Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nature ; 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898281

RESUMEN

De novo design of complex protein folds using solely computational means remains a substantial challenge1. Here we use a robust deep learning pipeline to design complex folds and soluble analogues of integral membrane proteins. Unique membrane topologies, such as those from G-protein-coupled receptors2, are not found in the soluble proteome, and we demonstrate that their structural features can be recapitulated in solution. Biophysical analyses demonstrate the high thermal stability of the designs, and experimental structures show remarkable design accuracy. The soluble analogues were functionalized with native structural motifs, as a proof of concept for bringing membrane protein functions to the soluble proteome, potentially enabling new approaches in drug discovery. In summary, we have designed complex protein topologies and enriched them with functionalities from membrane proteins, with high experimental success rates, leading to a de facto expansion of the functional soluble fold space.

2.
Nature ; 617(7959): 176-184, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37100904

RESUMEN

Physical interactions between proteins are essential for most biological processes governing life1. However, the molecular determinants of such interactions have been challenging to understand, even as genomic, proteomic and structural data increase. This knowledge gap has been a major obstacle for the comprehensive understanding of cellular protein-protein interaction networks and for the de novo design of protein binders that are crucial for synthetic biology and translational applications2-9. Here we use a geometric deep-learning framework operating on protein surfaces that generates fingerprints to describe geometric and chemical features that are critical to drive protein-protein interactions10. We hypothesized that these fingerprints capture the key aspects of molecular recognition that represent a new paradigm in the computational design of novel protein interactions. As a proof of principle, we computationally designed several de novo protein binders to engage four protein targets: SARS-CoV-2 spike, PD-1, PD-L1 and CTLA-4. Several designs were experimentally optimized, whereas others were generated purely in silico, reaching nanomolar affinity with structural and mutational characterization showing highly accurate predictions. Overall, our surface-centric approach captures the physical and chemical determinants of molecular recognition, enabling an approach for the de novo design of protein interactions and, more broadly, of artificial proteins with function.


Asunto(s)
Simulación por Computador , Aprendizaje Profundo , Unión Proteica , Proteínas , Humanos , Proteínas/química , Proteínas/metabolismo , Proteómica , Mapas de Interacción de Proteínas , Sitios de Unión , Biología Sintética
3.
Proc Natl Acad Sci U S A ; 119(43): e2206111119, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36252041

RESUMEN

De novo protein design enables the exploration of novel sequences and structures absent from the natural protein universe. De novo design also stands as a stringent test for our understanding of the underlying physical principles of protein folding and may lead to the development of proteins with unmatched functional characteristics. The first fundamental challenge of de novo design is to devise "designable" structural templates leading to sequences that will adopt the predicted fold. Here, we built on the TopoBuilder (TB) de novo design method, to automatically assemble structural templates with native-like features starting from string descriptors that capture the overall topology of proteins. Our framework eliminates the dependency of hand-crafted and fold-specific rules through an iterative, data-driven approach that extracts geometrical parameters from structural tertiary motifs. We evaluated the TopoBuilder framework by designing sequences for a set of five protein folds and experimental characterization revealed that several sequences were folded and stable in solution. The TopoBuilder de novo design framework will be broadly useful to guide the generation of artificial proteins with customized geometries, enabling the exploration of the protein universe.


Asunto(s)
Pliegue de Proteína , Proteínas , Modelos Moleculares , Ingeniería de Proteínas/métodos , Proteínas/química
4.
Nat Chem Biol ; 17(4): 492-500, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33398169

RESUMEN

De novo protein design has enabled the creation of new protein structures. However, the design of functional proteins has proved challenging, in part due to the difficulty of transplanting structurally complex functional sites to available protein structures. Here, we used a bottom-up approach to build de novo proteins tailored to accommodate structurally complex functional motifs. We applied the bottom-up strategy to successfully design five folds for four distinct binding motifs, including a bifunctionalized protein with two motifs. Crystal structures confirmed the atomic-level accuracy of the computational designs. These de novo proteins were functional as components of biosensors to monitor antibody responses and as orthogonal ligands to modulate synthetic signaling receptors in engineered mammalian cells. Our work demonstrates the potential of bottom-up approaches to accommodate complex structural motifs, which will be essential to endow de novo proteins with elaborate biochemical functions, such as molecular recognition or catalysis.


Asunto(s)
Ingeniería de Proteínas/métodos , Secuencias de Aminoácidos/genética , Sitios de Unión/genética , Catálisis , Ligandos , Modelos Moleculares , Unión Proteica/genética , Pliegue de Proteína , Proteínas/química
5.
PLoS Comput Biol ; 18(3): e1009178, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35294435

RESUMEN

Proteins are typically represented by discrete atomic coordinates providing an accessible framework to describe different conformations. However, in some fields proteins are more accurately represented as near-continuous surfaces, as these are imprinted with geometric (shape) and chemical (electrostatics) features of the underlying protein structure. Protein surfaces are dependent on their chemical composition and, ultimately determine protein function, acting as the interface that engages in interactions with other molecules. In the past, such representations were utilized to compare protein structures on global and local scales and have shed light on functional properties of proteins. Here we describe RosettaSurf, a surface-centric computational design protocol, that focuses on the molecular surface shape and electrostatic properties as means for protein engineering, offering a unique approach for the design of proteins and their functions. The RosettaSurf protocol combines the explicit optimization of molecular surface features with a global scoring function during the sequence design process, diverging from the typical design approaches that rely solely on an energy scoring function. With this computational approach, we attempt to address a fundamental problem in protein design related to the design of functional sites in proteins, even when structurally similar templates are absent in the characterized structural repertoire. Surface-centric design exploits the premise that molecular surfaces are, to a certain extent, independent of the underlying sequence and backbone configuration, meaning that different sequences in different proteins may present similar surfaces. We benchmarked RosettaSurf on various sequence recovery datasets and showcased its design capabilities by generating epitope mimics that were biochemically validated. Overall, our results indicate that the explicit optimization of surface features may lead to new routes for the design of functional proteins.


Asunto(s)
Ingeniería de Proteínas , Proteínas , Algoritmos , Biología Computacional/métodos , Conformación Proteica , Ingeniería de Proteínas/métodos , Proteínas/química , Electricidad Estática
6.
Nat Chem Biol ; 16(7): 725-730, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32284602

RESUMEN

Anti-CRISPR (Acr) proteins are powerful tools to control CRISPR-Cas technologies. However, the available Acr repertoire is limited to naturally occurring variants. Here, we applied structure-based design on AcrIIC1, a broad-spectrum CRISPR-Cas9 inhibitor, to improve its efficacy on different targets. We first show that inserting exogenous protein domains into a selected AcrIIC1 surface site dramatically enhances inhibition of Neisseria meningitidis (Nme)Cas9. Then, applying structure-guided design to the Cas9-binding surface, we converted AcrIIC1 into AcrIIC1X, a potent inhibitor of the Staphylococcus aureus (Sau)Cas9, an orthologue widely applied for in vivo genome editing. Finally, to demonstrate the utility of AcrIIC1X for genome engineering applications, we implemented a hepatocyte-specific SauCas9 ON-switch by placing AcrIIC1X expression under regulation of microRNA-122. Our work introduces designer Acrs as important biotechnological tools and provides an innovative strategy to safeguard CRISPR technologies.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Edición Génica/métodos , MicroARNs/genética , Ingeniería de Proteínas/métodos , Secuencia de Aminoácidos , Proteína 9 Asociada a CRISPR/metabolismo , Línea Celular Tumoral , Genoma Humano , Células HEK293 , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , MicroARNs/metabolismo , Modelos Moleculares , Mutagénesis Insercional , Neisseria meningitidis/enzimología , Neisseria meningitidis/genética , Plásmidos/química , Plásmidos/metabolismo , Dominios Proteicos , Estructura Secundaria de Proteína , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Staphylococcus aureus/enzimología , Staphylococcus aureus/genética
7.
PLoS Biol ; 17(2): e3000164, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30789898

RESUMEN

Throughout the last several decades, vaccination has been key to prevent and eradicate infectious diseases. However, many pathogens (e.g., respiratory syncytial virus [RSV], influenza, dengue, and others) have resisted vaccine development efforts, largely because of the failure to induce potent antibody responses targeting conserved epitopes. Deep profiling of human B cells often reveals potent neutralizing antibodies that emerge from natural infection, but these specificities are generally subdominant (i.e., are present in low titers). A major challenge for next-generation vaccines is to overcome established immunodominance hierarchies and focus antibody responses on crucial neutralization epitopes. Here, we show that a computationally designed epitope-focused immunogen presenting a single RSV neutralization epitope elicits superior epitope-specific responses compared to the viral fusion protein. In addition, the epitope-focused immunogen efficiently boosts antibodies targeting the palivizumab epitope, resulting in enhanced neutralization. Overall, we show that epitope-focused immunogens can boost subdominant neutralizing antibody responses in vivo and reshape established antibody hierarchies.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Epítopos/química , Receptores de Antígenos de Linfocitos B/inmunología , Proteínas Recombinantes de Fusión/química , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/química , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/genética , Clonación Molecular , Diseño Asistido por Computadora , Epítopos/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Inmunización/métodos , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Nanopartículas/química , Palivizumab/química , Palivizumab/inmunología , Receptores de Antígenos de Linfocitos B/química , Receptores de Antígenos de Linfocitos B/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/biosíntesis , Vacunas contra Virus Sincitial Respiratorio/genética , Homología Estructural de Proteína , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
8.
Chemistry ; 27(47): 12215-12223, 2021 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-34060672

RESUMEN

While the integration of supramolecular principles in catalysis attracts increasing attention, a direct comparative assessment of the resulting systems catalysts to work out distinct characteristics is often difficult. Herein is reported how the broad responsiveness of ether cyclizations to diverse inputs promises to fill this gap. Cyclizations in the confined, π-basic and Brønsted acidic interior of supramolecular capsules, for instance, are found to excel with speed (exceeding general Brønsted acid and hydrogen-bonding catalysts by far) and selective violations of the Baldwin rules (as extreme as the so far unique pnictogen-bonding catalysts). The complementary cyclization on π-acidic aromatic surfaces remains unique with regard to autocatalysis, which is shown to be chemo- and diastereoselective with regard to product-like co-catalysts but, so far, not enantioselective.


Asunto(s)
Éter , Catálisis , Ciclización , Enlace de Hidrógeno
9.
J Chem Phys ; 154(7): 074114, 2021 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-33607903

RESUMEN

Computational protein design has emerged as a powerful tool capable of identifying sequences compatible with pre-defined protein structures. The sequence design protocols, implemented in the Rosetta suite, have become widely used in the protein engineering community. To understand the strengths and limitations of the Rosetta design framework, we tested several design protocols on two distinct folds (SH3-1 and Ubiquitin). The sequence optimization, when started from native structures and natural sequences or polyvaline sequences, converges to sequences that are not recognized as belonging to the fold family of the target protein by standard bioinformatic tools, such as BLAST and Hmmer. The sequences generated from both starting conditions (native and polyvaline) are instead very similar to each other and recognized by Hmmer as belonging to the same "family." This demonstrates the capability of Rosetta to converge to similar sequences, even when sampling from distinct starting conditions, but, on the other hand, shows intrinsic inaccuracy of the scoring function that drifts toward sequences that lack identifiable natural sequence signatures. To address this problem, we developed a protocol embedding Rosetta Design simulations in a genetic algorithm, in which the sequence search is biased to converge to sequences that exist in nature. This protocol allows us to obtain sequences that have recognizable natural sequence signatures and, experimentally, the designed proteins are biochemically well behaved and thermodynamically stable.


Asunto(s)
Diseño de Fármacos , Proteínas/química , Secuencia de Aminoácidos , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Termodinámica
10.
PLoS Comput Biol ; 14(11): e1006623, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30452434

RESUMEN

The robust computational design of functional proteins has the potential to deeply impact translational research and broaden our understanding of the determinants of protein function and stability. The low success rates of computational design protocols and the extensive in vitro optimization often required, highlight the challenge of designing proteins that perform essential biochemical functions, such as binding or catalysis. One of the most simplistic approaches for the design of function is to adopt functional motifs in naturally occurring proteins and transplant them to computationally designed proteins. The structural complexity of the functional motif largely determines how readily one can find host protein structures that are "designable", meaning that are likely to present the functional motif in the desired conformation. One promising route to enhance the "designability" of protein structures is to allow backbone flexibility. Here, we present a computational approach that couples conformational folding with sequence design to embed functional motifs into heterologous proteins-Rosetta Functional Folding and Design (FunFolDes). We performed extensive computational benchmarks, where we observed that the enforcement of functional requirements resulted in designs distant from the global energetic minimum of the protein. An observation consistent with several experimental studies that have revealed function-stability tradeoffs. To test the design capabilities of FunFolDes we transplanted two viral epitopes into distant structural templates including one de novo "functionless" fold, which represent two typical challenges where the designability problem arises. The designed proteins were experimentally characterized showing high binding affinities to monoclonal antibodies, making them valuable candidates for vaccine design endeavors. Overall, we present an accessible strategy to repurpose old protein folds for new functions. This may lead to important improvements on the computational design of proteins, with structurally complex functional sites, that can perform elaborate biochemical functions related to binding and catalysis.


Asunto(s)
Biología Computacional/métodos , Ingeniería de Proteínas/métodos , Proteínas/química , Secuencias de Aminoácidos , Anticuerpos Monoclonales/química , Catálisis , Epítopos/química , Humanos , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Programas Informáticos
11.
bioRxiv ; 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38496615

RESUMEN

De novo design of complex protein folds using solely computational means remains a significant challenge. Here, we use a robust deep learning pipeline to design complex folds and soluble analogues of integral membrane proteins. Unique membrane topologies, such as those from GPCRs, are not found in the soluble proteome and we demonstrate that their structural features can be recapitulated in solution. Biophysical analyses reveal high thermal stability of the designs and experimental structures show remarkable design accuracy. The soluble analogues were functionalized with native structural motifs, standing as a proof-of-concept for bringing membrane protein functions to the soluble proteome, potentially enabling new approaches in drug discovery. In summary, we designed complex protein topologies and enriched them with functionalities from membrane proteins, with high experimental success rates, leading to a de facto expansion of the functional soluble fold space.

12.
Protein Sci ; 32(6): e4653, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37165539

RESUMEN

De novo protein design enhances our understanding of the principles that govern protein folding and interactions, and has the potential to revolutionize biotechnology through the engineering of novel protein functionalities. Despite recent progress in computational design strategies, de novo design of protein structures remains challenging, given the vast size of the sequence-structure space. AlphaFold2 (AF2), a state-of-the-art neural network architecture, achieved remarkable accuracy in predicting protein structures from amino acid sequences. This raises the question whether AF2 has learned the principles of protein folding sufficiently for de novo design. Here, we sought to answer this question by inverting the AF2 network, using the prediction weight set and a loss function to bias the generated sequences to adopt a target fold. Initial design trials resulted in de novo designs with an overrepresentation of hydrophobic residues on the protein surface compared to their natural protein family, requiring additional surface optimization. In silico validation of the designs showed protein structures with the correct fold, a hydrophilic surface and a densely packed hydrophobic core. In vitro validation showed that 7 out of 39 designs were folded and stable in solution with high melting temperatures. In summary, our design workflow solely based on AF2 does not seem to fully capture basic principles of de novo protein design, as observed in the protein surface's hydrophobic vs. hydrophilic patterning. However, with minimal post-design intervention, these pipelines generated viable sequences as assessed experimental characterization. Thus, such pipelines show the potential to contribute to solving outstanding challenges in de novo protein design.


Asunto(s)
Furilfuramida , Ingeniería de Proteínas , Ingeniería de Proteínas/métodos , Proteínas/química , Secuencia de Aminoácidos , Pliegue de Proteína
13.
ACS Chem Biol ; 18(6): 1259-1265, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37252896

RESUMEN

Protein-based therapeutics, such as monoclonal antibodies and cytokines, are important therapies for various pathophysiological conditions such as oncology, autoimmune disorders, and viral infections. However, the wide application of such protein therapeutics is often hindered by dose-limiting toxicities and adverse effects, namely, cytokine storm syndrome, organ failure, and others. Therefore, spatiotemporal control of the activities of these proteins is crucial to further expand their application. Here, we report the design and application of small-molecule-controlled switchable protein therapeutics by taking advantage of a previously engineered OFF-switch system. We used the Rosetta modeling suite to computationally optimize the affinity between B-cell lymphoma 2 (Bcl-2) protein and a previously developed computationally designed protein partner (LD3) to obtain a fast and efficient heterodimer disruption upon the addition of a competing drug (Venetoclax). The incorporation of the engineered OFF-switch system into anti-CTLA4, anti-HER2 antibodies, or an Fc-fused IL-15 cytokine demonstrated an efficient disruption in vitro, as well as fast clearance in vivo upon the addition of the competing drug Venetoclax. These results provide a proof-of-concept for the rational design of controllable biologics by introducing a drug-induced OFF-switch into existing protein-based therapeutics.


Asunto(s)
Anticuerpos Monoclonales , Sulfonamidas , Anticuerpos Monoclonales/uso terapéutico , Citocinas
14.
Cell Rep ; 42(4): 112389, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37058406

RESUMEN

Enterovirus A71 (EV-A71) causes hand, foot, and mouth disease outbreaks with neurological complications and deaths. We previously isolated an EV-A71 variant in the stool, cerebrospinal fluid, and blood of an immunocompromised patient who had a leucine-to-arginine substitution on the VP1 capsid protein, resulting in increased heparin sulfate binding. We show here that this mutation increases the virus's pathogenicity in orally infected mice with depleted B cells, which mimics the patient's immune status, and increases susceptibility to neutralizing antibodies. However, a double mutant with even greater heparin sulfate affinity is not pathogenic, suggesting that increased heparin sulfate affinity may trap virions in peripheral tissues and reduce neurovirulence. This research sheds light on the increased pathogenicity of variant with heparin sulfate (HS)-binding ability in individuals with decreased B cell immunity.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Humanos , Animales , Ratones , Enterovirus/genética , Enterovirus Humano A/genética , Antígenos Virales/metabolismo , Heparitina Sulfato/metabolismo , Heparina/metabolismo
15.
Protein Sci ; 31(9): e4400, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36040259

RESUMEN

Recent advances in protein-design methodology have led to a dramatic increase in reliability and scale. With these advances, dozens and even thousands of designed proteins are automatically generated and screened. Nevertheless, the success rate, particularly in design of functional proteins, is low and fundamental goals such as reliable de novo design of efficient enzymes remain beyond reach. Experimental analyses have consistently indicated that a major reason for design failure is inaccuracy and misfolding relative to the design conception. To address this challenge, we describe complementary methods to diagnose and ameliorate suboptimal regions in designed proteins: first, we develop a Rosetta atomistic computational mutation scanning approach to detect energetically suboptimal positions in designs (available on a web server https://pSUFER.weizmann.ac.il); second, we demonstrate that AlphaFold2 ab initio structure prediction flags regions that may misfold in designed enzymes and binders; and third, we focus FuncLib design calculations on suboptimal positions in a previously designed low-efficiency enzyme, improving its catalytic efficiency by 330-fold. Furthermore, applied to a de novo designed protein that exhibited limited stability, the same approach markedly improved stability and expressibility. Thus, foldability analysis and enhancement may dramatically increase the success rate in design of functional proteins.


Asunto(s)
Ingeniería de Proteínas , Proteínas , Catálisis , Conformación Proteica , Ingeniería de Proteínas/métodos , Proteínas/química , Reproducibilidad de los Resultados
16.
Nat Commun ; 12(1): 5754, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34599176

RESUMEN

Small-molecule responsive protein switches are crucial components to control synthetic cellular activities. However, the repertoire of small-molecule protein switches is insufficient for many applications, including those in the translational spaces, where properties such as safety, immunogenicity, drug half-life, and drug side-effects are critical. Here, we present a computational protein design strategy to repurpose drug-inhibited protein-protein interactions as OFF- and ON-switches. The designed binders and drug-receptors form chemically-disruptable heterodimers (CDH) which dissociate in the presence of small molecules. To design ON-switches, we converted the CDHs into a multi-domain architecture which we refer to as activation by inhibitor release switches (AIR) that incorporate a rationally designed drug-insensitive receptor protein. CDHs and AIRs showed excellent performance as drug responsive switches to control combinations of synthetic circuits in mammalian cells. This approach effectively expands the chemical space and logic responses in living cells and provides a blueprint to develop new ON- and OFF-switches.


Asunto(s)
Diseño Asistido por Computadora , Receptores de Droga/metabolismo , Biología Sintética/métodos , Células HEK293 , Humanos , Multimerización de Proteína/efectos de los fármacos , Receptores de Droga/agonistas , Receptores de Droga/antagonistas & inhibidores
17.
Science ; 368(6492)2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32409444

RESUMEN

De novo protein design has been successful in expanding the natural protein repertoire. However, most de novo proteins lack biological function, presenting a major methodological challenge. In vaccinology, the induction of precise antibody responses remains a cornerstone for next-generation vaccines. Here, we present a protein design algorithm called TopoBuilder, with which we engineered epitope-focused immunogens displaying complex structural motifs. In both mice and nonhuman primates, cocktails of three de novo-designed immunogens induced robust neutralizing responses against the respiratory syncytial virus. Furthermore, the immunogens refocused preexisting antibody responses toward defined neutralization epitopes. Overall, our design approach opens the possibility of targeting specific epitopes for the development of vaccines and therapeutic antibodies and, more generally, will be applicable to the design of de novo proteins displaying complex functional motifs.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Biología Computacional/métodos , Epítopos Inmunodominantes/química , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/química , Vacunas contra Virus Sincitial Respiratorio/química , Virus Sincitial Respiratorio Humano/inmunología , Secuencias de Aminoácidos , Humanos , Epítopos Inmunodominantes/inmunología , Conformación Proteica , Proteínas Recombinantes de Fusión/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología
18.
Org Lett ; 8(16): 3581-4, 2006 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-16869665

RESUMEN

[reaction: see text] Phosphoramidites based on BINOL readily react with trimethylaluminum in "noncoordinating" solvents, leading to the corresponding aminophosphine which is the real ligand in copper-catalyzed asymmetric transformations. This artifact explains the experimental differences in the asymmetric ring opening of meso bicyclic hydrazines using dialkylzinc or trialkylaluminum reagents as nucleophiles.

20.
Org Lett ; 6(12): 1959-62, 2004 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-15176793

RESUMEN

[structure: see text] Full conversion and enantioselectivities up to 83% have been obtained in the conjugate addition reactions of diethyl zinc to Michael acceptors catalyzed by well-defined (chiral) copper(I) aminoarenethiolates. Interesting differences between organozinc or Grignard reagents have been found: for cyclic enones R(2)Zn reagents afford better results, whereas earlier work showed that RMgX reagents react more selectively with acyclic enones.


Asunto(s)
Compuestos Organometálicos/química , Compuestos Organometálicos/síntesis química , Compuestos de Zinc/síntesis química , Alcoholes/síntesis química , Catálisis , Cetonas/síntesis química , Estructura Molecular , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA