Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Biochem ; 87: 131-157, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29494241

RESUMEN

Directed evolution is a powerful technique for generating tailor-made enzymes for a wide range of biocatalytic applications. Following the principles of natural evolution, iterative cycles of mutagenesis and screening or selection are applied to modify protein properties, enhance catalytic activities, or develop completely new protein catalysts for non-natural chemical transformations. This review briefly surveys the experimental methods used to generate genetic diversity and screen or select for improved enzyme variants. Emphasis is placed on a key challenge, namely how to generate novel catalytic activities that expand the scope of natural reactions. Two particularly effective strategies, exploiting catalytic promiscuity and rational design, are illustrated by representative examples of successfully evolved enzymes. Opportunities for extending these approaches to more complex biocatalytic systems are also considered.


Asunto(s)
Evolución Molecular Dirigida/métodos , Enzimas/genética , Enzimas/metabolismo , Animales , Biocatálisis , Diseño de Fármacos , Enzimas/química , Variación Genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Redes y Vías Metabólicas/genética , Modelos Moleculares , Ingeniería de Proteínas/métodos , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Selección Genética , Estereoisomerismo , Especificidad por Sustrato
2.
Annu Rev Biochem ; 82: 447-70, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23746259

RESUMEN

Diverse engineering strategies have been developed to create enzymes with novel catalytic activities. Among these, computational approaches hold particular promise. Enzymes have been computationally designed to promote several nonbiological reactions, including a Diels-Alder cycloaddition, proton transfer, multistep retroaldol transformations, and metal-dependent hydrolysis of phosphotriesters. Although their efficiencies (kcat/KM = 0.1-100 M(-1) s(-1)) are typically low compared with those of the best natural enzymes (10(6)-10(8) M(-1) s(-1)), these catalysts are excellent starting points for laboratory evolution. This review surveys recent progress in combining computational and evolutionary approaches to enzyme design, together with insights into enzyme function gained from studies of the engineered catalysts.


Asunto(s)
Biología Computacional/métodos , Evolución Molecular Dirigida/métodos , Ingeniería de Proteínas/métodos , Proteínas/química , Catálisis , Dominio Catalítico , Enzimas/química , Enzimas/metabolismo , Modelos Moleculares
3.
Nature ; 606(7912): 49-58, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35650353

RESUMEN

The ability to design efficient enzymes from scratch would have a profound effect on chemistry, biotechnology and medicine. Rapid progress in protein engineering over the past decade makes us optimistic that this ambition is within reach. The development of artificial enzymes containing metal cofactors and noncanonical organocatalytic groups shows how protein structure can be optimized to harness the reactivity of nonproteinogenic elements. In parallel, computational methods have been used to design protein catalysts for diverse reactions on the basis of fundamental principles of transition state stabilization. Although the activities of designed catalysts have been quite low, extensive laboratory evolution has been used to generate efficient enzymes. Structural analysis of these systems has revealed the high degree of precision that will be needed to design catalysts with greater activity. To this end, emerging protein design methods, including deep learning, hold particular promise for improving model accuracy. Here we take stock of key developments in the field and highlight new opportunities for innovation that should allow us to transition beyond the current state of the art and enable the robust design of biocatalysts to address societal needs.


Asunto(s)
Biocatálisis , Biotecnología , Ingeniería de Proteínas , Proteínas , Biotecnología/métodos , Biotecnología/tendencias , Ingeniería de Proteínas/métodos , Ingeniería de Proteínas/tendencias , Proteínas/química , Proteínas/metabolismo
4.
Nat Chem Biol ; 20(6): 761-769, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38308044

RESUMEN

Engineered biosynthetic assembly lines could revolutionize the sustainable production of bioactive natural product analogs. Although yeast display is a proven, powerful tool for altering the substrate specificity of gatekeeper adenylation domains in nonribosomal peptide synthetases (NRPSs), comparable strategies for other components of these megaenzymes have not been described. Here we report a high-throughput approach for engineering condensation (C) domains responsible for peptide elongation. We show that a 120-kDa NRPS module, displayed in functional form on yeast, can productively interact with an upstream module, provided in solution, to produce amide products tethered to the yeast surface. Using this system to screen a large C-domain library, we reprogrammed a surfactin synthetase module to accept a fatty acid donor, increasing catalytic efficiency for this noncanonical substrate >40-fold. Because C domains can function as selectivity filters in NRPSs, this methodology should facilitate the precision engineering of these molecular assembly lines.


Asunto(s)
Péptido Sintasas , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Péptido Sintasas/química , Especificidad por Sustrato , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Ingeniería de Proteínas/métodos , Ensayos Analíticos de Alto Rendimiento , Dominios Proteicos
5.
J Am Chem Soc ; 146(3): 1957-1966, 2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38264790

RESUMEN

Nitrene transfer reactions catalyzed by heme proteins have broad potential for the stereoselective formation of carbon-nitrogen bonds. However, competition between productive nitrene transfer and the undesirable reduction of nitrene precursors limits the broad implementation of such biocatalytic methods. Here, we investigated the reduction of azides by the model heme protein myoglobin to gain mechanistic insights into the factors that control the fate of key reaction intermediates. In this system, the reaction proceeds via a proposed nitrene intermediate that is rapidly reduced and protonated to give a reactive ferrous amide species, which we characterized by UV/vis and Mössbauer spectroscopies, quantum mechanical calculations, and X-ray crystallography. Rate-limiting protonation of the ferrous amide to produce the corresponding amine is the final step in the catalytic cycle. These findings contribute to our understanding of the heme protein-catalyzed reduction of azides and provide a guide for future enzyme engineering campaigns to create more efficient nitrene transferases. Moreover, harnessing the reduction reaction in a chemoenzymatic cascade provided a potentially practical route to substituted pyrroles.

6.
Chem Rev ; 122(9): 9145-9197, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35394752

RESUMEN

Proteins that self-assemble into polyhedral shell-like structures are useful molecular containers both in nature and in the laboratory. Here we review efforts to repurpose diverse protein cages, including viral capsids, ferritins, bacterial microcompartments, and designed capsules, as vaccines, drug delivery vehicles, targeted imaging agents, nanoreactors, templates for controlled materials synthesis, building blocks for higher-order architectures, and more. A deep understanding of the principles underlying the construction, function, and evolution of natural systems has been key to tailoring selective cargo encapsulation and interactions with both biological systems and synthetic materials through protein engineering and directed evolution. The ability to adapt and design increasingly sophisticated capsid structures and functions stands to benefit the fields of catalysis, materials science, and medicine.


Asunto(s)
Cápside , Ciencia de los Materiales , Cápside/química , Proteínas de la Cápside/química , Catálisis , Ingeniería de Proteínas
7.
Nat Prod Rep ; 40(9): 1479-1497, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37231979

RESUMEN

Covering: 1878 to early 2023Cyanophycin is a biopolymer consisting of a poly-aspartate backbone with arginines linked to each Asp sidechain through isopeptide bonds. Cyanophycin is made by cyanophycin synthetase 1 or 2 through ATP-dependent polymerization of Asp and Arg, or ß-Asp-Arg, respectively. It is degraded into dipeptides by exo-cyanophycinases, and these dipeptides are hydrolyzed into free amino acids by general or dedicated isodipeptidase enzymes. When synthesized, chains of cyanophycin coalesce into large, inert, membrane-less granules. Although discovered in cyanobacteria, cyanophycin is made by species throughout the bacterial kingdom, and cyanophycin metabolism provides advantages for toxic bloom forming algae and some human pathogens. Some bacteria have developed dedicated schemes for cyanophycin accumulation and use, which include fine temporal and spatial regulation. Cyanophycin has also been heterologously produced in a variety of host organisms to a remarkable level, over 50% of the host's dry mass, and has potential for a variety of green industrial applications. In this review, we summarize the progression of cyanophycin research, with an emphasis on recent structural studies of enzymes in the cyanophycin biosynthetic pathway. These include several unexpected revelations that show cyanophycin synthetase to be a very cool, multi-functional macromolecular machine.


Asunto(s)
Proteínas Bacterianas , Cianobacterias , Humanos , Proteínas Bacterianas/metabolismo , Cianobacterias/metabolismo , Péptido Sintasas/metabolismo , Dipéptidos/química
8.
Nat Chem Biol ; 17(10): 1101-1110, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34385683

RESUMEN

Cyanophycin is a natural biopolymer produced by a wide range of bacteria, consisting of a chain of poly-L-Asp residues with L-Arg residues attached to the ß-carboxylate sidechains by isopeptide bonds. Cyanophycin is synthesized from ATP, aspartic acid and arginine by a homooligomeric enzyme called cyanophycin synthetase (CphA1). CphA1 has domains that are homologous to glutathione synthetases and muramyl ligases, but no other structural information has been available. Here, we present cryo-electron microscopy and X-ray crystallography structures of cyanophycin synthetases from three different bacteria, including cocomplex structures of CphA1 with ATP and cyanophycin polymer analogs at 2.6 Å resolution. These structures reveal two distinct tetrameric architectures, show the configuration of active sites and polymer-binding regions, indicate dynamic conformational changes and afford insight into catalytic mechanism. Accompanying biochemical interrogation of substrate binding sites, catalytic centers and oligomerization interfaces combine with the structures to provide a holistic understanding of cyanophycin biosynthesis.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Péptido Sintasas/química , Péptido Sintasas/metabolismo , Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Modelos Moleculares , Péptido Sintasas/genética , Conformación Proteica
9.
Proc Natl Acad Sci U S A ; 117(48): 30362-30369, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33203677

RESUMEN

De novo protein design has succeeded in generating a large variety of globular proteins, but the construction of protein scaffolds with cavities that could accommodate large signaling molecules, cofactors, and substrates remains an outstanding challenge. The long, often flexible loops that form such cavities in many natural proteins are difficult to precisely program and thus challenging for computational protein design. Here we describe an alternative approach to this problem. We fused two stable proteins with C2 symmetry-a de novo designed dimeric ferredoxin fold and a de novo designed TIM barrel-such that their symmetry axes are aligned to create scaffolds with large cavities that can serve as binding pockets or enzymatic reaction chambers. The crystal structures of two such designs confirm the presence of a 420 cubic Ångström chamber defined by the top of the designed TIM barrel and the bottom of the ferredoxin dimer. We functionalized the scaffold by installing a metal-binding site consisting of four glutamate residues close to the symmetry axis. The protein binds lanthanide ions with very high affinity as demonstrated by tryptophan-enhanced terbium luminescence. This approach can be extended to other metals and cofactors, making this scaffold a modular platform for the design of binding proteins and biocatalysts.


Asunto(s)
Elementos de la Serie de los Lantanoides/química , Elementos de la Serie de los Lantanoides/metabolismo , Metaloproteínas/química , Metaloproteínas/metabolismo , Ingeniería de Proteínas , Sitios de Unión , Modelos Moleculares , Conformación Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad
10.
Angew Chem Int Ed Engl ; 62(49): e202315565, 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-37933996

RESUMEN

Albert Eschenmoser, one of the greatest organic chemists of the past hundred years, died on July 14, 2023 at the age of 97. The extraordinary breadth of his scientific contributions ranged from synthetic methodology, structure elucidation, and synthesis of natural products to the chemical etiology of biomolecular structures.

11.
J Am Chem Soc ; 144(38): 17567-17575, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36070491

RESUMEN

High-throughput engineering has the potential to revolutionize the customization of biosynthetic assembly lines for the sustainable production of pharmaceutically relevant natural product analogs. Here, we show that the substrate specificity of gatekeeper adenylation domains of nonribosomal peptide synthetases can be switched from an α-amino acid to an α-hydroxy acid in a single round of combinatorial mutagenesis and selection using yeast cell surface display. In addition to shedding light on how such proteins discriminate between amino and hydroxy groups, the remodeled domains function in a pathway context to produce α-hydroxy acid-containing linear peptides and cyclic depsipeptides with high efficiency. Site-specific replacement of backbone amines with oxygens by an engineered synthetase provides the means to probe and tune the activities of diverse peptide metabolites in a simple and predictable fashion.


Asunto(s)
Productos Biológicos , Depsipéptidos , Aminas , Aminoácidos/metabolismo , Hidroxiácidos , Péptido Sintasas/metabolismo , Especificidad por Sustrato
12.
Chembiochem ; 23(20): e202200332, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-35951442

RESUMEN

Although viruses have been successfully repurposed as vaccines, antibiotics, and anticancer therapeutics, they also raise concerns regarding genome integration and immunogenicity. Virus-like particles and non-viral protein cages represent a potentially safer alternative but often lack desired functionality. Here, we investigated the utility of a new enzymatic bioconjugation method, called lysine acylation using conjugating enzymes (LACE), to chemoenzymatically modify protein cages. We equipped two structurally distinct protein capsules with a LACE-reactive peptide tag and demonstrated their modification with diverse ligands. This modular approach combines the advantages of chemical conjugation and genetic fusion and allows for site-specific modification with recombinant proteins as well as synthetic peptides with facile control of the extent of labeling. This strategy has the potential to fine-tune protein containers of different shape and size by providing them with new properties that go beyond their biologically native functions.


Asunto(s)
Lisina , Péptidos , Lisina/metabolismo , Péptidos/metabolismo , Proteínas Recombinantes/genética , Acilación , Antibacterianos
13.
J Am Chem Soc ; 143(7): 2736-2740, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33570948

RESUMEN

Nonribosomal peptides (NRPs) are a therapeutically important class of secondary metabolites that are produced by modular synthetases in assembly-line fashion. We previously showed that a single Trp-to-Ser mutation in the initial Phe-loading adenylation domain of tyrocidine synthetase completely switches the specificity toward clickable analogues. Here we report that this minimally invasive strategy enables efficient functionalization of the bioactive NRP on the pathway level. In a reconstituted tyrocidine synthetase, the W227S point mutation permitted selective incorporation of Phe analogues with alkyne, halogen, and benzoyl substituents by the initiation module. The respective W2742S mutation in module 4 similarly permits efficient incorporation of these functionalized substrate analogues at position 4, expanding this strategy to elongation modules. Efficient incorporation of an alkyne handle at position 1 or 4 of tyrocidine A allowed site-selective one-step fluorescent labeling of the corresponding tyrocidine analogues by Cu(I)-catalyzed alkyne-azide cycloaddition. By combining synthetic biology with bioorthogonal chemistry, this approach holds great potential for NRP isolation and molecular target elucidation as well as combinatorial optimization of NRP therapeutics.


Asunto(s)
Péptidos/metabolismo , Alquinos/química , Azidas/química , Catálisis , Cobre/química , Reacción de Cicloadición , Colorantes Fluorescentes/química , Mutagénesis Sitio-Dirigida , Biosíntesis de Péptidos Independientes de Ácidos Nucleicos , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Péptidos/química , Tirocidina/análogos & derivados , Tirocidina/síntesis química
14.
Chembiochem ; 22(1): 92-99, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-32810341

RESUMEN

Nature employs a limited number of genetically encoded amino acids for the construction of functional proteins. By engineering components of the cellular translation machinery, however, it is now possible to genetically encode noncanonical building blocks with tailored electronic and structural properties. The ability to incorporate unique chemical functionality into proteins provides a powerful tool to probe mechanism and create novel function. In this minireview, we highlight several recent studies that illustrate how noncanonical amino acids have been used to capture and characterize reactive intermediates, fine-tune the catalytic properties of enzymes, and stabilize short-lived protein-protein complexes.


Asunto(s)
Proteínas/genética , Código Genético , Ingeniería de Proteínas , Proteínas/química
15.
Proc Natl Acad Sci U S A ; 115(21): 5432-5437, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29735682

RESUMEN

Viruses are remarkable nanomachines that efficiently hijack cellular functions to replicate and self-assemble their components within a complex biological environment. As all steps of the viral life cycle depend on formation of a protective proteinaceous shell that packages the DNA or RNA genome, bottom-up construction of virus-like nucleocapsids from nonviral materials could provide valuable insights into virion assembly and evolution. Such constructs could also serve as safe alternatives to natural viruses for diverse nano- and biotechnological applications. Here we show that artificial virus-like nucleocapsids can be generated-rapidly and surprisingly easily-by engineering and laboratory evolution of a nonviral protein cage formed by Aquifex aeolicus lumazine synthase (AaLS) and its encoding mRNA. Cationic peptides were appended to the engineered capsid proteins to enable specific recognition of packaging signals on cognate mRNAs, and subsequent evolutionary optimization afforded nucleocapsids with expanded spherical structures that encapsulate their own full-length RNA genome in vivo and protect the cargo molecules from nucleases. These findings provide strong experimental support for the hypothesis that subcellular protein-bounded compartments may have facilitated the emergence of ancient viruses.


Asunto(s)
Bacterias/enzimología , Bioingeniería , Proteínas de la Cápside/metabolismo , Evolución Molecular Dirigida , Complejos Multienzimáticos/metabolismo , Nucleocápside/metabolismo , Fragmentos de Péptidos/metabolismo , Bacterias/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Modelos Moleculares , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Nucleocápside/química , Nucleocápside/genética , Ensamble de Virus
16.
Chimia (Aarau) ; 75(4): 323-328, 2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33902803

RESUMEN

Well-defined containers constructed from multiple protein subunits are a unique class of nanomaterial useful in supramolecular chemistry and biology. These protein cages are widespread in nature, where they are responsible for a diversity of important tasks. As such, producing our own designer protein cages, complete with bespoke functionalities, is a promising avenue to new nanodevices, biotechnology and therapies. Herein, we describe how an artificial, computationally designed protein cage can be rationally engineered using supramolecular intuition to produce new functional capsules. Positive supercharging the interior cavity of this porous protein cage enables the efficient encapsulation of oligonucleotides by electrostatically-driven self-assembly. Moreover, the resulting cargo-loaded cages enter mammalian cells and release their cargo, for example siRNA which modulates gene expression. To expand the cargo scope of this proteinaceous container, a higher level of supramolecular complexity can also be introduced. Encapsulation of anionic surfactants affords protein-scaffolded micelles, which are capable of sequestering poorly water-soluble small molecules within their hydrophobic cores. These hybrid particles stably carry bioactive cargo and deliver it intracellularly, thereby increasing potency. Further development of these genetically-encoded materials is ongoing towards specific applications ranging from cell biology to medicine.


Asunto(s)
Sistemas de Liberación de Medicamentos , Proteínas , Cápsulas , ARN Interferente Pequeño
17.
Angew Chem Int Ed Engl ; 60(27): 15063-15068, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-33880851

RESUMEN

Changing the primary metal coordination sphere is a powerful strategy for tuning metalloprotein properties. Here we used amber stop codon suppression with engineered pyrrolysyl-tRNA synthetases, including two newly evolved enzymes, to replace the proximal histidine in myoglobin with Nδ -methylhistidine, 5-thiazoylalanine, 4-thiazoylalanine and 3-(3-thienyl)alanine. In addition to tuning the heme redox potential over a >200 mV range, these noncanonical ligands modulate the protein's carbene transfer activity with ethyl diazoacetate. Variants with increased reduction potential proved superior for cyclopropanation and N-H insertion, whereas variants with reduced Eo values gave higher S-H insertion activity. Given the functional importance of histidine in many enzymes, these genetically encoded analogues could be valuable tools for probing mechanism and enabling new chemistries.


Asunto(s)
Hemo/metabolismo , Metaloproteínas/metabolismo , Metano/análogos & derivados , Hemo/química , Ligandos , Metaloproteínas/química , Metano/química , Metano/metabolismo
18.
J Am Chem Soc ; 142(23): 10250-10254, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32427470

RESUMEN

Controlling regio- and stereoselectivity of aldol additions is generally challenging. Here we show that an artificial aldolase with high specificity for acetone as the aldol donor can be reengineered via single active site mutations to accept linear and cyclic aliphatic ketones with notable efficiency, regioselectivity, and stereocontrol. Biochemical and crystallographic data show how the mutated residues modulate the binding and activation of specific aldol donors, as well as their subsequent reaction with diverse aldehyde acceptors. Broadening the substrate scope of this evolutionarily naïve catalyst proved much easier than previous attempts to redesign natural aldolases, suggesting that such proteins may be excellent starting points for the development of customized biocatalysts for diverse practical applications.

19.
Chembiochem ; 21(1-2): 74-79, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31523885

RESUMEN

The enzyme lumazine synthase (LS) has been engineered to self-assemble into hollow-shell structures that encapsulate unnatural cargo proteins through complementary electrostatic interactions. Herein, we show that a negatively supercharged LS variant can also form organic-inorganic hybrids with gold nanomaterials. Simple mixing of LS pentamers with positively charged gold nanocrystals in aqueous buffer spontaneously affords protein-shelled gold cores. The procedure works well with differently sized and shaped gold nanocrystals, and the resulting shelled complexes exhibit dramatically enhanced colloidal stability over a wide range of pH (4.0-10.0) and at high ionic strength (up to 1 m NaCl). They are even stable over days upon dilution in buffer. Self-assembly of engineered LS shells in this way offers an easy and attractive alternative to commonly used ligand-exchange methods for stabilizing inorganic nanomaterials.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Complejos Multienzimáticos/biosíntesis , Coloides/química , Coloides/metabolismo , Oro/metabolismo , Concentración de Iones de Hidrógeno , Complejos Multienzimáticos/química , Concentración Osmolar , Tamaño de la Partícula , Porosidad , Ingeniería de Proteínas , Propiedades de Superficie
20.
J Am Chem Soc ; 141(24): 9432-9443, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31117660

RESUMEN

The structural and functional diversity of proteins combined with their genetic programmability has made them indispensable modern materials. Well-defined, hollow protein capsules have proven to be particularly useful due to their ability to compartmentalize macromolecules and chemical processes. To this end, viral capsids are common scaffolds and have been successfully repurposed to produce a suite of practical protein-based nanotechnologies. Recently, the recapitulation of viromimetic function in protein cages of nonviral origin has emerged as a strategy to both complement physical studies of natural viruses and produce useful scaffolds for diverse applications. In this perspective, we review recent progress toward generation of virus-like behavior in nonviral protein cages through rational engineering and directed evolution. These artificial systems can aid our understanding of the emergence of viruses from existing cellular components, as well as provide alternative approaches to tackle current problems, and open up new opportunities, in medicine and biotechnology.


Asunto(s)
Proteínas Bacterianas/metabolismo , Materiales Biomiméticos/metabolismo , Virus/química , Secuencia de Aminoácidos , Bacterias/química , Proteínas Bacterianas/química , Materiales Biomiméticos/química , Evolución Molecular Dirigida , Ingeniería de Proteínas , ARN/química , ARN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA