Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Structure ; 31(12): 1589-1603.e6, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-37776857

RESUMEN

Human thirty-eight-negative kinase-1 (TNK1) is implicated in cancer progression. The TNK1 ubiquitin-associated (UBA) domain binds polyubiquitin and plays a regulatory role in TNK1 activity and stability. No experimentally determined molecular structure of this unusual UBA domain is available. We fused the UBA domain to the 1TEL variant of the translocation ETS leukemia protein sterile alpha motif (TELSAM) crystallization chaperone and obtained crystals diffracting as far as 1.53 Å. GG and GSGG linkers allowed the UBA to reproducibly find a productive binding mode against its host 1TEL polymer and crystallize at protein concentrations as low as 0.2 mg/mL. Our studies support a mechanism of 1TEL fusion crystallization and show that 1TEL fusion crystals require fewer crystal contacts than traditional protein crystals. Modeling and experimental validation suggest the UBA domain may be selective for both the length and linkages of polyubiquitin chains.


Asunto(s)
Chaperonas Moleculares , Poliubiquitina , Humanos , Poliubiquitina/química , Unión Proteica , Cristalización , Estructura Terciaria de Proteína , Dominios Proteicos , Chaperonas Moleculares/metabolismo , Proteínas Fetales/metabolismo , Proteínas Tirosina Quinasas/metabolismo
2.
Open Biol ; 12(3): 210271, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35232248

RESUMEN

While conducting pilot studies into the usefulness of fusion to TELSAM polymers as a potential protein crystallization strategy, we observed novel properties in crystals of two TELSAM-target protein fusions, as follows. (i) A TELSAM-target protein fusion can crystallize more rapidly and with greater propensity than the same target protein alone. (ii) TELSAM-target protein fusions can be crystallized at low protein concentrations. This unprecedented observation suggests a route to crystallize proteins that can only be produced in microgram amounts. (iii) The TELSAM polymers themselves need not directly contact one another in the crystal lattice in order to form well-diffracting crystals. This novel observation is important because it suggests that TELSAM may be able to crystallize target proteins too large to allow direct inter-polymer contacts. (iv) Flexible TELSAM-target protein linkers can allow target proteins to find productive binding modes against the TELSAM polymer. (v) TELSAM polymers can adjust their helical rise to allow fused target proteins to make productive crystal contacts. (vi). Fusion to TELSAM polymers can stabilize weak inter-target protein crystal contacts. We report features of these TELSAM-target protein crystal structures and outline future work needed to validate TELSAM as a crystallization chaperone and determine best practices for its use.


Asunto(s)
Chaperonas Moleculares , Polímeros , Cristalización , Cristalografía por Rayos X , Chaperonas Moleculares/química , Polímeros/química
3.
Neuropeptides ; 92: 102231, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35180645

RESUMEN

Neuropeptide Y (NPY), peptide YY (PYY) and pancreatic polypeptide (PP) form the evolutionarily conserved pancreatic polypeptide family. While the fold is widely utilized in nature, crystal structures remain elusive, particularly for the human forms, with only the structure of a distant avian form of PP reported. Here we utilize a crystallization chaperone (antibody Fab fragment), specifically recognizing the amidated peptide termini, to solve the structures of human NPY and human PYY. Intriguingly, and despite limited sequence identity (~50%), the structure of human PYY closely resembles that of avian PP, highlighting the broad structural conservation of the fold throughout evolution. Specifically, the PYY structure is characterized by a C-terminal amidated α-helix, preceded by a backfolded poly-proline N-terminus, with the termini in close proximity to each other. In contrast, in the structure of human NPY the N-terminal component is disordered, while the helical component of the peptide is observed in a four-helix bundle type arrangement, consistent with a propensity for multimerization suggested by NMR studies.


Asunto(s)
Neuropéptido Y , Péptido YY , Humanos , Polipéptido Pancreático , Receptores de Neuropéptido Y
4.
J Mol Biol ; 433(16): 166909, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-33676924

RESUMEN

Structural studies of membrane proteins, especially small membrane proteins, are associated with well-known experimental challenges. Complexation with monoclonal antibody fragments is a common strategy to augment such proteins; however, generating antibody fragments that specifically bind a target protein is not trivial. Here we identify a helical epitope, from the membrane-proximal external region (MPER) of the gp41-transmembrane subunit of the HIV envelope protein, that is recognized by several well-characterized antibodies and that can be fused as a contiguous extension of the N-terminal transmembrane helix of a broad range of membrane proteins. To analyze whether this MPER-epitope tag might aid structural studies of small membrane proteins, we determined an X-ray crystal structure of a membrane protein target that does not crystallize without the aid of crystallization chaperones, the Fluc fluoride channel, fused to the MPER epitope and in complex with antibody. We also demonstrate the utility of this approach for single particle electron microscopy with Fluc and two additional small membrane proteins that represent different membrane protein folds, AdiC and GlpF. These studies show that the MPER epitope provides a structurally defined, rigid docking site for antibody fragments that is transferable among diverse membrane proteins and can be engineered without prior structural information. Antibodies that bind to the MPER epitope serve as effective crystallization chaperones and electron microscopy fiducial markers, enabling structural studies of challenging small membrane proteins.


Asunto(s)
Epítopos/química , Proteínas de la Membrana/química , Modelos Moleculares , Dominios y Motivos de Interacción de Proteínas , Cristalografía por Rayos X , Epítopos/inmunología , Humanos , Proteínas de la Membrana/inmunología , Microscopía Electrónica , Conformación Proteica , Relación Estructura-Actividad
5.
IUCrJ ; 8(Pt 2): 154-160, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33708392

RESUMEN

The production of diffraction-quality protein crystals is challenging and often requires bespoke, time-consuming and expensive strategies. A system has been developed in which the BCL6 BTB domain acts as a crystallization chaperone and promiscuous assembly block that may form the basis for affinity-capture crystallography. The protein of interest is expressed with a C-terminal tag that interacts with the BTB domain, and co-crystallization leads to its incorporation within a BTB-domain lattice. This strategy was used to solve the structure of the SH3 domain of human nebulin, a structure previously solved by NMR, at 1.56 Šresolution. This approach is simple and effective, requiring only routine protein complexation and crystallization screening, and should be applicable to a range of proteins.

6.
Protein Expr Purif ; 179: 105796, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33221505

RESUMEN

TREM2 has been identified by genomic analysis as a potential and novel target for the treatment of Alzheimer's disease. To enable structure-based screening of potential small molecule therapeutics, we sought to develop a robust crystallization platform for the TREM2 Ig-like domain. A systematic set of constructs containing the structural chaperone, maltose binding protein (MBP), fused to the Ig domain of TREM2, were evaluated in parallel expression and purification, followed by crystallization studies. Using protein crystallization and high-resolution diffraction as a readout, a MBP-TREM2 Ig fusion construct was identified that generates reproducible protein crystals diffracting at 2.0 Å, which makes it suitable for soaking of potential ligands. Importantly, analysis of crystal packing interfaces indicates that most of the surface of the TREM2 Ig domain is available for small molecule binding. A proof of concept co-crystallization study with a small library of fragments validated potential utility of this system for the discovery of new TREM2 therapeutics.


Asunto(s)
Cristalización/métodos , Glicoproteínas de Membrana , Chaperonas Moleculares , Receptores Inmunológicos , Proteínas Recombinantes de Fusión , Humanos , Proteínas de Unión a Maltosa/química , Proteínas de Unión a Maltosa/genética , Proteínas de Unión a Maltosa/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
7.
Protein Sci ; 28(4): 823-836, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30666745

RESUMEN

An antibody fragment that recognizes the tertiary structure of a target protein with high affinity can be utilized as a crystallization chaperone. Difficulties in establishing conformation-specific antibodies, however, limit the applicability of antibody fragment-assisted crystallization. Here, we attempted to establish an alternative method to promote the crystallization of target proteins using an already established anti-tag antibody. The monoclonal antibody NZ-1 recognizes the PA tag with an extremely high affinity. It was also established that the PA tag is accommodated in the antigen-binding pocket in a bent conformation, compatible with an insertion into loop regions on the target. We, therefore, explored the application of NZ-1 Fab as a crystallization chaperone that complexes with a target protein displaying a PA tag. Specifically, we inserted the PA tag into the ß-hairpins of the PDZ tandem fragment of a bacterial Site-2 protease. We crystallized the PA-inserted PDZ tandem mutants with the NZ-1 Fab and solved the co-crystal structure to analyze their interaction modes. Although the initial insertion designs produced only moderate-resolution structures, eliminating the solvent-accessible space between the NZ-1 Fab and target PDZ tandem improved the diffraction qualities remarkably. Our results demonstrate that the NZ-1-PA system efficiently promotes crystallization of the target protein. The present work also suggests that ß-hairpins are suitable sites for the PA insertion because the PA tag contains a Pro-Gly sequence with a propensity for a ß-turn conformation.


Asunto(s)
Anticuerpos Monoclonales/química , Bacterias/química , Proteínas Bacterianas/química , Fragmentos Fab de Inmunoglobulinas/química , Péptido Hidrolasas/química , Aquifex , Bacterias/enzimología , Cristalización/métodos , Cristalografía por Rayos X/métodos , Modelos Moleculares , Dominios PDZ , Conformación Proteica
8.
Structure ; 26(4): 635-639.e1, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29526432

RESUMEN

Crystallization of dual-topology fluoride (Fluc) channels requires small, soluble crystallization chaperones known as monobodies, which act as primary crystal lattice contacts. Previous structures of Flucs have been solved in the presence of monobodies that inhibit fluoride currents in single-channel electrophysiological recordings. These structures have revealed two-fold symmetric, doubly bound arrangements, with one monobody on each side of the membrane. The combined electrophysiological and structural observations raise the possibility that chaperone binding allosterically closes the channel, altering the structure from its conducting form. To address this, we identify and solve the structure with a different monobody that only partially blocks fluoride currents. The structure of the channel-monobody complex is asymmetric, with monobody bound to one side of the channel only. The channel conformation is nearly identical on the bound and uncomplexed sides, and to all previously solved structures, providing direct structural evidence that monobody binding does not induce local structural changes.


Asunto(s)
Proteínas Bacterianas/química , Bordetella pertussis/química , Fluoruros/química , Canales Iónicos/química , Chaperonas Moleculares/química , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Bordetella pertussis/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Fluoruros/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Transporte Iónico , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
J Mol Biol ; 430(3): 322-336, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29277294

RESUMEN

Monoclonal antibodies constitute one of the largest groups of drugs to treat cancers and immune disorders, and are guiding the design of vaccines against infectious diseases. Fragments antigen-binding (Fabs) have been preferred over monoclonal antibodies for the structural characterization of antibody-antigen complexes due to their relatively low flexibility. Nonetheless, Fabs often remain challenging to crystallize because of the surface characteristics of complementary determining regions and the residual flexibility in the hinge region between the variable and constant domains. Here, we used a variable heavy-chain (VHH) domain specific for the human kappa light chain to assist in the structure determination of three therapeutic Fabs that were recalcitrant to crystallization on their own. We show that this ligand alters the surface properties of the antibody-ligand complex and lowers its aggregation temperature to favor crystallization. The VHH crystallization chaperone also restricts the flexible hinge of Fabs to a narrow range of angles, and so independently of the variable region. Our findings contribute a valuable approach to antibody structure determination and provide biophysical insight into the principles that govern the crystallization of macromolecules.


Asunto(s)
Anticuerpos Monoclonales Humanizados/química , Cristalización/métodos , Cristalografía por Rayos X/métodos , Fragmentos Fab de Inmunoglobulinas/química , Cadenas kappa de Inmunoglobulina/química , Agregado de Proteínas , Anticuerpos de Dominio Único/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Alineación de Secuencia
10.
J Mol Biol ; 430(3): 337-347, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29273204

RESUMEN

Antibody Fab fragments have been exploited with significant success to facilitate the structure determination of challenging macromolecules as crystallization chaperones and as molecular fiducial marks for single particle cryo-electron microscopy approaches. However, the inherent flexibility of the "elbow" regions, which link the constant and variable domains of the Fab, can introduce disorder and thus diminish their effectiveness. We have developed a phage display engineering strategy to generate synthetic Fab variants that significantly reduces elbow flexibility, while maintaining their high affinity and stability. This strategy was validated using previously recalcitrant Fab-antigen complexes where introduction of an engineered elbow region enhanced crystallization and diffraction resolution. Furthermore, incorporation of the mutations appears to be generally portable to other synthetic antibodies and may serve as a universal strategy to enhance the success rates of Fabs as structure determination chaperones.


Asunto(s)
Antígenos/química , Microscopía por Crioelectrón/métodos , Fragmentos Fab de Inmunoglobulinas/química , Antígenos/ultraestructura , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/ultraestructura , Cristalización/métodos , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/ultraestructura , Biblioteca de Péptidos , Conformación Proteica , Ingeniería de Proteínas , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestructura
11.
J Mol Biol ; 430(14): 2128-2138, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29126898

RESUMEN

To untangle the complex signaling of the c-Jun N-terminal kinase (JNK) isoforms, we need tools that can selectively detect and inhibit individual isoforms. Because of the high similarity between JNK1, JNK2 and JNK3, it is very difficult to generate small-molecule inhibitors with this discriminatory power. Thus, we have recently selected protein binders from the designed ankyrin repeat protein (DARPin) library which were indeed isoform-specific inhibitors of JNK1 with low nanomolar potency. Here we provide the structural basis for their isotype discrimination and their inhibitory action. All our previous attempts to generate crystal structures of complexes had failed. We have now made use of a technology we recently developed which consists of rigid fusion of an additional special DARPin, which acts as a crystallization enhancer. This can be rigidly fused with different geometries, thereby generating a range of alternative crystal packings. The structures reveal the molecular basis for isoform specificity of the DARPins and their ability to prevent JNK activation and may thus form the basis of further investigation of the JNK family as well as novel approaches to drug design.


Asunto(s)
Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/química , Ingeniería de Proteínas/métodos , Inhibidores de Proteínas Quinasas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Secuencia de Aminoácidos , Repetición de Anquirina , Sitios de Unión , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Bibliotecas de Moléculas Pequeñas/química
12.
Front Microbiol ; 9: 3014, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30671027

RESUMEN

YabT is a serine/threonine kinase of the Hanks family from Bacillus subtilis, which lacks the canonical extracellular signal receptor domain but is anchored to the membrane through a C-terminal transmembrane helix. A previous study demonstrated that a basic juxtamembrane region corresponds to a DNA-binding motif essential for the activation of YabT trans-autophosphorylation. YabT is expressed during spore development and localizes to the asymmetric septum where it specifically phosphorylates essential proteins involved in genome maintenance, such as RecA, SsbA, and YabA. YabT has also been shown to phosphorylate proteins involved in protein synthesis, such as AbrB and Ef-Tu, suggesting a possible regulatory role in the progressive metabolic quiescence of the forespore. Finally, cross phosphorylations with other protein kinases implicate YabT in the regulation of numerous other cellular processes. Using an artificial protein scaffold as crystallization helper, we determined the first crystal structure of this DNA-dependent bacterial protein kinase. This allowed us to trap the active conformation of the kinase domain of YabT. Using NMR, we showed that the basic juxtamembrane region of YabT is disordered in the absence of DNA in solution, just like it is in the crystal, and that it is stabilized upon DNA binding. In comparison with its closest structural homolog, the mycobacterial kinase PknB allowed us to discuss the dimerization mode of YabT. Together with phosphorylation assays and DNA-binding experiments, this structural analysis helped us to gain new insights into the regulatory activation mechanism of YabT.

13.
Protein Sci ; 27(2): 561-567, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29052270

RESUMEN

The dual specificity phosphatase DUSP1 was the first mitogen activated protein kinase phosphatase (MKP) to be identified. It dephosphorylates conserved tyrosine and threonine residues in the activation loops of mitogen activated protein kinases ERK2, JNK1 and p38-alpha. Here, we report the crystal structure of the human DUSP1 catalytic domain at 2.49 Å resolution. Uniquely, the protein was crystallized as an MBP fusion protein in complex with a monobody that binds to MBP. Sulfate ions occupy the phosphotyrosine and putative phosphothreonine binding sites in the DUSP1 catalytic domain.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/química , Fosfatasa 1 de Especificidad Dual/metabolismo , Proteínas de Unión a Maltosa/metabolismo , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Fosfotreonina/química , Fosfotirosina/química , Conformación Proteica , Especificidad por Sustrato , Sulfatos/química
14.
Front Immunol ; 8: 1234, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29038656

RESUMEN

Marburg virus (MARV) is a highly lethal hemorrhagic fever virus that is increasingly re-emerging in Africa, has been imported to both Europe and the US, and is also a Tier 1 bioterror threat. As a negative sense RNA virus, MARV has error prone replication which can yield progeny capable of evading countermeasures. To evaluate this vulnerability, we sought to determine the epitopes of 4 llama single-domain antibodies (sdAbs or VHH) specific for nucleoprotein (NP), each capable of forming MARV monoclonal affinity reagent sandwich assays. Here, we show that all sdAb bound the C-terminal region of NP, which was produced recombinantly to derive X-ray crystal structures of the three best performing antibody-antigen complexes. The common epitope is a trio of alpha helices that form a novel asymmetric basin-like depression that accommodates each sdAb paratope via substantial complementarity-determining region (CDR) restructuring. Shared core contacts were complemented by unique accessory contacts on the sides and overlooks of the basin yielding very different approach routes for each sdAb to bind the antigen. The C-terminal region of MARV NP was unable to be crystallized alone and required engagement with sdAb to form crystals suggesting the antibodies acted as crystallization chaperones. While gross structural homology is apparent between the two most conserved helices of MARV and Ebolavirus, the positions and morphologies of the resulting basins were markedly different. Naturally occurring amino acid variations occurring in bat and human Marburgvirus strains all mapped to surfaces distant from the predicted sdAb contacts suggesting a vital role for the NP interface in virus replication. As an essential internal structural component potentially interfacing with a partner protein it is likely the C-terminal epitope remains hidden or "cryptic" until virion disruption occurs. Conservation of this epitope over 50 years of Marburgvirus evolution should make these sdAb useful foundations for diagnostics and therapeutics resistant to drift.

15.
Structure ; 25(10): 1611-1622.e4, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28919443

RESUMEN

Antibody fragments are frequently used as a "crystallization chaperone" to aid structural analysis of complex macromolecules that are otherwise crystallization resistant, but conventional fragment formats have not been designed for this particular application. By fusing an anti-parallel coiled-coil structure derived from the SARAH domain of human Mst1 kinase to the variable region of an antibody, we succeeded in creating a novel chimeric antibody fragment of ∼37 kDa, termed "Fv-clasp," which exhibits excellent crystallization compatibility while maintaining the binding ability of the original IgG molecule. The "clasp" and the engineered disulfide bond at the bottom of the Fv suppressed the internal mobility of the fragment and shielded hydrophobic residues, likely contributing to the high heat stability and the crystallizability of the Fv-clasp. Finally, Fv-clasp antibodies showed superior "chaperoning" activity over conventional Fab fragments, and facilitated the structure determination of an ectodomain fragment of integrin α6ß1.


Asunto(s)
Fragmentos Fab de Inmunoglobulinas/química , Proteínas Serina-Treonina Quinasas/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intracelular , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Estabilidad Proteica
16.
Bio Protoc ; 7(3): e2116, 2017 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34458442

RESUMEN

The SLC26 or SulP proteins constitute a large family of anion transporters that are ubiquitously expressed in pro- and eukaryotes. In human, SLC26 proteins perform important roles in ion homeostasis and malfunctioning of selected members is associated with diseases. This protocol details the production and crystallization of a prokaryotic SLC26 homolog, termed SLC26Dg, from Deinococcus geothermalis. Following these instructions we obtained well-folded and homogenous material of the membrane protein SLC26Dg and the nanobody Nb5776 that enabled us to crystallize the complex and determine its structure ( Geertsma et al., 2015 ). The procedure may be adapted to purify and crystallize other membrane protein complexes.

17.
Protein Sci ; 25(12): 2268-2276, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27595817

RESUMEN

Fv antibody fragments have been used as co-crystallization partners in structural biology, particularly in membrane protein crystallography. However, there are inherent technical issues associated with the large-scale production of soluble, functional Fv fragments through conventional methods in various expression systems. To circumvent these problems, we developed a new method, in which a single synthetic polyprotein consisting of a variable light (VL ) domain, an intervening removable affinity tag (iRAT), and a variable heavy (VH ) domain is expressed by a Gram-positive bacterial secretion system. This method ensures stoichiometric expression of VL and VH from the monocistronic construct followed by proper folding and assembly of the two variable domains. The iRAT segment can be removed by a site-specific protease during the purification process to yield tag-free Fv fragments suitable for crystallization trials. In vitro refolding step is not required to obtain correctly folded Fv fragments. As a proof of concept, we tested the iRAT-based production of multiple Fv fragments, including a crystallization chaperone for a mammalian membrane protein as well as FDA-approved therapeutic antibodies. The resulting Fv fragments were functionally active and crystallized in complex with the target proteins. The iRAT system is a reliable, rapid and broadly applicable means of producing milligram quantities of Fv fragments for structural and biochemical studies.


Asunto(s)
Brevibacillus/metabolismo , Anticuerpos de Cadena Única , Brevibacillus/genética , Cristalografía por Rayos X/métodos , Humanos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/aislamiento & purificación
18.
Protein Sci ; 25(3): 559-71, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26682969

RESUMEN

Although chaperone-assisted protein crystallization remains a comparatively rare undertaking, the number of crystal structures of polypeptides fused to maltose-binding protein (MBP) that have been deposited in the Protein Data Bank (PDB) has grown dramatically during the past decade. Altogether, 102 fusion protein structures were detected by Basic Local Alignment Search Tool (BLAST) analysis. Collectively, these structures comprise a range of sizes, space groups, and resolutions that are typical of the PDB as a whole. While most of these MBP fusion proteins were equipped with short inter-domain linkers to increase their rigidity, fusion proteins with long linkers have also been crystallized. In some cases, surface entropy reduction mutations in MBP appear to have facilitated the formation of crystals. A comparison of the structures of fused and unfused proteins, where both are available, reveals that MBP-mediated structural distortions are very rare.


Asunto(s)
Cristalización/métodos , Proteínas de Unión a Maltosa/química , Péptidos/química , Proteínas Recombinantes de Fusión/química , Secuencia de Aminoácidos , Animales , Clonación Molecular/métodos , Cristalografía/métodos , Entropía , Humanos , Proteínas de Unión a Maltosa/genética , Proteínas de Unión a Maltosa/aislamiento & purificación , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/aislamiento & purificación , Mutación , Péptidos/genética , Péptidos/aislamiento & purificación , Conformación Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación
19.
Methods Enzymol ; 557: 219-45, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25950967

RESUMEN

Phage display selections generate high-affinity synthetic reagents that can be used as tools in structural characterization of membrane proteins. Currently, most selection protocols are performed with membrane protein targets in detergents. However, there are numerous technical issues associated with this, primarily that detergents are poor mimics of the native lipid environment. Here, we describe a set of protocols for phage display selection that involves reconstituting membrane proteins in nanodiscs, which are small discoidal particles consisting of lipids enclosed by membrane scaffold proteins. The nanodisc format enabled us to expand the capabilities of competitive and subtractive phage display selection steps, and generation of high-quality synthetic reagents for membrane proteins in native-like lipid environment.


Asunto(s)
Fragmentos de Inmunoglobulinas/química , Lípidos de la Membrana/química , Proteínas de la Membrana/química , Nanoestructuras/química , Biblioteca de Péptidos , Animales , Biotinilación , Detergentes/química , Humanos , Indicadores y Reactivos , Magnetismo/métodos , Imanes/química , Modelos Moleculares , Estreptavidina/química
20.
J Biomol Screen ; 19(6): 829-38, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24695620

RESUMEN

Antibodies are powerful research tools that can be used in many areas of biology to probe, measure, and perturb various biological structures. Successful drug discovery is dependent on the correct identification of a target implicated in disease, coupled with the successful selection, optimization, and development of a candidate drug. Because of their specific binding characteristics, with regard to specificity, affinity, and avidity, coupled with their amenability to protein engineering, antibodies have become a key tool in drug discovery, enabling the quantification, localization, and modulation of proteins of interest. This review summarizes the application of antibodies and other protein affinity reagents as specific research tools within the drug discovery process.


Asunto(s)
Anticuerpos/química , Descubrimiento de Drogas/métodos , Proteómica/métodos , Animales , Animales Modificados Genéticamente , Afinidad de Anticuerpos , Cristalización , Epítopos/química , Humanos , Inmunoglobulina G/química , Chaperonas Moleculares/química , Fenotipo , Ingeniería de Proteínas/métodos , ARN/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA