Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Microb Cell Fact ; 22(1): 259, 2023 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-38104077

RESUMEN

BACKGROUND: Komagataella phaffii (Pichia pastoris) is a methylotrophic commercially important non-conventional species of yeast that grows in a fermentor to exceptionally high densities on simple media and secretes recombinant proteins efficiently. Genetic engineering strategies are being explored in this organism to facilitate cost-effective biomanufacturing. Small, stable artificial chromosomes in K. phaffii could offer unique advantages by accommodating multiple integrations of extraneous genes and their promoters without accumulating perturbations of native chromosomes or exhausting the availability of selection markers. RESULTS: Here, we describe a linear "nano"chromosome (of 15-25 kb) that, according to whole-genome sequencing, persists in K. phaffii over many generations with a copy number per cell of one, provided non-homologous end joining is compromised (by KU70-knockout). The nanochromosome includes a copy of the centromere from K. phaffii chromosome 3, a K. phaffii-derived autonomously replicating sequence on either side of the centromere, and a pair of K. phaffii-like telomeres. It contains, within its q arm, a landing zone in which genes of interest alternate with long (approx. 1-kb) non-coding DNA chosen to facilitate homologous recombination and serve as spacers. The landing zone can be extended along the nanochromosome, in an inch-worming mode of sequential gene integrations, accompanied by recycling of just two antibiotic-resistance markers. The nanochromosome was used to express PDI, a gene encoding protein disulfide isomerase. Co-expression with PDI allowed the production, from a genomically integrated gene, of secreted murine complement factor H, a plasma protein containing 40 disulfide bonds. As further proof-of-principle, we co-expressed, from a nanochromosome, both PDI and a gene for GFP-tagged human complement factor H under the control of PAOX1 and demonstrated that the secreted protein was active as a regulator of the complement system. CONCLUSIONS: We have added K. phaffii to the list of organisms that can produce human proteins from genes carried on a stable, linear, artificial chromosome. We envisage using nanochromosomes as repositories for numerous extraneous genes, allowing intensive engineering of K. phaffii without compromising its genome or weakening the resulting strain.


Asunto(s)
Pichia , Saccharomycetales , Humanos , Animales , Ratones , Pichia/genética , Pichia/metabolismo , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Saccharomycetales/genética , Recombinación Homóloga , Cromosomas
2.
J Biol Chem ; 294(52): 20148-20163, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31719147

RESUMEN

Activation and suppression of the complement system compete on every serum-exposed surface, host or foreign. Potentially harmful outcomes of this competition depend on surface molecules through mechanisms that remain incompletely understood. Combining surface plasmon resonance (SPR) with atomic force microscopy (AFM), here we studied two complement system proteins at the single-molecule level: C3b, the proteolytically activated form of C3, and factor H (FH), the surface-sensing C3b-binding complement regulator. We used SPR to monitor complement initiation occurring through a positive-feedback loop wherein surface-deposited C3b participates in convertases that cleave C3, thereby depositing more C3b. Over multiple cycles of flowing factor B, factor D, and C3 over the SPR chip, we amplified C3b from ∼20 to ∼220 molecules·µm-2 AFM revealed C3b clusters of up to 20 molecules and solitary C3b molecules deposited up to 200 nm away from the clusters. A force of 0.17 ± 0.02 nanonewtons was needed to pull a single FH molecule, anchored to the AFM probe, from its complex with surface-attached C3b. The extent to which FH molecules stretched before detachment varied widely among complexes. Performing force-distance measurements with FH(D1119G), a variant lacking one of the C3b-binding sites and causing atypical hemolytic uremic syndrome, we found that it detached more uniformly and easily. In further SPR experiments, KD values between FH and C3b on a custom-made chip surface were 5-fold tighter than on commercial chips and similar to those on erythrocytes. These results suggest that the chemistry at the surface on which FH acts drives conformational adjustments that are functionally critical.


Asunto(s)
Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Microscopía de Fuerza Atómica , Resonancia por Plasmón de Superficie , Activación de Complemento , Complemento C3b/química , Complemento C3d/química , Complemento C3d/metabolismo , Factor H de Complemento/química , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Cinética , Unión Proteica
3.
Cell Microbiol ; 21(7): e13030, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30965383

RESUMEN

An effective vaccine is a priority for malaria control and elimination. The leading candidate in the Plasmodium falciparum blood stage is PfRh5. PfRh5 assembles into trimeric complex with PfRipr and PfCyRPA in the parasite, and this complex is essential for erythrocyte invasion. In this study, we show that antibodies specific for PfRh5 and PfCyRPA prevent trimeric complex formation. We identify the EGF-7 domain on PfRipr as a neutralising epitope and demonstrate that antibodies against this region act downstream of complex formation to prevent merozoite invasion. Antibodies against the C-terminal region of PfRipr were more inhibitory than those against either PfRh5 or PfCyRPA alone, and a combination of antibodies against PfCyRPA and PfRipr acted synergistically to reduce invasion. This study supports prioritisation of PfRipr for development as part of a next-generation antimalarial vaccine.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Antígenos de Protozoos/genética , Proteínas Portadoras/genética , Malaria Falciparum/tratamiento farmacológico , Proteínas Protozoarias/genética , Anticuerpos Neutralizantes/inmunología , Proteínas Portadoras/antagonistas & inhibidores , Eritrocitos/efectos de los fármacos , Eritrocitos/inmunología , Humanos , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/farmacología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Merozoítos/efectos de los fármacos , Merozoítos/inmunología , Plasmodium falciparum/inmunología , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/inmunología
4.
J Am Soc Nephrol ; 29(6): 1649-1661, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29588430

RESUMEN

Background C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway of complement activation, and treatment options for C3G remain limited. Complement factor H (FH) is a potent regulator of the alternative pathway and might offer a solution, but the mass and complexity of FH makes generation of full-length FH far from trivial. We previously generated a mini-FH construct, with FH short consensus repeats 1-5 linked to repeats 18-20 (FH1-5^18-20), that was effective in experimental C3G. However, the serum t1/2 of FH1-5^18-20 was significantly shorter than that of serum-purified FH.Methods We introduced the oligomerization domain of human FH-related protein 1 (denoted by R1-2) at the carboxy or amino terminus of human FH1-5^18-20 to generate two homodimeric mini-FH constructs (FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2, respectively) in Chinese hamster ovary cells and tested these constructs using binding, fluid-phase, and erythrocyte lysis assays, followed by experiments in FH-deficient Cfh-/- mice.Results FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2 homodimerized in solution and displayed avid binding profiles on clustered C3b surfaces, particularly FHR1-2^1-5^18-20 Each construct was >10-fold more effective than FH at inhibiting cell surface complement activity in vitro and restricted glomerular basement membrane C3 deposition in vivo significantly better than FH or FH1-5^18-20 FH1-5^18-20^R1-2 had a C3 breakdown fragment binding profile similar to that of FH, a >5-fold increase in serum t1/2 compared with that of FH1-5^18-20, and significantly better retention in the kidney than FH or FH1-5^18-20Conclusions FH1-5^18-20^R1-2 may have utility as a treatment option for C3G or other complement-mediated diseases.


Asunto(s)
Complemento C3/metabolismo , Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Factor H de Complemento/farmacocinética , Glomerulonefritis Membranoproliferativa/metabolismo , Animales , Factor H de Complemento/síntesis química , Factor H de Complemento/genética , Vía Alternativa del Complemento , Cricetinae , Membrana Basal Glomerular/metabolismo , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Semivida , Ratones , Unión Proteica , Ingeniería de Proteínas
5.
J Biol Chem ; 292(32): 13345-13360, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28637873

RESUMEN

Spontaneous activation enables the complement system to respond very rapidly to diverse threats. This activation is efficiently suppressed by complement factor H (CFH) on self-surfaces but not on foreign surfaces. The surface selectivity of CFH, a soluble protein containing 20 complement-control protein modules (CCPs 1-20), may be compromised by disease-linked mutations. However, which of the several functions of CFH drives this self-surface selectivity remains unknown. To address this, we expressed human CFH mutants in Pichia pastoris We found that recombinant I62-CFH (protective against age-related macular degeneration) and V62-CFH functioned equivalently, matching or outperforming plasma-derived CFH, whereas R53H-CFH, linked to atypical hemolytic uremic syndrome (aHUS), was defective in C3bBb decay-accelerating activity (DAA) and factor I cofactor activity (CA). The aHUS-linked CCP 19 mutant D1119G-CFH had virtually no CA on (self-like) sheep erythrocytes (ES) but retained DAA. The aHUS-linked CCP 20 mutant S1191L/V1197A-CFH (LA-CFH) had dramatically reduced CA on ES but was less compromised in DAA. D1119G-CFH and LA-CFH both performed poorly at preventing complement-mediated hemolysis of ES PspCN, a CFH-binding Streptococcus pneumoniae protein domain, binds CFH tightly and increases accessibility of CCPs 19 and 20. PspCN did not improve the DAA of any CFH variant on ES Conversely, PspCN boosted the CA, on ES, of I62-CFH, R53H-CFH, and LA-CFH and also enhanced hemolysis protection by I62-CFH and LA-CFH. We conclude that CCPs 19 and 20 are critical for efficient CA on self-surfaces but less important for DAA. Exposing CCPs 19 and 20 with PspCN and thus enhancing CA on self-surfaces may reverse deficiencies of some CFH variants.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Activación de Complemento , Degeneración Macular/genética , Mutación , Sustitución de Aminoácidos , Animales , Síndrome Hemolítico Urémico Atípico/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , C3 Convertasa de la Vía Alternativa del Complemento/química , C3 Convertasa de la Vía Alternativa del Complemento/genética , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Complemento C3d/química , Complemento C3d/genética , Complemento C3d/metabolismo , Factor H de Complemento/química , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Factor I de Complemento/química , Factor I de Complemento/genética , Factor I de Complemento/metabolismo , Eritrocitos/química , Hemólisis , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/metabolismo , Degeneración Macular/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Oveja Doméstica , Solubilidad , Streptococcus pneumoniae/metabolismo , Propiedades de Superficie
6.
J Immunol ; 196(2): 866-76, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26643478

RESUMEN

The serum proteins factor H (FH), consisting of 20 complement control protein modules (CCPs), and its splice product FH-like protein 1 (FHL-1; consisting of CCPs 1-7) are major regulators of the alternative pathway (AP) of complement activation. The engineered version of FH, miniFH, contains only the N- and C-terminal portions of FH linked by an optimized peptide and shows ∼ 10-fold higher ex vivo potency. We explored the hypothesis that regulatory potency is enhanced by unmasking of a ligand-binding site in the C-terminal CCPs 19-20 that is cryptic in full-length native FH. Therefore, we produced an FH variant lacking the central domains 10-15 (FHΔ10-15). To explore how avidity affects regulatory strength, we generated a duplicated version of miniFH, termed midiFH. We compared activities of FHΔ10-15 and midiFH to miniFH, FH, and FHL-1. Relative to FH, FHΔ10-15 exhibited an altered binding profile toward C3 activation products and a 5-fold-enhanced complement regulation on a paroxysmal nocturnal hemoglobinuria patient's erythrocytes. Contrary to dogma, FHL-1 and FH exhibited equal regulatory activity, suggesting that the role of FHL-1 in AP regulation has been underestimated. Unexpectedly, a substantially increased avidity for complement opsonins, as seen in midiFH, did not potentiate the inhibitory potential on host cells. In conclusion, comparisons of engineered and native FH-based regulators have identified features that determine high AP regulatory activity on host cells. Unrestricted availability of FH CCPs 19-20 and an optimal spatial orientation between the N- and C-terminal FH regions are key.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/inmunología , Factor H de Complemento/inmunología , Inactivadores del Complemento/farmacología , Vía Alternativa del Complemento/inmunología , Proteínas Recombinantes/farmacología , Secuencia de Aminoácidos , Factor H de Complemento/química , Inactivadores del Complemento/síntesis química , Inactivadores del Complemento/inmunología , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Unión Proteica , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/inmunología
7.
J Immunol ; 196(3): 1239-48, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26700768

RESUMEN

The human complement system is the frontline defense mechanism against invading pathogens. The coexistence of humans and microbes throughout evolution has produced ingenious molecular mechanisms by which microorganisms escape complement attack. A common evasion strategy used by diverse pathogens is the hijacking of soluble human complement regulators to their surfaces to afford protection from complement activation. One such host regulator is factor H (FH), which acts as a negative regulator of complement to protect host tissues from aberrant complement activation. In this report, we show that Plasmodium falciparum merozoites, the invasive form of the malaria parasites, actively recruit FH and its alternative spliced form FH-like protein 1 when exposed to human serum. We have mapped the binding site in FH that recognizes merozoites and identified Pf92, a member of the six-cysteine family of Plasmodium surface proteins, as its direct interaction partner. When bound to merozoites, FH retains cofactor activity, a key function that allows it to downregulate the alternative pathway of complement. In P. falciparum parasites that lack Pf92, we observed changes in the pattern of C3b cleavage that are consistent with decreased regulation of complement activation. These results also show that recruitment of FH affords P. falciparum merozoites protection from complement-mediated lysis. Our study provides new insights on mechanisms of immune evasion of malaria parasites and highlights the important function of surface coat proteins in the interplay between complement regulation and successful infection of the host.


Asunto(s)
Activación de Complemento/inmunología , Factor H de Complemento/inmunología , Evasión Inmune/inmunología , Malaria Falciparum/inmunología , Western Blotting , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Merozoítos/inmunología
8.
Mol Cell Proteomics ; 15(8): 2730-43, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27250206

RESUMEN

The slow but spontaneous and ubiquitous formation of C3(H2O), the hydrolytic and conformationally rearranged product of C3, initiates antibody-independent activation of the complement system that is a key first line of antimicrobial defense. The structure of C3(H2O) has not been determined. Here we subjected C3(H2O) to quantitative cross-linking/mass spectrometry (QCLMS). This revealed details of the structural differences and similarities between C3(H2O) and C3, as well as between C3(H2O) and its pivotal proteolytic cleavage product, C3b, which shares functionally similarity with C3(H2O). Considered in combination with the crystal structures of C3 and C3b, the QCMLS data suggest that C3(H2O) generation is accompanied by the migration of the thioester-containing domain of C3 from one end of the molecule to the other. This creates a stable C3b-like platform able to bind the zymogen, factor B, or the regulator, factor H. Integration of available crystallographic and QCLMS data allowed the determination of a 3D model of the C3(H2O) domain architecture. The unique arrangement of domains thus observed in C3(H2O), which retains the anaphylatoxin domain (that is excised when C3 is enzymatically activated to C3b), can be used to rationalize observed differences between C3(H2O) and C3b in terms of complement activation and regulation.


Asunto(s)
Complemento C3/química , Complemento C3b/química , Espectrometría de Masas/métodos , Reactivos de Enlaces Cruzados , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica , Dominios Proteicos
9.
PLoS Pathog ; 11(2): e1004670, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25723550

RESUMEN

During blood stage Plasmodium falciparum infection, merozoites invade uninfected erythrocytes via a complex, multistep process involving a series of distinct receptor-ligand binding events. Understanding each element in this process increases the potential to block the parasite's life cycle via drugs or vaccines. To investigate specific receptor-ligand interactions, they were systematically blocked using a combination of genetic deletion, enzymatic receptor cleavage and inhibition of binding via antibodies, peptides and small molecules, and the resulting temporal changes in invasion and morphological effects on erythrocytes were filmed using live cell imaging. Analysis of the videos have shown receptor-ligand interactions occur in the following sequence with the following cellular morphologies; 1) an early heparin-blockable interaction which weakly deforms the erythrocyte, 2) EBA and PfRh ligands which strongly deform the erythrocyte, a process dependant on the merozoite's actin-myosin motor, 3) a PfRh5-basigin binding step which results in a pore or opening between parasite and host through which it appears small molecules and possibly invasion components can flow and 4) an AMA1-RON2 interaction that mediates tight junction formation, which acts as an anchor point for internalization. In addition to enhancing general knowledge of apicomplexan biology, this work provides a rational basis to combine sequentially acting merozoite vaccine candidates in a single multi-receptor-blocking vaccine.


Asunto(s)
Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Plasmodium falciparum/patogenicidad , Receptores de Superficie Celular/metabolismo , Animales , Antígenos de Protozoos/metabolismo , Basigina/metabolismo , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Forma de la Célula , Células Cultivadas , Eritrocitos/metabolismo , Eritrocitos/patología , Interacciones Huésped-Parásitos/fisiología , Ligandos , Malaria Falciparum/metabolismo , Proteínas de la Membrana/metabolismo , Merozoítos/metabolismo , Merozoítos/patología , Plasmodium falciparum/metabolismo , Unión Proteica , Proteínas Protozoarias/metabolismo , Conejos , Transducción de Señal
10.
J Immunol ; 195(10): 4986-98, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26459349

RESUMEN

In an attempt to evade annihilation by the vertebrate complement system, many microbes capture factor H (FH), the key soluble complement-regulating protein in human plasma. However, FH is normally an active complement suppressor exclusively on self-surfaces and this selective action of FH is pivotal to self versus non-self discrimination by the complement system. We investigated whether the bacterially captured FH becomes functionally enhanced and, if so, how this is achieved at a structural level. We found, using site-directed and truncation mutagenesis, surface plasmon resonance, nuclear magnetic resonance spectroscopy, and cross-linking and mass spectrometry, that the N-terminal domain of Streptococcus pneumoniae protein PspC (PspCN) not only binds FH extraordinarily tightly but also holds it in a previously uncharacterized conformation. Functional enhancement arises from exposure of a C-terminal cryptic second binding site in FH for C3b, the activation-specific fragment of the pivotal complement component, C3. This conformational change of FH doubles its affinity for C3b and increases 5-fold its ability to accelerate decay of the binary enzyme (C3bBb) responsible for converting C3 to C3b in an amplification loop. Despite not sharing critical FH-binding residues, PspCNs from D39 and Tigr4 S. pneumoniae exhibit similar FH-anchoring and enhancing properties. We propose that these bacterial proteins mimic molecular markers of self-surfaces, providing a compelling hypothesis for how FH prevents complement-mediated injury to host tissue while lacking efficacy on virtually all other surfaces. In hemolysis assays with 2-aminoethylisothiouronium bromide-treated erythrocytes that recapitulate paroxysmal nocturnal hemoglobinuria, PspCN enhanced protection of cells by FH, suggesting a new paradigm for therapeutic complement suppression.


Asunto(s)
Proteínas Bacterianas/química , Complemento C3b/química , Factor H de Complemento/química , Streptococcus pneumoniae/química , Proteínas Bacterianas/inmunología , Complemento C3b/inmunología , Factor H de Complemento/inmunología , Hemoglobinuria Paroxística/inmunología , Humanos , Estructura Terciaria de Proteína , Streptococcus pneumoniae/inmunología
11.
J Biol Chem ; 289(1): 450-63, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24214979

RESUMEN

To survive and replicate within the human host, malaria parasites must invade erythrocytes. Invasion can be mediated by the P. falciparum reticulocyte-binding homologue protein 4 (PfRh4) on the merozoite surface interacting with complement receptor type 1 (CR1, CD35) on the erythrocyte membrane. The PfRh4 attachment site lies within the three N-terminal complement control protein modules (CCPs 1-3) of CR1, which intriguingly also accommodate binding and regulatory sites for the key complement activation-specific proteolytic products, C3b and C4b. One of these regulatory activities is decay-accelerating activity. Although PfRh4 does not impact C3b/C4b binding, it does inhibit this convertase disassociating capability. Here, we have employed ELISA, co-immunoprecipitation, and surface plasmon resonance to demonstrate that CCP 1 contains all the critical residues for PfRh4 interaction. We fine mapped by homologous substitution mutagenesis the PfRh4-binding site on CCP 1 and visualized it with a solution structure of CCPs 1-3 derived by NMR and small angle x-ray scattering. We cross-validated these results by creating an artificial PfRh4-binding site through substitution of putative PfRh4-interacting residues from CCP 1 into their homologous positions within CCP 8; strikingly, this engineered binding site had an ∼30-fold higher affinity for PfRh4 than the native one in CCP 1. These experiments define a candidate site on CR1 by which P. falciparum merozoites gain access to human erythrocytes in a non-sialic acid-dependent pathway of merozoite invasion.


Asunto(s)
Proteínas de la Membrana/metabolismo , Merozoítos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Receptores de Complemento 3b/metabolismo , Sitios de Unión , Complemento C3b/química , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C4b/química , Complemento C4b/genética , Complemento C4b/metabolismo , Eritrocitos/química , Eritrocitos/metabolismo , Eritrocitos/parasitología , Células HEK293 , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Merozoítos/química , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Plasmodium falciparum/química , Plasmodium falciparum/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Dispersión del Ángulo Pequeño , Resonancia por Plasmón de Superficie , Difracción de Rayos X
12.
Biochem Soc Trans ; 43(5): 812-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26517887

RESUMEN

Complement control protein modules (CCPs) occur in numerous functionally diverse extracellular proteins. Also known as short consensus repeats (SCRs) or sushi domains each CCP contains approximately 60 amino acid residues, including four consensus cysteines participating in two disulfide bonds. Varying in length and sequence, CCPs adopt a ß-sandwich type fold and have an overall prolate spheroidal shape with N- and C-termini lying close to opposite poles of the long axis. CCP-containing proteins are important as cytokine receptors and in neurotransmission, cell adhesion, blood clotting, extracellular matrix formation, haemoglobin metabolism and development, but CCPs are particularly well represented in the vertebrate complement system. For example, factor H (FH), a key soluble regulator of the alternative pathway of complement activation, is made up entirely from a chain of 20 CCPs joined by short linkers. Collectively, therefore, the 20 CCPs of FH must mediate all its functional capabilities. This is achieved via collaboration and division of labour among these modules. Structural studies have illuminated the dynamic architectures that allow FH and other CCP-rich proteins to perform their biological functions. These are largely the products of a highly varied set of intramolecular interactions between CCPs. The CCP can act as building block, spacer, highly versatile recognition site or dimerization mediator. Tandem CCPs may form composite binding sites or contribute to flexible, rigid or conformationally 'switchable' segments of the parent proteins.


Asunto(s)
Enzimas Activadoras de Complemento/química , Activación de Complemento , Proteínas Inactivadoras de Complemento/química , Diseño de Fármacos , Modelos Moleculares , Ingeniería de Proteínas , Animales , Sitios de Unión , Enzimas Activadoras de Complemento/genética , Enzimas Activadoras de Complemento/metabolismo , Factor H de Complemento/química , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Inactivadores del Complemento/química , Inactivadores del Complemento/metabolismo , Inactivadores del Complemento/farmacología , Proteínas Inactivadoras de Complemento/genética , Proteínas Inactivadoras de Complemento/metabolismo , Humanos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
13.
J Immunol ; 190(11): 5712-21, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23616575

RESUMEN

Inadequate control of the complement system is the underlying or aggravating factor in many human diseases. Whereas treatment options that specifically target the alternative pathway (AP) of complement activation are considered highly desirable, no such option is available in the clinic. In this study, we present a successful example of protein engineering, guided by structural insight on the complement regulator factor H (FH), yielding a novel complement-targeted therapeutic (mini-FH) with clinical potential. Despite a 70% reduction in size, mini-FH retained and in some respects exceeded the regulatory activity and cell surface-recognition properties of its parent protein FH, including the recently described recognition of sites of oxidative stress. Importantly, the chosen design extended the functional spectrum of the inhibitor, as mini-FH showed increased binding to the surface-bound opsonins iC3b and C3dg when compared with FH. Thus, mini-FH is equipped with a unique and clinically valuable triple-targeting profile toward diseased host cells, through its binding to sites of ongoing complement activation, markers of oxidative damage, and host surface-specific polyanions. When assessed in a clinically relevant AP-mediated disease model of paroxysmal nocturnal hemoglobinuria, mini-FH largely outperformed FH and indicated advantages over clinically evaluated AP inhibitors. Thus, the rational engineering of a streamlined FH construct not only provided insight into the function of a key complement regulator, but also yielded a novel inhibitor that combines a triple-targeting approach with high AP-specific inhibitory activity (IC50 ~ 40 nM), which may pave the way toward new options for the treatment of complement-mediated diseases.


Asunto(s)
Factor H de Complemento/química , Factores Inmunológicos/química , Ingeniería de Proteínas , Secuencia de Aminoácidos , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Factor H de Complemento/farmacología , Vía Alternativa del Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Hemólisis/efectos de los fármacos , Hemólisis/inmunología , Humanos , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Unión Proteica/inmunología , Proteínas Recombinantes de Fusión
14.
J Am Soc Nephrol ; 25(11): 2425-33, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24722444

RESUMEN

Complement C3 activation is a characteristic finding in membranoproliferative GN (MPGN). This activation can be caused by immune complex deposition or an acquired or inherited defect in complement regulation. Deficiency of complement factor H has long been associated with MPGN. More recently, heterozygous genetic variants have been reported in sporadic cases of MPGN, although their functional significance has not been assessed. We describe a family with MPGN and acquired partial lipodystrophy. Although C3 nephritic factor was shown in family members with acquired partial lipodystrophy, it did not segregate with the renal phenotype. Genetic analysis revealed a novel heterozygous mutation in complement factor H (R83S) in addition to known risk polymorphisms carried by individuals with MPGN. Patients with MPGN had normal levels of factor H, and structural analysis of the mutant revealed only subtle alterations. However, functional analysis revealed profoundly reduced C3b binding, cofactor activity, and decay accelerating activity leading to loss of regulation of the alternative pathway. In summary, this family showed a confluence of common and rare functionally significant genetic risk factors causing disease. Data from our analysis of these factors highlight the role of the alternative pathway of complement in MPGN.


Asunto(s)
Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Vía Alternativa del Complemento/genética , Eritrocitos/inmunología , Glomerulonefritis Membranoproliferativa/genética , Glomerulonefritis Membranoproliferativa/inmunología , Enfermedades Renales/genética , Animales , Factor H de Complemento/química , Factor H de Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Cristalografía por Rayos X , Eritrocitos/citología , Salud de la Familia , Femenino , Haplotipos , Enfermedades por Deficiencia de Complemento Hereditario , Heterocigoto , Humanos , Enfermedades Renales/inmunología , Masculino , Linaje , Polimorfismo Genético , Estructura Terciaria de Proteína , Ovinos , Relación Estructura-Actividad
15.
J Comput Chem ; 35(7): 518-25, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24323885

RESUMEN

In this study, we examine the feasibility and limitations of describing the motional behavior of three-domain proteins in which the domains are linearly connected. In addition to attempting the determination of the internal and overall reorientational correlation times, we investigate the existence of correlations in the motions between the three domains. Since in linearly arranged three-domain proteins, there are typically no experimental data that can directly report on motional correlation between the first and the third domain, we address this question by dynamics simulations. Two limiting cases occur: (1) for weak repulsive potentials and (2) when strong repulsive potentials are applied between sequential domains. The motions of the terminal domains become correlated in the case of strong interdomain repulsive potentials when these potentials do not allow the angle between the sequential domains to be smaller than about 60°. Using the model-free (MF) and extended MF formalisms of Lipari and Szabo, we find that the motional behavior can be separated into two components; the first component represents the concerted overall motion of the three domains, and the second describes the independent component of the motion of each individual domain. We find that this division of the motional behavior of the protein is maintained only when their timescales are distinct and can be made when the angles between sequential domains remain between 60° and 160°. In this work, we identify and quantify interdomain motional correlations.


Asunto(s)
Proteínas/química , Algoritmos , Simulación del Acoplamiento Molecular , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Estructura Terciaria de Proteína
16.
J Am Soc Nephrol ; 24(1): 53-65, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23204401

RESUMEN

Factor H (fH) and properdin both modulate complement; however, fH inhibits activation, and properdin promotes activation of the alternative pathway of complement. Mutations in fH associate with several human kidney diseases, but whether inhibiting properdin would be beneficial in these diseases is unknown. Here, we found that either genetic or pharmacological blockade of properdin, which we expected to be therapeutic, converted the mild C3 GN of an fH-mutant mouse to a lethal C3 GN with features of human dense deposit disease. We attributed this phenotypic change to a differential effect of properdin on the dynamics of alternative pathway complement activation in the fluid phase and the cell surface in the fH-mutant mice. Thus, in fH mutation-related C3 glomerulopathy, additional factors that impact the activation of the alternative pathway of complement critically determine the nature and severity of kidney pathology. These results show that therapeutic manipulation of the complement system requires rigorous disease-specific target validation.


Asunto(s)
Glomerulonefritis Membranoproliferativa/genética , Enfermedades Renales/genética , Properdina/deficiencia , Animales , Complemento C3/metabolismo , Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Vía Alternativa del Complemento , Modelos Animales de Enfermedad , Glomerulonefritis Membranoproliferativa/metabolismo , Glomerulonefritis Membranoproliferativa/patología , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Glomérulos Renales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Mutación
17.
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
18.
J Biol Chem ; 287(39): 32381-93, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22843697

RESUMEN

Paralogs NDE1 (nuclear distribution element 1) and NDEL1 (NDE-like 1) are essential for mitosis and neurodevelopment. Both proteins are predicted to have similar structures, based upon high sequence similarity, and they co-complex in mammalian cells. X-ray diffraction studies and homology modeling suggest that their N-terminal regions (residues 8-167) adopt continuous, extended α-helical coiled-coil structures, but no experimentally derived information on the structure of their C-terminal regions or the architecture of the full-length proteins is available. In the case of NDE1, no biophysical data exists. Here we characterize the structural architecture of both full-length proteins utilizing negative stain electron microscopy along with our established paradigm of chemical cross-linking followed by tryptic digestion, mass spectrometry, and database searching, which we enhance using isotope labeling for mixed NDE1-NDEL1. We determined that full-length NDE1 forms needle-like dimers and tetramers in solution, similar to crystal structures of NDEL1, as well as chain-like end-to-end polymers. The C-terminal domain of each protein, required for interaction with key protein partners dynein and DISC1 (disrupted-in-schizophrenia 1), includes a predicted disordered region that allows a bent back structure. This facilitates interaction of the C-terminal region with the N-terminal coiled-coil domain and is in agreement with previous results showing N- and C-terminal regions of NDEL1 and NDE1 cooperating in dynein interaction. It sheds light on recently identified mutations in the NDE1 gene that cause truncation of the encoded protein. Additionally, analysis of mixed NDE1-NDEL1 complexes demonstrates that NDE1 and NDEL1 can interact directly.


Asunto(s)
Proteínas Portadoras/química , Proteínas Asociadas a Microtúbulos/química , Modelos Moleculares , Pliegue de Proteína , Multimerización de Proteína/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
19.
J Exp Med ; 204(10): 2277-83, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17893204

RESUMEN

Nearly 50 million people worldwide suffer from age-related macular degeneration (AMD), which causes severe loss of central vision. A single-nucleotide polymorphism in the gene for the complement regulator factor H (FH), which causes a Tyr-to-His substitution at position 402, is linked to approximately 50% of attributable risks for AMD. We present the crystal structure of the region of FH containing the polymorphic amino acid His402 in complex with an analogue of the glycosaminoglycans (GAGs) that localize the complement regulator on the cell surface. The structure demonstrates direct coordination of ligand by the disease-associated polymorphic residue, providing a molecular explanation of the genetic observation. This glycan-binding site occupies the center of an extended interaction groove on the regulator's surface, implying multivalent binding of sulfated GAGs. This finding is confirmed by structure-based site-directed mutagenesis, nuclear magnetic resonance-monitored binding experiments performed for both H402 and Y402 variants with this and another model GAG, and analysis of an extended GAG-FH complex.


Asunto(s)
Envejecimiento/fisiología , Factor H de Complemento/química , Factor H de Complemento/metabolismo , Sitios de Unión , Factor H de Complemento/genética , Cristalografía por Rayos X , Productos del Gen gag/química , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Ligandos , Modelos Moleculares , Mutación/genética , Polisacáridos/química , Polisacáridos/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Sacarosa/análogos & derivados , Sacarosa/química , Sacarosa/metabolismo , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA