Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Front Immunol ; 13: 931210, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091034

RESUMO

Atypical hemolytic uremic syndrome (aHUS) an important form of a thrombotic microangiopathy (TMA) that can frequently lead to acute kidney injury (AKI). An important subset of aHUS is the anti-factor H associated aHUS. This variant of aHUS can occur due to deletion of the complement factor H genes, CFHR1 and CFHR3, along with the presence of anti-factor H antibodies. However, it is a point of interest to note that not all patients with anti-factor H associated aHUS have a CFHR1/R3 deletion. Factor-H has a vital role in the regulation of the complement system, specifically the alternate pathway. Therefore, dysregulation of the complement system can lead to inflammatory or autoimmune diseases. Patients with this disease respond well to treatment with plasma exchange therapy along with Eculizumab and immunosuppressant therapy. Anti-factor H antibody associated aHUS has a certain genetic predilection therefore there is focus on further advancements in the diagnosis and management of this disease. In this article we discuss the baseline characteristics of patients with anti-factor H associated aHUS, their triggers, various treatment modalities and future perspectives.


Assuntos
Injúria Renal Aguda , Síndrome Hemolítico-Urêmica Atípica , Proteínas do Sistema Complemento , Injúria Renal Aguda/genética , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/terapia , Anticorpos/genética , Anticorpos/imunologia , Síndrome Hemolítico-Urêmica Atípica/complicações , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/imunologia , Síndrome Hemolítico-Urêmica Atípica/terapia , Proteínas Sanguíneas/genética , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/antagonistas & inibidores , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Humanos , Troca Plasmática
2.
Cells ; 10(12)2021 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-34944087

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a rare disorder characterized by dysregulation of the alternate pathway. The diagnosis of aHUS is one of exclusion, which complicates its early detection and corresponding intervention to mitigate its high rate of mortality and associated morbidity. Heterozygous mutations in complement regulatory proteins linked to aHUS are not always phenotypically active, and may require a particular trigger for the disease to manifest. This list of triggers continues to expand as more data is aggregated, particularly centered around COVID-19 and pediatric vaccinations. Novel genetic mutations continue to be identified though advancements in technology as well as greater access to cohorts of interest, as in diacylglycerol kinase epsilon (DGKE). DGKE mutations associated with aHUS are the first non-complement regulatory proteins associated with the disease, drastically changing the established framework. Additional markers that are less understood, but continue to be acknowledged, include the unique autoantibodies to complement factor H and complement factor I which are pathogenic drivers in aHUS. Interventional therapeutics have undergone the most advancements, as pharmacokinetic and pharmacodynamic properties are modified as needed in addition to their as biosimilar counterparts. As data continues to be gathered in this field, future advancements will optimally decrease the mortality and morbidity of this disease in children.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Fator H do Complemento/genética , Fator I do Complemento/genética , Diacilglicerol Quinase/genética , Mutação , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Autoanticorpos/imunologia , COVID-19/genética , COVID-19/imunologia , Vacinas contra COVID-19/efeitos adversos , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/uso terapêutico , Criança , Fator H do Complemento/imunologia , Fator I do Complemento/imunologia , Diacilglicerol Quinase/imunologia , Humanos , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Tratamento Farmacológico da COVID-19
3.
Front Immunol ; 12: 690821, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34177949

RESUMO

Complement factor B (FB) mutant variants are associated with excessive complement activation in kidney diseases such as atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy and membranoproliferative glomerulonephritis (MPGN). Patients with aHUS are currently treated with eculizumab while there is no specific treatment for other complement-mediated renal diseases. In this study the phenotype of three FB missense variants, detected in patients with aHUS (D371G and E601K) and MPGN (I242L), was investigated. Patient sera with the D371G and I242L mutations induced hemolysis of sheep erythrocytes. Mutagenesis was performed to study the effect of factor D (FD) inhibition on C3 convertase-induced FB cleavage, complement-mediated hemolysis, and the release of soluble C5b-9 from glomerular endothelial cells. The FD inhibitor danicopan abrogated C3 convertase-associated FB cleavage to the Bb fragment in patient serum, and of the FB constructs, D371G, E601K, I242L, the gain-of-function mutation D279G, and the wild-type construct, in FB-depleted serum. Furthermore, the FD-inhibitor blocked hemolysis induced by the D371G and D279G gain-of-function mutants. In FB-depleted serum the D371G and D279G mutants induced release of C5b-9 from glomerular endothelial cells that was reduced by the FD-inhibitor. These results suggest that FD inhibition can effectively block complement overactivation induced by FB gain-of-function mutations.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Ativação do Complemento , Fator B do Complemento/genética , Fator D do Complemento/antagonistas & inibidores , Glomerulonefrite Membranoproliferativa/imunologia , Animais , Síndrome Hemolítico-Urêmica Atípica/genética , Criança , Convertases de Complemento C3-C5/imunologia , Complemento C3b/imunologia , Fator B do Complemento/imunologia , Fator D do Complemento/imunologia , Células Endoteliais/imunologia , Eritrócitos , Feminino , Glomerulonefrite Membranoproliferativa/genética , Hemólise , Humanos , Lactente , Glomérulos Renais/citologia , Masculino , Pessoa de Meia-Idade , Mutação , Fenótipo , Coelhos , Ovinos
4.
Methods Mol Biol ; 2227: 1-20, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33847926

RESUMO

The complement cascade is an evolutionary ancient innate immune defense system, playing a major role in the defense against infections. Its function in maintaining host homeostasis on activated cells has been emphasized by the crucial role of its overactivation in ever growing number of diseases, such as atypical hemolytic uremic syndrome (aHUS), autoimmune diseases as systemic lupus erythematosus (SLE), C3 glomerulopathies (C3GN), age-related macular degeneration (AMD), graft rejection, Alzheimer disease, and cancer, to name just a few. The last decade of research on complement has extended its implication in many pathological processes, offering new insights to potential therapeutic targets and asserting the necessity of reliable, sensitive, specific, accurate, and reproducible biomarkers to decipher complement role in pathology. We need to evaluate accurately which pathway or role should be targeted pharmacologically, and optimize treatment efficacy versus toxicity. This chapter is an introduction to the role of complement in human diseases and the use of complement-related biomarkers in the clinical practice. It is a part of a book intending to give reliable and standardized methods to evaluate complement according to nowadays needs and knowledge.


Assuntos
Proteínas do Sistema Complemento/análise , Testes Diagnósticos de Rotina/métodos , Padrões de Prática Médica , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Ativação do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Desenvolvimento de Medicamentos , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Humanos , Nefropatias/diagnóstico , Nefropatias/imunologia , Nefropatias/metabolismo , Degeneração Macular/diagnóstico , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Neoplasias/diagnóstico , Neoplasias/imunologia , Neoplasias/metabolismo
5.
Methods Mol Biol ; 2227: 69-81, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33847932

RESUMO

Impairment of the complement regulatory protein Factor H (FH) is implicated in the physiopathological mechanisms of different diseases like atypical hemolytic and uremic syndrome and C3 glomerulopathies. It may be due to genetic abnormalities or acquired with the development of autoantibodies. FH has several ligands; therefore, the exploration of its functions requires to perform different tests. Among them, two hemolytic tests are very useful because they give specific and complementary information about FH functions. The first one is dedicated to explore the FH capacity to dissociate the alternative pathway C3 convertase, whereas the second one is designed to explore the capacity of FH to bind cell surfaces and to protect them from complement attack. This chapter describes the procedures to perform these two hemolytic tests, exploring in a complementary way the FH functionality.


Assuntos
Fator H do Complemento/análise , Fator H do Complemento/fisiologia , Ensaio de Atividade Hemolítica de Complemento/métodos , Animais , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C3b/análise , Complemento C3b/metabolismo , Citaferese/métodos , Eritrócitos/citologia , Eritrócitos/metabolismo , Humanos , Nefropatias/sangue , Nefropatias/diagnóstico , Nefropatias/imunologia , Ratos , Ovinos
6.
Methods Mol Biol ; 2227: 83-96, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33847933

RESUMO

The complement system is a key part of innate immunity. However, if the system becomes dysregulated, damage to healthy host cells can occur, especially to the glomerular cells of the kidney. The convertases of the alternative pathway of the complement system play a crucial role in complement activation. In healthy conditions, their activity is strictly regulated. In patients with diseases caused by complement alternative pathway dysregulation, such as C3 glomerulopathy and atypical hemolytic uremic syndrome, factors can be present in the blood that disturb this delicate balance, leading to convertase overactivity. Such factors include C3 nephritic factors, which are autoantibodies against the C3 convertase that prolong its activity, or genetic variants resulting in a stabilized convertase complex. This chapter describes a method in which the activity and stability of the alternative pathway convertases can be measured to detect aberrant serum factors causing convertase overactivity.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Ensaio de Atividade Hemolítica de Complemento/métodos , Via Alternativa do Complemento , Animais , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Ativação do Complemento , Complemento C3/imunologia , Fator Nefrítico do Complemento 3/análise , Fator Nefrítico do Complemento 3/imunologia , Convertases de Complemento C3-C5/análise , Via Alternativa do Complemento/imunologia , Glomerulonefrite/sangue , Glomerulonefrite/diagnóstico , Glomerulonefrite/imunologia , Cobaias , Humanos , Coelhos
7.
Methods Mol Biol ; 2227: 97-105, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33847934

RESUMO

The three pathways of the complement system converge toward the cleavage of the central complement component C3 into its activated fragments, with C3b being able to bind covalently to the activating surface. The endothelial cells are among the major targets for complement attack in pathological conditions, as the atypical hemolytic uremic syndrome. Therefore, study of complement C3 deposition on endothelial cells by flow cytometry is a sensitive test to measure complement activation. This test can be used as a research or clinical tool to test complement activation induced by patients' sera or to test the functional consequences of newly discovered complement mutations as well as different triggers of endothelial cells injury.


Assuntos
Complemento C3/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Citometria de Fluxo/métodos , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Células Cultivadas , Ativação do Complemento/fisiologia , Células Endoteliais/patologia , Humanos , Ligação Proteica
8.
Biomolecules ; 11(2)2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33671302

RESUMO

The complement system is part of the innate immune response, where it provides immediate protection from infectious agents and plays a fundamental role in homeostasis. Complement dysregulation occurs in several diseases, where the tightly regulated proteolytic cascade turns offensive. Prominent examples are atypical hemolytic uremic syndrome, paroxysmal nocturnal hemoglobinuria and Alzheimer's disease. Therapeutic intervention targeting complement activation may allow treatment of such debilitating diseases. In this review, we describe a panel of complement targeting nanobodies that allow modulation at different steps of the proteolytic cascade, from the activation of the C1 complex in the classical pathway to formation of the C5 convertase in the terminal pathway. Thorough structural and functional characterization has provided a deep mechanistic understanding of the mode of inhibition for each of the nanobodies. These complement specific nanobodies are novel powerful probes for basic research and offer new opportunities for in vivo complement modulation.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento , Nanomedicina/métodos , Anticorpos de Domínio Único/química , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C1/química , Convertases de Complemento C3-C5/química , Epitopos/química , Hemoglobinúria Paroxística/tratamento farmacológico , Hemoglobinúria Paroxística/imunologia , Humanos , Imunidade Inata , Imunoglobulina G/imunologia , Inflamação , Conformação Molecular , Ligação Proteica , Proteólise
9.
J Am Soc Nephrol ; 32(5): 1227-1235, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33712527

RESUMO

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS), a severe thrombotic microangiopathy, is often related to complement dysregulation, but the pathomechanisms remain unknown in at least 30% of patients. Researchers have described autoantibodies to complement factor H of the IgG class in 10% of patients with aHUS but have not reported anti-factor H autoantibodies of the IgM class. METHODS: In 186 patients with thrombotic microangiopathy clinically presented as aHUS, we searched for anti-factor H autoantibodies of the IgM class and those of the IgG and IgA classes. We used immunochromatography to purify anti-factor H IgM autoantibodies and immunoenzymatic methods and a competition assay with mapping mAbs to characterize interaction with the target protein. RESULTS: We detected anti-factor H autoantibodies of the IgM class in seven of 186 (3.8%) patients with thrombotic microangiopathy presented as aHUS. No association was observed between anti-factor H IgM and homozygous deletions involving CFHR3-CFHR1. A significantly higher proportion of patients with bone marrow transplant-related thrombotic microangiopathy had anti-factor H IgM autoantibodies versus other patients with aHUS: three of 20 (15%) versus four of 166 (2.4%), respectively. The identified IgM autoantibodies recognize the SCR domain 19 of factor H molecule in all patients and interact with the factor H molecule, inhibiting its binding to C3b. CONCLUSIONS: Detectable autoantibodies to factor H of the IgM class may be present in patients with aHUS, and their frequency is six-fold higher in thrombotic microangiopathy forms associated with bone marrow transplant. The autoantibody interaction with factor H's active site may support an autoimmune mechanism in some cases previously considered to be of unknown origin.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/sangue , Autoanticorpos/sangue , Fator H do Complemento/imunologia , Imunoglobulina M/imunologia , Adolescente , Adulto , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
11.
Int Urol Nephrol ; 53(10): 2067-2080, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33389509

RESUMO

The complement system plays a significant role within the pathological process of C3 glomerulopathy (C3GP) and atypical hemolytic uremic syndrome (aHUS). In daily practice, clinicians should differentiate the subgroups of C3GP because of they should apply different treatment modalities. In the past, C3GP was considered as a part of membranoproliferative glomerulonephritis (MPGN). MPGN is defined as glomerular capillary thickening secondary to the synthesis of the new glomerular basement membrane and mesangial cellular hyperplasia with mesangial matrix expansion. Atypical hemolytic uremic syndrome is an ultra-rare disease that can be outlined by the triad of Coombs negative microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. Recent advances demonstrated that these diseases share common abnormalities of the control of the alternative complement system. Therefore, nowadays, most researchers advocate that there may be overlap in the pathogenesis of C3GP and aHUS. This review will provide recent novel mechanisms and treatment options in these diseases. For the purposes that we mentioned above and to help clinicians, we aimed to describe the etiology, pathophysiology, and treatment of C3GP and aHUS in this comprehensive review.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C3 , Via Alternativa do Complemento , Nefropatias/imunologia , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/terapia , Humanos , Nefropatias/diagnóstico , Nefropatias/terapia
12.
CEN Case Rep ; 10(2): 255-260, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33386505

RESUMO

Hemolytic uremic syndrome (HUS), a cause of pediatric acute kidney injury (AKI), has a spectrum of extra-renal manifestations. While neurological and gastrointestinal system involvement is common, cardiac involvement is rare. This is more so with pericardial involvement, though it has been reported in a handful of HUS cases associated with shiga toxin-producing Escherichia coli (STEC HUS). However, this complication has scarcely been reported in atypical HUS (aHUS) where there is alternate complement abnormality or DKGE (diacylglycerol kinase epsilon) mutation. We describe two children diagnosed with anti-complement factor H (CFH) antibody-associated aHUS who had pericardial involvement. Two boys, one 10-year-old and another 8-year-old, presented with pallor, oliguria and hypertension. They both had microangiopathic haemolytic anemia, thrombocytopenia and AKI suggestive of HUS. Complement workup revealed elevated anti-CFH antibody titres. With a diagnosis of anti-CFH antibody aHUS, they were started on plasmapheresis, pulse methylprednisolone and cyclophosphamide. The first case developed cardiac tamponade during the second week of hospital stay for which he needed pigtail drainage and further immunosuppression with rituximab. He gradually improved and pigtail was removed. The second case presented with pericardial effusion which subsequently resolved during the course of treatment. Thus, our patients developed pericardial effusion, with one of them progressing to life-threatening cardiac tamponade. Therefore, it is prudent that we are aware of this complication while treating children with aHUS.


Assuntos
Anticorpos Anti-Idiotípicos/sangue , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Fator H do Complemento/imunologia , Derrame Pericárdico/complicações , Síndrome Hemolítico-Urêmica Atípica/imunologia , Criança , Humanos , Masculino
14.
Front Immunol ; 12: 789897, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35069568

RESUMO

Complement factor I (FI) is a central inhibitor of the complement system, and impaired FI function increases complement activation, contributing to diseases such as age-related macular degeneration (AMD) and atypical hemolytic uremic syndrome (aHUS). Genetic variation in complement factor I (CFI) has been identified in both AMD and aHUS, with more than half of these variants leading to reduced FI secretion levels. For many of the variants with normal FI secretion, however, functional implications are not yet known. Here we studied 11 rare missense variants, with FI secretion levels comparable to wildtype, but a predicted damaging effects based on the Combined Annotation Dependent Depletion (CADD) score. Three variants (p.Pro50Ala, p.Arg339Gln, and p.Ser570Thr) were analyzed in plasma and serum samples of carriers affected by AMD. All 11 variants (nine for the first time in this study) were recombinantly expressed and the ability to degrade C3b was studied with the C3b degradation assay. The amount of degradation was determined by measuring the degradation product iC3b with ELISA. Eight of 11 (73%) mutant proteins (p.Pro50Ala, p.Arg339Gln, p.Ile340Thr, p.Gly342Glu, p.Gly349Arg, p.Arg474Gln, p.Gly487Cys, and p.Gly512Ser) showed significantly impaired C3b degradation, and were therefore classified as likely pathogenic. Our data indicate that genetic variants in CFI with a CADD score >20 are likely to affect FI function, and that monitoring iC3b in a degradation assay is a useful tool to establish the pathogenicity of CFI variants in functional studies.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Fator I do Complemento , Degeneração Macular , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C3b/imunologia , Complemento C3b/metabolismo , Fator I do Complemento/genética , Fator I do Complemento/imunologia , Fator I do Complemento/metabolismo , Feminino , Humanos , Degeneração Macular/sangue , Degeneração Macular/genética , Degeneração Macular/imunologia , Masculino
16.
Clin Immunol ; 221: 108616, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33148511

RESUMO

In complement-driven thrombotic microangiopathies, failure to regulate complement activation leads to end-organ damage. The modified Ham (mHam) test measures complement-mediated killing of a nucleated cell in vitro but lacks a confirmatory assay and reliable positive controls. We demonstrate that C5b-9 accumulation on the surface of TF1 PIGAnull cells correlates with cell killing in the mHam. We also show that Sialidase treatment of cells or addition of Shiga toxin 1 to human serum serve as a more reliable positive control for the mHam than cobra venom factor or lipopolysaccharide. Simultaneously performing the mHam and measuring C5b-9 accumulation either in GVB++ or GVB0 MgEGTA buffer with the addition of complement pathway specific inhibitors (anti-C5 antibody or a factor D inhibitor, ACH-145951) can be used to localize defects in complement regulation. As more targeted complement inhibitors become available, these assays may aid in the selection of personalized treatments for patients with complement-mediated diseases.


Assuntos
Síndrome Antifosfolipídica/imunologia , Síndrome Hemolítico-Urêmica Atípica/imunologia , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/farmacologia , Adulto , Bioensaio , Linhagem Celular Tumoral , Complemento C3c/imunologia , Complemento C4b/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Venenos Elapídicos/farmacologia , Feminino , Humanos , Lipopolissacarídeos/farmacologia , Masculino , Pessoa de Meia-Idade , Neuraminidase/farmacologia , Fragmentos de Peptídeos/imunologia , Toxina Shiga I/farmacologia
17.
Mol Immunol ; 128: 175-187, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33137606

RESUMO

It has long been known that the complement cascade is activated in various forms of glomerulonephritis. In many of these diseases, immune-complexes deposit in the glomeruli and activate the classical pathway. Researchers have also identified additional mechanisms by which complement is activated in the kidney, including diseases in which the alternative and lectin pathways are activated. The kidney appears to be particularly susceptible to activation of the alternative pathway, and this pathway has been implicated as a primary driver of atypical hemolytic uremic syndrome, C3 glomerulopathy, anti-neutrophil cytoplasmic antibody-associated vasculitis, as well as some forms of immune-complex glomerulonephritis. In this paper we review the shared and distinct mechanisms by which complement is activated in these different diseases. We also review the opportunities for using therapeutic complement inhibitors to treat kidney diseases.


Assuntos
Proteínas do Sistema Complemento/imunologia , Nefropatias/imunologia , Animais , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/imunologia , Complexo Antígeno-Anticorpo/imunologia , Síndrome Hemolítico-Urêmica Atípica/imunologia , Ativação do Complemento/imunologia , Inativadores do Complemento/imunologia , Glomerulonefrite/imunologia , Humanos , Rim/imunologia
18.
Immunobiology ; 225(5): 152000, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32962820

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is caused mainly by complement dysregulation. Although various defects in the complement system explaining pathophysiology have been described in recent years, the etiology still remains unclear in about thirty percent of cases. In exploring other causes, similar to anti- complement factor H (anti-CFH) antibody associated HUS, we hypothesized that anti-complement factor I (anti-CFI) antibody could play a role in aHUS. Further, we tried to describe the clinical profile and outcome of those with high anti CFI antibody titers. Eleven of thirty five children (31 %) diagnosed with aHUS from July 2017 to December 2018 had high IgG anti-CFI antibody titers. Median age was 10 months (6, 33) with no sex difference. Thirty-six percent (4/11) had nephrotic-range proteinuria. C3 was low in 8 children (72.7 %) with mean C3 (68.1 ±â€¯14.7 mg/dL). Plasmapheresis was done in 2 children who promptly responded, suggesting the possible role of anti-CFI antibody in pathogenesis of aHUS in these patients. Further studies examining role of anti-CFI antibodies in aHUS is warranted with longitudinal and genetic studies.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Fator I do Complemento/imunologia , Imunoglobulina G/sangue , Síndrome Hemolítico-Urêmica Atípica/sangue , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino
19.
J Thromb Haemost ; 18(9): 2110-2117, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32608159

RESUMO

COVID-19 is frequently accompanied by a hypercoagulable inflammatory state with microangiopathic pulmonary changes that can precede the diffuse alveolar damage characteristic of typical acute respiratory distress syndrome (ARDS) seen in other severe pathogenic infections. Parallels with systemic inflammatory disorders such as atypical hemolytic uremic syndrome (aHUS) have implicated the complement pathway in the pathogenesis of COVID-19, and particularly the anaphylatoxins C3a and C5a released from cleavage of C3 and C5, respectively. C5a is a potent cell signalling protein that activates a cytokine storm-a hyper-inflammatory phenomenon-within hours of infection and the innate immune response. However, excess C5a can result in a pro-inflammatory environment orchestrated through a plethora of mechanisms that propagate lung injury, lymphocyte exhaustion, and an immune paresis. Furthermore, disruption of the homeostatic interactions between complement and extrinsic and intrinsic coagulation pathways contributes to a net pro-coagulant state in the microvasculature of critical organs. Fatal COVID-19 has been associated with a systemic inflammatory response accompanied by a pro-coagulant state and organ damage, particularly microvascular thrombi in the lungs and kidneys. Pathologic studies report strong evidence of complement activation. C5 blockade reduces inflammatory cytokines and their manifestations in animal studies, and has shown benefits in patients with aHUS, prompting investigation of this approach in the treatment of COVID-19. This review describes the role of the complement pathway and particularly C5a and its aberrations in highly pathogenic virus infections, and therefore its potential as a therapeutic target in COVID-19.


Assuntos
Coagulação Sanguínea , COVID-19/imunologia , Ativação do Complemento , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Inflamação/metabolismo , Animais , Síndrome Hemolítico-Urêmica Atípica/complicações , Síndrome Hemolítico-Urêmica Atípica/imunologia , COVID-19/complicações , COVID-19/patologia , Inativadores do Complemento/farmacologia , Citocinas/metabolismo , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/imunologia , Homeostase , Humanos , Imunidade Inata , Pneumopatias , Lesão Pulmonar , Transdução de Sinais
20.
Adv Chronic Kidney Dis ; 27(2): 120-127.e4, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32553244

RESUMO

Atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G) are associated with loss of regulation of the alternative pathway of complement and its resulting overactivation. As rare diseases, genetic variants leading to aHUS and C3G were previously analysed in relatively low patient numbers. To improve this analysis, data were pooled from six centres. Totals of 610 rare variants for aHUS and 82 for C3G were presented in an interactive database for 13 genes. Using allele frequency comparisons with the Exome Aggregation Consortium as a reference genome, the patients with aHUS showed significantly more protein-altering ultrarare variants (allele frequency <0.01%) in five genes CFH, CFI, CD46, C3, and DGKE. In patients with C3G, the corresponding association was only found for C3 and CFH. Protein structure analyses of these five proteins showed distinct differences in the positioning of these variants in C3 and FH. For aHUS, variants were clustered at the C-terminus of FH and implicated changes in the binding of FH to host cell surfaces. For C3G, variants were clustered at the N-terminal C3b binding site of FH and implicated changes in the fluid-phase regulation of C3b. We discuss the utility of the Web database as a patient resource for clinicians.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Complemento C3/genética , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/imunologia , Síndrome Hemolítico-Urêmica Atípica/fisiopatologia , Frequência do Gene , Predisposição Genética para Doença , Doenças da Deficiência Hereditária de Complemento/diagnóstico , Doenças da Deficiência Hereditária de Complemento/imunologia , Humanos , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...