Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33859044

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of blindness among the elderly. Canonical disease models suggest that defective interactions between complement factor H (CFH) and cell surface heparan sulfate (HS) result in increased alternative complement pathway activity, cytolytic damage, and tissue inflammation in the retina. Although these factors are thought to contribute to increased disease risk, multiple studies indicate that noncanonical mechanisms that result from defective CFH and HS interaction may contribute to the progression of AMD as well. A total of 60 ciliated sensory neurons in the nematode Caenorhabditis elegans detect chemical, olfactory, mechanical, and thermal cues in the environment. Here, we find that a C. elegans CFH homolog localizes on CEP mechanosensory neuron cilia where it has noncanonical roles in maintaining inversin/NPHP-2 within its namesake proximal compartment and preventing inversin/NPHP-2 accumulation in distal cilia compartments in aging adults. CFH localization and maintenance of inversin/NPHP-2 compartment integrity depend on the HS 3-O sulfotransferase HST-3.1 and the transmembrane proteoglycan syndecan/SDN-1. Defective inversin/NPHP-2 localization in mouse and human photoreceptors with CFH mutations indicates that these functions and interactions may be conserved in vertebrate sensory neurons, suggesting that previously unappreciated defects in cilia structure may contribute to the progressive photoreceptor dysfunction associated with CFH loss-of-function mutations in some AMD patients.


Asunto(s)
Factor H de Complemento/metabolismo , Heparitina Sulfato/metabolismo , Retina/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Cilios/metabolismo , Factor H de Complemento/fisiología , Heparitina Sulfato/fisiología , Degeneración Macular/metabolismo , Degeneración Macular/fisiopatología , Neuronas/metabolismo , Factores de Transcripción/metabolismo
2.
Methods Mol Biol ; 2227: 69-81, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33847932

RESUMEN

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.


Asunto(s)
Factor H de Complemento/análisis , Factor H de Complemento/fisiología , Ensayo de Actividad Hemolítica de Complemento/métodos , Animales , Síndrome Hemolítico Urémico Atípico/sangre , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/inmunología , Complemento C3b/análisis , Complemento C3b/metabolismo , Citaféresis/métodos , Eritrocitos/citología , Eritrocitos/metabolismo , Humanos , Enfermedades Renales/sangre , Enfermedades Renales/diagnóstico , Enfermedades Renales/inmunología , Ratas , Ovinos
3.
Clin J Am Soc Nephrol ; 16(6): 942-956, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33536243

RESUMEN

In the past 20 years, we have witnessed tremendous advances in our ability to diagnose and treat genetic diseases of the kidney caused by complement dysregulation. Staggering progress was realized toward a better understanding of the genetic underpinnings and pathophysiology of many forms of atypical hemolytic uremic syndrome (aHUS) and C3-dominant glomerulopathies that are driven by complement system abnormalities. Many of these seminal discoveries paved the way for the design and characterization of several innovative therapies, some of which have already radically improved patients' outcomes. This review offers a broad overview of the exciting developments that have occurred in the recent past, with a particular focus on single-gene (or Mendelian), complement-driven aHUS and C3-dominant glomerulopathies that should be of interest to both nephrologists and kidney researchers. The discussion is restricted to genes with robust associations with both aHUS and C3-dominant glomerulopathies (complement factor H, complement component 3, complement factor H-related proteins) or only aHUS (complement factor B, complement factor I, and membrane cofactor protein). Key questions and challenges are highlighted, along with potential avenues for future directions.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Complemento C3/genética , Factor B del Complemento/genética , Enfermedades Renales/genética , Complemento C3/fisiología , Factor B del Complemento/fisiología , Factor H de Complemento/genética , Factor H de Complemento/fisiología , Humanos
4.
J Am Soc Nephrol ; 31(2): 241-256, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31980588

RESUMEN

Sequence and copy number variations in the human CFHR-Factor H gene cluster comprising the complement genes CFHR1, CFHR2, CFHR3, CFHR4, CFHR5, and Factor H are linked to the human kidney diseases atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy. Distinct genetic and chromosomal alterations, deletions, or duplications generate hybrid or mutant CFHR genes, as well as hybrid CFHR-Factor H genes, and alter the FHR and Factor H plasma repertoire. A clear association between the genetic modifications and the pathologic outcome is emerging: CFHR1, CFHR3, and Factor H gene alterations combined with intact CFHR2, CFHR4, and CFHR5 genes are reported in atypical hemolytic uremic syndrome. But alterations in each of the five CFHR genes in the context of an intact Factor H gene are described in C3 glomerulopathy. These genetic modifications influence complement function and the interplay of the five FHR proteins with each other and with Factor H. Understanding how mutant or hybrid FHR proteins, Factor H::FHR hybrid proteins, and altered Factor H, FHR plasma profiles cause pathology is of high interest for diagnosis and therapy.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Complemento C3/análisis , Glomerulonefritis Membranoproliferativa/genética , Síndrome Hemolítico Urémico Atípico/etiología , Factor H de Complemento/química , Factor H de Complemento/genética , Factor H de Complemento/fisiología , Variaciones en el Número de Copia de ADN , Predisposición Genética a la Enfermedad , Variación Genética , Glomerulonefritis Membranoproliferativa/etiología , Humanos , Riñón/patología , Familia de Multigenes
6.
J Thromb Thrombolysis ; 48(1): 95-102, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30915671

RESUMEN

Coagulation factor XIII (FXIII) covalently crosslinks pre-formed fibrin clots preventing their premature fibrinolysis. In plasma, FXIII circulates as a zymogenic heterotetramer composed of catalytic FXIII-A subunits, and carrier/regulatory FXIII-B subunits. FXIII-A is a well characterized component of this complex, and has been associated with several pleiotropic roles outside coagulation as well. In comparison only protective/regulatory roles towards the FXIII-A subunit have been identified for FXIII-B. Strong homology between FXIII-B and complement regulator Complement factor H suggests a putative role of FXIII-B in complement activation. In the current study we have analyzed the similarities and yet functional divergence of these two proteins using in silico sequence alignment and structural analysis. We have evaluated complement activation post reconstitution of FXIII components into FXIII deficient and CFH deficient plasma. We have also transiently expressed FXIII-B in SH-SY5Y cell lines and evaluated its effect on the endogenous complement activation. Our investigations show no effect of FXIII-B subunit on the rate of complement activation. Therefore we conclude that at a physiological level, FXIII-B subunit plays no role in the complement system, although a vestigial function in altered pathological states might still exist.


Asunto(s)
Activación de Complemento , Factor XIII/química , Recolección de Muestras de Sangre , Línea Celular , Factor H de Complemento/química , Factor H de Complemento/fisiología , Simulación por Computador , Factor XIII/fisiología , Humanos , Estructura Molecular , Dominios Proteicos , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Homología Estructural de Proteína , Transfección
7.
Behav Brain Res ; 364: 233-244, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30731099

RESUMEN

BACKGROUND: At present, the harm of new-type drug, methamphetamine (METH), has gradually exceeded that of the traditional opioid drugs, and METH abuse has become a serious public health and social problem. In our previous study, complement factor H (CFH) was found to be upregulated in the sera of METH-addicted patients and rats and in certain brain regions in the rats. METHODS: We used ELISA and immunofluorescence to confirm the changes in CFH in the serum and hippocampus of a METH behavioral sensitization mouse model, and C1q expression was also detected by immunofluorescence in the hippocampus. We aimed to elucidate the involvement of CFH and C1q in the mechanism of METH addiction. We also detected the distribution of various small molecules by matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) in select brain regions: the nucleus accumbens, the hippocampus and the ventral tegmental area. RESULTS: The expression of CFH was upregulated in the serum and hippocampus of METH behavioral sensitization model mice, consistent with our previous research on conditioned place preference rats. In contrast, C1q decreased dramatically in the mossy fibers of the hippocampus. The results of small-molecule imaging by MALDI-MSI showed that the levels of K+, antioxidants, neurotransmitters, and ATP metabolism-related molecules were altered in different regions. CONCLUSIONS: These results indicate the involvement of the complement system in the mechanism of METH addiction and validate the presence of oxidative stress, energy metabolism changes during addiction. This suggests the utility of further investigation into the above aspects.


Asunto(s)
Trastornos Relacionados con Anfetaminas/metabolismo , Conducta Animal/efectos de los fármacos , Factor H de Complemento/metabolismo , Animales , Conducta Adictiva/metabolismo , Encéfalo/metabolismo , Complemento C1q/metabolismo , Factor H de Complemento/fisiología , Condicionamiento Clásico/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Aprendizaje/efectos de los fármacos , Masculino , Metanfetamina/efectos adversos , Metanfetamina/metabolismo , Metanfetamina/farmacología , Ratones , Ratones Endogámicos C57BL , Imagen Molecular/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
8.
Am J Pathol ; 189(4): 826-838, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30711487

RESUMEN

Single-nucleotide polymorphisms and rare mutations in factor H (FH; official name, CFH) are associated with age-related macular degeneration and atypical hemolytic uremic syndrome, a form of thrombotic microangiopathy. Mice with the FH W1206R mutation (FHR/R) share features with human atypical hemolytic uremic syndrome. Herein, we report that FHR/R mice exhibited retinal vascular occlusion and ischemia. Retinal fluorescein angiography demonstrated delayed perfusion and vascular leakage in FHR/R mice. Optical coherence tomography imaging of FHR/R mice showed retinal degeneration, edema, and detachment. Histologic analysis of FHR/R mice revealed retinal thinning, vessel occlusion, as well as degeneration of photoreceptors and retinal pigment epithelium. Immunofluorescence showed albumin leakage from blood vessels into the neural retina, and electron microscopy demonstrated vascular endothelial cell irregularity with narrowing of retinal and choroidal vessels. Knockout of C6, a component of the membrane attack complex, prevented the aforementioned retinal phenotype in FHR/R mice, consistent with membrane attack complex-mediated pathogenesis. Pharmacologic blockade of C5 also rescued retinas of FHR/R mice. This FHR/R mouse strain represents a model for retinal vascular occlusive disorders and ischemic retinopathy. The results suggest complement dysregulation can contribute to retinal vascular occlusion and that an anti-C5 antibody might be helpful for C5-mediated thrombotic retinal diseases.


Asunto(s)
Factor H de Complemento/fisiología , Isquemia/etiología , Mutación , Neovascularización Patológica/etiología , Enfermedades de la Retina/etiología , Epitelio Pigmentado de la Retina/patología , Trombosis/etiología , Animales , Factor H de Complemento/genética , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Epitelio Pigmentado de la Retina/metabolismo , Trombosis/metabolismo , Trombosis/patología
9.
Prog Retin Eye Res ; 62: 38-57, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28928087

RESUMEN

Age-Related Macular Degeneration (AMD) is a complex multifactorial disease characterized in its early stages by lipoprotein accumulations in Bruch's Membrane (BrM), seen on fundoscopic exam as drusen, and in its late forms by neovascularization ("wet") or geographic atrophy of the Retinal Pigmented Epithelial (RPE) cell layer ("dry"). Genetic studies have strongly supported a relationship between the alternative complement cascade, in particular the common H402 variant in Complement Factor H (CFH) and development of AMD. However, the functional significance of the CFH Y402H polymorphism remains elusive. In this article, we critically review the literature surrounding the functional significance of this polymorphism. Furthermore, based on our group's studies we propose a model in which CFH H402 affects CFH binding to heparan sulfate proteoglycans leading to accelerated lipoprotein accumulation in BrM and drusen progression. We also review the literature on the role of other complement components in AMD pathobiologies, including C3a, C5a and the membrane attack complex (MAC), and on transgenic mouse models developed to interrogate in vivo the effects of the CFH Y402H polymorphism.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Degeneración Macular/genética , Polimorfismo de Nucleótido Simple , Factor H de Complemento/genética , Factor H de Complemento/fisiología , Estudios de Asociación Genética , Humanos , Degeneración Macular/patología , Drusas Retinianas/metabolismo , Drusas Retinianas/patología
10.
Invest Ophthalmol Vis Sci ; 56(6): 3427-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26030099

RESUMEN

PURPOSE: Variants of complement factor H (Cfh) affecting short consensus repeats (SCRs) 6 to 8 increase the risk of age-related macular degeneration. Our aim was to explore the effect of expressing a Cfh variant on the in vivo susceptibility of the retina and RPE to oxidative stress and inflammation, using chimeric Cfh transgenic mice (chCfhTg). METHODS: The chCfhTg and age-matched C57BL/6J (B6) mice were subjected to oxidative stress by either normal aging, or by exposure to a combination of oral hydroquinone (0.8% HQ) and increased light. Eyes were collected for immunohistochemistry of RPE-choroid flat mounts and of retinal sections, ELISA, electron microscopy, and RPE/microglia gene expression analysis. RESULTS: Aging mice to 2 years led to an increased accumulation of basal laminar deposits, subretinal microglia/macrophages (MG/MΦ) staining for CD16 and for malondialdehyde (MDA), and MDA-modified proteins in the retina in chCfhTg compared to B6 mice. The chCfhTg mice maintained on HQ diet and increased light showed greater deposition of basal laminar deposits, more accumulation of fundus spots suggestive of MG/MΦ, and increased deposition of C3d in the sub-RPE space, compared to controls. In addition, chCfhTg mice demonstrated upregulation of NLRP3, IP-10, CD68, and TREM-2 in the RNA isolates from RPE/MG/MΦ. CONCLUSIONS: Expression of a Cfh transgene introducing a variant in SCRs 6 to 8 was sufficient to lead to increased retinal/RPE susceptibility to oxidative stress, a proinflammatory MG/MΦ phenotype, and a proinflammatory RPE/MG/MΦ gene expression profile in a transgenic mouse model. Our data suggest that altered interactions of Cfh with MDA-modified proteins may be relevant in explaining the effects of the Cfh variant.


Asunto(s)
Factor H de Complemento/genética , Microglía/citología , Estrés Oxidativo/genética , Retina/metabolismo , Envejecimiento/fisiología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Proteínas Portadoras/metabolismo , Quimiocina CXCL10/metabolismo , Factor H de Complemento/fisiología , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/fisiopatología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR , Estrés Oxidativo/fisiología , Receptores Inmunológicos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
11.
Proc Natl Acad Sci U S A ; 112(23): E3040-9, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-25991857

RESUMEN

Complement factor H (CFH) is a major susceptibility gene for age-related macular degeneration (AMD); however, its impact on AMD pathobiology is unresolved. Here, the role of CFH in the development of AMD pathology in vivo was interrogated by analyzing aged Cfh(+/-) and Cfh(-/-) mice fed a high-fat, cholesterol-enriched diet. Strikingly, decreased levels of CFH led to increased sub-retinal pigmented epithelium (sub-RPE) deposit formation, specifically basal laminar deposits, following high-fat diet. Mechanistically, our data show that deposits are due to CFH competition for lipoprotein binding sites in Bruch's membrane. Interestingly and despite sub-RPE deposit formation occurring in both Cfh(+/-) and Cfh(-/-) mice, RPE damage accompanied by loss of vision occurred only in old Cfh(+/-) mice. We demonstrate that such pathology is a function of excess complement activation in Cfh(+/-) mice versus complement deficiency in Cfh(-/-) animals. Due to the CFH-dependent increase in sub-RPE deposit height, we interrogated the potential of CFH as a previously unidentified regulator of Bruch's membrane lipoprotein binding and show, using human Bruch's membrane explants, that CFH removes endogenous human lipoproteins in aged donors. Thus, advanced age, high-fat diet, and decreased CFH induce sub-RPE deposit formation leading to complement activation, which contributes to RPE damage and visual function impairment. This new understanding of the complicated interactions of CFH in AMD-like pathology provides an improved foundation for the development of targeted therapies for AMD.


Asunto(s)
Factor H de Complemento/fisiología , Degeneración Macular/fisiopatología , Animales , Coroides/metabolismo , Coroides/patología , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Dieta Alta en Grasa , Degeneración Macular/patología , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
12.
Semin Thromb Hemost ; 40(5): 544-50, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24967890

RESUMEN

Molecular linkages between von Willebrand factor (VWF) and the alternative complement pathway (AP) have recently been discovered. Endothelial cell (EC)-anchored ultra-large (UL) VWF multimeric strings function as an activating surface for the AP. C3 (in active C3b form) binds to the EC-anchored ULVWF strings, and promotes the assembly of C3bBb (C3 convertase) and C3bBbC3b (C5 convertase). These linkages may help to explain enigmatic clinical problems related to thrombotic microangiopathies, including some cases of refractory thrombotic thrombocytopenic purpura (TTP), TTP associated with only mild-modest deficiencies of ADAMTS-13, the provocation (or exacerbation) of acute episodes in patients with the atypical hemolytic uremic syndrome, and thrombosis in paroxysmal nocturnal hemoglobinuria. Recent experiments have also demonstrated that complement factor H performs a dual role: participating in regulation of the AP by binding to EC-anchored ULVWF strings; and functioning as a reductase to decrease the size of soluble VWF multimers.


Asunto(s)
Vía Alternativa del Complemento/fisiología , Microangiopatías Trombóticas/fisiopatología , Factor de von Willebrand/fisiología , Síndrome Hemolítico Urémico Atípico/fisiopatología , Factor H de Complemento/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Hemoglobinuria Paroxística/fisiopatología , Humanos , Púrpura Trombocitopénica Trombótica/fisiopatología , Trombosis/fisiopatología , Microangiopatías Trombóticas/genética
13.
Semin Thromb Hemost ; 40(4): 503-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24799304

RESUMEN

Hemolytic uremic syndrome (HUS), the most common cause of acute renal failure in childhood, is mainly caused by infection with Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC). Besides its cytotoxic activity, Stx has been shown to interact with the complement system. Complement breakdown products have been found in serum of HUS patients suggesting complement activation and in vitro studies have demonstrated that Stx2 directly activates complement leading to formation of terminal complement complex. Furthermore, Stx2 has been found to bind to factor H (FH) resulting in a reduced cofactor activity on the cell surface. Binding of Stx2 has also been shown for other members of the FH family, namely FH-like protein 1 and FH-related protein 1. Both proteins also compete with FH for Stx binding, so that in the presence of FHR-1 less FH is bound to Stx and therefore more is available for endothelial cell protection. In addition, Stx2 has been demonstrated to downregulate the membrane-bound regulator CD59 on the surface of glomerular endothelial and tubulus epithelial cells on protein and at the mRNA level. In conclusion, Stx modulates complement regulator proteins leading to an impaired control and thus to enhanced complement activation. Its implication in the pathogenesis of EHEC-induced HUS in vivo and whether complement blockage might be a therapeutic option still has to be elucidated.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Escherichia coli Enterohemorrágica , Síndrome Hemolítico-Urémico/genética , Síndrome Hemolítico-Urémico/inmunología , Toxina Shiga II/química , Toxina Shiga/química , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD59/metabolismo , Membrana Celular/metabolismo , Activación de Complemento , Factor H de Complemento/fisiología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Glomérulos Renales/inmunología , Glomérulos Renales/metabolismo , Mutación
14.
Mol Immunol ; 59(2): 188-93, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24632373

RESUMEN

Anti-phospholipid syndrome (APS) is a complex autoimmune disease, associated with recurrent venous and arterial thrombosis in various tissues. APS is associated with specific antibodies against plasma beta-2 glycoprotein 1 (ß2-GP1), and these antibodies react with ß2-GP1 bound to negatively charged phospholipids (e.g. cardiolipin) on cell membranes. Some APS patients also have autoantibodies to complement factor H (FH), a homologue of ß2-GP1, which also binds to anionic phospholipids. ß2-GP1 has earlier been shown to inhibit the intrinsic (contact) activated blood coagulation pathway, promoted by anionic phospholipids. Here we examine whether FH could have similar anti-thrombotic properties. In vitro experiments with surface-bound phospholipids and human plasma, in the presence of FH, confirm this hitherto unreported property of FH.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Coagulación Sanguínea/fisiología , beta 2 Glicoproteína I/inmunología , Anticoagulantes/farmacología , Síndrome Antifosfolípido/inmunología , Autoanticuerpos/inmunología , Coagulación Sanguínea/efectos de los fármacos , Cardiolipinas/inmunología , Factor H de Complemento/inmunología , Factor H de Complemento/farmacología , Factor H de Complemento/fisiología , Factor XII/antagonistas & inhibidores , Factor XII/metabolismo , Humanos , Ácidos Fosfatidicos/inmunología , Fosfatidilserinas/inmunología , Fosfolípidos , Trombosis/inmunología
15.
Semin Nephrol ; 33(6): 575-85, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24161042

RESUMEN

Activation of the complement system is tightly regulated by plasma and cell-associated complement regulatory proteins (CRPs), such as factor H (fH), decay-accelerating factor, and membrane cofactor protein. Animal models of disease have provided considerable insights into the important roles for CRPs in the kidney. Mice deficient in fH have excessive fluid phase C3 activation and inactivation, leading to deposition of inactivated C3b in glomerular capillary walls (GCW), comparable with dense deposit disease. In contrast, when fH lacks C-terminal surface targeting regions, local activation on the GCW leads to a disease reminiscent of thrombotic microangiopathy. The uniquely rodent protein, CR1-related y (Crry), has features analogous to human membrane cofactor protein. Defective Crry leads to unrestricted alternative pathway activation in the tubulointerstitium, resulting in pathologic features ranging from thrombotic microangiopathy (TMA), acute kidney injury, and tubulointerstitium nephritis. In the presence of initiators of the classic or lectin pathways, commonly in the form of immune complexes in human glomerular diseases, complement regulation is stressed, with the potential for recruitment of the spontaneously active alternative pathway. The threshold for this activation is set by CRPs; pathology is more likely when complement regulation is defective. Within the endocapillary region of the GCW, fH is key, while decay-accelerating factor and Crry are protective on mesangial cells and podocytes. Arguably, acquired alterations in these CRPs is a more common event, extending from pathologic states of cellular injury or production of inhibitory antibodies, to physiological fine tuning of the adaptive immune response.


Asunto(s)
Activación de Complemento/fisiología , Proteínas del Sistema Complemento/fisiología , Glomerulonefritis Membranosa/inmunología , Nefritis Lúpica/inmunología , Microangiopatías Trombóticas/inmunología , Animales , Factor H de Complemento/fisiología , Modelos Animales de Enfermedad , Humanos , Ratones
16.
J Am Soc Nephrol ; 24(11): 1820-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23907509

RESUMEN

Dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) are widely recognized subtypes of C3 glomerulopathy. These ultra-rare renal diseases are characterized by fluid-phase dysregulation of the alternative complement pathway that leads to deposition of complement proteins in the renal glomerulus. Disease triggers are unknown and because targeted treatments are lacking, progress to end stage renal failure is a common final outcome. We studied soluble CR1, a potent regulator of complement activity, to test whether it restores complement regulation in C3 glomerulopathy. In vitro studies using sera from patients with DDD showed that soluble CR1 prevents dysregulation of the alternative pathway C3 convertase, even in the presence of C3 nephritic factors. In mice deficient in complement factor H and transgenic for human CR1, soluble CR1 therapy stopped alternative pathway activation, resulting in normalization of serum C3 levels and clearance of iC3b from glomerular basement membranes. Short-term use of soluble CR1 in a pediatric patient with end stage renal failure demonstrated its safety and ability to normalize activity of the terminal complement pathway. Overall, these data indicate that soluble CR1 re-establishes regulation of the alternative complement pathway and provide support for a limited trial to evaluate soluble CR1 as a treatment for DDD and C3GN.


Asunto(s)
Complemento C3/análisis , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Receptores de Complemento 3b/uso terapéutico , Animales , Niño , Factor H de Complemento/fisiología , Vía Alternativa del Complemento , Glomerulonefritis Membranoproliferativa/inmunología , Humanos , Ratones
17.
J Immunol ; 191(4): 1775-84, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23863906

RESUMEN

Staphylococcus aureus is a major human pathogen causing more than a tenth of all septicemia cases and often superficial and deep infections in various tissues. One of the immune evasion strategies of S. aureus is to secrete proteins that bind to the central complement opsonin C3b. One of these, extracellular complement binding protein (Ecb), is known to interfere directly with functions of C3b. Because C3b is also the target of the physiological plasma complement regulator, factor H (FH), we studied the effect of Ecb on the complement regulatory functions of FH. We show that Ecb enhances acquisition of FH from serum onto staphylococcal surfaces. Ecb and FH enhance mutual binding to C3b and also the function of each other in downregulating complement activation. Both Ecb and the C-terminal domains 19-20 of FH bind to the C3d part of C3b. We show that the mutual enhancing effect of Ecb and FH on binding to C3b depends on binding of the FH domain 19 to the C3d part of C3b next to the binding site of Ecb on C3d. Our results show that Ecb, FH, and C3b form a tripartite complex. Upon exposure of serum-sensitive Haemophilus influenzae to human serum, Ecb protected the bacteria, and this effect was enhanced by the addition of the C-terminal domains 19-20 of FH. This finding indicates that the tripartite complex formation could give additional protection to bacteria and that S. aureus is thereby able to use host FH and bacterial Ecb in a concerted action to eliminate C3b at the site of infection.


Asunto(s)
Proteínas Bacterianas/fisiología , Complemento C3b/metabolismo , Factor H de Complemento/fisiología , Proteínas Inactivadoras de Complemento/fisiología , Vía Alternativa del Complemento , Evasión Inmune/inmunología , Staphylococcus aureus/inmunología , Factores de Virulencia/fisiología , Proteínas Bacterianas/química , Sitios de Unión , Complemento C3b/antagonistas & inhibidores , Complemento C3b/química , Factor H de Complemento/química , Factor H de Complemento/genética , Proteínas Inactivadoras de Complemento/química , Haemophilus influenzae/inmunología , Humanos , Inmunidad Innata , Modelos Moleculares , Complejos Multiproteicos , Fragmentos de Péptidos/metabolismo , Mutación Puntual , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Suero/inmunología , Infecciones Estafilocócicas , Factores de Virulencia/química
18.
PLoS One ; 8(7): e68616, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23844226

RESUMEN

We previously reported that aged mice lacking complement factor H (CFH) exhibit visual defects and structural changes in the retina. However, it is not known whether this phenotype is age-related or is the consequence of disturbed development. To address this question we investigated the effect of Cfh gene deletion on the retinal phenotype of young and mid-age mice. Cfh(-/-) mouse eyes exhibited thickening of the retina and reduced nuclear density, but relatively normal scotopic and photopic electroretinograms. At 12 months there was evidence of subtle astroglial activation in the Cfh(-/-) eyes, and significant elevation of the complement regulator, decay-accelerating factor (DAF) in Müller cells. In the retinal pigment epithelium (RPE) of young control and Cfh(-/-) animals mitochondria and melanosomes were oriented basally and apically respectively, whereas the apical positioning of melanosomes was significantly perturbed in the mid-age Cfh(-/-) RPE. We conclude that deletion of Cfh in the mouse leads to defects in the retina that precede any marked loss of visual function, but which become progressively more marked as the animals age. These observations are consistent with a lifelong role for CFH in retinal homeostasis.


Asunto(s)
Envejecimiento , Factor H de Complemento/fisiología , Retina/fisiopatología , Agudeza Visual/fisiología , Animales , Antígenos CD55/metabolismo , Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Electrorretinografía , Potenciales Evocados Visuales/genética , Potenciales Evocados Visuales/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Melanosomas/metabolismo , Melanosomas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Estimulación Luminosa , Retina/metabolismo , Retina/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Agudeza Visual/genética , Vías Visuales/fisiopatología
19.
Biochemistry ; 52(23): 3949-62, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23701234

RESUMEN

Factor H (FH) is a soluble regulator of the proteolytic cascade at the core of the evolutionarily ancient vertebrate complement system. Although FH consists of a single chain of similar protein modules, it has a demanding job description. Its chief role is to prevent complement-mediated injury to healthy host cells and tissues. This entails recognition of molecular patterns on host surfaces combined with control of one of nature's most dangerous examples of a positive-feedback loop. In this way, FH modulates, where and when needed, an amplification process that otherwise exponentially escalates the production of the pro-inflammatory, pro-phagocytic, and pro-cytolytic cleavage products of complement proteins C3 and C5. Mutations and single-nucleotide polymorphisms in the FH gene and autoantibodies against FH predispose individuals to diseases, including age-related macular degeneration, dense-deposit disease, and atypical hemolytic uremic syndrome. Moreover, deletions or variations of genes for FH-related proteins also influence the risk of disease. Numerous pathogens hijack FH and use it for self-defense. As reviewed herein, a molecular understanding of FH function is emerging. While its functional oligomeric status remains uncertain, progress has been achieved in characterizing its three-dimensional architecture and, to a lesser extent, its intermodular flexibility. Models are proposed, based on the reconciliation of older data with a wealth of recent evidence, in which a latent circulating form of FH is activated by its principal target, C3b tethered to a self-surface. Such models suggest hypotheses linking sequence variations to pathophysiology, but improved, more quantitative, functional assays and rigorous data analysis are required to test these ideas.


Asunto(s)
Activación de Complemento , Factor H de Complemento/fisiología , Secuencias de Aminoácidos , Sitios de Unión , Complemento C3b/química , Complemento C3b/fisiología , Factor H de Complemento/química , Humanos , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
20.
Adv Exp Med Biol ; 735: 173-87, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23402026

RESUMEN

The complement system is a central homeostatic system of the vertebrate organism and part f innate immunity. When activated, complement has multiple functions and drives homeostasis and the elimination of infectious microbes (Walport MJ (2001) N Engl J Med 344:1140-1144; Zipfel PF, Skerka C (2009) Nat Rev Immunol 9:729-740). Several inflammatory disorders are caused by defective complement action, and the growing, detailed understanding of the underlying pathophysiological principles translate into therapy with complement inhibitors. As complement inhibitors have been pproved for treatment of the complement-mediated disorders hemolytic uremic syndrome (HUS) and paroxysmal nocturnal hemoglobinuria (PNH), there is a growing interest to extended and improve the options for other complement-mediated diseases. Here, we summarize the current understanding and concepts how defective complement action at biological surfaces lead to pathology and disease, and how this understanding can be used for the development of surface targeting complement inhibitors.


Asunto(s)
Inactivadores del Complemento/uso terapéutico , Proteínas del Sistema Complemento/fisiología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/fisiopatología , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/fisiopatología , Animales , Factor H de Complemento/fisiología , Proteínas del Sistema Complemento/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas con Dominio LIM/fisiología , Peroxidación de Lípido/efectos de los fármacos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/fisiopatología , Malondialdehído/metabolismo , Proteínas Musculares/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...