, obviates the phenotype of Cfh(-/-) mice. Thus, uncontrolled C3 activation in vivo is essential for the development of MPGN associated with deficiency of factor H.
- Published
- 2002
- Full Text
- View/download PDF
32. Factor H–Related Protein 1 Drives Disease Susceptibility and Prognosis in C3 Glomerulopathy
- Author
-
Márquez-Tirado, Bárbara, Gutiérrez-Tenorio, Josué, Tortajada, Agustín, Lucientes Continente, Laura, Caravaca-Fontán, Fernando, Malik, Talat H., Roldán Montero, Raquel, Elías, Sandra, Saiz Gonzalez, Ana, Fernández-Juarez, Gema, Sánchez-Corral, Pilar, Pickering, Matthew C., Praga, Manuel, Rodríguez de Córdoba, Santiago, Goicoechea de Jorge, Elena, Ministerio de Ciencia e Innovación (España), European Commission, Comunidad de Madrid, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Gutiérrez-Tenorio, Josué [0000-0001-8590-5455], Tortajada, Agustín [0000-0002-2131-2594], Lucientes, Laura [0000-0001-5596-370X], Caravaca-Fontán, Fernando [0000-0002-5830-9663], Saiz Gonzalez, Ana [0000-0001-7110-450X], Fernández-Juárez, Gema [0000-0001-6641-7763], Sánchez-Corral, Pilar [0000-0003-4212-1233], Pickering, Matthew C. [0000-0002-1153-0192], Praga, Manuel [0000-0001-9270-1071], Rodríguez de Córdoba, Santiago [0000-0001-6401-1874], Goicoechea de Jorge, Elena [0000-0002-4978-2483], Gutiérrez-Tenorio, Josué, Tortajada, Agustín, Lucientes, Laura, Caravaca-Fontán, Fernando, Saiz Gonzalez, Ana, Fernández-Juárez, Gema, Sánchez-Corral, Pilar, Pickering, Matthew C., Praga, Manuel, Rodríguez de Córdoba, Santiago, and Goicoechea de Jorge, Elena
- Subjects
Glomerular disease ,Disease susceptibility ,DNA Copy Number Variations ,Factor H-related protein 1 ,Complement ,Genetic renal disease ,Glomerulonephritis, IGA ,General Medicine ,Blood Proteins ,Complement C3 ,Prognosis ,Basic Research ,Nephrology ,Complement Factor H ,Complement C3b Inactivator Proteins ,Humans ,C3 glomerulopathy ,Disease Susceptibility - Abstract
17 p.-8 fig., Background: C3 glomerulopathy (C3G) is a heterogeneous group of chronic renal diseases characterized predominantly by glomerular C3 deposition and complement dysregulation. Mutations in factor H-related (FHR) proteins resulting in duplicated dimerization domains are prototypical of C3G, although the underlying pathogenic mechanism is unclear., Methods: Using in vitro and in vivo assays, we performed extensive characterization of an FHR-1 mutant with a duplicated dimerization domain. To assess the FHR-1 mutant's association with disease susceptibility and renal prognosis, we also analyzed CFHR1 copy number variations and FHR-1 plasma levels in two Spanish C3G cohorts and in a control population., Results: Duplication of the dimerization domain conferred FHR-1 with an increased capacity to interact with C3-opsonized surfaces, which resulted in an excessive activation of the alternative pathway. This activation does not involve C3b binding competition with factor H. These findings support a scenario in which mutant FHR-1 binds to C3-activated fragments and recruits native C3 and C3b; this leads to formation of alternative pathway C3 convertases, which increases deposition of C3b molecules, overcoming FH regulation. This suggests that a balanced FHR-1/FH ratio is crucial to control complement amplification on opsonized surfaces. Consistent with this conceptual framework, we show that the genetic deficiency of FHR-1 or decreased FHR-1 in plasma confers protection against developing C3G and associates with better renal outcome., Conclusions: Our findings explain how FHR-1 mutants with duplicated dimerization domains result in predisposition to C3G. They also provide a pathogenic mechanism that may be shared by other diseases, such as IgA nephropathy or age-related macular degeneration, and identify FHR-1 as a potential novel therapeutic target in C3G., E. Goicoechea de Jorge is supported by Ministerio de Ciencia e Innovación grant RTI2018-095955-B-100 and the European Union’s Horizon 2020 Framework Programme grant 899163. J. Gutiérrez-Tenorio is supported by Ministerio de Ciencia e Innovación grant BES-2015-073833. L. Lucientes Continente is supported by the Autonomous Region of Madrid grant S2017/BMD-3673. G. Fernández-Juarez, P. Sánchez-Corral, B. Márquez-Tirado, and M. Praga are supported by the Instituto de Salud Carlos III and the European Union’s European Regional Development Fund grants PI19/01695, PI19/00970, and PI19/01624, respectively. M.C. Pickering is a Wellcome Trust Senior Fellow in Clinical Science (212252/Z/18/Z). S. Rodríguez de Córdoba is supported by the Ministerio de Economía y Competitividad grant PID2019-104912RB-100 and Autonomous Region of Madrid grant S2017/BMD-3673.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.