Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 465
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 205(6): 1535-1539, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32769119

RESUMEN

C8α-γ deficiency was examined in four unrelated African Americans. Two individuals were compound heterozygotes for a previously reported point mutation in exon 9. mRNA from the remaining six C8A alleles contained a 10 nt insertion between nt 992 and 993 corresponding to the junction between exons 6 and 7. This suggested that C8α-γ deficiency in these individuals was caused by a splicing defect. Genomic sequencing revealed a G→A point mutation in intron 6, upstream of the exon 7 acceptor site. This mutation converts a GG to an AG, generates a consensus 3' splice site that shifts the reading frame, and creates a premature stop codon downstream. To verify that the point mutation caused a splicing defect, we tested wild-type and mutant mRNA substrates, containing 333 nt of the C8α intron 6/exon 7 boundary, in an in vitro splicing assay. This assay generated spliced RNA containing the 10 bp insertion observed in the C8α mRNA of affected patients. In addition, in mutant RNA substrates, the new 3' splice site was preferentially recognized compared with wild-type. Preferential selection of the mutant splice site likely reflects its positioning adjacent to a polypyrimidine tract that is stronger than that adjacent to the wild-type site. In summary, we have identified a G→A mutation in intron 6 of C8A as a predominant cause of C8α-γ deficiency in African Americans. This mutation creates a new and preferred 3' splice site, results in a 10 nt insertion in mRNA, shifts the reading frame, and produces a premature stop codon downstream.


Asunto(s)
Negro o Afroamericano , Complemento C8/genética , Síndromes de Inmunodeficiencia/genética , Mutación Puntual/genética , Empalme de Proteína/genética , Sitios de Empalme de ARN/genética , ARN Mensajero/genética , Alelos , Estudios de Asociación Genética , Heterocigoto , Humanos , Análisis de Secuencia de ADN
2.
Brain ; 144(2): 528-552, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33382892

RESUMEN

The complement system is part of the innate immune system that comprises several small proteins activated by sequential cleavages. The majority of these complement components, such as components 3a (C3a) and C5a, are chemotactic and pro-inflammatory. However, in this study, we revealed an inhibitory role of complement component 8 gamma (C8G) in neuroinflammation. In patients with Alzheimer's disease, who exhibit strong neuroinflammation, we found higher C8G levels in brain tissue, CSF, and plasma. Our novel findings also showed that the expression level of C8G increases in the inflamed mouse brain, and that C8G is mainly localized to brain astrocytes. Experiments using recombinant C8G protein and shRNA-mediated knockdown showed that C8G inhibits glial hyperactivation, neuroinflammation, and cognitive decline in acute and chronic animal models of Alzheimer's disease. Additionally, we identified sphingosine-1-phosphate receptor 2 (S1PR2) as a novel interaction protein of C8G and demonstrated that astrocyte-derived C8G interacts with S1PR2 to antagonize the pro-inflammatory action of S1P in microglia. Taken together, our results reveal the previously unrecognized role of C8G as a neuroinflammation inhibitor. Our findings pave the way towards therapeutic containment of neuroinflammation in Alzheimer's disease and related neurological diseases.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Complemento C8/inmunología , Encefalitis/inmunología , Enfermedad de Alzheimer/inmunología , Animales , Astrocitos/inmunología , Células Cultivadas , Complemento C8/líquido cefalorraquídeo , Masculino , Ratones Endogámicos C57BL , Microglía/inmunología , Subunidades de Proteína/inmunología , Receptores de Esfingosina-1-Fosfato/inmunología
3.
Eur J Pediatr ; 181(5): 1997-2004, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35118517

RESUMEN

Diagnosis of primary complement deficiencies requires a high index of suspicion. Thus, susceptible patients are often underdiagnosed and untreated. Here, we present a multicenter experience with two novel inborn errors of the classical complement system. This is a retrospective multicenter analysis of computerized medical records of children (<18 years) admitted in the period between 2012 and 2018 at Shaare Zedek Medical Center in Jerusalem and Edmond and Lily Safra Children's Hospital, Tel-Hashomer Medical Center, in Ramat Gan, Israel. Patients were genetically diagnosed by a complementary immune workup. We identified 5 patients (3 males) from four different families harboring two novel mutations in the complement components C6-C8. Genetic mutations were identified by whole-exome sequencing or by sequencing of the coding exons of a single gene based on the findings in the immune workup. Clinical manifestations consisted of meningitis with or without meningococcemia. The immune workup demonstrated nearly absent levels of CH50, compatible with a complement pathway defect. Diagnosis delay ranged between 0 and 30 years. CONCLUSION: Awareness of risk factors for primary complement deficiencies, even at the first infectious episode, should facilitate prompt immune and genetic workup, commencing diagnosis and proper treatment for the patient and family. WHAT IS KNOWN: • Deficiencies in the classical terminal complement components increase susceptibility to invasive meningococcal infections. • Recurrent meningococcal infections mandate a diagnostic workup of the complement system. WHAT IS NEW: • Genetic workup can be utilized for prompt diagnosis of complement deficiencies. • High rates of consanguinity, even in the presence of a single meningococcal infection, should promote immune and genetic workups.


Asunto(s)
Meningitis Meningocócica , Infecciones Meningocócicas , Neisseria meningitidis , Niño , Complemento C6 , Complemento C8/genética , Proteínas del Sistema Complemento/genética , Femenino , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Masculino , Estudios Retrospectivos
4.
Proteins ; 88(11): 1482-1492, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32548853

RESUMEN

Proteins are the active players in performing essential molecular activities throughout biology, and their dynamics has been broadly demonstrated to relate to their mechanisms. The intrinsic fluctuations have often been used to represent their dynamics and then compared to the experimental B-factors. However, proteins do not move in a vacuum and their motions are modulated by solvent that can impose forces on the structure. In this paper, we introduce a new structural concept, which has been called the structural compliance, for the evaluation of the global and local deformability of the protein structure in response to intramolecular and solvent forces. Based on the application of pairwise pulling forces to a protein elastic network, this structural quantity has been computed and sometimes is even found to yield an improved correlation with the experimental B-factors, meaning that it may serve as a better metric for protein flexibility. The inverse of structural compliance, namely the structural stiffness, has also been defined, which shows a clear anticorrelation with the experimental data. Although the present applications are made to proteins, this approach can also be applied to other biomolecular structures such as RNA. This present study considers only elastic network models, but the approach could be applied further to conventional atomic molecular dynamics. Compliance is found to have a slightly better agreement with the experimental B-factors, perhaps reflecting its bias toward the effects of local perturbations, in contrast to mean square fluctuations. The code for calculating protein compliance and stiffness is freely accessible at https://jerniganlab.github.io/Software/PACKMAN/Tutorials/compliance.


Asunto(s)
Complemento C8/química , Proteínas Fúngicas/química , Lectinas/química , Redes Neurales de la Computación , Programas Informáticos , Agaricales/química , Fenómenos Biomecánicos , Elasticidad , Humanos , Internet , Simulación de Dinámica Molecular
5.
Med Microbiol Immunol ; 209(2): 109-124, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31781935

RESUMEN

The complement system is a critical component of both innate and adaptive immune responses. It has both protective and pathogenic roles in viral infections. There are no studies regarding the role of complement system in Chandipura virus (CHPV) infection. The current study has investigated the role of complement pathways in the in vitro neutralization of CHPV in Vero E6 cells. Using normal human serum (NHS), heat-inactivated serum (HIS), human serum deficient of complement factor, respective reconstituted serum, assays like in vitro neutralization, real-time PCR, and flow cytometry-based tissue culture-based limited dose assay (TC-LDA) were carried out for assessing the activation of different complement pathways. NHS from 9/10 donors showed complement dependent neutralization, reduction in viral load and decrease in percentage of CHPV-positive cells compared to their HIS counterparts. EGTA or EDTA pretreatment experiments indicated that CHPV neutralization proceeds through the alternative pathway of the complement activation. Our data showed a strong dependence on C3 for the in vitro neutralization of CHPV. Disparity in CHPV neutralization levels between factor B-deficient and reconstituted sera could be attributed to amplification loop/"tick-over" mechanism. Assays using C3, C5, and C8 deficient sera indicated that complement-mediated CHPV neutralization and suppression of CHPV infectivity are primarily through C3 and C5, and not dependent on downstream complement factor C8. With no specific anti-viral treatment/vaccine against Chandipura, the current data, elucidating role of human complement system in the neutralization of CHPV, may help in designing effective therapeutics.


Asunto(s)
Vía Alternativa del Complemento , Proteínas del Sistema Complemento/fisiología , Vesiculovirus/inmunología , Animales , Chlorocebus aethiops , Complemento C3/metabolismo , Complemento C3/fisiología , Complemento C5/metabolismo , Complemento C5/fisiología , Complemento C8/metabolismo , Complemento C8/fisiología , Factor B del Complemento/metabolismo , Factor B del Complemento/fisiología , Proteínas del Sistema Complemento/metabolismo , Ácido Edético , Ácido Egtácico , Humanos , Pruebas de Neutralización , Suero/inmunología , Suero/virología , Células Vero , Vesiculovirus/fisiología , Replicación Viral/inmunología
6.
Immunity ; 30(5): 684-95, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19446473

RESUMEN

Perforin, a pore-forming protein secreted by cytotoxic lymphocytes, is indispensable for destroying virus-infected cells and for maintaining immune homeostasis. Perforin polymerizes into transmembrane channels that inflict osmotic stress and facilitate target cell uptake of proapoptotic granzymes. Despite this, the mechanism through which perforin monomers self-associate remains unknown. Our current study establishes the molecular basis for perforin oligomerization and pore assembly. We show that after calcium-dependent membrane binding, direct ionic attraction between the opposite faces of adjacent perforin monomers was necessary for pore formation. By using mutagenesis, we identified the opposing charges on residues Arg213 (positive) and Glu343 (negative) to be critical for intermolecular interaction. Specifically, disrupting this interaction had no effect on perforin synthesis, folding, or trafficking in the killer cell, but caused a marked kinetic defect of oligomerization at the target cell membrane, severely disrupting lysis and granzyme B-induced apoptosis. Our study provides important insights into perforin's mechanism of action.


Asunto(s)
Complemento C8/metabolismo , Perforina/metabolismo , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/fisiología , Estructuras Celulares/metabolismo , Eritrocitos/fisiología , Granzimas/metabolismo , Humanos , Células Jurkat , Mutación/genética , Perforina/química , Perforina/genética , Porosidad , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ovinos
7.
Fish Shellfish Immunol ; 72: 658-669, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29146450

RESUMEN

Complement component C8, which mediates membrane attack complex formation and bacterial lysis, plays important roles in the complement system. The cDNA sequences of the C8α, C8ß and C8γ genes were cloned from half-smooth tongue sole (Cynoglossus semilaevis). Full-length cDNA of CsC8α (C8α of C. semilaevis), CsC8ß and CsC8γ was 1990, 2219 and 886 bp, respectively, which contained open reading frames of 1797, 1749 and 666 bp, encoding 598, 582 and 221 amino acids, respectively. The deduced proteins of CsC8α, CsC8ß and CsC8γ showed the closest amino acid similarity to C8α (73%) of Siniperca chuatsi, C8ß (76%) of Oryzias latipes and C8γ (72%) of Takifugu rubripes, respectively. The highest expression level of CsC8α, CsC8ß and CsC8γ among the 13 normal tissues was observed in liver tissue, followed by much lower levels in other tissues. After infection with Vibrio anguillarum, CsC8α, CsC8ß and CsC8γ were significantly up-regulated in all of the detected tissues, including the intestine, liver, gill, head kidney, blood and spleen. Then, a recombinant expression plasmid was constructed, and the recombinant CsC8α protein was expressed in GS115 pichia pastoris yeast. Furthermore, to investigate the biological functions of recombinant CsC8α, an antibacterial assay was performed, and the results showed that recombinant CsC8α obviously inhibited growth of V. anguillarum, Edwardsiella tarda and Vibrio parahaemolyticus. Taken together, these results suggest that CsC8α, CsC8ß and CsC8γ may play important roles in the immune defense of C. semilaevis.


Asunto(s)
Complemento C8/genética , Complemento C8/inmunología , Enfermedades de los Peces/inmunología , Peces Planos/genética , Peces Planos/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Humoral/genética , Secuencia de Aminoácidos , Animales , Infecciones Bacterianas/inmunología , Secuencia de Bases , Complemento C8/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica , Micosis/inmunología , Filogenia , Alineación de Secuencia/veterinaria
8.
Fish Shellfish Immunol ; 81: 1-9, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29981471

RESUMEN

The redlip mullet (Liza haematocheila) is one of the most economically important fish in Korea and other East Asian countries; it is susceptible to infections by pathogens such as Lactococcus garvieae, Argulus spp., Trichodina spp., and Vibrio spp. Learning about the mechanisms of the complement system of the innate immunity of redlip mullet is important for efforts towards eradicating pathogens. Here, we report a comprehensive study of the terminal complement complex (TCC) components that form the membrane attack complex (MAC) through in-silico characterization and comparative spatial and temporal expression profiling. Five conserved domains (TSP1, LDLa, MACPF, CCP, and FIMAC) were detected in the TCC components, but the CCP and FIMAC domains were absent in MuC8ß and MuC9. Expression analysis of four TCC genes from healthy redlip mullets showed the highest expression levels in the liver, whereas limited expression was observed in other tissues; immune-induced expression in the head kidney and spleen revealed significant responses against Lactococcus garvieae and poly I:C injection, suggesting their involvement in MAC formation in response to harmful pathogenic infections. Furthermore, the response to poly I:C may suggest the role of TCC components in the breakdown of the membrane of enveloped viruses. These findings may help to elucidate the mechanisms behind the complement system of the teleosts innate immunity.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/genética , Inmunidad Innata , Smegmamorpha/inmunología , Animales , Complemento C6/genética , Complemento C6/inmunología , Complemento C7/genética , Complemento C7/inmunología , Complemento C8/genética , Complemento C8/inmunología , Complemento C9/genética , Complemento C9/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Perfilación de la Expresión Génica , Lactococcus , Lipopolisacáridos , Hígado/inmunología , Poli I-C/farmacología , Smegmamorpha/genética , Bazo/inmunología
9.
Microb Pathog ; 112: 182-189, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28963011

RESUMEN

Leptospirosis is a severe worldwide zoonotic disease caused by pathogenic Leptospira spp. It has been demonstrated that pathogenic leptospires are resistant to the bactericidal activity of normal human serum while saprophytic strains are susceptible. Pathogenic strains have the ability to bind soluble complement regulators and these activities are thought to contribute to bacterial immune evasion. One strategy used by some pathogens to evade the complement cascade, which is not well explored, is to block the terminal pathway. We have, thus, examined whether leptospires are able to interact with components of the terminal complement pathway. ELISA screening using anti-leptospires serum has shown that the pathogenic, virulent strain L. interrogans L1-130 can bind to immobilized human C8 (1 µg). However, virulent and saprophyte L. biflexa strains showed the ability to interact with C8 and C9, when these components were employed at physiological concentration (50 µg/mL), but the virulent strain seemed more competent. Lsa23, a putative leptospiral adhesin only present in pathogenic strains, interacts with C8 and C9 in a dose-dependent mode, suggesting that this protein could mediate the binding of virulent Leptospira with these components. To our knowledge, this is the first work reporting the binding of Leptospira to C8 and C9 terminal complement components, suggesting that the inhibition of this pathway is part of the strategy used by leptospires to evade the innate immunity.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Leptospira interrogans/inmunología , Leptospira interrogans/metabolismo , Leptospirosis/inmunología , Dominios y Motivos de Interacción de Proteínas , Adhesinas Bacterianas , Proteínas Bacterianas/genética , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Complemento C7/metabolismo , Complemento C8/metabolismo , Complemento C9/metabolismo , Vectores Genéticos , Humanos , Evasión Inmune , Inmunidad Innata , Leptospira interrogans/genética , Leptospira interrogans/patogenicidad , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas Recombinantes
10.
Fish Shellfish Immunol ; 62: 107-115, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28027983

RESUMEN

The complement components C8α and C8ß mediate the formation of the membrane attack complex (MAC) to resist pathogenic bacteria and play important roles in innate immunity. Full-length complement C8α (Pv-C8α) and C8ß (Pv-C8ß) cDNA were identified in the darkbarbel catfish Pelteobagrus vachellii, and their mRNA expression levels were analyzed after ammonia-N and pathogen treatment. The Pv-C8α gene contained 1983 bp, including a 1794-bp open reading frame (ORF) encoding 598 amino acids. The Pv-C8ß gene contained 1952 bp, including a 1761-bp ORF encoding 587 amino acids. Pv-C8α and Pv-C8ß had the highest amino acid identity with rainbow trout Oncorhynchus mykiss C8α (62%) and Japanese flounder Paralichthys olivaceus C8ß (83%), respectively. Sequence analysis indicated that both Pv-C8α and Pv-C8ß contained a thrombospondin type-1 (TSP1) domain, a low-density lipoprotein receptor class A (LDLR-A) domain, a membrane attack complex/perforin (MACPF) domain and an epidermal growth factor-like (EGF-like) domain. In addition, Pv-C8α and Pv-C8ß were mainly distributed in the liver, head kidney, spleen, and eggs. Under ammonia-N stress, the Pv-C8α and Pv-C8ß mRNA levels significantly decreased (P < 0.05), with minimum levels, respectively, attained at 24 and 48 h in the liver, 48 and 24 h in the head kidney, and 24 and 24 h in the spleen. After Aeromonas hydrophila challenge, the Pv-C8α and Pv-C8ß mRNA levels significantly increased (P < 0.05), with maximum levels, respectively, attained at 48 and 24 h in the liver, 24 and 48 h in the head kidney, and 48 and 48 h in the spleen. The present study indicated that Pv-C8α and Pv-C8ß exhibited important immune responses to infection and that ammonia-N in water decreased the immune responses of Pv-C8α and Pv-C8ß.


Asunto(s)
Amoníaco/toxicidad , Bagres , Complemento C8/genética , Enfermedades de los Peces/genética , Proteínas de Peces/genética , Infecciones por Bacterias Gramnegativas/veterinaria , Aeromonas hydrophila/fisiología , Secuencia de Aminoácidos , Animales , Clonación Molecular , Complemento C8/química , Complemento C8/metabolismo , ADN Complementario/genética , ADN Complementario/metabolismo , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Proteínas de Peces/química , Proteínas de Peces/metabolismo , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/microbiología , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Aleatoria , Alineación de Secuencia/veterinaria
11.
Int J Mol Sci ; 17(3): 345, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-27005612

RESUMEN

The complement components C8α, C8ß and C9 have important roles in the innate immune system against invading microorganisms. Partial cDNA sequences of the Pf_C8α, Pf_C8ß and Pf_C9 genes (Pf: abbreviation of Pelteobagrus fulvidraco) were cloned from yellow catfish. The Pf_C8α, Pf_C8ß and Pf_C9 genes showed the greatest amino acid similarity to C8α (54%) and C8ß (62%) of zebrafish and to C9 (52%) of grass carp, respectively. Ontogenetic expression analyses using real-time quantitative PCR suggested that the three genes may play crucial roles during embryonic and early larval development. The mRNA expressions of the three genes were all at the highest levels in liver tissue, and at lower or much lower levels in 16 other tissues, demonstrating that the liver is the primary site for the protein synthesis of Pf_C8α, Pf_C8ß and Pf_C9. Injection of Aeromonas hydrophila led to up-regulation of the three genes in the spleen, head kidney, kidney, liver and blood tissues, indicating that the three genes may contribute to the host's defense against invading pathogenic microbes. An increased understanding of the functions of the Pf_C8α, Pf_C8ß and Pf_C9 genes in the innate immunity of yellow catfish will help enhance production of this valuable freshwater species.


Asunto(s)
Aeromonas hydrophila , Bagres/inmunología , Complemento C8/genética , Complemento C9/genética , Proteínas de Peces/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Bagres/genética , Bagres/microbiología , Enfermedades de los Peces/microbiología , Regulación de la Expresión Génica , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Especificidad de Órganos , Filogenia , Homología de Secuencia de Aminoácido
12.
Apoptosis ; 20(4): 433-43, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25735751

RESUMEN

The loss of photoreceptors is the defining characteristic of many retinal degenerative diseases, but the mechanisms that regulate photoreceptor cell death are not fully understood. Here we have used the 661W cone photoreceptor cell line to ask whether exposure to the terminal complement complex C5b-9 induces cell death and/or modulates the sensitivity of these cells to other cellular stressors. 661W cone photoreceptors were exposed to complete normal human serum following antibody blockade of CD59. Apoptosis induction was assessed morphologically, by flow cytometry, and on western blotting by probing for cleaved PARP and activated caspase-3. Necroptosis was assessed by flow cytometry and Sirtuin 2 inhibition using 2-cyano-3-[5-(2,5-dichlorophenyl)-2-furyl]-N-5-quinolinylacrylamide (AGK2). The sensitivity of 661W cells to ionomycin, staurosporine, peroxide and chelerythrine was also investigated, with or without prior formation of C5b-9. 661W cells underwent apoptotic cell death following exposure to C5b-9, as judged by poly(ADP-ribose) polymerase 1 cleavage and activation of caspase-3. We also observed apoptotic cell death in response to staurosporine, but 661W cells were resistant to both ionomycin and peroxide. Interestingly, C5b-9 significantly increased 661W sensitivity to staurosporine-induced apoptosis and necroptosis. These studies show that low levels of C5b-9 on 661W cells can induce apoptosis, and that C5b-9 specifically sensitizes 661W cells to certain apoptotic and necroptotic pathways. Our observations provide new insight into the potential role of the complement system in photoreceptor loss, with implications for the molecular aetiology of retinal disease.


Asunto(s)
Apoptosis , Complemento C5b/metabolismo , Complemento C6/metabolismo , Complemento C7/metabolismo , Complemento C8/metabolismo , Complemento C9/metabolismo , Células Fotorreceptoras/citología , Células Fotorreceptoras/metabolismo , Caspasa 3/metabolismo , Línea Celular , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Humanos , Necrosis
13.
Am J Nephrol ; 41(1): 48-56, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25662584

RESUMEN

BACKGROUND: Diabetic nephropathy is the main cause of end-stage renal disease and has reached epidemic proportions. METHODS: Comprehensive genomic profiling (RNAseq) was employed in the ZS (F1 hybrids of Zucker and spontaneously hypertensive heart failure) model of diabetic nephropathy. Controls were lean littermates. RESULTS: Diabetic nephropathy in obese, diabetic ZS was accelerated by a single episode of renal ischemia (DI). This rapid renal decline was accompanied by the activation of the renal complement system in DI, and to a lesser extent in sham-operated diabetic rats (DS). In DI there were significant increases in renal mRNA encoding C3, C4, C5, C6, C8, and C9 over sham-operated lean normal controls (LS). Moreover, mRNAs encoding the receptors for the anaphylatoxins C3a and C5a were also significantly increased in DI compared to LS. The classic complement pathway was activated in diabetic kidneys with significant increases of C1qa, C1qb, and C1qc mRNAs in DI over LS. In addition, critical regulators of complement activation were significantly attenuated in DI and DS. These included mRNAs encoding CD55, decay accelerating factor, and CD59, which inhibit the membrane attack complex. C3, C4, and C9 proteins were demonstrated in renal tubules and glomeruli. The complement RNAseq data were incorporated into a gene network showing interactions among C3-generating renal tubular cells and other immune competent migratory cells. CONCLUSIONS: We conclude that local activation of the complement system mediates renal injury in diabetic nephropathy.


Asunto(s)
Proteínas del Sistema Complemento/genética , Nefropatías Diabéticas/genética , Isquemia/complicaciones , Riñón/irrigación sanguínea , Riñón/metabolismo , ARN Mensajero/metabolismo , Animales , Antígenos CD55/genética , Antígenos CD59/genética , Hipoxia de la Célula/fisiología , Células Cultivadas , Complemento C1q/genética , Complemento C3/genética , Complemento C4/genética , Complemento C5/genética , Complemento C6/genética , Complemento C8/genética , Complemento C9/genética , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Riñón/patología , Túbulos Renales/citología , Masculino , Obesidad/complicaciones , Ratas , Receptor de Anafilatoxina C5a/genética , Receptores Acoplados a Proteínas G/genética
14.
Biomacromolecules ; 15(7): 2389-97, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24983134

RESUMEN

Integrating catalytic functions into polymeric vesicles through enzyme entrapment is appealing for bioreactor fabrication, yet there are critical issues regarding the regulation of solute transport through membranes and enzyme loading without denaturation. Polyion complex vesicles (PICsomes) with semipermeable membranes and the propensity to form in water can overcome these issues; however, cross-linking is required for sufficient physiological stability. Herein, we report the first successful fabrication of non-cross-linked PICsomes with sufficient stability at physiological salinity and temperature by tuning the hydrophobicity of the aliphatic side chains in the pendant group of the constituent polyelectrolytes. Dynamic light scattering and transmission electron microscopy revealed that the intervesicular fusion and disintegration of the PICsomes was prevented and a narrow distribution was maintained at physiological salinity and temperatures. Furthermore, their application as enzymatic nanoreactors was verified even in the presence of proteases. As such, the potential utility of the PICsomes in biomedical fields was established.


Asunto(s)
Iones/química , Nanopartículas/química , Polímeros/química , Cloruro de Sodio/química , Temperatura , Fenómenos Químicos , Complemento C8/química , Enzimas/química , Espectroscopía de Resonancia Magnética , Microscopía Electrónica de Transmisión , Nanotecnología
15.
Biotechnol Lett ; 36(2): 319-25, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24101243

RESUMEN

Antimicrobial peptides are active against a diverse spectrum of microorganisms. Using a bioinformatics method, six potential novel antimicrobial peptides, A1, C1, A2, A3, C2 and A4, were identified in the C8α complement component. The corresponding genes were then cloned into a new vector as fusions with the self-cleavage protein N(pro) protein mutant EDDIE gene. The expressed or synthetic peptides, A1, A2, A3 and A4, showed antimicrobial activities against several bacteria, while peptides C1 and C2 did not. Peptides A1 to A4 showed no hemolytic activities over 3 h when at 500 µg/ml. Thus, A1, A2, A3 and A4, derived from the C8α complement system, are novel antimicrobial peptides.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Bacterias/efectos de los fármacos , Complemento C8/farmacología , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/toxicidad , Complemento C8/genética , Complemento C8/toxicidad , Eritrocitos/efectos de los fármacos , Hemólisis , Humanos , Pruebas de Sensibilidad Microbiana , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/toxicidad
16.
Chem Biol Interact ; 395: 110998, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38614317

RESUMEN

Complement component 8gamma (C8γ), a member of the lipocalin protein family, is suggested to act as a carrier protein for various chemicals. Although C8γ has been identified in both humans and rodents for some time, our understanding of the species differences in its chemical binding properties remains limited. In the present study, with the aim to elucidate the potential role of C8γ as a carrier protein in both humans and mice, we conducted a radioligand binding assay to examine the chemical binding properties of human C8γ (hC8γ) and mouse C8γ (mC8γ). Scatchard analysis revealed that [14C]TPT bound to hC8γ with an equilibrium dissociation constant (Kd) of 64.2 ± 32.4 nM, comparable to that of [14C]TPT to mC8γ. Competitive ligand-binding assays demonstrated binding of TPT and TBT to hC8γ, while diphenyltin, dibutyltin, monophenyltin, monobutyltin, and tetrabutyltin did not exhibit binding. These results suggest that for effective binding to C8γ, chemicals must possess substituents of appropriate bulkiness. Further analyses with other group 14 compounds with triphenyl substituents revealed that a central metal atom, rather than a central non-metal or semi-metal atom, is crucial for specific binding to both hC8γ and mC8γ. Overall our findings imply that C8γ may play a role in the physiological or toxicological actions of group 14 metal compounds with tributyl or triphenyl substituents by binding to these chemicals in both humans and mice.


Asunto(s)
Unión Proteica , Animales , Humanos , Ratones , Complemento C8/metabolismo , Complemento C8/química , Compuestos Organometálicos/química , Compuestos Organometálicos/metabolismo , Unión Competitiva
18.
Fish Shellfish Immunol ; 35(3): 1055-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23876999

RESUMEN

Nile tilapia (Oreochromis niloticus), one of the most important groups of food fishes in the world, has frequently suffered from serious challenge from pathogens in recent years. Immune responses of Nile tilapia should be understood to protect the aquaculture industry of this fish. The complement system has an important function in recognizing bacteria, opsonizing these pathogens by phagocytes, or killing them by direct lysis. In this study, two Nile tilapia complement component genes, complement component 1 inhibitor (C1INH) and complement component 8ß subunit (C8ß), were cloned and their expression characteristics were analyzed. C1INH cDNA was found containing a 1791 bp open reading frame (ORF) encoding a putative protein with 597 amino acids, a 101 bp 5'-untranslated region (UTR) and a 236 bp 3'-UTR. The predicted protein structure for this gene consisted of two Ig-like domains and glycosyl hydrolase family-9 active site signature 2. The C8ß cDNA consisted of a 1761 bp ORF encoding 587 amino acids, a 15 bp 5'-UTR and a 170 bp 3'-UTR. The predicted protein of C8ß contained three motifs, thrombospondin type-1 repeat, membrane attack complex/perforin domain, and LDL-receptor class A. Expression analysis revealed that these two complement genes were highly expressed in the liver, however, were weakly expressed in the gill, heart, brain, kidney, intestine, spleen and dorsal muscle tissues. The present study provided insights into the complement system and immune functions of Nile tilapia.


Asunto(s)
Cíclidos/metabolismo , Proteínas Inactivadoras del Complemento 1/metabolismo , Complemento C8/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Proteínas Inactivadoras del Complemento 1/genética , Complemento C8/genética , ADN Complementario , Regulación de la Expresión Génica , Datos de Secuencia Molecular , Alineación de Secuencia , Especificidad de la Especie
19.
Fish Shellfish Immunol ; 35(6): 1993-2000, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24076167

RESUMEN

Complement-mediated cytolysis is the important effect of immune response, which results from the assembly of terminal complement components (C5b-9). Among them, α subunit of C8 (C8α) is the first protein that traverses the lipid bilayer, and then initiates the recruitment of C9 molecules to form pore on target membranes. In this article, a full-length cDNA of C8α (CpC8α) is identified from the whitespotted bamboo shark (Chiloscyllium plagiosum) by RACE. The CpC8α cDNA is 2183 bp in length, encoding a protein of 591 amino acids. The deduced CpC8α exhibits 89%, 49% and 44% identity with nurse shark, frog and human orthologs, respectively. Sequence alignment indicates that the C8α is well conserved during the evolution process from sharks to mammals, with the same modular architecture as well as the identical cysteine composition in the mature protein. Phylogenetic analysis places CpC8α and nurse shark C8α in cartilaginous fish clade, in parallel with the teleost taxa, to form the C8α cluster with higher vertebrates. Hydrophobicity analysis also indicates a similar hydrophobicity of CpC8α to mammals. Finally, expression analysis revealed CpC8α transcripts were constitutively highly expressed in shark liver, with much less expression in other tissues. The well conserved structure and properties suggests an analogous function of CpC8α to mammalian C8α, though it remains to be confirmed by further study.


Asunto(s)
Complemento C8/genética , Proteínas de Peces/genética , Tiburones/genética , Tiburones/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , China , Clonación Molecular , Complemento C8/química , Complemento C8/metabolismo , ADN Complementario/genética , ADN Complementario/metabolismo , Proteínas de Peces/química , Proteínas de Peces/metabolismo , Masculino , Datos de Secuencia Molecular , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Alineación de Secuencia
20.
Orbit ; 32(6): 384-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23895036

RESUMEN

Haemangiopericytoma (HPC) is a rare soft tissue tumour of fibroblastic origin and is part of the solitary fibrous tumour spectrum. The tumour is generally considered to be benign, but can behave clinically as if sarcomatous -- with relentless infiltrative local growth. HPC generally presents in adulthood (median age 45 years for orbital disease) and is equally frequent in both sexes. HPC can arise in any site in the body and presents as a slowly growing, painless mass. We report a case of a 20 year old African male seen at Kikuyu Eye Unit, Kenya, with a 12 year history of a gradually enlarging, painless orbital mass. The patient underwent skin-sparing orbital exenteration with complete tumour excision; histology confirmed diagnosis of HPC.


Asunto(s)
Hemangiopericitoma/patología , Neoplasias Orbitales/patología , Actinas/metabolismo , Antígenos CD34/metabolismo , Biomarcadores de Tumor/metabolismo , Complemento C8/metabolismo , Hemangiopericitoma/metabolismo , Hemangiopericitoma/cirugía , Humanos , Masculino , Evisceración Orbitaria , Neoplasias Orbitales/metabolismo , Neoplasias Orbitales/cirugía , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA