Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Cell Mol Life Sci ; 79(6): 312, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35604495

RESUMEN

The human signaling molecules Tie1 and Tie2 receptor tyrosine kinases (RTKs) play important pathophysiological roles in many diseases, including different cancers. The activity of Tie1 is mediated mainly through the downstream angiopoietin-1 (Ang1)-dependent activation of Tie2, rendering both Tie 1 and the Tie1/Tie2/Ang1 axis attractive putative targets for therapeutic intervention. However, the development of inhibitors that target Tie1 and an understanding of their effect on Tie2 and on the Tie1/Tie2/Ang1 axis remain unfulfilled tasks, due, largely, to the facts that Tie1 is an orphan receptor and is difficult to produce and use in the quantities required for immune antibody library screens. In a search for a selective inhibitor of this orphan receptor, we sought to exploit the advantages (e.g., small size that allows binding to hidden epitopes) of non-immune nanobodies and to simultaneously overcome their limitations (i.e., low expression and stability). We thus performed expression, stability, and affinity screens of yeast-surface-displayed naïve and predesigned synthetic (non-immune) nanobody libraries against the Tie1 extracellular domain. The screens yielded a nanobody with high expression and good affinity and specificity for Tie1, thereby yielding preferential binding for Tie1 over Tie2. The stability, selectivity, potency, and therapeutic potential of this synthetic nanobody were profiled using in vitro and cell-based assays. The nanobody triggered Tie1-dependent inhibition of RTK (Tie2, Akt, and Fak) phosphorylation and angiogenesis in endothelial cells, as well as suppression of human glioblastoma cell viability and migration. This study opens the way to developing nanobodies as therapeutics for different cancers associated with Tie1 activation.


Asunto(s)
Neoplasias , Anticuerpos de Dominio Único , Angiopoyetina 1 , Células Endoteliales/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Fosforilación , Receptor TIE-1/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Anticuerpos de Dominio Único/farmacología
2.
Cell Mol Life Sci ; 79(2): 113, 2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35099616

RESUMEN

Induction of bone formation by Wnt ligands is inhibited when sclerostin (Scl), an osteocyte-produced antagonist, binds to its receptors, the low-density lipoprotein receptor-related proteins 5 or 6 (LRP5/6). Recently, it was shown that enhanced inhibition is achieved by Scl binding to the co-receptor LRP4. However, it is not clear if the binding of Scl to LRP4 facilitates Scl binding to LRP5/6 or inhibits the Wnt pathway in an LRP5/6-independent manner. Here, using the yeast display system, we demonstrate that Scl exhibits a stronger binding affinity for LRP4 than for LRP6. Moreover, we found stronger Scl binding to LRP6 in the presence of LRP4. We further show that a Scl mutant (SclN93A), which tightly binds LRP4 but not LRP6, does not inhibit the Wnt pathway on its own. We demonstrate that SclN93A competes with Scl for a common binding site on LRP4 and antagonizes Scl inhibition of the Wnt signaling pathway in osteoblasts in vitro. Finally, we demonstrate that 2 weeks of bi-weekly subcutaneous injections of SclN93A fused to the fragment crystallizable (Fc) domain of immunoglobulin (SclN93AFc), which retains the antagonistic activity of the mutant, significantly increases bone formation rate and enhances trabecular volumetric bone fraction, trabecular number, and bone length in developing mice. Our data show that LRP4 serves as an anchor that facilitates Scl-LRP6 binding and that inhibition of the Wnt pathway by Scl depends on its prior binding to LRP4. We further provide evidence that compounds that inhibit Scl-LRP4 interactions offer a potential strategy to promote anabolic bone functions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Osteogénesis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Unión Competitiva/efectos de los fármacos , Unión Competitiva/genética , Células Cultivadas , Femenino , Células HEK293 , Humanos , Proteínas Relacionadas con Receptor de LDL/antagonistas & inhibidores , Proteínas Relacionadas con Receptor de LDL/química , Proteínas Relacionadas con Receptor de LDL/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Mutantes/química , Proteínas Mutantes/farmacología , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas/genética , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/química
3.
Proteins ; 90(1): 45-57, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34293212

RESUMEN

Deep mutational scanning provides unprecedented wealth of quantitative data regarding the functional outcome of mutations in proteins. A single experiment may measure properties (eg, structural stability) of numerous protein variants. Leveraging the experimental data to gain insights about unexplored regions of the mutational landscape is a major computational challenge. Such insights may facilitate further experimental work and accelerate the development of novel protein variants with beneficial therapeutic or industrially relevant properties. Here we present a novel, machine learning approach for the prediction of functional mutation outcome in the context of deep mutational screens. Using sequence (one-hot) features of variants with known properties, as well as structural features derived from models thereof, we train predictive statistical models to estimate the unknown properties of other variants. The utility of the new computational scheme is demonstrated using five sets of mutational scanning data, denoted "targets": (a) protease specificity of APPI (amyloid precursor protein inhibitor) variants; (b-d) three stability related properties of IGBPG (immunoglobulin G-binding ß1 domain of streptococcal protein G) variants; and (e) fluorescence of GFP (green fluorescent protein) variants. Performance is measured by the overall correlation of the predicted and observed properties, and enrichment-the ability to predict the most potent variants and presumably guide further experiments. Despite the diversity of the targets the statistical models can generalize variant examples thereof and predict the properties of test variants with both single and multiple mutations.


Asunto(s)
Análisis Mutacional de ADN/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Aprendizaje Automático , Mutación/genética , Proteínas , Algoritmos , Biología Computacional/métodos , Modelos Estadísticos , Mapas de Interacción de Proteínas , Proteínas/química , Proteínas/genética , Proteínas/metabolismo
4.
Bioconjug Chem ; 33(5): 795-806, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35446024

RESUMEN

Tissue inhibitors of metalloproteinases (TIMPs) are natural inhibitors of the matrix metalloproteinase (MMP) family of proteins, whose members are key regulators of the proteolysis of extracellular matrix components and hence of multiple biological processes. In particular, imbalanced activity of matrix metalloproteinase-14 (MMP-14) may lead to the development of cancer and cardiovascular and other diseases. This study aimed to engineer TIMP2, one of the four homologous TIMPs, as a potential therapeutic by virtue of its ability to bind to the active-site Zn2+ of MMP-14. However, the susceptibility to degradation of TIMP2 and its small size, which results in a short circulation half-life, limit its use as a therapeutic. PEGylation was thus used to improve the pharmacokinetic profile of TIMP2. PEGylation of the MMP-targeting N-terminal domain of TIMP2 (N-TIMP2), via either cysteine or lysine residues, resulted in a significant decrease in N-TIMP2 affinity toward MMP-14 or multisite conjugation and conjugate heterogeneity, respectively. Our strategy designed to address this problem was based on incorporating a noncanonical amino acid (NCAA) into N-TIMP2 to enable site-specific mono-PEGylation. The first step was to incorporate the NCAA propargyl lysine (PrK) at position S31 in N-TIMP2, which does not interfere with the N-TIMP2-MMP-14 binding interface. Thereafter, site-specific PEGylation was achieved via a click chemistry reaction between N-TIMP2-S31PrK and PEG-azide-20K. Inhibition studies showed that PEGylated N-TIMP2-S31PrK did indeed retain its inhibitory activity toward MMP-14. The modified protein also showed improved serum stability vs non-PEGylated N-TIMP2. In vivo pharmacokinetic studies in mice revealed a significant 8-fold increase in the elimination half-life of PEGylated N-TIMP2 vs the non-PEGylated protein. This study shows that site-specific bioorthogonal mono-PEGylation extends the half-life of N-TIMP2 without impairing its biological activity, thereby highlighting the advantage of this strategy for generating potent PEGylated proteins.


Asunto(s)
Lisina , Metaloproteinasa 14 de la Matriz , Inhibidor Tisular de Metaloproteinasa-2 , Animales , Semivida , Lisina/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasas de la Matriz , Ratones , Polietilenglicoles/química , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
5.
Cell Mol Life Sci ; 78(23): 7185-7198, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34643743

RESUMEN

A review of the multidisciplinary scientific literature reveals a large variety of amyloid-ß (Aß) oligomeric species, differing in molecular weight, conformation and morphology. These species, which may assemble via either on- or off-aggregation pathways, exhibit differences in stability, function and neurotoxicity, according to different experimental settings. The conformations of the different Aß species are stabilized by intra- and inter-molecular hydrogen bonds and by electrostatic and hydrophobic interactions, all depending on the chemical and physical environment (e.g., solvent, ions, pH) and interactions with other molecules, such as lipids and proteins. This complexity and the lack of a complete understanding of the relationship between the different Aß species and their toxicity is currently dictating the nature of the inhibitor (or inducer)-based approaches that are under development for interfering with (or inducing) the formation of specific species and Aß oligomerization, and for interfering with the associated downstream neurotoxic effects. Here, we review the principles that underlie the involvement of different Aß oligomeric species in neurodegeneration, both in vitro and in preclinical studies. In addition, we provide an overview of the existing inhibitors (or inducers) of Aß oligomerization that serve as potential therapeutics for neurodegenerative diseases. The review, which covers the exciting studies that have been published in the past few years, comprises three main parts: 1) on- and off-fibrillar assembly mechanisms and Aß structural polymorphism; 2) interactions of Aß with other molecules and cell components that dictate the Aß aggregation pathway; and 3) targeting the on-fibrillar Aß assembly pathway as a therapeutic approach.


Asunto(s)
Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Amiloide/química , Enfermedades Neurodegenerativas/patología , Fragmentos de Péptidos/metabolismo , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/terapia , Fragmentos de Péptidos/genética , Agregación Patológica de Proteínas/patología , Conformación Proteica
6.
Cell Mol Life Sci ; 78(5): 2145-2155, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32844279

RESUMEN

BIM is a key apoptotic protein, participating in diverse cellular processes. Interestingly, recent studies have hypothesized that BIM is associated with the extensive neuronal cell death encountered in protein misfolding diseases, such as Alzheimer's disease. Here, we report that the core pro-apoptotic domain of BIM, the BIM-BH3 motif, forms ubiquitous amyloid fibrils. The BIM-BH3 fibrils exhibit cytotoxicity, disrupt mitochondrial functions, and modulate the structures and dynamics of mitochondrial membrane mimics. Interestingly, a slightly longer peptide in which BIM-BH3 was flanked by four additional residues, widely employed as a model of the pro-apoptotic core domain of BIM, did not form fibrils, nor exhibited cell disruptive properties. The experimental data suggest a new mechanistic role for the BIM-BH3 domain, and demonstrate, for the first time, that an apoptotic peptide forms toxic amyloid fibrils.


Asunto(s)
Amiloide/química , Apoptosis , Proteína 11 Similar a Bcl2/química , Dominios Proteicos , Secuencia de Aminoácidos , Amiloide/metabolismo , Amiloide/ultraestructura , Proteína 11 Similar a Bcl2/genética , Proteína 11 Similar a Bcl2/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Dicroismo Circular , Complejo IV de Transporte de Electrones/metabolismo , Humanos , Microscopía Electrónica , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Homología de Secuencia de Aminoácido
7.
Biochem J ; 478(5): 1139-1157, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33600567

RESUMEN

Crosstalk of the oncogenic matrix metalloproteinase-9 (MMP9) and one of its ligands, CD44, involves cleavage of CD44 by the MMP9 catalytic domain, with the CD44-MMP9 interaction on the cell surface taking place through the MMP9 hemopexin domain (PEX). This interaction promotes cancer cell migration and invasiveness. In concert, MMP9-processed CD44 induces the expression of MMP9, which degrades ECM components and facilitates growth factor release and activation, cancer cell invasiveness, and metastasis. Since both MMP9 and CD44 contribute to cancer progression, we have developed a new strategy to fully block this neoplastic process by engineering a multi-specific inhibitor that simultaneously targets CD44 and both the catalytic and PEX domains of MMP9. Using a yeast surface display technology, we first obtained a high-affinity inhibitor for the MMP9 catalytic domain, which we termed C9, by modifying a natural non-specific MMP inhibitor, N-TIMP2. We then conjugated C9 via a flexible linker to PEX, thereby creating a multi-specific inhibitor (C9-PEX) that simultaneously targets the MMP9 catalytic and PEX domains and CD44. It is likely that, via its co-localization with CD44, C9-PEX may compete with MMP9 localization on the cell surface, thereby inhibiting MMP9 catalytic activity, reducing MMP9 cellular levels, interfering with MMP9 homodimerization, and reducing the activation of downstream MAPK/ERK pathway signaling. The developed platform could be extended to other oncogenic MMPs as well as to other important target proteins, thereby offering great promise for creating novel multi-specific therapeutics for cancer and other diseases.


Asunto(s)
Hemopexina/antagonistas & inhibidores , Receptores de Hialuranos/antagonistas & inhibidores , Metaloproteinasa 9 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Movimiento Celular , Proliferación Celular , Hemopexina/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Células MCF-7 , Metaloproteinasa 9 de la Matriz/metabolismo , Fosforilación , Transducción de Señal
8.
J Am Chem Soc ; 143(41): 17261-17275, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34609866

RESUMEN

Protein-protein interactions (PPIs) have evolved to display binding affinities that can support their function. As such, cognate and noncognate PPIs could be highly similar structurally but exhibit huge differences in binding affinities. To understand this phenomenon, we study three homologous protease-inhibitor PPIs that span 9 orders of magnitude in binding affinity. Using state-of-the-art methodology that combines protein randomization, affinity sorting, deep sequencing, and data normalization, we report quantitative binding landscapes consisting of ΔΔGbind values for the three PPIs, gleaned from tens of thousands of single and double mutations. We show that binding landscapes of the three complexes are strikingly different and depend on the PPI evolutionary optimality. We observe different patterns of couplings between mutations for the three PPIs with negative and positive epistasis appearing most frequently at hot-spot and cold-spot positions, respectively. The evolutionary trends observed here are likely to be universal to other biological complexes in the cell.


Asunto(s)
Mapeo de Interacción de Proteínas
9.
PLoS Biol ; 16(8): e2002979, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30142160

RESUMEN

There is currently a demand for new highly efficient and specific drugs to treat osteoporosis, a chronic bone disease affecting millions of people worldwide. We have developed a combinatorial strategy for engineering bispecific inhibitors that simultaneously target the unique combination of c-FMS and αvß3 integrin, which act in concert to facilitate bone resorption by osteoclasts. Using functional fluorescence-activated cell sorting (FACS)-based screening assays of random mutagenesis macrophage colony-stimulating factor (M-CSF) libraries against c-FMS and αvß3 integrin, we engineered dual-specific M-CSF mutants with high affinity to both receptors. These bispecific mutants act as functional antagonists of c-FMS and αvß3 integrin activation and hence of osteoclast differentiation in vitro and osteoclast activity in vivo. This study thus introduces a versatile platform for the creation of new-generation therapeutics with high efficacy and specificity for osteoporosis and other bone diseases. It also provides new tools for studying molecular mechanisms and the cell signaling pathways that mediate osteoclast differentiation and function.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/prevención & control , Integrina alfaVbeta3/antagonistas & inhibidores , Factor Estimulante de Colonias de Macrófagos/farmacología , Osteoclastos/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Animales , Sitios de Unión , Conservadores de la Densidad Ósea/química , Conservadores de la Densidad Ósea/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Humanos , Integrina alfaVbeta3/química , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Factor Estimulante de Colonias de Macrófagos/química , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Simulación del Acoplamiento Molecular , Mutación , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Ingeniería de Proteínas , Dominios y Motivos de Interacción de Proteínas , Receptor de Factor Estimulante de Colonias de Macrófagos/química , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
10.
Biochem J ; 477(9): 1701-1719, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32296833

RESUMEN

To facilitate investigations of protein-protein interactions (PPIs), we developed a novel platform for quantitative mapping of protein binding specificity landscapes, which combines the multi-target screening of a mutagenesis library into high- and low-affinity populations with sophisticated next-generation sequencing analysis. Importantly, this method generates accurate models to predict affinity and specificity values for any mutation within a protein complex, and requires only a few experimental binding affinity measurements using purified proteins for calibration. We demonstrated the utility of the approach by mapping quantitative landscapes for interactions between the N-terminal domain of the tissue inhibitor of metalloproteinase 2 (N-TIMP2) and three matrix metalloproteinases (MMPs) having homologous structures but different affinities (MMP-1, MMP-3, and MMP-14). The binding landscapes for N-TIMP2/MMP-1 and N-TIMP2/MMP-3 showed the PPIs to be almost fully optimized, with most single mutations giving a loss of affinity. In contrast, the non-optimized PPI for N-TIMP2/MMP-14 was reflected in a wide range of binding affinities, where single mutations exhibited a far more attenuated effect on the PPI. Our new platform reliably and comprehensively identified not only hot- and cold-spot residues, but also specificity-switch mutations that shape target affinity and specificity. Thus, our approach provides a methodology giving an unprecedentedly rich quantitative analysis of the binding specificity landscape, which will broaden the understanding of the mechanisms and evolutionary origins of specific PPIs and facilitate the rational design of specific inhibitors for structurally similar target proteins.


Asunto(s)
Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Inhibidor Tisular de Metaloproteinasa-2/genética , Biología Computacional/métodos , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Metaloproteinasa 3 de la Matriz/metabolismo , Mutagénesis , Mutación , Ingeniería de Proteínas/métodos , Inhibidor Tisular de Metaloproteinasa-2/química , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
11.
Trends Biochem Sci ; 41(5): 421-433, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27061494

RESUMEN

Two alternative strategies are commonly used to study protein-protein interactions (PPIs) and to engineer protein-based inhibitors. In one approach, binders are selected experimentally from combinatorial libraries of protein mutants that are displayed on a cell surface. In the other approach, computational modeling is used to explore an astronomically large number of protein sequences to select a small number of sequences for experimental testing. While both approaches have some limitations, their combination produces superior results in various protein engineering applications. Such applications include the design of novel binders and inhibitors, the enhancement of affinity and specificity, and the mapping of binding epitopes. The combination of these approaches also aids in the understanding of the specificity profiles of various PPIs.


Asunto(s)
Evolución Molecular Dirigida/métodos , Biblioteca de Péptidos , Ingeniería de Proteínas/métodos , Proteínas/química , Secuencia de Aminoácidos , Bacteriófagos/genética , Bacteriófagos/metabolismo , Sitios de Unión , Humanos , Mutación , Mapeo Peptídico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas/genética , Proteínas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
12.
J Biol Chem ; 294(13): 5105-5120, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30700553

RESUMEN

Serine protease inhibitors of the Kunitz-bovine pancreatic trypsin inhibitor (BPTI) family are ubiquitous biological regulators of proteolysis. These small proteins are resistant to proteolysis, but can be slowly cleaved within the protease-binding loop by target proteases, thereby compromising their activity. For the human protease mesotrypsin, this cleavage is especially rapid. Here, we aimed to stabilize the Kunitz domain structure against proteolysis through disulfide engineering. Substitution within the Kunitz inhibitor domain of the amyloid precursor protein (APPI) that incorporated a new disulfide bond between residues 17 and 34 reduced proteolysis by mesotrypsin 74-fold. Similar disulfide engineering of tissue factor pathway inhibitor-1 Kunitz domain 1 (KD1TFPI1) and bikunin Kunitz domain 2 (KD2bikunin) likewise stabilized these inhibitors against mesotrypsin proteolysis 17- and 6.6-fold, respectively. Crystal structures of disulfide-engineered APPI and KD1TFPI1 variants in a complex with mesotrypsin at 1.5 and 2.0 Å resolution, respectively, confirmed the formation of well-ordered disulfide bonds positioned to stabilize the binding loop. Long all-atom molecular dynamics simulations of disulfide-engineered Kunitz domains and their complexes with mesotrypsin revealed conformational stabilization of the primed side of the inhibitor-binding loop by the engineered disulfide, along with global suppression of conformational dynamics in the Kunitz domain. Our findings suggest that the Cys-17-Cys-34 disulfide slows proteolysis by dampening conformational fluctuations in the binding loop and minimizing motion at the enzyme-inhibitor interface. The generalizable approach developed here for the stabilization against proteolysis of Kunitz domains, which can serve as important scaffolds for therapeutics, may thus find applications in drug development.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Aprotinina/metabolismo , Tripsina/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animales , Aprotinina/química , Aprotinina/genética , Cristalografía por Rayos X , Disulfuros/química , Disulfuros/metabolismo , Humanos , Modelos Moleculares , Conformación Proteica , Dominios Proteicos , Ingeniería de Proteínas , Proteolisis , Tripsina/química
13.
Molecules ; 25(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096693

RESUMEN

Receptor tyrosine kinases (RTKs) are major players in signal transduction, regulating cellular activities in both normal regeneration and malignancy. Thus, many RTKs, c-Kit among them, play key roles in the function of both normal and neoplastic cells, and as such constitute attractive targets for therapeutic intervention. We thus sought to manipulate the self-association of stem cell factor (SCF), the cognate ligand of c-Kit, and hence its suboptimal affinity and activation potency for c-Kit. To this end, we used directed evolution to engineer SCF variants having different c-Kit activation potencies. Our yeast-displayed SCF mutant (SCFM) library screens identified altered dimerization potential and increased affinity for c-Kit by specific SCF-variants. We demonstrated the delicate balance between SCF homo-dimerization, c-Kit binding, and agonistic potencies by structural studies, in vitro binding assays and a functional angiogenesis assay. Importantly, our findings showed that a monomeric SCF variant exhibited superior agonistic potency vs. the wild-type SCF protein and vs. other high-affinity dimeric SCF variants. Our data showed that action of the monomeric ligands in binding to the RTK monomers and inducing receptor dimerization and hence activation was superior to that of the wild-type dimeric ligand, which has a higher affinity to RTK dimers but a lower activation potential. The findings of this study on the binding and c-Kit activation of engineered SCF variants thus provides insights into the structure-function dynamics of ligands and RTKs.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/agonistas , Factor de Células Madre/farmacología , Línea Celular Tumoral , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/genética
14.
J Biol Chem ; 293(34): 13310-13326, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-29986882

RESUMEN

The pathophysiological functions of the signaling molecules matrix metalloproteinase-14 (MMP-14) and integrin αvß3 in various types of cancer are believed to derive from their collaborative activity in promoting invasion, metastasis, and angiogenesis, as shown in vitro and in vivo The two effectors act in concert in a cell-specific manner through the localization of pro-MMP-2 to the cell surface, where it is processed to intermediate and matured MMP-2. The matured MMP-2 product is localized to the cell surface via its binding to integrin αvß3 The MMP-14/MMP-2/integrin αvß3 axis thus constitutes an attractive putative target for therapeutic interventions, but the development of inhibitors that target this axis remains an unfulfilled task. To address the lack of such multitarget inhibitors, we have established a combinatorial approach that is based on flow cytometry screening of a yeast-displayed N-TIMP2 (N-terminal domain variant of tissue inhibitor of metalloproteinase-2) mutant library. On the basis of this screening, we generated protein monomers and a heterodimer that contain monovalent and bivalent binding epitopes to MMP-14 and integrin αvß3 Among these proteins, the bi-specific heterodimer, which bound strongly to both MMP-14 and integrin αvß3, exhibited superior ability to inhibit MMP-2 activation and displayed the highest inhibitory activity in cell-based models of a MMP-14-, MMP-2-, and integrin αvß3-dependent glioblastoma and of endothelial cell invasiveness and endothelial capillary tube formation. These assays enabled us to show the superiority of the combined target effects of the inhibitors and to investigate separately the role each of the three signaling molecules in various malignant processes.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Integrina alfaVbeta3/antagonistas & inhibidores , Metaloproteinasa 14 de la Matriz/química , Metaloproteinasa 2 de la Matriz/química , Neovascularización Patológica/tratamiento farmacológico , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Animales , Movimiento Celular , Técnicas Químicas Combinatorias , Glioblastoma/metabolismo , Glioblastoma/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Desnudos , Mutación , Dominios Proteicos , Inhibidor Tisular de Metaloproteinasa-2/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Biol Chem ; 293(33): 12663-12680, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29934309

RESUMEN

Human tissue kallikrein (KLK) proteases are hormone-like signaling molecules with important functions in cancer pathophysiology. KLK-related peptidase 6 (KLK6), specifically, is highly up-regulated in several types of cancer, where its increased activity promotes cancer invasion and metastasis. This characteristic suggests KLK6 as an attractive target for therapeutic interventions. However, inhibitors that specifically target KLK6 have not yet been reported, possibly because KLK6 shares a high sequence homology and structural similarity with other serine proteases and resists inhibition by many polypeptide inhibitors. Here, we present an innovative combinatorial approach to engineering KLK6 inhibitors via flow cytometry-based screening of a yeast-displayed mutant library of the human amyloid precursor protein Kunitz protease inhibitor domain (APPI), an inhibitor of other serine proteases, such as anionic and cationic trypsins. On the basis of this screening, we generated APPIM17L,I18F,S19F,F34V (APPI-4M), an APPI variant with a KLK6 inhibition constant (Ki ) of 160 pm and a turnover time of 10 days. To the best of our knowledge, APPI-4M is the most potent KLK6 inhibitor reported to date, displaying 146-fold improved affinity and 13-fold improved proteolytic stability compared with WT APPI (APPIWT). We further demonstrate that APPI-4M acts as a functional inhibitor in a cell-based model of KLK6-dependent breast cancer invasion. Finally, the crystal structures of the APPIWT/KLK6 and APPI-4M/KLK6 complexes revealed the structural and mechanistic bases for the improved KLK6 binding and proteolytic resistance of APPI-4M. We anticipate that APPI-4M will have substantial translational potential as both imaging agent and therapeutic.


Asunto(s)
Precursor de Proteína beta-Amiloide/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ingeniería Genética , Calicreínas/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Proteolisis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Calicreínas/química , Modelos Moleculares , Unión Proteica , Conformación Proteica , Células Tumorales Cultivadas
16.
Proteins ; 87(9): 738-747, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31017342

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by selective death of motor neurons in the brainstem, motor cortex, and spinal cord, leading to muscle atrophy and eventually to death. It is currently held that various oligomerization-inducing mutations in superoxide dismutase 1 (SOD1), an amyloid-forming protein, may be implicated in the familial form of this fast-progressing highly lethal neurodegenerative disease. A possible therapeutic approach could therefore lie in developing inhibitors to SOD1 mutants. By screening a focused mutagenesis library, mutated randomly in specific "stability patch" positions of the B1 domain of protein G (HTB1), we previously identified low affinity inhibitors of aggregation of SOD1G93A and SOD1G85R mutants. Herein, with the aim to generate a more potent inhibitor with higher affinity to SOD1 mutants, we employed an unbiased, random mutagenesis approach covering the entire sequence space of HTB1 to optimize as yet undefined positions for improved interactions with SOD1. Using affinity maturation screens in yeast, we identified a variant, which we designated HTB1M3 , that bound strongly to SOD1 misfolded mutants but not to wild-type SOD1. In-vitro aggregation assays indicated that in the presence of HTB1M3 misfolded SOD1 assembled into oligomeric species that were not toxic to NSC-34 neuronal cells. In addition, when NSC-34 cells were exposed to misfolded SOD1 mutants, either soluble or preaggregated, in the presence of HTB1M3 , this inhibitor prevented the prion-like propagation of SOD1 from one neuronal cell to another by blocking the penetration of SOD1 into the neuronal cells.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Línea Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Citometría de Flujo , Humanos , Ratones , Mutación , Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo , Pliegue de Proteína , Superóxido Dismutasa-1/genética
17.
Proteins ; 87(3): 185-197, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30520519

RESUMEN

We combined computational and experimental methods to interrogate the binding determinants of angiopoietin-2 (Ang2) to its receptor tyrosine kinase (RTK) Tie2-a central signaling system in angiogenesis, inflammation, and tumorigenesis. We used physics-based electrostatic and surface-area calculations to identify the subset of interfacial Ang2 and Tie2 residues that can affect binding directly. Using random and site-directed mutagenesis and yeast surface display (YSD), we validated these predictions and identified additional Ang2 positions that affected receptor binding. We then used burial-based calculations to classify the larger set of Ang2 residues that are buried in the Ang2 core, whose mutations can perturb the Ang2 structure and thereby affect interactions with Tie2 indirectly. Our analysis showed that the Ang2-Tie2 interface is dominated by nonpolar contributions, with only three Ang2 and two Tie2 residues that contribute electrostatically to intermolecular interactions. Individual interfacial residues contributed only moderately to binding, suggesting that engineering of this interface will require multiple mutations to reach major effects. Conversely, substitutions in substantially buried Ang2 residues were more prevalent in our experimental screen, reduced binding substantially, and are therefore more likely to have a deleterious effect that might contribute to oncogenesis. Computational analysis of additional RTK-ligand complexes, c-Kit-SCF and M-CSF-c-FMS, and comparison to previous YSD results, further show the utility of our combined methodology.


Asunto(s)
Complejos Multiproteicos/química , Mapas de Interacción de Proteínas/genética , Receptor TIE-2/química , Proteínas de Transporte Vesicular/química , Carcinogénesis/genética , Simulación por Computador , Humanos , Inflamación/genética , Ligandos , Complejos Multiproteicos/genética , Mutagénesis Sitio-Dirigida , Mutación/genética , Neovascularización Patológica/genética , Unión Proteica/genética , Proteínas Proto-Oncogénicas c-kit/química , Receptor TIE-2/genética , Transducción de Señal/genética , Factor de Células Madre/química , Proteínas de Transporte Vesicular/genética
18.
Biochem J ; 475(19): 3087-3103, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30213841

RESUMEN

Aggregation and accumulation of the 42-residue amyloid ß peptide (Aß42) in the extracellular matrix and within neuronal cells is considered a major cause of neuronal cell cytotoxicity and death in Alzheimer's disease (AD) patients. Therefore, molecules that bind to Aß42 and prevent its aggregation are therapeutically promising as AD treatment. Here, we show that a non-self-aggregating Aß42 variant carrying two surface mutations, F19S and L34P (Aß42DM), inhibits wild-type Aß42 aggregation and significantly reduces Aß42-mediated cell cytotoxicity. In addition, Aß42DM inhibits the uptake and internalization of extracellularly added pre-formed Aß42 aggregates into cells. This was the case in both neuronal and non-neuronal cells co-expressing Aß42 and Aß42DM or following pre-treatment of cells with extracellular soluble forms of the two peptides, even at high Aß42 to Aß42DM molar ratios. In cells, Aß42DM associates with Aß42, while in vitro, the two soluble recombinant peptides exhibit nano-molar binding affinity. Importantly, Aß42DM potently suppresses Aß42 amyloid aggregation in vitro, as demonstrated by thioflavin T fluorescence and transmission electron microscopy for detecting amyloid fibrils. Overall, we present a new approach for inhibiting Aß42 fibril formation both within and outside cells. Accordingly, Aß42DM should be evaluated in vivo for potential use as a therapeutic lead for treating AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Líquido Extracelular/metabolismo , Variación Genética/fisiología , Líquido Intracelular/metabolismo , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Amiloide/genética , Péptidos beta-Amiloides/genética , Línea Celular Tumoral , Supervivencia Celular/fisiología , Células HEK293 , Humanos , Fragmentos de Péptidos/genética , Agregación Patológica de Proteínas/genética , Resonancia por Plasmón de Superficie/métodos
19.
Biochem J ; 475(7): 1335-1352, 2018 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-29535275

RESUMEN

High structural and sequence similarity within protein families can pose significant challenges to the development of selective inhibitors, especially toward proteolytic enzymes. Such enzymes usually belong to large families of closely similar proteases and may also hydrolyze, with different rates, protein- or peptide-based inhibitors. To address this challenge, we employed a combinatorial yeast surface display library approach complemented with a novel pre-equilibrium, competitive screening strategy for facile assessment of the effects of multiple mutations on inhibitor association rates and binding specificity. As a proof of principle for this combined approach, we utilized this strategy to alter inhibitor/protease association rates and to tailor the selectivity of the amyloid ß-protein precursor Kunitz protease inhibitor domain (APPI) for inhibition of the oncogenic protease mesotrypsin, in the presence of three competing serine proteases, anionic trypsin, cationic trypsin and kallikrein-6. We generated a variant, designated APPIP13W/M17G/I18F/F34V, with up to 30-fold greater specificity relative to the parental APPIM17G/I18F/F34V protein, and 6500- to 230 000-fold improved specificity relative to the wild-type APPI protein in the presence of the other proteases tested. A series of molecular docking simulations suggested a mechanism of interaction that supported the biochemical results. These simulations predicted that the selectivity and specificity are affected by the interaction of the mutated APPI residues with nonconserved enzyme residues located in or near the binding site. Our strategy will facilitate a better understanding of the binding landscape of multispecific proteins and will pave the way for design of new drugs and diagnostic tools targeting proteases and other proteins.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Biblioteca de Péptidos , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Tripsina/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Unión Competitiva , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Especificidad por Sustrato , Tripsina/genética
20.
BMC Biol ; 16(1): 92, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-30119679

RESUMEN

BACKGROUND: Increased activity of the receptor tyrosine kinase Tie2 has been implicated in the promotion of pathological angiogenesis. This activity is mainly mediated through angiopoietin (Ang)1- and Ang2-dependent activation of integrins by Tie2, rendering the Ang/Tie2/integrin axis an attractive putative target for cancer therapeutics. RESULTS: To target this axis, we developed single domain, non-immunoglobulin high-affinity bi-specific protein inhibitors against both Tie2 and αvß3 integrin. We have previously engineered the Ang2-binding domain of Tie2 (Ang2-BD) as a Tie2 inhibitor. Here, we engineered an exposed loop in Ang2-BD to generate variants that include an integrin-binding Arg-Gly-Asp (RGD) motif and used flow cytometry screening of a yeast-displayed Ang2-BD RGD loop library to identify the integrin antagonists. The bi-specific antagonists targeting both Tie2 and αvß3 integrin inhibited adhesion and proliferation of endothelial cells cultured together with the αvß3 integrin ligand vitronectin, as well as endothelial cell invasion and tube formation. The bi-specific reagents inhibited downstream signaling by Tie2 intracellularly in response to its agonist Ang1 more effectively than the wild-type Ang2 BD that binds Tie2 alone. CONCLUSIONS: Collectively, this study-the first to describe inhibitors targeting all the known functions resulting from Tie2/integrin αvß3 cross-talk-has created new tools for studying Tie2- and integrin αvß3-dependent molecular pathways and provides the basis for the rational and combinatorial engineering of ligand-Tie2 and ligand-integrin αvß3 receptor interactions. Given the roles of these pathways in cancer angiogenesis and metastasis, this proof of principle study paves the route to create novel Tie2/integrin αvß3-targeting proteins for clinical use as imaging and therapeutic agents.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neovascularización Fisiológica/genética , Receptor TIE-2/antagonistas & inhibidores , Receptores de Vitronectina/genética , Ribonucleasa Pancreática/antagonistas & inhibidores , Inhibidores de la Angiogénesis/química , Animales , Ratones , Receptor TIE-2/química , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Receptores de Vitronectina/química , Receptores de Vitronectina/metabolismo , Ribonucleasa Pancreática/química , Ribonucleasa Pancreática/genética , Ribonucleasa Pancreática/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA