Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Mol Immunol ; 168: 10-16, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38368725

ABSTRACT

Complement alternative pathway (AP) dysregulation drives C3 glomerulopathy (C3G), a rare renal disorder characterized by glomerular C3 deposition and glomerular damage, for which no effective treatments are available. Blockade of complement C3 is emerging as a viable therapeutic option. In an earlier study we showed that SLN500, a small interfering RNA targeting liver C3 synthesis, was able to limit AP dysregulation and glomerular C3d deposits in mice with partial factor H (FH) deficiency (Cfh+/- mice). Here, we assessed the pharmacological effects of SLN501 - an optimized SLN500 version - in mice with complete FH deficiency (Cfh-/- mice) that exhibit a more severe C3G phenotype. SLN501 effectively prevented liver C3 synthesis, thus limiting AP dysregulation, glomerular C3d deposits and the development of ultrastructural alterations. These data provide firm evidence of the use of siRNA-mediated liver C3 gene silencing as a potential therapy for treating C3G patients with either partial or complete FH loss of function.


Subject(s)
Complement Factor H/deficiency , Glomerulonephritis, Membranoproliferative , Hereditary Complement Deficiency Diseases , Kidney Diseases , Humans , Animals , Mice , Complement C3/genetics , Complement C3/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Complement Factor H/genetics , Complement Factor H/therapeutic use , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/drug therapy , Glomerulonephritis, Membranoproliferative/metabolism , Complement Pathway, Alternative
2.
Exp Biol Med (Maywood) ; 247(2): 77-86, 2022 01.
Article in English | MEDLINE | ID: mdl-34775843

ABSTRACT

Age-related macular degeneration is the leading cause of blindness in the elderly. The Y402H polymorphism in complement factor H promotes disease-like pathogenesis, and a Cfh+/- murine model can replicate this phenotype, but only after two years. We reasoned that by combining CFH deficiency with cigarette smoke exposure, we might be able to accelerate disease progression to facilitate preclinical research in this disease. Wild-type and Cfh+/- mice were exposed to nose-only cigarette smoke for three months. Retinal tissue morphology and visual function were evaluated by optical coherence tomography, fundus photography and autofluorescence, and electroretinogram. Retinal pigment epithelial cell phenotype and ultrastructure were evaluated by immunofluorescence staining and transmission electron microscopy. Cfh+/- smoking mice showed a dome-like protruding lesion at the ellipsoid zone (drusen-like deposition), many retinal hyper-autofluorescence spots, and a marked decrease in A- and B-wave amplitudes. Compared with non-smoking mice, wild-type and Cfh+/- smoking mice showed sub-retinal pigment epithelium complement protein 3 deposition, activation of microglia, metabolic waste accumulation, and impairment of tight junctions. Microglia cells migrated into the photoreceptor outer segment layer in Cfh+/- smoking mice showed increased activation. Our results suggest that exposing Cfh+/- mice to smoking leads to earlier onset of age-related macular degeneration than in other animal models, which may facilitate preclinical research into the pathophysiology and treatment of this disease.


Subject(s)
Complement Factor H/deficiency , Hereditary Complement Deficiency Diseases/metabolism , Kidney Diseases/metabolism , Macular Degeneration/metabolism , Smoking/metabolism , Animals , Complement Factor H/genetics , Complement Factor H/metabolism , Disease Models, Animal , Hereditary Complement Deficiency Diseases/genetics , Kidney Diseases/genetics , Macular Degeneration/etiology , Macular Degeneration/genetics , Mice , Mice, Knockout , Smoking/adverse effects , Smoking/genetics
3.
Front Immunol ; 12: 752916, 2021.
Article in English | MEDLINE | ID: mdl-34956184

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 (mFH1-5^18-20^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.


Subject(s)
Complement C3/immunology , Complement Factor H/immunology , Animals , Complement Factor H/deficiency , Kidney/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Front Immunol ; 12: 681098, 2021.
Article in English | MEDLINE | ID: mdl-34054871

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.


Subject(s)
Complement C3/immunology , Complement C3/metabolism , Complement Factor H/biosynthesis , Complement Factor H/deficiency , Protein Disulfide-Isomerases/metabolism , Recombinant Proteins/metabolism , Animals , Gene Expression , Immunomodulation , Mice , Mice, Knockout , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Yeasts/genetics , Yeasts/metabolism
5.
Pediatr Nephrol ; 36(6): 1365-1375, 2021 06.
Article in English | MEDLINE | ID: mdl-32529325

ABSTRACT

Deficiency of Complement Factor H Related (CFHR) plasma proteins and Autoantibody Positive Hemolytic Uremic Syndrome (DEAP-HUS) is a subtype of atypical hemolytic uremic syndrome, known to be associated with significant morbidity. Its pathogenesis is linked to the production of IgG autoantibodies against complement factor H, a regulator of the alternative complement pathway. The binding of the autoantibodies to the C terminal of complement factor H interferes with its regulatory function, leading to increased activation of the alternative complement pathway and consequent endothelial cellular damage. Early diagnosis and initiation of appropriate therapy is reported to lead to favorable outcomes. Institution of plasma exchange therapy within 24 h of diagnosis has been shown to rapidly lower antibody levels, leading to clinical improvement. Adjunctive immunosuppression therapy suppresses antibody production and helps in maintaining long-term clinical remission of the disease. Available data advocates a treatment regimen that combines plasma therapy (preferably plasma exchange) and immunosuppression to halt disease process and sustain long-term disease remission.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Autoantibodies , Complement Factor H , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/therapy , Complement Factor H/deficiency , Humans , Immunosuppression Therapy , Plasma Exchange
6.
Sci Rep ; 10(1): 10320, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32587311

ABSTRACT

Polymorphisms in the Complement Factor H (CFH) gene, coding for the Factor H protein (FH), can increase the risk for age-related macular degeneration (AMD). AMD-associated CFH risk variants, Y402H in particular, impair FH function leading to complement overactivation. Whether this alone suffices to trigger AMD pathogenesis remains unclear. In AMD, retinal homeostasis is compromised due to the dysfunction of retinal pigment epithelium (RPE) cells. To investigate the impact of endogenous FH loss on RPE cell balance, we silenced CFH in human hTERT-RPE1 cells. FH reduction led to accumulation of C3, at both RNA and protein level and increased RPE vulnerability toward oxidative stress. Mild hydrogen-peroxide exposure in combination with CFH knock-down led to a reduction of glycolysis and mitochondrial respiration, paralleled by an increase in lipid peroxidation, which is a key aspect of AMD pathogenesis. In parallel, cell viability was decreased. The perturbations of energy metabolism were accompanied by transcriptional deregulation of several glucose metabolism genes as well as genes modulating mitochondrial stability. Our data suggest that endogenously produced FH contributes to transcriptional and metabolic homeostasis and protects RPE cells from oxidative stress, highlighting a novel role of FH in AMD pathogenesis.


Subject(s)
Epithelial Cells/pathology , Macular Degeneration/genetics , Retinal Pigment Epithelium/pathology , Cell Line , Cell Survival/genetics , Complement Factor H/deficiency , Complement Factor H/genetics , Energy Metabolism/genetics , Gene Knockdown Techniques , Glycolysis/genetics , Humans , Lipid Peroxidation/genetics , Macular Degeneration/pathology , Oxidative Stress/genetics , Retinal Pigment Epithelium/cytology
7.
J Clin Invest ; 130(8): 4039-4054, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32369457

ABSTRACT

Hepatocellular carcinoma (HCC) is difficult to detect, carries a poor prognosis, and is one of few cancers with an increasing yearly incidence. Molecular defects in complement factor H (CFH), a critical regulatory protein of the complement alternative pathway (AP), are typically associated with inflammatory diseases of the eye and kidney. Little is known regarding the role of CFH in controlling complement activation within the liver. While studying aging CFH-deficient (fH-/-) mice, we observed spontaneous hepatic tumor formation in more than 50% of aged fH-/- males. Examination of fH-/- livers (3-24 months) for evidence of complement-mediated inflammation revealed widespread deposition of complement-activation fragments throughout the sinusoids, elevated transaminase levels, increased hepatic CD8+ and F4/80+ cells, overexpression of hepatic mRNA associated with inflammatory signaling pathways, steatosis, and increased collagen deposition. Immunostaining of human HCC biopsies revealed extensive deposition of complement fragments within the tumors. Investigating the Cancer Genome Atlas also revealed that increased CFH mRNA expression is associated with improved survival in patients with HCC, whereas mutations are associated with worse survival. These results indicate that CFH is critical for controlling complement activation in the liver, and in its absence, AP activation leads to chronic inflammation and promotes hepatic carcinogenesis.


Subject(s)
Carcinoma, Hepatocellular , Complement Factor H/deficiency , Gene Expression Regulation, Neoplastic , Hereditary Complement Deficiency Diseases , Kidney Diseases , Liver Neoplasms , Liver , Neoplasm Proteins , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Complement Factor H/genetics , Complement Factor H/metabolism , Hereditary Complement Deficiency Diseases/genetics , Hereditary Complement Deficiency Diseases/metabolism , Hereditary Complement Deficiency Diseases/pathology , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism
8.
FEBS Lett ; 594(16): 2543-2555, 2020 08.
Article in English | MEDLINE | ID: mdl-31943152

ABSTRACT

Dengue disease is an inflammatory-driven pathology, and complement overactivation is linked to disease severity and vascular leakage. Additionally, dysregulation of complement alternative pathway (AP) components has been described, such as upregulation of complement factor D and downregulation of complement factor H (FH), which activate and inhibit the AP, respectively. Thus, the pathology of severe dengue could in part result from AP dysfunction, even though complement and AP activation usually provide protection against viral infections. In dengue virus-infected macrophages and endothelial cells (ECs), the site of replication and target for vascular pathology, respectively, the AP is activated. The AP activation, reduced FH and vascular leakage seen in dengue disease in part parallels other complement AP pathologies associated with FH deficiency, such as atypical haemolytic uraemic syndrome (aHUS). aHUS can be therapeutically targeted with inhibitors of complement terminal activity, raising the idea that strategies such as inhibition of complement or delivery of FH or other complement regulatory components to EC may be beneficial to combat the vascular leakage seen in severe dengue.


Subject(s)
Complement Factor D/immunology , Complement Factor H/immunology , Complement Pathway, Alternative , Dengue Virus/immunology , Dengue/immunology , Animals , Atypical Hemolytic Uremic Syndrome/immunology , Atypical Hemolytic Uremic Syndrome/pathology , Complement Factor H/deficiency , Dengue/pathology , Endothelial Cells/immunology , Endothelial Cells/pathology , Hereditary Complement Deficiency Diseases/immunology , Hereditary Complement Deficiency Diseases/pathology , Humans , Kidney Diseases/immunology , Kidney Diseases/pathology , Macrophages/immunology , Macrophages/pathology
9.
Front Immunol ; 10: 1607, 2019.
Article in English | MEDLINE | ID: mdl-31354740

ABSTRACT

Complement factor H (CFH) has a pivotal role in regulating alternative complement activation through its ability to inhibit the cleavage of the central complement component C3, which links innate and humoral immunity. However, insights into the role of CFH in B cell biology are limited. Here, we demonstrate that deficiency of CFH in mice leads to altered splenic B cell development characterized by the accumulation of marginal zone (MZ) B cells. Furthermore, B cells in Cfh-/- mice exhibit enhanced B cell receptor (BCR) signaling as evaluated by increased levels of phosphorylated Bruton's tyrosine kinase (pBTK) and phosphorylated spleen tyrosine kinase (pSYK). We show that enhanced BCR activation is associated with uncontrolled C3 consumption in the spleen and elevated complement receptor 2 (CR2, also known as CD21) levels on the surface of mature splenic B cells. Moreover, aged Cfh-/- mice developed splenomegaly with distorted spleen architecture and spontaneous B cell-dependent autoimmunity characterized by germinal center hyperactivity and a marked increase in anti-double stranded DNA (dsDNA) antibodies. Taken together, our data indicate that CFH, through its function as a complement repressor, acts as a negative regulator of BCR signaling and limits autoimmunity.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Complement Factor H/genetics , Spleen/immunology , Spleen/metabolism , Animals , Autoimmunity , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Biomarkers , Complement Factor H/deficiency , Complement Factor H/immunology , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
10.
Acta Med Port ; 32(2): 158-161, 2019 Feb 28.
Article in Portuguese | MEDLINE | ID: mdl-30896397

ABSTRACT

We report a case of an 18-month-old boy with H factor deficiency with atypical presentation: recurrent acute otitis media and several maternal family members with autoimmune disorders (vitiligo, thyroiditis and immune trombocytopenia). Blood tests revealed low C3 and AH50, as well as low properdin and H factor. I factor was normal. CFH gene molecular test confirmed the H factor deficiency diagnosis. This child had none of the typical manifestations of this disorder, namely Neisseria meningitidis infection or renal disease (glomerulonephritis and atypical haemolytic uremic syndrome). Autoimmune family history and correct interpretation of blood tests' results were crucial for this diagnosis.


Apresenta-se um caso clínico de um rapaz de 18 meses com défice de fator H com apresentação clínica atípica ­ otite média aguda recorrente e história familiar da linhagem materna com doença autoimune (vitiligo, tiroidite e púrpura trombocitopénica imune). Analiticamente apresentava C3 e AH50 diminuídos, assim como properdina e fator H baixos. O fator I era normal. O estudo molecular do gene CFH confirmou o diagnóstico de défice de fator H. Esta criança não teve nenhuma das manifestações típicas, nomeadamente doença invasiva por Neisseria meningitidis ou doença renal (glomerulonefrite e síndrome hemolítica urémica atípica). A história familiar de autoimunidade e a correta interpretação dos achados laboratoriais foram fundamentais para o diagnóstico.


Subject(s)
Complement C3/deficiency , Complement Factor H/deficiency , Kidney Diseases/diagnosis , Otitis Media/etiology , Acute Disease , Autoimmune Diseases , Complement Factor H/genetics , Hereditary Complement Deficiency Diseases , Humans , Infant , Kidney Diseases/complications , Male , Pedigree , Properdin/deficiency , Recurrence
11.
Eur J Immunol ; 48(5): 791-802, 2018 05.
Article in English | MEDLINE | ID: mdl-29389016

ABSTRACT

Natural IgM binds to glomerular epitopes in several progressive kidney diseases. Previous work has shown that IgM also binds within the glomerulus after ischemia/reperfusion (I/R) but does not fully activate the complement system. Factor H is a circulating complement regulatory protein, and congenital or acquired deficiency of factor H is a strong risk factor for several types of kidney disease. We hypothesized that factor H controls complement activation by IgM in the kidney after I/R, and that heterozygous factor H deficiency would permit IgM-mediated complement activation and injury at this location. We found that mice with targeted heterozygous deletion of the gene for factor H developed more severe kidney injury after I/R than wild-type controls, as expected, but that complement activation within the glomeruli remained well controlled. Furthermore, mice that are unable to generate soluble IgM were not protected from renal I/R, even in the setting of heterozygous factor H deficiency. These results demonstrate that factor H is important for limiting injury in the kidney after I/R, but it is not critical for controlling complement activation by immunoglobulin within the glomerulus in this setting. IgM binds to glomerular epitopes after I/R, but it is not a significant source of injury.


Subject(s)
Acute Kidney Injury/pathology , Complement Activation/immunology , Complement Factor H/deficiency , Complement Factor H/immunology , Immunoglobulin M/immunology , Kidney Diseases/immunology , Kidney Glomerulus/immunology , Reperfusion Injury/immunology , Acute Kidney Injury/genetics , Animals , Complement Factor H/genetics , Complement Pathway, Alternative/immunology , Epitopes/immunology , Hereditary Complement Deficiency Diseases , Immunoglobulin M/deficiency , Kidney Diseases/genetics , Kidney Glomerulus/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/pathology
12.
Iran J Kidney Dis ; 12(6): 376-381, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30595568

ABSTRACT

Complement C3 glomerulopathy refers to a disease process in which abnormal control of complement activation or degradation results in predominant C3 fragment deposition within the glomerulus and causes glomerular damage. Abnormal control of the complement alternative pathway is a well-established risk factor for the occurrence of C3 glomerulonephritis. It is the first reported case in Iran with multiple mutations in complement factor H, with one of these mutations we have expected in hemolytic uremic syndrome rather than C3 glomerulopathy Genetic analysis showed that the molecular abnormalities of factor H led to complement factor H malfunction that were polymorphous and not restricted to the C-terminal domains of the protein.


Subject(s)
Atypical Hemolytic Uremic Syndrome/etiology , Complement C3/genetics , Glomerulonephritis/genetics , Kidney Glomerulus/pathology , Adult , Complement Activation , Complement Factor H/deficiency , Complement Factor H/genetics , Complement Pathway, Alternative , Female , Glomerulonephritis/complications , Glomerulonephritis/pathology , Humans , Iran , Mutation
13.
J Pediatr Hematol Oncol ; 40(1): e41-e44, 2018 01.
Article in English | MEDLINE | ID: mdl-28538511

ABSTRACT

We herein reported a 4-month-old boy with transplantation-associated atypical hemolytic uremic syndrome (TA-aHUS) who was successfully treated with eculizumab. The patient diagnosed with type 3 of familial hemophagocytic lymphohistiocytosis underwent cord blood transplantation. After transplantation, he developed TA-aHUS, but plasma exchanges were unsuccessful. We identified deletions in CFH-related gene 1 (del-CFHR1) by the multiplex ligation-dependent probe amplification testing procedure and CFH autoantibodies. Eculizumab has been administered to the patient, with a marked improvement being achieved in thrombocytopenia. He has been well except for the persistent microhematuria for a year after transplantation. Uncontrolled complement activation might be involved in the pathophysiology of TA-aHUS.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Atypical Hemolytic Uremic Syndrome/drug therapy , Cord Blood Stem Cell Transplantation/adverse effects , Atypical Hemolytic Uremic Syndrome/etiology , Autoantibodies/immunology , Complement Factor H/deficiency , Complement Factor H/immunology , Hereditary Complement Deficiency Diseases , Humans , Infant , Kidney Diseases , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/therapy , Male , Plasma Exchange , Treatment Outcome
14.
Saudi J Kidney Dis Transpl ; 28(6): 1427-1431, 2017.
Article in English | MEDLINE | ID: mdl-29265059

ABSTRACT

The hemolytic uremic syndrome (HUS) is a heterogeneous group of similar entities characterized by microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure (ARF) and is an important cause of ARF in childhood. Mutations have been reported in the complement regulatory protein factor H in both sporadic and familial HUS and have been identified in 10-20% of cases. Inherited HUS is unusual. We report the occurrence of HUS in two siblings after delivery, complicated with ARF and with a good outcome.


Subject(s)
Acute Kidney Injury/etiology , Complement Factor H/deficiency , Hemolytic-Uremic Syndrome/etiology , Kidney Diseases/complications , Puerperal Disorders/etiology , Acute Kidney Injury/diagnosis , Acute Kidney Injury/therapy , Adult , Complement Factor H/genetics , Female , Genetic Predisposition to Disease , Glucocorticoids/therapeutic use , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/therapy , Hereditary Complement Deficiency Diseases , Humans , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Kidney Diseases/therapy , Phenotype , Plasma Exchange , Polymorphism, Genetic , Postpartum Period , Pregnancy , Puerperal Disorders/diagnosis , Puerperal Disorders/therapy , Recurrence , Risk Factors , Siblings , Treatment Outcome
15.
Kidney Int ; 91(6): 1386-1397, 2017 06.
Article in English | MEDLINE | ID: mdl-28139294

ABSTRACT

C3 glomerulopathy is a potentially life-threatening disease of the kidney caused by dysregulated alternative pathway complement activation. The specific complement mediator(s) responsible for kidney injury in C3 glomerulopathy are yet to be defined and no specific therapy is currently available. We previously developed a mouse model of lethal C3 glomerulopathy with factor H and properdin gene double mutations. Therefore, we used this model to examine the role of C5 and C5a receptor (C5aR) in the pathogenesis of the disease. Disease severity in these factor H/properdin double-mutant mice was found to be correlated with plasma C5 levels, and prophylactic anti-C5 mAb therapy was effective in preventing lethal C3 glomerulopathy. When given to these double-mutant mice that had already developed active disease with severe proteinuria, anti-C5 mAb treatment also prevented death in half of the mice. Deficiency of C5aR significantly reduced disease severity, suggesting that C5aR-mediated inflammation contributed to C3 glomerulopathy. Thus, C5 and C5aR have a critical role in C3 glomerulopathy. Hence, early intervention targeting these pathways may be an effective therapeutic strategy for patients with C3 glomerulopathy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Complement C3/metabolism , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/drug effects , Glomerulonephritis/prevention & control , Kidney/drug effects , Renal Insufficiency/prevention & control , Animals , Complement C3/immunology , Complement C5/immunology , Complement C5/metabolism , Complement Factor H/deficiency , Complement Factor H/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Properdin/deficiency , Properdin/genetics , Proteinuria/immunology , Proteinuria/metabolism , Proteinuria/prevention & control , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Renal Insufficiency/genetics , Renal Insufficiency/immunology , Renal Insufficiency/metabolism , Signal Transduction/drug effects , Time Factors
16.
Article in English | MEDLINE | ID: mdl-27929404

ABSTRACT

To evaluate the association among complement factor H-related (CFHRs) gene deficiency, complement factor H (CFH) autoantibodies, and atypical hemolytic uremic syndrome (aHUS) susceptibility. EMBASE, PubMed, and the ISI Web of Science databases were searched for all eligible studies on the relationship among CFHRs deficiency, anti-FH autoantibodies, and aHUS risk. Eight case-control studies with 927 cases and 1182 controls were included in this study. CFHR1 deficiency was significantly associated with an increased risk of aHUS (odds ratio (OR) = 3.61, 95% confidence interval (95% CI), 1.96, 6.63, p < 0.001), while no association was demonstrated in individuals with only CFHR1/R3 deficiency (OR = 1.32, 95% CI, 0.50, 3.50, p = 0.56). Moreover, a more significant correlation was observed in people with both FH-anti autoantibodies and CFHR1 deficiency (OR = 11.75, 95% CI, 4.53, 30.44, p < 0.001) in contrast to those with only CFHR1 deficiency. In addition, the results were essentially consistent among subgroups stratified by study quality, ethnicity, and gene detection methods. The present meta-analysis indicated that CFHR1 deletion was significantly associated with the risk of aHUS, particularly when combined with anti-FH autoantibodies, indicating that potential interactions among CFHR1 deficiency and anti-FH autoantibodies might impact the risk of aHUS.


Subject(s)
Atypical Hemolytic Uremic Syndrome/epidemiology , Complement Factor H/deficiency , Complement Factor H/immunology , Kidney Diseases/epidemiology , Kidney Diseases/genetics , Autoantibodies , Complement Factor H/genetics , Hereditary Complement Deficiency Diseases , Humans , Odds Ratio , Risk , Sequence Deletion
17.
Pediatr Nephrol ; 31(12): 2375-2378, 2016 12.
Article in English | MEDLINE | ID: mdl-27744619

ABSTRACT

BACKGROUND: Rational options for the treatment of end-stage renal disease (ESRD) due to atypical hemolytic uremic syndrome (aHUS) in children are still open to discussion. In the case of human complement factor H (CFH) deficiency, the choice is either kidney transplantation in combination with eculizumab, a humanized anti-C5 monoclonal antibody, or a combined liver-kidney transplantation. CASE-DIAGNOSIS/TREATMENT: A child with a homozygous CFH deficiency underwent a successful liver-kidney transplantation. CFH levels normalized within days. After 6 years of follow-up, the graft function (Cockroft clearance 100 ml min-1 1.73 m-2) and the liver functions were normal. RESULTS AND CONCLUSIONS: The results of this long-term follow-up confirm that combined liver-kidney transplantation remains a reasonable option in patients with ESRD due to aHUS when an identified genetic abnormality of the C3 convertase regulator synthesized in the liver has been identified.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/surgery , Complement Factor H/deficiency , Kidney Diseases/genetics , Kidney Transplantation/methods , Liver Transplantation/methods , Atypical Hemolytic Uremic Syndrome/etiology , Complement Factor H/genetics , Consanguinity , Hereditary Complement Deficiency Diseases , Humans , Infant , Kidney Diseases/complications , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery , Male , Plasma Exchange , Treatment Outcome
18.
Sci Rep ; 6: 28445, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27345360

ABSTRACT

Podocyte loss is the initial event in the development of glomerulosclerosis, the structural hallmark of progressive proteinuric nephropathies. Understanding mechanisms underlying glomerular injury is the key challenge for identifying novel therapeutic targets. In mice with protein-overload induced by bovine serum albumin (BSA), we evaluated whether the alternative pathway (AP) of complement mediated podocyte depletion and podocyte-dependent parietal epithelial cell (PEC) activation causing glomerulosclerosis. Factor H (Cfh(-/-)) or factor B-deficient mice were studied in comparison with wild-type (WT) littermates. WT+BSA mice showed podocyte depletion accompanied by glomerular complement C3 and C3a deposits, PEC migration to capillary tuft, proliferation, and glomerulosclerosis. These changes were more prominent in Cfh(-/-) +BSA mice. The pathogenic role of AP was documented by data that factor B deficiency preserved glomerular integrity. In protein-overload mice, PEC dysregulation was associated with upregulation of CXCR4 and GDNF/c-Ret axis. In vitro studies provided additional evidence of a direct action of C3a on proliferation and CXCR4-related migration of PECs. These effects were enhanced by podocyte-derived GDNF. In patients with proteinuric nephropathy, glomerular C3/C3a paralleled PEC activation, CXCR4 and GDNF upregulation. These results indicate that mechanistically uncontrolled AP complement activation is not dispensable for podocyte-dependent PEC activation resulting in glomerulosclerosis.


Subject(s)
Complement C3a/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Adult , Animals , Cattle , Cell Proliferation , Cells, Cultured , Complement Factor B/deficiency , Complement Factor B/genetics , Complement Factor H/deficiency , Complement Factor H/genetics , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Kidney/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Podocytes/cytology , Podocytes/metabolism , Proteinuria/etiology , Serum Albumin, Bovine/administration & dosage , Up-Regulation , Young Adult
19.
Exp Eye Res ; 147: 138-143, 2016 06.
Article in English | MEDLINE | ID: mdl-27181225

ABSTRACT

Extra-cellular deposition including amyloid beta (Aß) is a feature of retinal ageing. It has been documented for Bruch's membrane (BM) where Aß is elevated in complement factor H knockout mice (Cfh(-/-)) proposed as a model for age related macular degeneration. However, arterial deposition in choroidal vessels prior to perfusion across BM has not been examined. Aß is associated with tau phosphorylation and these are linked in blood vessels in Alzheimers Disease where they can drive perivascular pathology. Here we ask if Aß, tau and phosphorylated tau are features of ageing in choroidal vessels in 12 month C57 BL/6 and Cfh(-/-) mice, using immune staining and Western blot analysis. Greater levels of Aß and phosphorylated tau are found in choroidal vessels in Cfh(-/-) mice. Western blot revealed a 40% increase in Aß in Cfh(-/-) over C57 BL/6 mice. Aß deposits coat around 55% of the luminal wall in Cfh(-/-) compared to only about 40% in C57 BL/6. Total tau was similar in both groups, but phosphorylated tau increased by >100% in Cfh(-/-) compared to C57 BL/6 and covered >75% of the luminal wall compared to 50% in C57 BL/6. Hence, phosphorylated tau is a marked choroidal feature in this mouse model. Aß deposition was clumped in Cfh(-/-) mice and likely to influence blood flow dynamics. Disturbed flow is associated with atherogenesis and may be related to the accumulation of membrane attack complex recently identified between choroidal vessels in those at high risk of macular degeneration due to complement factor H polymorphisms.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood Vessels/metabolism , Choroid/metabolism , Complement Factor H/deficiency , Retinal Degeneration/metabolism , tau Proteins/metabolism , Aging/physiology , Animals , Blotting, Western , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Kidney Int ; 89(4): 823-32, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26924054

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.


Subject(s)
Complement C3/metabolism , Complement Factor H/deficiency , Glomerulonephritis/metabolism , Kidney Diseases/metabolism , Macrophage-1 Antigen/metabolism , Animals , CD11b Antigen/genetics , Complement Factor H/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Hereditary Complement Deficiency Diseases , Mice, Inbred C57BL , Myeloid Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...