Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
EMBO J ; 40(4): e106174, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33459420

ABSTRACT

Cross-linking mass spectrometry has developed into an important method to study protein structures and interactions. The in-solution cross-linking workflows involve time and sample consuming steps and do not provide sensible solutions for differentiating cross-links obtained from co-occurring protein oligomers, complexes, or conformers. Here we developed a cross-linking workflow combining blue native PAGE with in-gel cross-linking mass spectrometry (IGX-MS). This workflow circumvents steps, such as buffer exchange and cross-linker concentration optimization. Additionally, IGX-MS enables the parallel analysis of co-occurring protein complexes using only small amounts of sample. Another benefit of IGX-MS, demonstrated by experiments on GroEL and purified bovine heart mitochondria, is the substantial reduction of undesired over-length cross-links compared to in-solution cross-linking. We next used IGX-MS to investigate the complement components C5, C6, and their hetero-dimeric C5b6 complex. The obtained cross-links were used to generate a refined structural model of the complement component C6, resembling C6 in its inactivated state. This finding shows that IGX-MS can provide new insights into the initial stages of the terminal complement pathway.


Subject(s)
Complement C5/metabolism , Complement C6/metabolism , Complement System Proteins/metabolism , Cross-Linking Reagents/chemistry , Mass Spectrometry/methods , Mitochondria, Heart/metabolism , Animals , Cattle , Complement C5/chemistry , Complement C6/chemistry , Complement System Proteins/chemistry
2.
Nat Commun ; 9(1): 5316, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552328

ABSTRACT

The membrane attack complex (MAC) is one of the immune system's first responders. Complement proteins assemble on target membranes to form pores that lyse pathogens and impact tissue homeostasis of self-cells. How MAC disrupts the membrane barrier remains unclear. Here we use electron cryo-microscopy and flicker spectroscopy to show that MAC interacts with lipid bilayers in two distinct ways. Whereas C6 and C7 associate with the outer leaflet and reduce the energy for membrane bending, C8 and C9 traverse the bilayer increasing membrane rigidity. CryoEM reconstructions reveal plasticity of the MAC pore and demonstrate how C5b6 acts as a platform, directing assembly of a giant ß-barrel whose structure is supported by a glycan scaffold. Our work provides a structural basis for understanding how ß-pore forming proteins breach the membrane and reveals a mechanism for how MAC kills pathogens and regulates cell functions.


Subject(s)
Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/ultrastructure , Cryoelectron Microscopy/methods , Lipid Bilayers/chemistry , Complement C6/chemistry , Complement C6/metabolism , Complement C6/ultrastructure , Complement C7/chemistry , Complement C7/metabolism , Complement C7/ultrastructure , Complement C8/chemistry , Complement C8/metabolism , Complement C8/ultrastructure , Complement C9/chemistry , Complement C9/metabolism , Complement C9/ultrastructure , Complement Membrane Attack Complex/metabolism , Humans , Image Processing, Computer-Assisted , Lipid Bilayers/metabolism , Liposomes , Models, Molecular , Polysaccharides/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Spectrum Analysis/methods
3.
Fish Shellfish Immunol ; 34(5): 1360-5, 2013 May.
Article in English | MEDLINE | ID: mdl-23422818

ABSTRACT

Aeromonas hydrophila, a widespread bacterium in the aquatic environment, causes haemorrhagic septicemia in fish. In the last decade, the disease has caused mass mortality and tremendous economic loss in cultured grass carp in the mainland China. The complement component C6 is a constituent of a biochemical cascade that serves as a major effector of the human innate and adaptor immunity, and eliminates infected cells. The objective of this study was to identify single nucleotide polymorphisms (SNPs) in the C6 gene and to assess their association with A. hydrophila resistance in grass carp. A resource population consisting of 186 susceptible and 191 resistant grass carp was constructed. The gcC6 genomic sequence is composed of 9292 bp, containing 18 exons and 17 introns. The promoter sequence of gcC6 gene contained several consensus sequences for hepatic-specific transcription factors. We sequenced a total of 9744 bp of the C6 gene from a diverse population of grass carp and identified 8 SNPs that were genotyped in the resource population. Statistical analysis revealed a lack of association between any individual SNPs and resistance to A. hydrophila in grass carp. The SNPs 1214G>A, 1380G>C, 2095A>C and 2167T>C were linked together (r(2) > 0.8). The haplotype GCCC generated with these four SNPs was associated marginally with resistance to A. hydrophila in grass carp. These findings suggest a lack of strong association of the C6 polymorphisms with the A. hydrophila resistance in grass carp.


Subject(s)
Aeromonas hydrophila/physiology , Carps , Complement C6/genetics , Fish Diseases/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Gram-Negative Bacterial Infections/veterinary , Animals , Base Sequence , China , Cloning, Molecular , Complement C6/chemistry , Complement C6/metabolism , DNA, Complementary/analysis , Disease Resistance , Fish Diseases/microbiology , Fish Proteins/chemistry , Fish Proteins/metabolism , Genotype , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Haplotypes , Introns , Molecular Sequence Data , Polymerase Chain Reaction/veterinary , Polymorphism, Single Nucleotide , RNA, Messenger/analysis , Sequence Alignment , Sequence Analysis, Protein/veterinary
4.
J Biol Chem ; 287(13): 10210-10222, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22267737

ABSTRACT

The complement membrane attack complex (MAC) is formed by the sequential assembly of C5b with four homologous proteins as follows: one copy each of C6, C7, and C8 and 12-14 copies of C9. Together these form a lytic pore in bacterial membranes. C6 through C9 comprise a MAC-perforin domain flanked by 4-9 "auxiliary" domains. Here, we report the crystal structure of C6, the first and longest of the pore proteins to be recruited by C5b. Comparisons with the structures of the C8αßγ heterodimer and perforin show that the central domain of C6 adopts a "closed" (perforin-like) state that is distinct from the "open" conformations in C8. We further show that C6, C8α, and C8ß contain three homologous subdomains ("upper," "lower," and "regulatory") related by rotations about two hinge points. In C6, the regulatory segment includes four auxiliary domains that stabilize the closed conformation, inhibiting release of membrane-inserting elements. In C8ß, rotation of the regulatory segment is linked to an opening of the central ß-sheet of its clockwise partner, C8α. Based on these observations, we propose a model for initiation and unidirectional propagation of the MAC in which the auxiliary domains play key roles: in the assembly of the C5b-8 initiation complex; in driving and regulating the opening of the ß-sheet of the MAC-performin domain of each new recruit as it adds to the growing pore; and in stabilizing the final pore. Our model of the assembled pore resembles those of the cholesterol-dependent cytolysins but is distinct from that recently proposed for perforin.


Subject(s)
Complement C6/chemistry , Complement Membrane Attack Complex , Models, Biological , Models, Molecular , Complement C6/metabolism , Complement System Proteins/chemistry , Complement System Proteins/metabolism , Crystallography, X-Ray , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
5.
Int J Biochem Cell Biol ; 39(12): 2314-23, 2007.
Article in English | MEDLINE | ID: mdl-17707681

ABSTRACT

Angiogenesis, or neovascularization, is tightly orchestrated by endogenous regulators that promote or inhibit the process. The fine-tuning of these pro- and anti-angiogenic elements (the angiogenic balance) helps establish the homeostasis in tissues, and any aberration leads to pathologic conditions. The type I thrombospondin repeats are a family of protein structural elements involved in the control of angiogenesis, and some proteins containing these repeats have been identified as negative regulators of angiogenesis. Here we identify a set of 11 novel, anti-angiogenic 18-20-amino acid peptides that are derived from proteins that belong to the CCN protein family and contain type I thrombospondin motifs. We have named these peptides spondinstatin-1, cyrostatin, connectostatin, nephroblastostatin, wispostatin-2, wispostatin-3, netrinstatin-5C, netrinstatin-5D, adamtsostatin-like-4, fibulostatin-6.1, and complestatin-C6 to reflect their origin. We have shown that these peptides inhibit proliferation and migration of human umbilical vein endothelial cells in vitro. By conducting a clustering analysis of the amino acid sequences using sequence similarity criteria and of the experimental results using a hierarchical clustering algorithm, we have demonstrated that there is an underlying correlation between the sequence and activity of the identified peptides. This combination of experimental and computational approaches introduces a novel systematic framework for studying peptide activity, identifying novel peptides with anti-angiogenic activity, and designing mimetic peptides with tailored properties.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Peptide Fragments/pharmacology , Thrombospondin 1/chemistry , ADAMTS Proteins , Amino Acid Sequence , Angiogenesis Inhibitors/chemical synthesis , CCN Intercellular Signaling Proteins , Cells, Cultured , Cluster Analysis , Complement C6/chemistry , Connective Tissue Growth Factor , Cysteine-Rich Protein 61 , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Humans , Immediate-Early Proteins/chemistry , Immunoglobulins/chemistry , Insulin-Like Growth Factor Binding Proteins/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Molecular Sequence Data , Netrin Receptors , Peptide Fragments/chemical synthesis , Peptide Fragments/genetics , Receptors, Cell Surface/chemistry , Repressor Proteins , Sequence Homology, Amino Acid , Thrombospondins/chemistry , Transcription Factors/chemistry
6.
Mol Immunol ; 44(10): 2756-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17257682

ABSTRACT

Complement component C6 is one of five terminal complement components incorporated into the membrane attack complex. Complete deficiency of C6 (C6Q0) leads to an increased susceptibility to Neisseria meningitidis infections, and affected individuals typically present with recurrent meningococcal disease. There is a relatively high prevalence of C6Q0 in the Western Cape, South Africa and three frameshift mutations have previously been described to be responsible for C6Q0 in this area-879delG, 1195delC, and 1936delG (current nomenclature). We have now genotyped a further nine genetically independent individuals with C6Q0, confirming previous reports that the most common defect in the Western Cape is 879delG. Moreover, we report the first identification of the 878delA mutation within the Western Cape, which has previously only been reported in individuals of African descent living in the United States or Europe. We also investigated the genotype of an Irish C6Q0 individual and her sibling, and report two previously undescribed mutations. One mutation alters a tyrosine codon to a stop codon within exon 10. The second mutation is within the 5' donor splice site of intron 3, and would, in all probability, disrupt splicing. These two mutations were shown to segregate independently. We also discuss the nomenclature for reporting C6 and C7 gene mutations, as the current nomenclature does not follow the recognised guidelines.


Subject(s)
Complement C6/genetics , Genetic Predisposition to Disease , Meningococcal Infections/genetics , Neisseria meningitidis , Complement C6/chemistry , Complement C6/deficiency , Humans , Mutation , Pedigree , South Africa , Tyrosine/chemistry , Tyrosine/genetics
7.
Mol Immunol ; 43(8): 1080-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16271768

ABSTRACT

The sixth complement protein (C6) is an essential component of the membrane attack complex (MAC); the end product of the lytic pathway of complement activation. The MAC complex constitutes a supramolecular assembly containing the five precursor proteins C5b, C6, C7, C8, and C9. Once assembled on the target surface it forms transmembrane channels that cause membrane damage and cytolysis of complement-opsonized pathogens. Besides mediating direct pathogen elimination, exposure of cells to sublytic doses of MAC can trigger diverse cellular responses such as, cell activation, induction of apoptosis, cell cycle re-entry and proliferation in various biological settings. The terminal complement components (C6-C9) are structurally related proteins, differing in size and complexity. In order to study their evolution, we report here the cloning and molecular characterization of C6 component in rainbow trout. The deduced amino acid sequence of trout C6 exhibits 55 and 44% identity with zebra fish and human orthologs, respectively. The 'domain' architecture of trout C6 resembles that of mammalian counterparts, and the cysteine backbone is also conserved. Finally, trout C6 gene appears to exist as a single copy in the trout genome, and is expressed in a wide range of trout tissues.


Subject(s)
Complement C6/genetics , Oncorhynchus mykiss/genetics , Phylogeny , Amino Acid Sequence , Animals , Cloning, Molecular , Complement C6/chemistry , DNA, Complementary/genetics , Gene Expression Profiling , Genome , Molecular Sequence Data , Oncorhynchus mykiss/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA
8.
J Biol Chem ; 280(11): 10636-45, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15598652

ABSTRACT

The complement protein C5 initiates assembly of the membrane attack complex. This remarkable process results in lysis of target cells and is fundamental to mammalian defense against infection. The 150-amino acid residue domain at the C terminus of C5 (C5-C345C) is pivotal to C5 function. It interacts with enzymes that convert C5 to C5b, the first step in the assembly of the membrane attack complex; it also binds to the membrane attack complex components C6 and C7 with high affinity. Here a recombinant version of this C5-C345C domain is shown to adopt the oligosaccharide/oligonucleotide binding fold, with two helices packed against a five-stranded beta-barrel. The structure is compared with those from the netrin-like module family that have a similar fold. Residues critical to the interaction with C5-convertase cluster on a mobile, hydrophobic inter-strand loop that protrudes from the open face of the beta-barrel. The opposite, helix-dominated face of C5-C345C carries a pair of exposed hydrophobic side chains adjacent to a striking negatively charged patch, consistent with affinity for positively charged factor I modules in C6 and C7. Modeling of homologous domains from complement proteins C3 and C4, which do not participate in membrane attack complex assembly, suggests that this provisionally identified C6/C7-interacting face is indeed specific to C5.


Subject(s)
Complement C5/chemistry , Amino Acid Sequence , Cell Membrane/metabolism , Complement C6/chemistry , Complement C7/chemistry , Escherichia coli/metabolism , Genetic Vectors , Humans , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Oligonucleotides/chemistry , Oligosaccharides/chemistry , Point Mutation , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Static Electricity , Stereoisomerism , Surface Plasmon Resonance , Time Factors
9.
J Immunol ; 173(7): 4547-52, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383587

ABSTRACT

Studies reported over 30 years ago revealed that latent, nonactivated C5 binds specifically and reversibly to C6 and C7. These reversible reactions are distinct from the essentially nonreversible associations with activated C5b that occur during assembly of the membrane attack complex, but they likely involve some, perhaps many, of the same molecular contacts. We recently reported that these reversible reactions are mediated by the C345C (NTR) domain at the C terminus of the C5 alpha-chain. Earlier work by others localized the complementary binding sites to a tryptic fragment of C6 composed entirely of two adjacent factor I modules (FIMs), and to a larger fragment of C7 composed of its homologous FIMs as well as two adjoining short consensus repeat modules. In this work, we expressed the tandem FIMs from C7 in bacteria. The mobility on SDS-polyacrylamide gels, lack of free sulfhydryl groups, and atypical circular dichroism spectrum of the recombinant product rC7-FIMs were all consistent with a native structure. Using surface plasmon resonance, we found that rC7-FIMs binds specifically to both C5 and the rC5-C345C domain with K(D) approximately 50 nM, and competes with C7 for binding to C5, as expected for an active domain. These results indicate that, like C6, the FIMs alone in C7 mediate reversible binding to C5. Based on available evidence, we suggest a model for an irreversible membrane attack complex assembly in which the C7 FIMs, but not those in C6, are bound to the C345C domain of C5 within the fully assembled complex.


Subject(s)
Complement C5/metabolism , Complement C7/metabolism , Complement Factor I/metabolism , Recombinant Proteins/metabolism , Amino Acid Motifs/genetics , Circular Dichroism , Complement C3/chemistry , Complement C6/chemistry , Complement C6/genetics , Complement C6/metabolism , Complement C7/chemistry , Complement C7/genetics , Complement Factor I/chemistry , Complement Factor I/genetics , Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/metabolism , Disulfides/chemistry , Escherichia coli/genetics , Humans , Netrin Receptors , Oxidation-Reduction , Protein Binding/genetics , Protein Binding/immunology , Protein Structure, Tertiary/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Cell Surface/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Surface Plasmon Resonance
10.
J Biol Chem ; 274(45): 31811-8, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542204

ABSTRACT

In order to elucidate the function of complement component C6, truncated C6 molecules were expressed recombinantly. These were either deleted of the factor I modules (FIMs) (C6des-748-913) or both complement control protein (CCP) modules and FIMs (C6des-611-913). C6des-748-913 exhibited approximately 60-70% of the hemolytic activity of full-length C6 when assayed for Alternative Pathway activity, but when measured for the Classical Pathway, C6des-748-914 was only 4-6% as effective as C6. The activity difference between C6 and C6des-748-913 for the two complement pathways can be explained by a greater stability of newly formed metastable C5b* when produced by the Alternative Pathway compared with that made by the Classical Pathway. The half-lives of metastable C5b* and the decay of (125)I-C5b measured from cells used to activate the Alternative Pathway were found to be about 5-12-fold longer than those same parameters derived from cells that had activated the Classical Pathway. (125)I-C5 binds reversibly to C6 in an ionic strength-dependent fashion, but (125)I-C5 binds only weakly to C6des-FIMs and not at all to C6des-CCP/FIMs. Therefore, although the FIMs are not required absolutely for C6 activity, these modules promote interaction of C6 with C5 enabling a more efficient bimolecular coupling ultimately leading to the formation of the C5b-6 complex.


Subject(s)
Complement C6/physiology , Complement Factor I/physiology , Animals , CHO Cells , Complement C6/chemistry , Complement C9/chemistry , Complement C9/physiology , Complement Factor I/chemistry , Complement Pathway, Alternative , Complement Pathway, Classical , Cricetinae , Humans , Rabbits , Sheep , Structure-Activity Relationship
11.
J Mol Evol ; 49(2): 282-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10441679

ABSTRACT

The plasma complement system comprises several activation pathways that share a common terminal route involving the assembly of the terminal complement complex (TCC), formed by C5b-C9. The order of emergence of the homologous components of TCC (C6, C7, C8alpha, C8beta, and C9) has been determined by phylogenetic analyses of their amino acid sequences. Using all the sequence data available for C6-C9 proteins, as well as for perforins, the results suggested that these TCC components originated from a single ancestral gene and that C6 and C7 were the earliest to emerge. Our evidence supports the notion that the ancestral gene had a complex modular composition. A series of gene duplications in combination with a tendency to lose modules resulted in successive complement proteins with decreasing modular complexity. C9 and perforin apparently are the result of different selective conditions to acquire pore-forming function. Thus C9 and perforin are examples of evolutionary parallelism.


Subject(s)
Complement C6/genetics , Complement C7/genetics , Complement C8/genetics , Complement C9/genetics , Evolution, Molecular , Phylogeny , Amino Acid Sequence , Animals , Complement C5/genetics , Complement C5b , Complement C6/chemistry , Complement C7/chemistry , Complement C8/chemistry , Complement C9/chemistry , Humans , Membrane Glycoproteins/genetics , Mice , Molecular Sequence Data , Perforin , Pore Forming Cytotoxic Proteins , Rabbits , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Time
12.
Biochim Biophys Acta ; 1342(1): 13-8, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9366265

ABSTRACT

Complement component C6 is known to contain two factor I modules in tandem at its C-terminus. To localize the disulfide bridges in those domains, native C6 was cleaved with trypsin, followed by subtilisin. The resulting digests were separated by reversed-phase HPLC, and all of the potential cystine-containing fragments were detected by a fluorescence assay and amino acid composition analyses. Final identification of the disulfide bonds was achieved by Edman degradation of the corresponding peptides. From the data gained a 1-3, 2-9, 4-7, 5-10, 6-8 pattern was determined (Cys752-Cys802, Cys763-Cys780, Cys765-Cys816, Cys772-Cys795, Cys841-Cys852, Cys846-Cys898, Cys859-Cys876, Cys861-Cys911, Cys867-Cys891). These findings are compared with the strongly related complement components C7 and factor I.


Subject(s)
Complement C6/chemistry , Cysteine , Cystine , Fibrinogen/chemistry , Amino Acid Sequence , Binding Sites , Chromatography, High Pressure Liquid , Disulfides , Humans , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Subtilisins , Trypsin
13.
J Immunol ; 157(8): 3648-57, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8871666

ABSTRACT

Combined subtotal deficiency of C6 and C7, in which both proteins are expressed at very low levels, has been observed in homozygous form in two families. A defect at the 5' splice donor site of intron 15 of the C6 gene explains the low molecular weight of the C6 protein and is probably responsible for its low expressed concentration. The C7 defect is more enigmatic: the protein is of normal molecular weight, low circulating concentration, and altered isoelectric point. An Arg > Ser codon substitution in exon 11 is the only molecular alteration within the mature C7 protein. These defects are associated with a characteristic set of polymorphic DNA markers in the C6/C7 region, forming a distinct haplotype. The haplotype has been found in combination with a number of other haplotypes containing defective genes that lead either to C6 or C7 deficiency, but with different consequences. Where it is combined with a C6-deficient gene, the serum C7 levels can be surprisingly high, possibly because there is no C6 generating C56 to consume the C7. In contrast, where the C7 genes are both defective (but still partially functional), there may be a profound deficit of circulating C7 because there is ample C6 to produce C56 and consume the already small amount of C7. Each molecular defect has also been found in isolation and has the expected effect.


Subject(s)
Complement C6/deficiency , Complement C6/genetics , Complement C7/deficiency , Complement C7/genetics , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Complement C6/chemistry , DNA/genetics , DNA Primers/genetics , Exons , Female , Genetic Markers , Haplotypes , Homozygote , Humans , Introns , Male , Molecular Sequence Data , Molecular Weight , Pedigree , Point Mutation , Polymerase Chain Reaction , Polymorphism, Genetic , RNA Splicing
14.
Immunology ; 75(1): 10-6, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1537585

ABSTRACT

A sensitive ELISA assay was used to quantitate serum complement component C6 concentrations. Levels in the range 0.3-3 micrograms/ml were measured in samples from eight individuals (four separate pedigrees) and two subjects with subtotal combined C6/C7 deficiency who have been reported previously. We defined C6 levels in this range as subtotal C6 deficiency (C6SD). In contrast, C6 deficiency with levels below 0.03 micrograms/ml was defined as C6Q0. C6Q0 has been found in 29 unrelated cases which have already been reported. Investigations of the properties of the C6 found in the C6SD subjects showed it to be haemolytically active and able to incorporate into the terminal complement complex. The protein had a relative molecular weight (Mr) of approximately 86% of normal C6 and this Mr was identical to that of the C6 of one combined deficient subject. The Mr of the C6 of the other combined deficient subject was previously estimated as 79% of the Mr of normal C6. Isoelectric focusing (IEF) analysis with band development by haemolytic overlay revealed that all C6SD samples produced an identical weak C6 band pattern anodal to normal C6A bands. The C7 IEF patterns of the two combined deficient subjects were identical, and the C6 IEF patterns of both were identical to those of the C6SD subjects. Thus the C6 of the combined deficient subjects is probably the same abnormal protein found in the C6SD individuals. None of the C6SD or combined deficient subjects have had meningococcal disease and it may be that low C6 levels afford some protection.


Subject(s)
Complement C6/chemistry , Complement C6/deficiency , Adult , Child , Complement C6/analysis , Complement C7/analysis , Complement Membrane Attack Complex/analysis , Enzyme-Linked Immunosorbent Assay , Female , Hemolysis/immunology , Humans , Immunoblotting , Isoelectric Focusing , Male , Middle Aged , Molecular Weight , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...