Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Annu Rev Pathol ; 12: 25-52, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-27959629

RESUMEN

The vertebrate complement system consists of sequentially interacting proteins that provide for a rapid and powerful host defense. Nearly 60 proteins comprise three activation pathways (classical, alternative, and lectin) and a terminal cytolytic pathway common to all. Attesting to its potency, nearly half of the system's components are engaged in its regulation. An emerging theme over the past decade is that variations in these inhibitors predispose to two scourges of modern humans. One, occurring most often in childhood, is a rare but deadly thrombomicroangiopathy called atypical hemolytic uremic syndrome. The other, age-related macular degeneration, is the most common form of blindness in the elderly. Their seemingly unrelated clinical presentations and pathologies share the common theme of overactivity of the complement system's alternative pathway. This review summarizes insights gained from contemporary genetics for understanding how dysregulation of this powerful innate immune system leads to these human diseases.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/fisiopatología , Vía Alternativa del Complemento/genética , Proteínas del Sistema Complemento/genética , Degeneración Macular/fisiopatología , Genotipo , Humanos
2.
J Am Soc Nephrol ; 27(6): 1665-77, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26471127

RESUMEN

C3 glomerulopathy is a recently described form of CKD. C3GN is a subtype of C3 glomerulopathy characterized by predominant C3 deposits in the glomeruli and is commonly the result of acquired or genetic abnormalities in the alternative pathway (AP) of the complement system. We identified and characterized the first mutation of the C3 gene (p. I734T) in two related individuals diagnosed with C3GN. Immunofluorescence and electron microscopy studies showed C3 deposits in the subendothelial space, associated with unusual deposits located near the complement receptor 1 (CR1)-expressing podocytes. In vitro, this C3 mutation exhibited decreased binding to CR1, resulting in less CR1-dependent cleavage of C3b by factor 1. Both patients had normal plasma C3 levels, and the mutant C3 interacted with factor B comparably to wild-type (WT) C3 to form a C3 convertase. Binding of mutant C3 to factor H was normal, but mutant C3 was less efficiently cleaved by factor I in the presence of factor H, leading to enhanced C3 fragment deposition on glomerular cells. In conclusion, our results reveal that a CR1 functional deficiency is a mechanism of intraglomerular AP dysregulation and could influence the localization of the glomerular C3 deposits.


Asunto(s)
Complemento C3/genética , Glomerulonefritis Membranoproliferativa/genética , Receptores de Complemento 3b/genética , Adulto , Factor H de Complemento/genética , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Linaje
3.
Invest Ophthalmol Vis Sci ; 56(11): 6873-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26501415

RESUMEN

PURPOSE: Age-related macular degeneration (AMD) has a substantial genetic risk component, as evidenced by the risk from common genetic variants uncovered in the first genome-wide association studies. More recently, it has become apparent that rare genetic variants also play an independent role in AMD risk. We sought to determine if rare variants in complement factor H (CFH) played a role in AMD risk. METHODS: We had previously collected DNA from a large population of patients with advanced age-related macular degeneration (A-AMD) and controls for targeted deep sequencing of candidate AMD risk genes. In this analysis, we tested for an increased burden of rare variants in CFH in 1665 cases and 752 controls from this cohort. RESULTS: We identified 65 missense, nonsense, or splice-site mutations with a minor allele frequency ≤ 1%. Rare variants with minor allele frequency ≤ 1% (odds ratio [OR] = 1.5, P = 4.4 × 10⁻²), 0.5% (OR = 1.6, P = 2.6 × 10⁻²), and all singletons (OR = 2.3, P = 3.3 × 10⁻²) were enriched in A-AMD cases. Moreover, we observed loss-of-function rare variants (nonsense, splice-site, and loss of a conserved cysteine) in 10 cases and serum levels of FH were decreased in all 5 with an available sample (haploinsufficiency). Further, rare variants in the major functional domains of CFH were increased in cases (OR = 3.2; P = 1.4 × 10⁻³) and the magnitude of the effect correlated with the disruptive nature of the variant, location in an active site, and inversely with minor allele frequency. CONCLUSIONS: In this large A-AMD cohort, rare variants in the CFH gene were enriched and tended to be located in functional sites or led to low serum levels. These data, combined with those indicating a similar, but even more striking, increase in rare variants found in CFI, strongly implicate complement activation in A-AMD etiopathogenesis as CFH and CFI interact to inhibit the alternative pathway.


Asunto(s)
Predisposición Genética a la Enfermedad , Variación Genética , Degeneración Macular/genética , Anciano , Codón sin Sentido , Factor H de Complemento/análisis , Factor H de Complemento/genética , Frecuencia de los Genes , Estudios de Asociación Genética , Humanos , Degeneración Macular/sangre , Mutación Missense , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
4.
Hum Mol Genet ; 24(13): 3861-70, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25788521

RESUMEN

To assess a potential diagnostic and therapeutic biomarker for age-related macular degeneration (AMD), we sequenced the complement factor I gene (CFI) in 2266 individuals with AMD and 1400 without, identifying 231 individuals with rare genetic variants. We evaluated the functional impact by measuring circulating serum factor I (FI) protein levels in individuals with and without rare CFI variants. The burden of very rare (frequency <1/1000) variants in CFI was strongly associated with disease (P = 1.1 × 10(-8)). In addition, we examined eight coding variants with counts ≥5 and saw evidence for association with AMD in three variants. Individuals with advanced AMD carrying a rare CFI variant had lower mean FI compared with non-AMD subjects carrying a variant (P < 0.001). Further new evidence that FI levels drive AMD risk comes from analyses showing individuals with a CFI rare variant and low FI were more likely to have advanced AMD (P = 5.6 × 10(-5)). Controlling for covariates, low FI increased the risk of advanced AMD among those with a variant compared with individuals without advanced AMD with a rare CFI variant (OR 13.6, P = 1.6 × 10(-4)), and also compared with control individuals without a rare CFI variant (OR 19.0, P = 1.1 × 10(-5)). Thus, low FI levels are strongly associated with rare CFI variants and AMD. Enhancing FI activity may be therapeutic and measuring FI provides a screening tool for identifying patients who are most likely to benefit from complement inhibitory therapy.


Asunto(s)
Factor I de Complemento/genética , Fibrinógeno/metabolismo , Degeneración Macular/genética , Factor I de Complemento/metabolismo , Femenino , Variación Genética , Humanos , Degeneración Macular/metabolismo , Degeneración Macular/patología , Masculino
5.
Anal Biochem ; 477: 78-85, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25660530

RESUMEN

Complement is a major effector arm of the innate immune system that responds rapidly to pathogens or altered self. The central protein of the system, C3, participates in an amplification loop that can lead to rapid complement deposition on a target and, if excessive, can result in host tissue damage. Currently, complement activation is routinely monitored by assessing total C3 levels, which is an indirect and relatively insensitive method. An alternative approach would be to measure downstream C3 activation products such as C3a and iC3b. However, in vitro activation can produce falsely elevated levels of these biomarkers. To circumvent this issue, a lateral flow immunoassay system was developed that measures iC3b in whole blood, plasma, and serum and avoids in vitro activation by minimizing sample handling. This assay system returns results within 15 min and specifically measures iC3b while having minimal cross-reactivity to other C3 split products. While evaluating the potential of this assay, it was observed that circulating iC3b levels can distinguish healthy individuals from those with complement activation-associated diseases. This tool is engineered to provide an improved method to assess complement activation at point of care and could facilitate studies to monitor disease progression in a variety of inflammatory conditions.


Asunto(s)
Análisis Químico de la Sangre/métodos , Activación de Complemento , Inmunoensayo/métodos , Hemorragia Cerebral/sangre , Hemorragia Cerebral/inmunología , Complemento C3/inmunología , Complemento C3b/inmunología , Humanos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Factores de Tiempo
6.
Blood ; 125(15): 2359-69, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25608561

RESUMEN

The pathogenesis of atypical hemolytic uremic syndrome (aHUS) is strongly linked to dysregulation of the alternative pathway of the complement system. Mutations in complement genes have been identified in about two-thirds of cases, with 5% to 15% being in C3. In this study, 23 aHUS-associated genetic changes in C3 were characterized relative to their interaction with the control proteins factor H (FH), membrane cofactor protein (MCP; CD46), and complement receptor 1 (CR1; CD35). In surface plasmon resonance experiments, 17 mutant recombinant proteins demonstrated a defect in binding to FH and/or MCP, whereas 2 demonstrated reduced binding to CR1. In the majority of cases, decreased binding affinity translated to a decrease in proteolytic inactivation (known as cofactor activity) of C3b via FH and MCP. These results were used to map the putative binding regions of C3b involved in the interaction with MCP and CR1 and interrogated relative to known FH binding sites. Seventy-six percent of patients with C3 mutations had low C3 levels that correlated with disease severity. This study expands our knowledge of the functional consequences of aHUS-associated C3 mutations relative to the interaction of C3 with complement regulatory proteins mediating cofactor activity.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Mapas de Interacción de Proteínas , Síndrome Hemolítico Urémico Atípico/patología , Sitios de Unión , Estudios de Cohortes , Complemento C3/análisis , Femenino , Humanos , Masculino , Modelos Moleculares , Mutación
7.
Mol Immunol ; 61(2): 118-125, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25034031

RESUMEN

Age-related macular degeneration (AMD) is a major cause of visual impairment in the western world. It is characterized by the presence of lipoproteinaceous deposits (drusen) in the inner layers of the retina. Immunohistochemistry studies identified deposition of complement proteins in the drusen as well as in the choroid. In the last decade, genetic studies have linked both common and rare variants in genes of the complement system to increased risk of development of AMD. Here, we review the variants described to date and discuss the functional implications of dysregulation of the alternative pathway of complement in AMD.


Asunto(s)
Proteínas del Sistema Complemento/genética , Predisposición Genética a la Enfermedad , Variación Genética , Degeneración Macular/genética , Degeneración Macular/inmunología , Humanos
8.
Hum Mol Genet ; 23(19): 5283-93, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24847005

RESUMEN

We sequenced the whole exome of 35 cases and 7 controls from 9 age-related macular degeneration (AMD) families in whom known common genetic risk alleles could not explain their high disease burden and/or their early-onset advanced disease. Two families harbored novel rare mutations in CFH (R53C and D90G). R53C segregates perfectly with AMD in 11 cases (heterozygous) and 1 elderly control (reference allele) (LOD = 5.07, P = 6.7 × 10(-7)). In an independent cohort, 4 out of 1676 cases but none of the 745 examined controls or 4300 NHBLI Exome Sequencing Project (ESP) samples carried the R53C mutation (P = 0.0039). In another family of six siblings, D90G similarly segregated with AMD in five cases and one control (LOD = 1.22, P = 0.009). No other sample in our large cohort or the ESP had this mutation. Functional studies demonstrated that R53C decreased the ability of FH to perform decay accelerating activity. D90G exhibited a decrease in cofactor-mediated inactivation. Both of these changes would lead to a loss of regulatory activity, resulting in excessive alternative pathway activation. This study represents an initial application of the whole-exome strategy to families with early-onset AMD. It successfully identified high impact alleles leading to clearer functional insight into AMD etiopathogenesis.


Asunto(s)
Factor H de Complemento/genética , Exoma , Variación Genética , Degeneración Macular/genética , Alelos , Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Femenino , Frecuencia de los Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Cinética , Escala de Lod , Degeneración Macular/metabolismo , Masculino , Modelos Moleculares , Linaje , Polimorfismo de Nucleótido Simple , Unión Proteica , Conformación Proteica
9.
J Plant Regist ; 8(2): 217-220, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25580180

RESUMEN

Zak ERA8 (ENHANCED RESPONSE to ABA8) (Reg. No. GP-966, PI 669443) is a unique line derived from soft white spring wheat (Triticum aestivum L.) cultivar Zak that has increased seed dormancy but after-ripens within 10 to 16 wk. The goal in developing this germplasm was to use increased seed dormancy to improve tolerance to preharvest sprouting, a problem that can cause severe economic losses. This germplasm was developed by USDA-ARS, Pullman, WA, in collaboration with Washington State University. Zak ERA8was tested under experimental number 60.1.27.10. The ERA8mutation was generated by chemical mutagenesis followed by selection for the inability to germinate on abscisic acid (ABA) concentrations too low to inhibit wild-type Zak seed germination. The semidominant Zak ERA8 line has been backcrossed twice to wild-type Zak. Following the first backcross, Zak ERA8 showed similar morphological and grain quality traits to the original Zak cultivar.

10.
Theor Appl Genet ; 126(3): 791-803, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23212773

RESUMEN

As a strategy to increase the seed dormancy of soft white wheat, mutants with increased sensitivity to the plant hormone abscisic acid (ABA) were identified in mutagenized grain of soft white spring wheat "Zak". Lack of seed dormancy is correlated with increased susceptibility to preharvest sprouting in wheat, especially those cultivars with white kernels. ABA induces seed dormancy during embryo maturation and inhibits the germination of mature grain. Three mutant lines called Zak ERA8, Zak ERA19A, and Zak ERA19B (Zak ENHANCED RESPONSE to ABA) were recovered based on failure to germinate on 5 µM ABA. All three mutants resulted in increased ABA sensitivity over a wide range of concentrations such that a phenotype can be detected at very low ABA concentrations. Wheat loses sensitivity to ABA inhibition of germination with extended periods of dry after-ripening. All three mutants recovered required more time to after-ripen sufficiently to germinate in the absence of ABA and to lose sensitivity to 5 µM ABA. However, an increase in ABA sensitivity could be detected after as long as 3 years of after-ripening using high ABA concentrations. The Zak ERA8 line showed the strongest phenotype and segregated as a single semi-dominant mutation. This mutation resulted in no obvious decrease in yield and is a good candidate gene for breeding preharvest sprouting tolerance.


Asunto(s)
Ácido Abscísico/farmacología , Latencia en las Plantas/efectos de los fármacos , Reguladores del Crecimiento de las Plantas/farmacología , Triticum/genética , Relación Dosis-Respuesta a Droga , Genotipo , Mutación , Fenotipo , Semillas/efectos de los fármacos , Semillas/genética
11.
Euphytica ; 188(1): 35-49, 2012 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25431501

RESUMEN

This paper describes the isolation of wheat mutants in the hard red spring Scarlet resulting in reduced sensitivity to the plant hormone abscisic acid (ABA) during seed germination. ABA induces seed dormancy during embryo maturation and inhibits the germination of mature seeds. Wheat sensitivity to ABA gradually decreases with dry after-ripening. Scarlet grain normally fails to germinate when fully dormant, shows ABA sensitive germination when partially after-ripened, and becomes ABA insensitive when after-ripened for 8-12 months. Scarlet ABA-insensitive (ScABI) mutants were isolated based on the ability to germinate on 5 µM ABA after only 3 weeks of after-ripening, a condition under which Scarlet would fail to germinate. Six independent seed-specific mutants were recovered. ScABI 1, ScABI2, ScABI3 and ScABI4 are able to germinate more efficiently than Scarlet at up to 25 µM ABA. The two strongest ABA insensitive lines, ScABI3 and ScABI4, both proved to be partly dominant suggesting that they result from gain-of-function mutations. The ScABI1, ScABI2, ScABI3, ScABI4, and ScABI5 mutants after-ripen more rapidly than Scarlet. Thus, ABA insensi-tivity is associated with decreased grain dormancy in Scarlet wheat. This suggests that ABA sensitivity is an important factor controlling grain dormancy in wheat, a trait that impacts seedling emergence and pre-harvest sprouting resistance.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...