Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(27): 15620-15631, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32576689

RESUMEN

Repulsive guidance molecules (RGMs) are cell surface proteins that regulate the development and homeostasis of many tissues and organs, including the nervous, skeletal, and immune systems. They control fundamental biological processes, such as migration and differentiation by direct interaction with the Neogenin (NEO1) receptor and function as coreceptors for the bone morphogenetic protein (BMP)/growth differentiation factor (GDF) family. We determined crystal structures of all three human RGM family members in complex with GDF5, as well as the ternary NEO1-RGMB-GDF5 assembly. Surprisingly, we show that all three RGMs inhibit GDF5 signaling, which is in stark contrast to RGM-mediated enhancement of signaling observed for other BMPs, like BMP2. Despite their opposite effect on GDF5 signaling, RGMs occupy the BMP type 1 receptor binding site similar to the observed interactions in RGM-BMP2 complexes. In the NEO1-RGMB-GDF5 complex, RGMB physically bridges NEO1 and GDF5, suggesting cross-talk between the GDF5 and NEO1 signaling pathways. Our crystal structures, combined with structure-guided mutagenesis of RGMs and BMP ligands, binding studies, and cellular assays suggest that RGMs inhibit GDF5 signaling by competing with GDF5 type 1 receptors. While our crystal structure analysis and in vitro binding data initially pointed towards a simple competition mechanism between RGMs and type 1 receptors as a possible basis for RGM-mediated GDF5 inhibition, further experiments utilizing BMP2-mimicking GDF5 variants clearly indicate a more complex mechanism that explains how RGMs can act as a functionality-changing switch for two structurally and biochemically similar signaling molecules.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas Ligadas a GPI/metabolismo , Factor 5 de Diferenciación de Crecimiento/metabolismo , Proteína de la Hemocromatosis/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/ultraestructura , Moléculas de Adhesión Celular Neuronal/ultraestructura , Cristalografía por Rayos X , Proteínas Ligadas a GPI/ultraestructura , Factor 5 de Diferenciación de Crecimiento/ultraestructura , Proteína de la Hemocromatosis/ultraestructura , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Proteínas del Tejido Nervioso/ultraestructura , Dominios Proteicos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Transducción de Señal
2.
Protein Expr Purif ; 186: 105918, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34044133

RESUMEN

Bone morphogenetic protein 2 (BMP21) is a highly interesting therapeutic growth factor due to its strong osteogenic/osteoinductive potential. However, its pronounced aggregation tendency renders recombinant and soluble production troublesome and complex. While prokaryotic expression systems can provide BMP2 in large amounts, the typically insoluble protein requires complex denaturation-renaturation procedures with medically hazardous reagents to obtain natively folded homodimeric BMP2. Based on a detailed aggregation analysis of wildtype BMP2, we designed a hydrophilic variant of BMP2 additionally containing an improved heparin binding site (BMP2-2Hep-7M). Consecutive optimization of BMP2-2Hep-7M expression and purification enabled production of soluble dimeric BMP2-2Hep-7M in high yield in E. coli. This was achieved by a) increasing protein hydrophilicity via introducing seven point mutations within aggregation hot spots of wildtype BMP2 and a longer N-terminus resulting in higher affinity for heparin, b) by employing E. coli strain SHuffle® T7, which enables the structurally essential disulfide-bond formation in BMP2 in the cytoplasm, c) by using BMP2 variant characteristic soluble expression conditions and application of l-arginine as solubility enhancer. The BMP2 variant BMP2-2Hep-7M shows strongly attenuated although not completely eliminated aggregation tendency.


Asunto(s)
Proteína Morfogenética Ósea 2 , Proteínas Recombinantes de Fusión , Sitios de Unión/genética , Proteína Morfogenética Ósea 2/química , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/aislamiento & purificación , Proteína Morfogenética Ósea 2/metabolismo , Escherichia coli/genética , Heparina/metabolismo , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Solubilidad
3.
Mol Pharmacol ; 95(2): 169-182, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30409791

RESUMEN

Organic cation transporters OCT1 (SLC22A1) and OCT2 (SLC22A2) are critically involved in absorption and excretion of diverse cationic drugs. Because drug-drug interactions at these transporters may induce adverse drug effects in patients, in vitro testing during drug development for interaction with the human transporters is mandatory. Recent data performed with rat OCT1 (rOCT1) suggest that currently performed in vitro tests assuming one polyspecific binding site are insufficient. Here we measured the binding and transport of model substrate 1-methyl-4-phenylpyridinium+ (MPP+) by cell-free-expressed fusion proteins of rOCT1 and rOCT1 mutants with green fluorescent protein that had been reconstituted into nanodiscs or proteoliposomes. The nanodiscs were formed with major scaffold protein (MSP) and different phospholipids, whereas the proteoliposomes were formed with a mixture of cholesterol, phosphatidylserine, and phosphatidylcholine. In nanodiscs formed with 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine or cholesterol, phosphatidylserine, and phosphatidylcholine, two low-affinity MPP+ binding sites and one high-affinity MPP+ binding site per transporter monomer were determined. Mutagenesis revealed that tryptophan 218 and aspartate 475 in neighboring positions in the modeled outward-open cleft contribute to one low-affinity binding site, whereas arginine 440 located distantly in the cleft is critical for MPP+ binding to another low-affinity site. Comparing MPP+ binding with MPP+ transport suggests that the low-affinity sites are involved in MPP+ transport, whereas high-affinity MPP+ binding influences transport allosterically. The data will be helpful in the interpretation of future crystal structures and provides a rationale for future in vitro testing that is more sophisticated and reliable, leading to the generation of pharmacophore models with high predictive power.


Asunto(s)
1-Metil-4-fenilpiridinio/metabolismo , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Animales , Sitios de Unión , Proteínas Fluorescentes Verdes/metabolismo , Mutagénesis/fisiología , Fosfolípidos/metabolismo , Proteolípidos/metabolismo , Ratas
4.
Mol Pharmacol ; 93(4): 402-415, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29339398

RESUMEN

The effects of mutations in the modeled outward-open cleft of rat organic cation transporter 1 (rOCT1) on affinities of substrates and inhibitors were investigated. Human embryonic kidney 293 cells were stably transfected with rOCT1 or rOCT1 mutants, and uptake of the substrates 1-methyl-4-phenylpyridinium+ (MPP+) and tetraethylammonium+ (TEA+) or inhibition of MPP+ uptake by the nontransported inhibitors tetrabutylammonium+ (TBuA+), tetrapentylammonium+ (TPeA+), and corticosterone was measured. Uptake measurements were performed on confluent cell layers using a 2-minute incubation or in dissociated cells using incubation times of 1, 5, or 10 seconds. With both methods, different apparent Michaelis-Menten constant (Km) values, different IC50 values, and varying effects of mutations were determined. In addition, varying IC50 values for the inhibition of MPP+ uptake and varying effects of mutations were obtained when different MPP+ concentrations far below the apparent Km value were used for uptake measurements. Eleven mutations were investigated by measuring initial uptake in dissociated cells and employing 0.1 µM MPP+ for uptake during inhibition experiments. Altered affinities for substrates and/or inhibitors were observed when Phe160, Trp218, Arg440, Leu447, and Asp475 were mutated. The mutations resulted in changes of apparent Km values for TEA+ and/or MPP+ Mutation of Trp218 and Asp475 led to altered IC50 values for TBuA+, TPeA+, and corticosterone, whereas the mutation of Phe160 and Leu447 changed the IC50 values for two inhibitors. Thereby amino acids in the outward-facing conformation of rOCT1 could be identified that interact with structurally different inhibitors and probably also with different substrates.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Mutagénesis/efectos de los fármacos , 1-Metil-4-fenilpiridinio/metabolismo , 1-Metil-4-fenilpiridinio/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Mutagénesis/fisiología , Compuestos de Amonio Cuaternario/metabolismo , Compuestos de Amonio Cuaternario/farmacología , Ratas , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología , Xenopus laevis
5.
Acta Biochim Biophys Sin (Shanghai) ; 50(1): 12-36, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29293886

RESUMEN

Transforming growth factor-ß (TGF-ß) family members, which include TGF-ßs, activins and bone morphogenetic proteins, are pleiotropic cytokines that elicit cell type-specific effects in a highly context-dependent manner in many different tissues. These secreted protein ligands signal via single-transmembrane Type I and Type II serine/threonine kinase receptors and intracellular SMAD transcription factors. Deregulation in signaling has been implicated in a broad array of diseases, and implicate the need for intricate fine tuning in cellular signaling responses. One important emerging mechanism by which TGF-ß family receptor signaling intensity, duration, specificity and diversity are regulated and/or mediated is through cell surface co-receptors. Here, we provide an overview of the co-receptors that have been identified for TGF-ß family members. While some appear to be specific to TGF-ß family members, others are shared with other pathways and provide possible ways for signal integration. This review focuses on novel functions of TGF-ß family co-receptors, which continue to be discovered.


Asunto(s)
Activinas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Activinas/química , Animales , Proteínas Morfogenéticas Óseas/química , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Receptores de Factores de Crecimiento Transformadores beta/química , Factor de Crecimiento Transformador beta/química
6.
Plant Physiol ; 172(3): 1911-1927, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27688623

RESUMEN

Nonspecific lipid transfer proteins reversibly bind different types of lipid molecules in a hydrophobic cavity. They facilitate phospholipid transfer between membranes in vitro, play a role in cuticle and possibly in suberin formation, and might be involved in plant pathogen defense signaling. This study focuses on the role of the lipid transfer protein AtLTPI-4 in crown gall development. Arabidopsis (Arabidopsis thaliana) crown gall tumors, which develop upon infection with the virulent Agrobacterium tumefaciens strain C58, highly expressed AtLTPI-4 Crown galls of the atltpI-4 loss-of-function mutant were much smaller compared with those of wild-type plants. The gene expression pattern and localization of the protein to the plasma membrane pointed to a function of AtLTPI-4 in cell wall suberization. Since Arabidopsis crown galls are covered by a suberin-containing periderm instead of a cuticle, we analyzed the suberin composition of crown galls and found a reduction in the amounts of long-chain fatty acids (C18:0) in the atltpI-4 mutant. To demonstrate the impact of AtLtpI-4 on extracellular lipid composition, we expressed the protein in Arabidopsis epidermis cells. This led to a significant increase in the very-long-chain fatty acids C24 and C26 in the cuticular wax fraction. Homology modeling and lipid-protein-overlay assays showed that AtLtpI-4 protein can bind these very-long-chain fatty acids. Thus, AtLtpI-4 protein may facilitate the transfer of long-chain as well as very-long-chain fatty acids into the apoplast, depending on the cell type in which it is expressed. In crown galls, which endogenously express AtLtpI-4, it is involved in suberin formation.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Proteínas Portadoras/metabolismo , Lípidos/biosíntesis , Proteínas de Transferencia de Fosfolípidos/metabolismo , Tumores de Planta , Arabidopsis/citología , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Proteínas de Arabidopsis/química , Proteínas Portadoras/química , Membrana Celular/metabolismo , Pared Celular/metabolismo , Ácidos Grasos/metabolismo , Modelos Moleculares , Mutación/genética , Especificidad de Órganos , Proteínas de Transferencia de Fosfolípidos/química , Epidermis de la Planta/metabolismo , Epidermis de la Planta/ultraestructura , Plantas Modificadas Genéticamente , Unión Proteica , Transporte de Proteínas , Nicotiana/genética , Tricomas/metabolismo , Tricomas/ultraestructura
7.
Biomacromolecules ; 18(3): 695-708, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28211679

RESUMEN

The regenerative potential of bone is strongly impaired in pathological conditions, such as nonunion fractures. To support bone regeneration various scaffolds have been developed in the past, which have been functionalized with osteogenic growth factors such as bone morphogenetic proteins (BMPs). However, most of them required supra-physiological levels of these proteins leading to burst releases, thereby causing severe side effects. Site-specific, covalent coupling of BMP2 to implant materials might be an optimal strategy in order to overcome these problems. Therefore, we created a BMP-2 variant (BMP2-K3Plk) containing a noncanonical amino acid (propargyl-l-lysine) substitution introduced by genetic code expansion that allows for site-specific and covalent immobilization onto polymeric scaffold materials. To directly compare different coupling strategies, we also produced a BMP2 variant containing an additional cysteine residue (BMP2-A2C) allowing covalent coupling by thioether formation. The BMP2-K3Plk mutant was coupled to functionalized beads by a copper-catalyzed azide-alkyne cycloaddition (CuAAC) either directly or via a short biotin-PEG linker both with high specificity. After exposing the BMP-coated beads to C2C12 cells, ALP expression appeared locally restricted in close proximity to these beads, showing that both coupled BMP2 variants trigger cell differentiation. The advantage of our approach over non-site-directed immobilization techniques is the ability to produce fully defined osteogenic surfaces, allowing for lower BMP2 loads and concomitant higher bioactivities, for example, due to controlled orientation toward BMP2 receptors. Such products might provide superior bone healing capabilities with potential safety advantages as of homogeneous product outcome.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Proteínas Inmovilizadas/química , Andamios del Tejido/química , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Materiales Biocompatibles/química , Proteína Morfogenética Ósea 2/química , Regeneración Ósea/fisiología , Huesos/citología , Huesos/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Clonación Molecular , Humanos , Osteogénesis/fisiología , Polímeros/química
8.
Mol Pharmacol ; 90(5): 508-521, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27555600

RESUMEN

Na+-d-glucose cotransporter 1 (SGLT1) is rate-limiting for glucose absorption in the small intestine. Shortly after intake of glucose-rich food, SGLT1 abundance in the luminal membrane of the small intestine is increased. This upregulation occurs via glucose-induced acceleration of the release of SGLT1-containing vesicles from the trans-Golgi network (TGN), which is regulated by a domain of protein RS1 (RSC1A1) named RS1-Reg. Dependent on phosphorylation, RS1-Reg blocks release of vesicles containing SGLT1 or concentrative nucleoside transporter 1. The hypothesis has been raised that RS1-Reg binds to different receptor proteins at the TGN, which trigger release of vesicles with different transporters. To identify the presumed receptor proteins, two-hybrid screening was performed. Interaction with ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme of polyamine synthesis, was observed and verified by immunoprecipitation. Binding of RS1-Reg mutants to ODC1 was characterized using surface plasmon resonance. Inhibition of ODC1 activity by RS1-Reg mutants and the ODC1 inhibitor difluoromethylornithine (DFMO) was measured in the absence and presence of glucose. In addition, short-term effects of DFMO, RS1-Reg mutants, the ODC1 product putrescine, and/or glucose on SGLT1 expressed in oocytes of Xenopus laevis were investigated. High-affinity binding of RS1-Reg to ODC1 was demonstrated, and evidence for a glucose binding site in ODC1 was provided. Binding of RS1-Reg to ODC1 inhibits the enzymatic activity at low intracellular glucose, which is blunted at high intracellular glucose. The data suggest that generation of putrescine by ODC1 at the TGN stimulates release of SGLT1-containing vesicles. This indicates a biomedically important role of ODC1 in regulation of glucose homeostasis.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Exocitosis/efectos de los fármacos , Glucosa/farmacología , Proteínas de Transporte de Monosacáridos/metabolismo , Ornitina Descarboxilasa/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Eflornitina/farmacología , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Humanos , Inmunoprecipitación , Espacio Intracelular/metabolismo , Cinética , Metilglucósidos/farmacología , Modelos Biológicos , Proteínas de Transporte de Monosacáridos/química , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Florizina/farmacología , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Proteínas Recombinantes/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Resonancia por Plasmón de Superficie , Xenopus laevis
9.
J Biol Chem ; 290(39): 24007-20, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26254468

RESUMEN

Growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are oocyte-specific growth factors with central roles in mammalian reproduction, regulating species-specific fecundity, ovarian follicular somatic cell differentiation, and oocyte quality. In the human, GDF9 is produced in a latent form, the mechanism of activation being an open question. Here, we produced a range of recombinant GDF9 and BMP15 variants, examined their in silico and physical interactions and their effects on ovarian granulosa cells (GC) and oocytes. We found that the potent synergistic actions of GDF9 and BMP15 on GC can be attributed to the formation of a heterodimer, which we have termed cumulin. Structural modeling of cumulin revealed a dimerization interface identical to homodimeric GDF9 and BMP15, indicating likely formation of a stable complex. This was confirmed by generation of recombinant heterodimeric complexes of pro/mature domains (pro-cumulin) and covalent mature domains (cumulin). Both pro-cumulin and cumulin exhibited highly potent bioactivity on GC, activating both SMAD2/3 and SMAD1/5/8 signaling pathways and promoting proliferation and expression of a set of genes associated with oocyte-regulated GC differentiation. Cumulin was more potent than pro-cumulin, pro-GDF9, pro-BMP15, or the two combined on GC. However, on cumulus-oocyte complexes, pro-cumulin was more effective than all other growth factors at notably improving oocyte quality as assessed by subsequent day 7 embryo development. Our results support a model of activation for human GDF9 dependent on cumulin formation through heterodimerization with BMP15. Oocyte-secreted cumulin is likely to be a central regulator of fertility in mono-ovular mammals.


Asunto(s)
Proteína Morfogenética Ósea 15/metabolismo , Células de la Granulosa/metabolismo , Factor 9 de Diferenciación de Crecimiento/metabolismo , Oocitos/metabolismo , Animales , Proteína Morfogenética Ósea 15/genética , Femenino , Células de la Granulosa/citología , Factor 9 de Diferenciación de Crecimiento/genética , Humanos , Ratones , Oocitos/citología , Multimerización de Proteína/fisiología , Transducción de Señal/fisiología , Proteínas Smad/genética , Proteínas Smad/metabolismo
10.
Eur J Immunol ; 45(7): 2122-33, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25900449

RESUMEN

TCRs of invariant NKT (iNKT) cells bind α-galactosylceramide (αGC) loaded CD1d in a highly conserved fashion and show a characteristic TCR gene usage: An "invariant" α chain with a canonical AV14/AJ18 rearrangement in mice (AV24/AJ18 in humans) is paired with ß chains containing characteristic Vß segments. In the rat, a multimember AV14 gene family increases the variability within this system. This study characterizes CD1d binding of rat AV14 gene segments in TCR transductants as well as CD1d binding and iNKT TCR expression of expanded polyclonal F344 rat iNKT populations. It defines an important role of position 93 at the V-J transition for TCR avidity and species cross-reactivity of the rat iNKT TCR. Furthermore, for the first time we identified variability within the fourth hypervariable loop (HV4) of the α chain as a modulator of CD1d:αGC binding in rat and mouse. Additionally, we confirmed the importance of the CDR2ß for CD1d:αGC binding, but also show that the CDR3ß may even have opposite effects on binding depending on the pairing α chain. Altogether, we characterized naturally occurring sources of variability for the iNKT TCR and speculate that they rather level than increase the largely germline encoded differences of iNKT TCR ligand avidity.


Asunto(s)
Antígenos CD1d/inmunología , Modelos Moleculares , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Animales , Citometría de Flujo , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Ratas , Ratas Endogámicas F344
11.
Chembiochem ; 17(22): 2123-2128, 2016 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-27595549

RESUMEN

Driving macrophage (Mϕ) polarization into the M2 phenotype provides potential against inflammatory diseases. Interleukin-4 (IL-4) promotes polarization into the M2-Mϕ phenotype, but its systemic use is constrained by dose-limiting toxicity. Consequently, we developed IL-4-decorated surfaces aiming at sustained and localized activity. IL-4 muteins were generated by genetic code expansion; Lys42 was replaced by unnatural amino acids (uAAs). Both muteins showed cell-stimulation ability and binding affinity to IL4Rα similar to those of wt-IL-4. Copper-catalyzed (CuAAC) and copper-free strain-promoted (SPAAC) 1,3-dipolar azide-alkyne cycloadditions were used to site-selectively anchor IL-4 to agarose surfaces. These surfaces had sustained IL-4 activity, as demonstrated by TF-1 cell proliferation and M2, but not M1, polarization of M-CSF-generated human Mϕ. The approach provides a blueprint for the engineering of cytokine-activated surfaces profiled for sustained and spatially controlled activity.


Asunto(s)
Interleucina-4/química , Macrófagos/metabolismo , Alquinos/química , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/metabolismo , Azidas/química , Catálisis , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Dicroismo Circular , Cobre/química , Reacción de Cicloadición , Código Genético , Células HEK293 , Humanos , Interferón gamma/farmacología , Interleucina-4/genética , Interleucina-4/metabolismo , Lipopolisacáridos/farmacología , Lisina/análogos & derivados , Lisina/química , Lisina/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Datos de Secuencia Molecular , Monocitos/citología , Mutagénesis Sitio-Dirigida , Sefarosa/química , Propiedades de Superficie
12.
BMC Biol ; 13: 77, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26385096

RESUMEN

BACKGROUND: Bone morphogenetic protein (BMP)-2 and growth and differentiation factor (GDF)-5 are two related transforming growth factor (TGF)-ß family members with important functions in embryonic development and tissue homeostasis. BMP-2 is best known for its osteoinductive properties whereas GDF-5-as evident from its alternative name, cartilage derived morphogenetic protein 1-plays an important role in the formation of cartilage. In spite of these differences both factors signal by binding to the same subset of BMP receptors, raising the question how these different functionalities are generated. The largest difference in receptor binding is observed in the interaction with the type I receptor BMPR-IA. GDF-5, in contrast to BMP-2, shows preferential binding to the isoform BMPR-IB, which is abrogated by a single amino acid (A57R) substitution. The resulting variant, GDF-5 R57A, represents a "BMP-2 mimic" with respect to BMP receptor binding. In this study we thus wanted to analyze whether the two growth factors can induce distinct signals via an identically composed receptor. RESULTS: Unexpectedly and dependent on the cellular context, GDF-5 R57A showed clear differences in its activity compared to BMP-2. In ATDC-5 cells, both ligands induced alkaline phosphatase (ALP) expression with similar potency. But in C2C12 cells, the BMP-2 mimic GDF-5 R57A (and also wild-type GDF-5) clearly antagonized BMP-2-mediated ALP expression, despite signaling in both cell lines occurring solely via BMPR-IA. The BMP-2- antagonizing properties of GDF-5 and GDF-5 R57A could also be observed in vivo when implanting BMP-2 and either one of the two GDF-5 ligands simultaneously at heterotopic sites. CONCLUSIONS: Although comparison of the crystal structures of the GDF-5 R57A:BMPR-IAEC- and BMP-2:BMPR-IAEC complex revealed small ligand-specific differences, these cannot account for the different signaling characteristics because the complexes seem identical in both differently reacting cell lines. We thus predict an additional component, most likely a not yet identified GDF-5-specific co-receptor, which alters the output of the signaling complexes. Hence the presence or absence of this component then switches GDF-5's signaling capabilities to act either similar to BMP-2 or as a BMP-2 antagonist. These findings might shed new light on the role of GDF-5, e.g., in cartilage maintenance and/or limb development in that it might act as an inhibitor of signaling events initiated by other BMPs.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Factor 5 de Diferenciación de Crecimiento/metabolismo , Línea Celular Tumoral , Humanos , Ligandos , Unión Proteica , Conformación Proteica , Transducción de Señal
13.
J Biol Chem ; 287(37): 31561-73, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22810231

RESUMEN

Organic cation transporters are membrane potential-dependent facilitative diffusion systems. Functional studies, extensive mutagenesis, and homology modeling indicate the following mechanism. A transporter conformation with a large outward-open cleft binds extracellular substrate, passes a state in which the substrate is occluded, turns to a conformation with an inward-open cleft, releases substrate, and subsequently turns back to the outward-open state. In the rat organic cation transporter (rOct1), voltage- and ligand-dependent movements of fluorescence-labeled cysteines were measured by voltage clamp fluorometry. For fluorescence detection, cysteine residues were introduced in extracellular parts of cleft-forming transmembrane α-helices (TMHs) 5, 8, and 11. Following expression of the mutants in Xenopus laevis oocytes, cysteines were labeled with tetramethylrhodamine-6-maleimide, and voltage-dependent conformational changes were monitored by voltage clamp fluorometry. One cysteine was introduced in the central domain of TMH 11 replacing glycine 478. This domain contains two amino acids that are involved in substrate binding and two glycine residues (Gly-477 and Gly-478) allowing for helix bending. Cys-478 could be modified with the transported substrate analog [2-(trimethylammonium)-ethyl]methanethiosulfonate but was inaccessible to tetramethylrhodamine-6-maleimide. Voltage-dependent movements at the indicator positions of TMHs 5, 8, and 11 were altered by substrate applications indicating large conformational changes during transport. The G478C exchange decreased transporter turnover and blocked voltage-dependent movements of TMHs 5 and 11. [2-(Trimethylammonium)-ethyl]methanethiosulfonate modification of Cys-478 blocked substrate binding, transport activity, and movement of TMH 8. The data suggest that Gly-478 is located within a mechanistically important hinge domain of TMH 11 in which substrate binding induces transport-related structural changes.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Activación del Canal Iónico/fisiología , Animales , Sitios de Unión , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Células HEK293 , Humanos , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Especificidad por Sustrato/fisiología , Xenopus laevis
14.
Biochim Biophys Acta ; 1824(3): 422-32, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22197591

RESUMEN

D-Serine dehydratase from Escherichia coli is a member of the ß-family (fold-type II) of the pyridoxal 5'-phosphate-dependent enzymes, catalyzing the conversion of D-serine to pyruvate and ammonia. The crystal structure of monomeric D-serine dehydratase has been solved to 1.97Å-resolution for an orthorhombic data set by molecular replacement. In addition, the structure was refined in a monoclinic data set to 1.55Å resolution. The structure of DSD reveals a larger pyridoxal 5'-phosphate-binding domain and a smaller domain. The active site of DSD is very similar to those of the other members of the ß-family. Lys118 forms the Schiff base to PLP, the cofactor phosphate group is liganded to a tetraglycine cluster Gly279-Gly283, and the 3-hydroxyl group of PLP is liganded to Asn170 and N1 to Thr424, respectively. In the closed conformation the movement of the small domain blocks the entrance to active site of DSD. The domain movement plays an important role in the formation of the substrate recognition site and the catalysis of the enzyme. Modeling of D-serine into the active site of DSD suggests that the hydroxyl group of D-serine is coordinated to the carboxyl group of Asp238. The carboxyl oxygen of D-serine is coordinated to the hydroxyl group of Ser167 and the amide group of Leu171 (O1), whereas the O2 of the carboxyl group of D-serine is hydrogen-bonded to the hydroxyl group of Ser167 and the amide group of Thr168. A catalytic mechanism very similar to that proposed for L-serine dehydratase is discussed.


Asunto(s)
Proteínas Bacterianas/química , Escherichia coli/química , Hidroliasas/química , Fosfato de Piridoxal/química , Secuencia de Aminoácidos , Aminoácidos , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Escherichia coli/enzimología , Hidroliasas/aislamiento & purificación , Hidroliasas/metabolismo , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Fosfato de Piridoxal/metabolismo
15.
J Transl Med ; 11: 4, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23294516

RESUMEN

BACKGROUND: Animal models of human inflammatory diseases have limited predictive quality for human clinical trials for various reasons including species specific activation mechanisms and the immunological background of the animals which markedly differs from the genetically heterogeneous and often aged patient population. OBJECTIVE: Development of an animal model allowing for testing therapeutics targeting pathways involved in the development of Atopic Dermatitis (AD) with better translatability to the patient. METHODS: NOD-scid IL2R γnull mice engrafted with human peripheral blood mononuclear cells (hPBMC) derived from patients suffering from AD and healthy volunteers were treated with IL-4 and the antagonistic IL-4 variant R121/Y124D (Pitrakinra). Levels of human (h)IgE, amount of B-, T- and plasma- cells and ratio of CD4 : CD8 positive cells served as read out for induction and inhibition of cell proliferation and hIgE secretion. Results were compared to in vitro analysis. RESULTS: hIgE secretion was induced by IL-4 and inhibited by the IL-4 antagonist Pitrakinra in vivo when formulated with methylcellulose. B-cells proliferated in response to IL-4 in vivo; the effect was abrogated by Pitrakinra. IL-4 shifted CD4 : CD8 ratios in vitro and in vivo when hPBMC derived from healthy volunteers were used. Pitrakinra reversed the effect. Human PBMC derived from patients with AD remained inert and engrafted mice reflected the individual responses observed in vitro. CONCLUSION: NOD-scid IL2R γnull mice engrafted with human PBMC reflect the immunological history of the donors and provide a complementary tool to in vitro studies. Thus, studies in this model might provide data with better translatability from bench to bedside.


Asunto(s)
Subunidad gamma Común de Receptores de Interleucina/genética , Monocitos/metabolismo , Transducción de Señal , Animales , Citometría de Flujo , Humanos , Inmunoglobulina E/metabolismo , Interleucina-4/metabolismo , Metilcelulosa/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
16.
Molecules ; 18(10): 11658-82, 2013 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-24071977

RESUMEN

Bone Morphogenetic Proteins (BMPs) are secreted protein hormones that act as morphogens and exert essential roles during embryonic development of tissues and organs. Signaling by BMPs occurs via hetero-oligomerization of two types of serine/threonine kinase transmembrane receptors. Due to the small number of available receptors for a large number of BMP ligands ligand-receptor promiscuity presents an evident problem requiring additional regulatory mechanisms for ligand-specific signaling. Such additional regulation is achieved through a plethora of extracellular antagonists, among them members of the Chordin superfamily, that modulate BMP signaling activity by binding. The key-element in Chordin-related antagonists for interacting with BMPs is the von Willebrand type C (VWC) module, which is a small domain of about 50 to 60 residues occurring in many different proteins. Although a structure of the VWC domain of the Chordin-member Crossveinless 2 (CV2) bound to BMP-2 has been determined by X-ray crystallography, the molecular mechanism by which the VWC domain binds BMPs has remained unclear. Here we present the NMR structure of the Danio rerio CV2 VWC1 domain in its unbound state showing that the key features for high affinity binding to BMP-2 is a pre-oriented peptide loop.


Asunto(s)
Proteínas Activadoras de GTPasa/química , Proteínas de Pez Cebra/química , Pez Cebra , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Proteína Morfogenética Ósea 2/química , Secuencia Conservada , Cistina/química , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
ACS Omega ; 8(28): 24841-24852, 2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37483220

RESUMEN

Interleukin-4 (IL-4) plays a key role in atopic diseases. It coordinates T-helper cell differentiation to subtype 2, thereby directing defense toward humoral immunity. Together with Interleukin-13, IL-4 further induces immunoglobulin class switch to IgE. Antibodies of this type activate mast cells and basophilic and eosinophilic granulocytes, which release pro-inflammatory mediators accounting for the typical symptoms of atopic diseases. IL-4 and IL-13 are thus major targets for pharmaceutical intervention strategies to treat atopic diseases. Besides neutralizing antibodies against IL-4, IL-13, or its receptors, IL-4 antagonists can present valuable alternatives. Pitrakinra, an Escherichia coli-derived IL-4 antagonist, has been evaluated in clinical trials for asthma treatment in the past; however, deficits such as short serum lifetime and potential immunogenicity among others stopped further development. To overcome such deficits, PEGylation of therapeutically important proteins has been used to increase the lifetime and proteolytic stability. As an alternative, glycoengineering is an emerging strategy used to improve pharmacokinetics of protein therapeutics. In this study, we have established different strategies to attach glycan moieties to defined positions in IL-4. Different chemical attachment strategies employing thiol chemistry were used to attach a glucose molecule at amino acid position 121, thereby converting IL-4 into a highly effective antagonist. To enhance the proteolytic stability of this IL-4 antagonist, additional glycan structures were introduced by glycoengineering utilizing eucaryotic expression. IL-4 antagonists with a combination of chemical and biosynthetic glycoengineering could be useful as therapeutic alternatives to IL-4 neutralizing antibodies already used to treat atopic diseases.

18.
Sci Rep ; 13(1): 22428, 2023 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-38104237

RESUMEN

Bone morphogenetic protein (BMP15) and growth differentiation factor (GDF9) are critical for ovarian follicular development and fertility and are associated with litter size in mammals. These proteins initially exist as pre-pro-mature proteins, that are subsequently cleaved into biologically active forms. Thus, the molecular forms of GDF9 and BMP15 may provide the key to understanding the differences in litter size determination in mammals. Herein, we compared GDF9 and BMP15 forms in mammals with high (pigs) and low to moderate (sheep) and low (red deer) ovulation-rate. In all species, oocyte lysates and secretions contained both promature and mature forms of BMP15 and GDF9. Whilst promature and mature GDF9 levels were similar between species, deer produced more BMP15 and exhibited, together with sheep, a higher promature:mature BMP15 ratio. N-linked glycosylation was prominant in proregion and mature GDF9 and in proregion BMP15 of pigs, and present in proregion GDF9 of sheep. There was no evidence of secreted native homo- or hetero-dimers although a GDF9 dimer in red deer oocyte lysate was detected. In summary, GDF9 appeared to be equally important in all species regardless of litter size, whilst BMP15 levels were highest in strict monovulatory species.


Asunto(s)
Proteína Morfogenética Ósea 15 , Factor 9 de Diferenciación de Crecimiento , Tamaño de la Camada , Animales , Femenino , Embarazo , Proteína Morfogenética Ósea 15/genética , Ciervos , Fertilidad , Factor 9 de Diferenciación de Crecimiento/genética , Oocitos/metabolismo , Ovulación , Ovinos , Porcinos
19.
Bioconjug Chem ; 23(7): 1396-405, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22681442

RESUMEN

Interleukin-4 (IL-4) is a prototypical regulator protein of the immune system that is crucial for the pathogenesis and maintenance of asthma and other atopic diseases. It, together with IL-13, uses the IL-4 receptor α chain (IL-4Rα) to signal into immune and other cells. An IL-4 mutein acting as a dual IL-4/IL-13 receptor antagonist is in clinical development. Here, it is described how IL-4 muteins containing a single engineered cysteine with a free thiol can be prepared and used for site-specific chemical modification. The muteins were initially expressed in E. coli, refolded, and purified, but not in a fully reduced nonconjugated form. Attempts to reduce the cysteine chemically failed because the native disulfide bonds of IL-4 were also reduced under similar conditions. Therefore, an enzymatic procedure was developed to reduce glutathionylated IL-4 cysteine muteins employing glutaredoxin and reduced glutathione. Cysteine muteins engineered at four different positions around the IL-4Rα binding site were enzymatically reduced at different rates. All muteins were prepared with free thiol in reasonable yield and were modified by N-ethylmaleimide (NEM) or maleimido-PEG. The effect on IL-4Rα binding of cysteine substitution and of the site-specific modification by glutathione, N-ethylmaleimide (NEM), or a branched 2.36 kDa poly(ethylene glycol) (PEG) will be discussed.


Asunto(s)
Cisteína/metabolismo , Glutatión Reductasa/metabolismo , Glutatión/metabolismo , Interleucina-4/química , Interleucina-4/metabolismo , Ingeniería de Proteínas , Compuestos de Sulfhidrilo/análisis , Cisteína/química , Humanos , Modelos Moleculares , Estructura Molecular , Compuestos de Sulfhidrilo/química
20.
J Biol Chem ; 285(53): 41614-26, 2010 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-20952383

RESUMEN

Sclerostin is expressed by osteocytes and has catabolic effects on bone. It has been shown to antagonize bone morphogenetic protein (BMP) and/or Wnt activity, although at present the underlying mechanisms are unclear. Consistent with previous findings, Sclerostin opposed direct Wnt3a-induced but not direct BMP7-induced responses when both ligand and antagonist were provided exogenously to cells. However, we found that when both proteins are expressed in the same cell, sclerostin can antagonize BMP signaling directly by inhibiting BMP7 secretion. Sclerostin interacts with both the BMP7 mature domain and pro-domain, leading to intracellular retention and proteasomal degradation of BMP7. Analysis of sclerostin knock-out mice revealed an inhibitory action of sclerostin on Wnt signaling in both osteoblasts and osteocytes in cortical and cancellous bones. BMP7 signaling was predominantly inhibited by sclerostin in osteocytes of the calcaneus and the cortical bone of the tibia. Our results suggest that sclerostin exerts its potent bone catabolic effects by antagonizing Wnt signaling in a paracrine and autocrine manner and antagonizing BMP signaling selectively in the osteocytes that synthesize simultaneously both sclerostin and BMP7 proteins.


Asunto(s)
Proteína Morfogenética Ósea 7/química , Proteínas Morfogenéticas Óseas/química , Marcadores Genéticos/fisiología , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Alelos , Animales , Proteínas Morfogenéticas Óseas/fisiología , Femenino , Glicoproteínas , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Noqueados , Transducción de Señal , Resonancia por Plasmón de Superficie , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA