Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(10): 1299-1310, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31534238

RESUMEN

Resisting and tolerating microbes are alternative strategies to survive infection, but little is known about the evolutionary mechanisms controlling this balance. Here genomic analyses of anatomically modern humans, extinct Denisovan hominins and mice revealed a TNFAIP3 allelic series with alterations in the encoded immune response inhibitor A20. Each TNFAIP3 allele encoded substitutions at non-catalytic residues of the ubiquitin protease OTU domain that diminished IκB kinase-dependent phosphorylation and activation of A20. Two TNFAIP3 alleles encoding A20 proteins with partial phosphorylation deficits seemed to be beneficial by increasing immunity without causing spontaneous inflammatory disease: A20 T108A;I207L, originating in Denisovans and introgressed in modern humans throughout Oceania, and A20 I325N, from an N-ethyl-N-nitrosourea (ENU)-mutagenized mouse strain. By contrast, a rare human TNFAIP3 allele encoding an A20 protein with 95% loss of phosphorylation, C243Y, caused spontaneous inflammatory disease in humans and mice. Analysis of the partial-phosphorylation A20 I325N allele in mice revealed diminished tolerance of bacterial lipopolysaccharide and poxvirus inoculation as tradeoffs for enhanced immunity.


Asunto(s)
Infecciones por Poxviridae/inmunología , Poxviridae/fisiología , Dominios Proteicos/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Alelos , Animales , Extinción Biológica , Humanos , Inmunidad , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense/genética , Fosforilación
2.
J Immunol ; 209(9): 1746-1759, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36162872

RESUMEN

α1-Antitrypsin (AAT), a serine protease inhibitor, is the third most abundant protein in plasma. Although the best-known function of AAT is irreversible inhibition of elastase, AAT is an acute-phase reactant and is increasingly recognized to have a panoply of other functions, including as an anti-inflammatory mediator and a host-protective molecule against various pathogens. Although a canonical receptor for AAT has not been identified, AAT can be internalized into the cytoplasm and is known to affect gene regulation. Because AAT has anti-inflammatory properties, we examined whether AAT binds the cytoplasmic glucocorticoid receptor (GR) in human macrophages. We report the finding that AAT binds to GR using several approaches, including coimmunoprecipitation, mass spectrometry, and microscale thermophoresis. We also performed in silico molecular modeling and found that binding between AAT and GR has a plausible stereochemical basis. The significance of this interaction in macrophages is evinced by AAT inhibition of LPS-induced NF-κB activation and IL-8 production as well as AAT induction of angiopoietin-like 4 protein, which are, in part, dependent on GR. Furthermore, this AAT-GR interaction contributes to a host-protective role against mycobacteria in macrophages. In summary, this study identifies a new mechanism for the gene regulation, anti-inflammatory, and host-defense properties of AAT.


Asunto(s)
Receptores de Glucocorticoides , alfa 1-Antitripsina , Humanos , alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina , Angiopoyetinas/metabolismo , Angiopoyetinas/uso terapéutico , Antiinflamatorios/uso terapéutico , Interleucina-8/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , FN-kappa B/metabolismo , Elastasa Pancreática/metabolismo , Receptores de Glucocorticoides/metabolismo , Inhibidores de Serina Proteinasa
3.
Int J Mol Sci ; 25(3)2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38339082

RESUMEN

Serine proteases are members of a large family of hydrolytic enzymes in which a particular serine residue in the active site performs an essential role as a nucleophile, which is required for their proteolytic cleavage function. The array of functions performed by serine proteases is vast and includes, among others, the following: (i) the ability to fight infections; (ii) the activation of blood coagulation or blood clot lysis systems; (iii) the activation of digestive enzymes; and (iv) reproduction. Serine protease activity is highly regulated by multiple families of protease inhibitors, known collectively as the SERine Protease INhibitor (SERPIN). The serpins use a conformational change mechanism to inhibit proteases in an irreversible way. The unusual conformational change required for serpin function provides an elegant opportunity for allosteric regulation by the binding of cofactors, of which the most well-studied is heparin. The goal of this review is to discuss some of the clinically relevant serine protease-serpin interactions that may be enhanced by heparin or other negatively charged polysaccharides. The paired serine protease-serpin in the framework of heparin that we review includes the following: thrombin-antithrombin III, plasmin-anti-plasmin, C1 esterase/kallikrein-C1 esterase inhibitor, and furin/TMPRSS2 (serine protease Transmembrane Protease 2)-alpha-1-antitrypsin, with the latter in the context of COVID-19 and prostate cancer.


Asunto(s)
Serpinas , Serpinas/metabolismo , Heparina/química , Serina Proteasas , Inhibidores de Serina Proteinasa/metabolismo , Anticoagulantes , Trombina/metabolismo
4.
J Biol Chem ; 298(7): 102119, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35691342

RESUMEN

The metal-dependent M17 aminopeptidases are conserved throughout all kingdoms of life. This large enzyme family is characterized by a conserved binuclear metal center and a distinctive homohexameric arrangement. Recently, we showed that hexamer formation in Plasmodium M17 aminopeptidases was controlled by the metal ion environment, although the functional necessity for hexamer formation is still unclear. To further understand the mechanistic role of the hexameric assembly, here we undertook an investigation of the structure and dynamics of the M17 aminopeptidase from Plasmodium falciparum, PfA-M17. We describe a novel structure of PfA-M17, which shows that the active sites of each trimer are linked by a dynamic loop, and loop movement is coupled with a drastic rearrangement of the binuclear metal center and substrate-binding pocket, rendering the protein inactive. Molecular dynamics simulations and biochemical analyses of PfA-M17 variants demonstrated that this rearrangement is inherent to PfA-M17, and that the transition between the active and inactive states is metal dependent and part of a dynamic regulatory mechanism. Key to the mechanism is a remodeling of the binuclear metal center, which occurs in response to a signal from the neighboring active site and serves to moderate the rate of proteolysis under different environmental conditions. In conclusion, this work identifies a precise mechanism by which oligomerization contributes to PfA-M17 function. Furthermore, it describes a novel role for metal cofactors in the regulation of enzymes, with implications for the wide range of metalloenzymes that operate via a two-metal ion catalytic center, including DNA processing enzymes and metalloproteases.


Asunto(s)
Aminopeptidasas , Plasmodium falciparum/enzimología , Aminopeptidasas/química , Aminopeptidasas/metabolismo , Dominio Catalítico , Metales/metabolismo , Plasmodium falciparum/metabolismo
5.
Biochem Soc Trans ; 51(3): 1361-1375, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37294003

RESUMEN

Alpha-1-antitrypsin (AAT), a serine protease inhibitor (serpin), is increasingly recognized to inhibit SARS-CoV-2 infection and counter many of the pathogenic mechanisms of COVID-19. Herein, we reviewed the epidemiologic evidence, the molecular mechanisms, and the clinical evidence that support this paradigm. As background to our discussion, we first examined the basic mechanism of SARS-CoV-2 infection and contend that despite the availability of vaccines and anti-viral agents, COVID-19 remains problematic due to viral evolution. We next underscored that measures to prevent severe COVID-19 currently exists but teeters on a balance and that current treatment for severe COVID-19 remains grossly suboptimal. We then reviewed the epidemiologic and clinical evidence that AAT deficiency increases risk of COVID-19 infection and of more severe disease, and the experimental evidence that AAT inhibits cell surface transmembrane protease 2 (TMPRSS2) - a host serine protease required for SARS-CoV-2 entry into cells - and that this inhibition may be augmented by heparin. We also elaborated on the panoply of other activities of AAT (and heparin) that could mitigate severity of COVID-19. Finally, we evaluated the available clinical evidence for AAT treatment of COVID-19.


Asunto(s)
COVID-19 , Deficiencia de alfa 1-Antitripsina , Humanos , Heparina , Epidemiología Molecular , SARS-CoV-2
6.
Proc Natl Acad Sci U S A ; 117(36): 22341-22350, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32855302

RESUMEN

Conformational diversity and self-cross-reactivity of antigens have been correlated with evasion from neutralizing antibody responses. We utilized single cell B cell sequencing, biolayer interferometry and X-ray crystallography to trace mutation selection pathways where the antibody response must resolve cross-reactivity between foreign and self-proteins bearing near-identical contact surfaces, but differing in conformational flexibility. Recurring antibody mutation trajectories mediate long-range rearrangements of framework (FW) and complementarity determining regions (CDRs) that increase binding site conformational diversity. These antibody mutations decrease affinity for self-antigen 19-fold and increase foreign affinity 67-fold, to yield a more than 1,250-fold increase in binding discrimination. These results demonstrate how conformational diversity in antigen and antibody does not act as a barrier, as previously suggested, but rather facilitates high affinity and high discrimination between foreign and self.


Asunto(s)
Anticuerpos , Diversidad de Anticuerpos/genética , Autoantígenos , Reordenamiento Génico de Linfocito B/genética , Mutación/genética , Animales , Anticuerpos/química , Anticuerpos/genética , Anticuerpos/metabolismo , Afinidad de Anticuerpos/genética , Autoanticuerpos/química , Autoanticuerpos/genética , Autoanticuerpos/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Regiones Determinantes de Complementariedad/genética , Inmunidad Humoral/genética , Ratones , Modelos Moleculares , Conformación Proteica , Hipermutación Somática de Inmunoglobulina/genética
7.
J Biol Chem ; 296: 100447, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33617878

RESUMEN

The fibronectin type III (FN3) monobody domain is a promising non-antibody scaffold, which features a less complex architecture than an antibody while maintaining analogous binding loops. We previously developed FN3Con, a hyperstable monobody derivative with diagnostic and therapeutic potential. Prestabilization of the scaffold mitigates the stability-function trade-off commonly associated with evolving a protein domain toward biological activity. Here, we aimed to examine if the FN3Con monobody could take on antibody-like binding to therapeutic targets, while retaining its extreme stability. We targeted the first of the Adnectin derivative of monobodies to reach clinical trials, which was engineered by directed evolution for binding to the therapeutic target VEGFR2; however, this function was gained at the expense of large losses in thermostability and increased oligomerization. In order to mitigate these losses, we grafted the binding loops from Adnectin-anti-VEGFR2 (CT-322) onto the prestabilized FN3Con scaffold to produce a domain that successfully bound with high affinity to the therapeutic target VEGFR2. This FN3Con-anti-VEGFR2 construct also maintains high thermostability, including remarkable long-term stability, retaining binding activity after 2 years of storage at 36 °C. Further investigations into buffer excipients doubled the presence of monomeric monobody in accelerated stability trials. These data suggest that loop grafting onto a prestabilized scaffold is a viable strategy for the development of monobody domains with desirable biophysical characteristics and that FN3Con is therefore well-suited to applications such as the evolution of multiple paratopes or shelf-stable diagnostics and therapeutics.


Asunto(s)
Anticuerpos/metabolismo , Dominio de Fibronectina del Tipo III/genética , Anticuerpos/inmunología , Dominio de Fibronectina del Tipo III/inmunología , Fibronectinas/genética , Fibronectinas/inmunología , Fibronectinas/metabolismo , Ingeniería Genética/métodos , Humanos , Regiones de Fijación a la Matriz , Mutación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Unión Proteica/genética , Unión Proteica/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Mol Biol Evol ; 38(7): 2915-2929, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-33744972

RESUMEN

Serine protease inhibitors (serpins) are found in all kingdoms of life and play essential roles in multiple physiological processes. Owing to the diversity of the superfamily, phylogenetic analysis is challenging and prokaryotic serpins have been speculated to have been acquired from Metazoa through horizontal gene transfer due to their unexpectedly high homology. Here, we have leveraged a structural alignment of diverse serpins to generate a comprehensive 6,000-sequence phylogeny that encompasses serpins from all kingdoms of life. We show that in addition to a central "hub" of highly conserved serpins, there has been extensive diversification of the superfamily into many novel functional clades. Our analysis indicates that the hub proteins are ancient and are similar because of convergent evolution, rather than the alternative hypothesis of horizontal gene transfer. This work clarifies longstanding questions in the evolution of serpins and provides new directions for research in the field of serpin biology.


Asunto(s)
Evolución Molecular , Familia de Multigenes , Filogenia , Serpinas/genética , Animales , Bacterias/genética , Cordados/genética , Invertebrados/genética , Plantas/genética
9.
Mol Microbiol ; 116(2): 397-415, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33756056

RESUMEN

Endolysin enzymes from bacteriophage cause bacterial lysis by degrading the peptidoglycan cell wall. The streptococcal C1 phage endolysin PlyC, is the most potent endolysin described to date and can rapidly lyse group A, C, and E streptococci. PlyC is known to bind the Group A streptococcal cell wall, but the specific molecular target or the binding site within PlyC remain uncharacterized. Here we report for the first time, that the polyrhamnose backbone of the Group A streptococcal cell wall is the binding target of PlyC. We have also characterized the putative rhamnose binding groove of PlyC and found four key residues that were critical to either the folding or the cell wall binding action of PlyC. Based on our results, we suggest that the interaction between PlyC and the cell wall may not be a high-affinity interaction as previously proposed, but rather a high avidity one, allowing for PlyC's remarkable lytic activity. Resistance to our current antibiotics is reaching crisis levels and there is an urgent need to develop the antibacterial agents with new modes of action. A detailed understanding of this potent endolysin may facilitate future developments of PlyC as a tool against the rise of antibiotic resistance.


Asunto(s)
Bacteriófagos/metabolismo , Endopeptidasas/metabolismo , Peptidoglicano/metabolismo , Ramnosa/metabolismo , Streptococcus pyogenes/virología , Bacteriófagos/genética , Sitios de Unión/fisiología , Membrana Celular/metabolismo , Pared Celular/metabolismo , Endopeptidasas/genética , Simulación del Acoplamiento Molecular , Unión Proteica/fisiología , Streptococcus pyogenes/metabolismo
10.
Biochem J ; 478(17): 3319-3330, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34424335

RESUMEN

Angiotensinogen fine-tunes the tightly controlled activity of the renin-angiotensin system by modulating the release of angiotensin peptides that control blood pressure. One mechanism by which this modulation is achieved is via angiotensinogen's Cys18-Cys138 disulfide bond that acts as a redox switch. Molecular dynamics simulations of each redox state of angiotensinogen reveal subtle dynamic differences between the reduced and oxidised forms, particularly at the N-terminus. Surface plasmon resonance data demonstrate that the two redox forms of angiotensinogen display different binding kinetics to an immobilised anti-angiotensinogen monoclonal antibody. Mass spectrometry mapped the epitope for the antibody to the N-terminal region of angiotensinogen. We therefore provide evidence that the different redox forms of angiotensinogen can be detected by an antibody-based detection method.


Asunto(s)
Angiotensinógeno/química , Angiotensinógeno/metabolismo , Simulación de Dinámica Molecular , Resonancia por Plasmón de Superficie/métodos , Angiotensinógeno/genética , Angiotensinógeno/inmunología , Anticuerpos Monoclonales/inmunología , Presión Sanguínea/fisiología , Cisteína/metabolismo , Disulfuros/metabolismo , Epítopos/inmunología , Humanos , Cinética , Oxidación-Reducción , Unión Proteica , Conformación Proteica en Hélice alfa , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Sistema Renina-Angiotensina/fisiología
11.
BMC Bioinformatics ; 21(1): 165, 2020 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-32349657

RESUMEN

BACKGROUND: Network motifs are connectivity structures that occur with significantly higher frequency than chance, and are thought to play important roles in complex biological networks, for example in gene regulation, interactomes, and metabolomes. Network motifs may also become pivotal in the rational design and engineering of complex biological systems underpinning the field of synthetic biology. Distinguishing true motifs from arbitrary substructures, however, remains a challenge. RESULTS: Here we demonstrate both theoretically and empirically that implicit assumptions present in mainstream methods for motif identification do not necessarily hold, with the ramification that motif studies using these mainstream methods are less able to effectively differentiate between spurious results and events of true statistical significance than is often presented. We show that these difficulties cannot be overcome without revising the methods of statistical analysis used to identify motifs. CONCLUSIONS: Present-day methods for the discovery of network motifs, and, indeed, even the methods for defining what they are, are critically reliant on a set of incorrect assumptions, casting a doubt on the scientific validity of motif-driven discoveries. The implications of these findings are therefore far-reaching across diverse areas of biology.


Asunto(s)
Biología Computacional/métodos , Redes Reguladoras de Genes , Algoritmos , Regulación de la Expresión Génica , Humanos , Reproducibilidad de los Resultados
12.
J Immunol ; 200(12): 4134-4145, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29728507

RESUMEN

A structural characterization of the interaction between αß TCRs and cognate peptide-MHC (pMHC) is central to understanding adaptive T cell-mediated immunity. X-ray crystallography, although the source of much structural data, traditionally provides only a static snapshot of the protein. Given the emerging evidence for the important role of conformational dynamics in protein function, we interrogated 309 crystallographic structures of pMHC complexes using ensemble refinement, a technique that can extract dynamic information from the x-ray data. Focusing on a subset of human pMHC class I systems, we found that in many cases, ensemble methods were able to uncover previously hidden evidence of significant conformational plasticity, thereby revealing additional information that can build upon and significantly enhance functional interpretations that are based on a single static structure. Notable examples include the interpretation of differences in the disease association of HLA subtypes, the relationship between peptide prominence and TCR recognition, the role of conformational flexibility in vaccine design, and the discrimination between induced fit and conformational selection models of TCR binding. We show that the currently widespread practice of analyzing pMHC interactions via the study of a single crystallographic structure does not make use of pertinent and easily accessible information from x-ray data concerning alternative protein conformations. This new analysis therefore not only highlights the capacity for ensemble methods to significantly enrich the interpretation of decades of structural data but also provides previously missing information concerning the dynamics of existing characterized TCR-pMHC interactions.


Asunto(s)
Complejo Mayor de Histocompatibilidad/inmunología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Cristalografía por Rayos X/métodos , Humanos , Unión Proteica/inmunología , Conformación Proteica , Linfocitos T/inmunología
13.
Proc Natl Acad Sci U S A ; 114(15): 3897-3902, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28356519

RESUMEN

Ancestral protein reconstruction allows the resurrection and characterization of ancient proteins based on computational analyses of sequences of modern-day proteins. Unfortunately, many protein families are highly divergent and not suitable for sequence-based reconstruction approaches. This limitation is exemplified by the antigen receptors of jawed vertebrates (B- and T-cell receptors), heterodimers formed by pairs of Ig domains. These receptors are believed to have evolved from an extinct homodimeric ancestor through a process of gene duplication and diversification; however molecular evidence has so far remained elusive. Here, we use a structural approach and laboratory evolution to reconstruct such molecules and characterize their interaction with antigen. High-resolution crystal structures of reconstructed homodimeric receptors in complex with hen-egg white lysozyme demonstrate how nanomolar affinity binding of asymmetrical antigen is enabled through selective recruitment and structural plasticity within the receptor-binding site. Our results provide structural evidence in support of long-held theories concerning the evolution of antigen receptors, and provide a blueprint for the experimental reconstruction of protein ancestry in the absence of phylogenetic evidence.


Asunto(s)
Evolución Molecular , Filogenia , Receptores de Inmunoglobulina Polimérica/química , Animales , Cristalografía por Rayos X , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/química , Cadenas kappa de Inmunoglobulina/genética , Muramidasa/química , Receptores de Inmunoglobulina Polimérica/genética , Vertebrados/genética , Vertebrados/inmunología
14.
Int J Mol Sci ; 21(19)2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32992915

RESUMEN

The interaction between T cell receptor (TCR) and peptide (p)-Human Leukocyte Antigen (HLA) complexes is the critical first step in determining T cell responses. X-ray crystallographic studies of pHLA in TCR-bound and free states provide a structural perspective that can help understand T cell activation. These structures represent a static "snapshot", yet the nature of pHLAs and their interactions with TCRs are highly dynamic. This has been demonstrated for HLA class I molecules with in silico techniques showing that some interactions, thought to stabilise pHLA-I, are only transient and prone to high flexibility. Here, we investigated the dynamics of HLA class II molecules by focusing on three allomorphs (HLA-DR1, -DR11 and -DR15) that are able to present the same epitope and activate CD4+ T cells. A single TCR (F24) has been shown to recognise all three HLA-DR molecules, albeit with different affinities. Using molecular dynamics and crystallographic ensemble refinement, we investigate the molecular basis of these different affinities and uncover hidden roles for HLA polymorphic residues. These polymorphisms were responsible for the widening of the antigen binding cleft and disruption of pHLA-TCR interactions, underpinning the hierarchy of F24 TCR binding affinity, and ultimately T cell activation. We expanded this approach to all available pHLA-DR structures and discovered that all HLA-DR molecules were inherently rigid. Together with in vitro protein stability and peptide affinity measurements, our results suggest that HLA-DR1 possesses inherently high protein stability, and low HLA-DM susceptibility.


Asunto(s)
Antígenos/química , Antígenos HLA-DR/química , Receptores de Antígenos de Linfocitos T/química , Antígenos/inmunología , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/inmunología , Cristalografía por Rayos X , Células HEK293 , Antígenos HLA-DR/inmunología , Humanos , Receptores de Antígenos de Linfocitos T/inmunología
15.
Biochemistry ; 58(21): 2524-2533, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31058493

RESUMEN

Sunflower trypsin inhibitor (SFTI-1) is a 14 amino acid serine protease inhibitor. The dual antiparallel ß-sheet arrangement of SFTI-1 is stabilized by an N-terminal-C-terminal backbone cyclization and a further disulfide bridge to form a final bicyclic structure. This constrained structure is further rigidified by an extensive network of internal hydrogen bonds. Thus, the structure of SFTI-1 in solution resembles the protease-bound structure, reducing the entropic penalty upon protease binding. When cleaved at the scissile bond, it is thought that the rigidifying features of SFTI-1 maintain its structure, allowing the scissile bond to be reformed. The lack of structural plasticity for SFTI-1 is proposed to favor initial protease binding and continued occupancy in the protease active site, resulting in an equilibrium between the cleaved and uncleaved inhibitor in the presence of a protease. We have determined, at 1.15 Å resolution, the X-ray crystal structures of complexes between human kallikrein-related peptidase 4 (KLK4) and SFTI-FCQR(Asn14) and between KLK4 and an acyclic form of the same inhibitor, SFTI-FCQR(Asn14)[1,14], with the latter displaying a cleaved scissile bond. Structural analysis and MD simulations together reveal the roles of the altered contact sequence, intramolecular hydrogen bonding network, and backbone cyclization in altering the state of SFTI's scissile bond ligation at the protease active site. Taken together, the data presented reveal insights into the role of dynamics in the standard-mechanism inhibition and suggest that modifications on the non-contact strand may be a useful, underexplored approach for generating further potent or selective SFTI-based inhibitors against members of the serine protease family.


Asunto(s)
Calicreínas/química , Péptidos Cíclicos/química , Proteínas de Plantas/química , Inhibidores de Serina Proteinasa/química , Animales , Dominio Catalítico , Cristalografía por Rayos X , Ciclización , Escherichia coli/metabolismo , Humanos , Enlace de Hidrógeno , Calicreínas/antagonistas & inhibidores , Calicreínas/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Péptidos Cíclicos/metabolismo , Péptidos Cíclicos/farmacología , Proteínas de Plantas/farmacología , Unión Proteica , Conformación Proteica en Lámina beta , Inhibidores de Serina Proteinasa/farmacología , Spodoptera/citología , Spodoptera/metabolismo , Transfección
16.
Mol Microbiol ; 110(6): 879-896, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30230642

RESUMEN

Bacteriophage-encoded endolysins can recognize and bind specific bacteria, and act to cleave the glycosidic and/or amide bonds in the peptidoglycan (PG) bacterial cell wall. Cleavage of the cell wall generally results in the death of the bacteria. Their utility as bacteriolytic agents could be exploited for human and veterinary medicines as well as various biotechnological applications. As interest grows in the commercial uses of these proteins, there has been much effort to successfully employ rational design and engineering to produce endolysins with bespoke properties. In this review, we interrogate the current structural data and identify structural features that would be of benefit to engineering the activity and specificity of phage endolysins. We show that the growing body of structural data can be used to predict catalytic residues and mechanism of action from sequences of hypothetical endolysins, and probe the importance of secondary structure repeats in bacterial cell wall-binding domains.


Asunto(s)
Antibacterianos/química , Bacteriófagos/enzimología , Biocatálisis , N-Acetil Muramoil-L-Alanina Amidasa/química , Proteínas Virales/química , Bacteriólisis , Pared Celular/metabolismo , Simulación por Computador , Cinética , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Unión Proteica , Dominios Proteicos , Estructura Cuaternaria de Proteína , Especificidad por Sustrato
17.
Anal Chem ; 91(12): 7631-7638, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31117403

RESUMEN

Herein, we describe a fluorescent immunosensor designed by incorporating an unnatural amino acid fluorophore into the binding site of an EGFR-specific antibody fragment, resulting in quantifiable EGFR-dependent changes in peak fluorescence emission wavelength. To date, immunosensor design strategies have relied on binding-induced changes in fluorescence intensity that are prone to excitation source fluctuations and sample-dependent noise. In this study, we used a rational design approach to incorporate a polarity indicator (Anap) into specific positions of an anti-EGFR single chain antibody to generate an emission wavelength-dependent immunosensor. We found that when incorporated within the topological neighborhood of the antigen binding interface, the Anap emission wavelength is blue-shifted by EGFR-binding in a titratable manner, up to 20 nm, with nanomolar detection limits. This approach could be applicable to other antibody/antigen combinations for integration into a wide range of assay platforms (including homogeneous, solid-phase assay, or microfluidic assays) for one-step protein quantification.


Asunto(s)
Técnicas Biosensibles/métodos , Fragmentos de Inmunoglobulinas/química , Aminoácidos/genética , Aminoácidos/metabolismo , Anticuerpos/inmunología , Reacciones Antígeno-Anticuerpo , Receptores ErbB/genética , Receptores ErbB/inmunología , Colorantes Fluorescentes/química , Humanos , Inmunoensayo , Fragmentos de Inmunoglobulinas/inmunología , Límite de Detección , Polimorfismo de Nucleótido Simple
18.
Biochem Soc Trans ; 47(2): 701-711, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30902926

RESUMEN

Engineered proteins, especially enzymes, are now commonly used in many industries owing to their catalytic power, specific binding of ligands, and properties as materials and food additives. As the number of potential uses for engineered proteins has increased, the interest in engineering or designing proteins to have greater stability, activity and specificity has increased in turn. With any rational engineering or design pursuit, the success of these endeavours relies on our fundamental understanding of the systems themselves; in the case of proteins, their structure-dynamics-function relationships. Proteins are most commonly rationally engineered by targeting the residues that we understand to be functionally important, such as enzyme active sites or ligand-binding sites. This means that the majority of the protein, i.e. regions remote from the active- or ligand-binding site, is often ignored. However, there is a growing body of literature that reports on, and rationalises, the successful engineering of proteins at remote sites. This minireview will discuss the current state of the art in protein engineering, with a particular focus on engineering regions that are remote from active- or ligand-binding sites. As the use of protein technologies expands, exploiting the potential improvements made possible through modifying remote regions will become vital if we are to realise the full potential of protein engineering and design.


Asunto(s)
Ingeniería de Proteínas/métodos , Proteínas/metabolismo , Evolución Molecular Dirigida , Mutación/genética , Proteínas/genética
19.
Clin Chem ; 65(1): e1-e9, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30538125

RESUMEN

BACKGROUND: Genetic association studies have reported single-nucleotide polymorphisms (SNPs) at chromosome 19q13.3 to be associated with prostate cancer (PCa) risk. Recently, the rs61752561 SNP (Asp84Asn substitution) in exon 3 of the kallikrein-related peptidase 3 (KLK3) gene encoding prostate-specific antigen (PSA) was reported to be strongly associated with PCa risk (P = 2.3 × 10-8). However, the biological contribution of the rs61752561 SNP to PCa risk has not been elucidated. METHODS: Recombinant PSA protein variants were generated to assess the SNP-mediated biochemical changes by stability and substrate activity assays. PC3 cell-PSA overexpression models were established to evaluate the effect of the SNP on PCa pathogenesis. Genotype-specific correlation of the SNP with total PSA (tPSA) concentrations and free/total (F/T) PSA ratio were determined from serum samples. RESULTS: Functional analysis showed that the rs61752561 SNP affects PSA stability and structural conformation and creates an extra glycosylation site. This PSA variant had reduced enzymatic activity and the ability to stimulate proliferation and migration of PCa cells. Interestingly, the minor allele is associated with lower tPSA concentrations and high F/T PSA ratio in serum samples, indicating that the amino acid substitution may affect PSA immunoreactivity to the antibodies used in the clinical immunoassays. CONCLUSIONS: The rs61752561 SNP appears to have a potential role in PCa pathogenesis by changing the glycosylation, protein stability, and PSA activity and may also affect the clinically measured F/T PSA ratio. Accounting for these effects on tPSA concentration and F/T PSA ratio may help to improve the accuracy of the current PSA test.


Asunto(s)
Polimorfismo de Nucleótido Simple , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/genética , Anciano , Movimiento Celular , Proliferación Celular , Predisposición Genética a la Enfermedad , Glicosilación , Humanos , Masculino , Persona de Mediana Edad , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/patología , Proteolisis
20.
Horm Metab Res ; 51(12): 765-769, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31826271

RESUMEN

Thyroid peroxidase (TPO) is an enzyme that participates in thyroid hormone biosynthesis. TPO is also a major autoantigen in autoimmune thyroid diseases (AITD). In this review, we summarize the latest developments in the field of TPO research. We present the current understanding of immunodominant serologic determinants, frequency of TPO-specific autoantibodies in the population, as well as genetic and environmental factors contributing to their development. Moreover, we report recent progress in the clinical utilities of TPO autoantibody testing, including thyroid dysfunctions and extra-thyroidal disorders.


Asunto(s)
Yoduro Peroxidasa/inmunología , Enfermedades de la Tiroides/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/genética , Autoantígenos/inmunología , Humanos , Yoduro Peroxidasa/genética , Enfermedades de la Tiroides/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA