Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nucleic Acids Res ; 52(7): 3938-3949, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38477328

RESUMEN

In the hypothetical RNA world, ribozymes could have acted as modern aminoacyl-tRNA synthetases (ARSs) to charge tRNAs, thus giving rise to the peptide synthesis along with the evolution of a primitive translation apparatus. We previously reported a T-boxzyme, Tx2.1, which selectively charges initiator tRNA with N-biotinyl-phenylalanine (BioPhe) in situ in a Flexible In-vitro Translation (FIT) system to produce BioPhe-initiating peptides. Here, we performed in vitro selection of elongation-capable T-boxzymes (elT-boxzymes), using para-azido-l-phenylalanine (PheAZ) as an acyl-donor. We implemented a new strategy to enrich elT-boxzyme-tRNA conjugates that self-aminoacylated on the 3'-terminus selectively. One of them, elT32, can charge PheAZ onto tRNA in trans in response to its cognate anticodon. Further evolution of elT32 resulted in elT49, with enhanced aminoacylation activity. We have demonstrated the translation of a PheAZ-containing peptide in an elT-boxzyme-integrated FIT system, revealing that elT-boxzymes are able to generate the PheAZ-tRNA in response to the cognate anticodon in situ of a custom-made translation system. This study, together with Tx2.1, illustrates a scenario where a series of ribozymes could have overseen aminoacylation and co-evolved with a primitive RNA-based translation system.


Asunto(s)
Anticodón , Biosíntesis de Proteínas , ARN Catalítico , Aminoacil-ARN de Transferencia , ARN Catalítico/metabolismo , ARN Catalítico/genética , Anticodón/genética , Aminoacil-ARN de Transferencia/metabolismo , Aminoacil-ARN de Transferencia/genética , Fenilalanina/metabolismo , Fenilalanina/análogos & derivados , Aminoacil-ARNt Sintetasas/metabolismo , Aminoacil-ARNt Sintetasas/genética , Aminoacilación de ARN de Transferencia , Aminoacilación , Extensión de la Cadena Peptídica de Translación
2.
Biomacromolecules ; 25(1): 355-365, 2024 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-38051119

RESUMEN

RNA-binding proteins participate in diverse cellular processes, including DNA repair, post-transcriptional modification, and cancer progression through their interactions with RNAs, making them attractive for biotechnological applications. While nature provides an array of naturally occurring RNA-binding proteins, developing de novo RNA-binding peptides remains challenging. In particular, tailoring peptides to target single-stranded RNA with low complexity is difficult due to the inherent structural flexibility of RNA molecules. Here, we developed a codon-restricted mRNA display and identified multiple de novo peptides from a peptide library that bind to poly(C) and poly(A) RNA with KDs ranging from micromolar to submicromolar concentrations. One of the newly identified peptides is capable of binding to the cytosine-rich sequences of the oncogenic Cdk6 3'UTR RNA and MYU lncRNA, with affinity comparable to that of the endogenous binding protein. Hence, we present a novel platform for discovering de novo single-stranded RNA-binding peptides that offer promising avenues for regulating RNA functions.


Asunto(s)
Péptidos , ARN , ARN Mensajero/química , Péptidos/química , Codón , Proteínas de Unión al ARN/genética
3.
Angew Chem Int Ed Engl ; 62(36): e202307157, 2023 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-37450419

RESUMEN

Receptor tyrosine kinases (RTKs) are generally activated through their dimerization and/or oligomerization induced by their cognate ligands, and one such RTK hepatocyte growth factor (HGF) receptor, known as MET, plays an important role in tissue regeneration. Here we show the development of ubiquitin (Ub)-based protein ligand multimers, referred to as U-bodies, which act as surrogate agonists for MET and are derived from MET-binding macrocyclic peptides. Monomeric Ub constructs (U-body) were first generated by genetic implantation of a macrocyclic peptide pharmacophore into a structural loop of Ub (lasso-grafting) and subsequent optimization of its flanking spacer sequences via mRNA display. Such U-body constructs exhibit potent binding affinity to MET, thermal stability, and proteolytic stability. The U-body constructs also partially/fully inhibited or enhanced HGF-induced MET-phosphorylation. Their multimerization to dimeric, tetrameric, and octameric U-bodies linked by an appropriate peptide linker yielded potent MET activation activity and downstream cell proliferation-promoting activity. This work suggests that lasso-grafting of macrocycles to Ub is an effective approach to devising protein-based artificial RTK agonists and it can be useful in the development of a new class of biologics for various therapeutic applications.


Asunto(s)
Factor de Crecimiento de Hepatocito , Ubiquitina , Factor de Crecimiento de Hepatocito/metabolismo , Ubiquitina/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-met/metabolismo , Fosforilación , Péptidos/farmacología , Péptidos/metabolismo
4.
J Am Chem Soc ; 144(44): 20332-20341, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36282922

RESUMEN

Bioengineering of ribosomally synthesized and post-translationally modified peptides (RiPPs) is an emerging approach to explore the diversity of pseudo-natural product structures for drug discovery purposes. However, despite the initial advances in this area, bioactivity reprogramming of multienzyme RiPP biosynthetic pathways remains a major challenge. Here, we report a platform for de novo discovery of functional thiopeptides based on reengineered biosynthesis of lactazole A, a RiPP natural product assembled by five biosynthetic enzymes. The platform combines in vitro biosynthesis of lactazole-like thiopeptides and mRNA display to prepare and screen large (≥1012) combinatorial libraries of pseudo-natural products. We demonstrate the utility of the developed protocols in an affinity selection against Traf2- and NCK-interacting kinase (TNIK), a protein involved in several cancers, which yielded a plethora of candidate thiopeptides. Of the 11 synthesized compounds, 9 had high affinities for the target kinase (best KD = 1.2 nM) and 10 inhibited its enzymatic activity (best Ki = 3 nM). X-ray structural analysis of the TNIK/thiopeptide interaction revealed the unique mode of substrate-competitive inhibition exhibited by two of the discovered compounds. The thiopeptides internalized to the cytosol of HEK293H cells as efficiently as the known cell-penetrating peptide Tat (4-6 µM). Accordingly, the most potent compound, TP15, inhibited TNIK in HCT116 cells. Altogether, our platform enables the exploration of pseudo-natural thiopeptides with favorable pharmacological properties in drug discovery applications.


Asunto(s)
Productos Biológicos , Productos Biológicos/farmacología , Productos Biológicos/metabolismo , Procesamiento Proteico-Postraduccional , Péptidos/química , Vías Biosintéticas , Descubrimiento de Drogas
5.
Nat Chem Biol ; 16(6): 702-709, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32203413

RESUMEN

When the primitive translation system first emerged in the hypothetical RNA world, ribozymes could have been responsible for aminoacylation. Given that naturally occurring T-box riboswitches selectively sense the aminoacylation status of cognate tRNAs, we introduced a domain of random sequence into a T-box-tRNA conjugate and isolated ribozymes that were self-aminoacylating on the 3'-terminal hydroxyl group. One of them, named Tx2.1, recognizes the anticodon and D-loop of tRNA via interaction with its stem I domain, similarly to the parental T-box, and selectively charges N-biotinyl-L-phenylalanine (Bio-lPhe) onto the 3' end of the cognate tRNA in trans. We also demonstrated the ribosomal synthesis of a Bio-lPhe-initiated peptide in a Tx2.1-coupled in vitro translation system, in which Tx2.1 catalyzed specific tRNA aminoacylation in situ. This suggests that such ribozymes could have coevolved with a primitive translation system in the RNA world.


Asunto(s)
ARN Catalítico/genética , ARN Catalítico/metabolismo , Riboswitch/genética , Aminoacilación de ARN de Transferencia/efectos de los fármacos , Bacillus subtilis/enzimología , Secuencia de Bases , Biotina/química , Dominio Catalítico , Biblioteca de Genes , Modelos Genéticos , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Fenilalanina/química , Unión Proteica , Estreptavidina/metabolismo
6.
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
7.
Biochem Biophys Res Commun ; 516(2): 445-450, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31227214

RESUMEN

Transforming growth factor-ß 1 (TGFß1)-stimulated clone 22 (TSC22) family includes proteins containing a leucine zipper domain and a TSC-box that are highly conserved during evolution. Currently, limited data are available on the function of this protein family, especially of TSC-22 homologous gene-1 (THG-1)/TSC22 domain family member 4 (TSC22D4). Similar to other family members, THG-1 functions depending on its interaction with the partner proteins and it is suggested to mediate a broad range of biological processes. THG-1-specific binding molecules will be instrumental for elucidating its functions. Therefore, the Random non-standard Peptide Integrated Discovery (RaPID) system was modified using commercially available materials and used for selecting macrocyclic peptides (MCPs) that bind to THG-1. Several MCPs were identified to bind THG-1. Fluorescein- and biotin-tagged MCPs were synthesized and employed as THG-1 detection probes. Notably, a fluorescein-tagged MCP specifically detected THG-1-expressing cells. Biotin-tagged MCPs can be successfully used for Enzyme-Linked Protein Sorbent Assay (ELISA) like assay of THG-1 protein and affinity-precipitation of purified THG-1 and endogenous THG-1 in esophageal squamous cell carcinoma cell lysates. The modified RaPID system rapidly and successfully identified THG-1-binding MCPs in vitro and the synthesized THG-1 binding MCPs are useful alternatives acting for antibodies.


Asunto(s)
Compuestos Macrocíclicos/metabolismo , Péptidos/metabolismo , Factores de Transcripción/metabolismo , Fluoresceína/metabolismo , Células HEK293 , Humanos
8.
J Am Chem Soc ; 140(2): 566-569, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29278497

RESUMEN

Protein cages have recently emerged as an important platform for nanotechnology development. Of the naturally existing protein cages, viruses are among the most efficient nanomachines, overcoming various barriers to achieve component replication and efficient self-assembly in complex biological milieu. We have designed an artificial system that can carry out the most basic steps of viral particle assembly in vivo. Our strategy is based on patchwork capsids formed from Aquifex aeolicus lumazine synthase and a circularly permuted variant with appended cationic peptides. These two-component protein containers self-assemble in vivo, capturing endogenous RNA molecules in a size-selective manner. By varying the number and design of the RNA-binding peptides displayed on the lumenal surface, the length of guest RNA can be further controlled. Using a fluorescent aptamer, we also show that short-lived RNA species are captured by the protein cage. This platform has potential as a model system for investigating virus assembly, as well as developing RNA regulation or sampling tools to augment biotechnology.


Asunto(s)
Proteínas/química , ARN/química , Modelos Biológicos , Modelos Moleculares , Nanotecnología , Tamaño de la Partícula
9.
RNA ; 22(12): 1918-1928, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27852928

RESUMEN

RNA aptamers are structured motifs that bind to specific molecules. A growing number of RNAs bearing aptamer elements, whose functions are modulated by direct binding of metabolites, have been found in living cells. Recent studies have suggested that more small RNAs binding to metabolites likely exist and may be involved in diverse cellular processes. However, conventional methods are not necessarily suitable for the discovery of such RNA aptamer elements in small RNAs with lengths ranging from 50 to 200 nucleotides, due to the far more abundant tRNAs in this size range. Here, we describe a new in vitro selection method to uncover naturally occurring small RNAs capable of binding to a ligand of interest, referred to as small RNA transcriptomic SELEX (smaRt-SELEX). By means of this method, we identified a motif in human precursor microRNA 125a (hsa-pre-miR-125a) that interacts with folic acid. Mutation studies revealed that the terminal loop region of hsa-pre-miR-125a is important for this binding interaction. This method has potential for the discovery of new RNA aptamer elements or catalytic motifs in biological small RNA fractions.


Asunto(s)
Ácido Fólico/metabolismo , MicroARNs/metabolismo , Transcriptoma , Humanos , Técnica SELEX de Producción de Aptámeros
10.
Methods ; 106: 105-11, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27163863

RESUMEN

Detection of rare small RNA species whose sizes are overlapping with tRNAs often suffers from insufficient sensitivity due to the overwhelming abundance of tRNAs. We here report a method, named tRid (tRNA rid), for removing abundant tRNAs from small RNA fractions regardless of tRNA sequence species. By means of tRid, we are able to selectively enrich small RNAs which have been previously difficult to access due to mass existence of tRNAs in such fractions. A flexible tRNA-acylation ribozyme, known as flexizyme, is a key tool where the total tRNAs are aminoacylated with N-biotinylated phenylalanine regardless of tRNA sequences, and therefore the biotin-tagged tRNAs could be readily removed from the small RNA fractions by the use of streptavidin-immobilized magnetic beads. Next generation sequencing of the isolated small RNA fraction revealed that small RNAs with less than 200nt were effectively enriched, allowing us to identify previously unknown small RNAs in HeLa and E. coli.


Asunto(s)
ARN Catalítico/genética , ARN Pequeño no Traducido/aislamiento & purificación , ARN de Transferencia/genética , Aminoacil-ARNt Sintetasas , Secuenciación de Nucleótidos de Alto Rendimiento , Conformación de Ácido Nucleico , ARN Catalítico/química , ARN Pequeño no Traducido/genética , ARN de Transferencia/química , Aminoacilación de ARN de Transferencia/genética
11.
Nat Chem Biol ; 10(7): 555-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24907900

RESUMEN

The Watson-Crick base pairs between the 3'-terminal end of tRNAs and ribosomal RNA in the peptidyl transferase center are universally conserved. Here, we report that the introduction of compensatory mutations to Escherichia coli RNAs in this site leads to an orthogonal system independent of the wild-type counterpart, as demonstrated via the production of two peptide sequences from a single mRNA. This work thus identifies a new way to reprogram the genetic code.


Asunto(s)
Proteínas de Escherichia coli/genética , Escherichia coli/genética , Peptidil Transferasas/genética , Biosíntesis de Proteínas , ARN Ribosómico 23S/genética , ARN de Transferencia/genética , Emparejamiento Base , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Código Genético , Mutación , Peptidil Transferasas/metabolismo , Ingeniería de Proteínas , ARN Ribosómico 23S/metabolismo , ARN de Transferencia/metabolismo , Ribosomas/genética , Ribosomas/metabolismo
12.
Int J Mol Sci ; 16(3): 6513-31, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25803109

RESUMEN

Genetic code expansion and reprogramming methodologies allow us to incorporate non-canonical amino acids (ncAAs) bearing various functional groups, such as fluorescent groups, bioorthogonal functional groups, and post-translational modifications, into a desired position or multiple positions in polypeptides both in vitro and in vivo. In order to efficiently incorporate a wide range of ncAAs, several methodologies have been developed, such as orthogonal aminoacyl-tRNA-synthetase (AARS)-tRNA pairs, aminoacylation ribozymes, frame-shift suppression of quadruplet codons, and engineered ribosomes. More recently, it has been reported that an engineered translation system specifically utilizes an artificially built genetic code and functions orthogonally to naturally occurring counterpart. In this review we summarize recent advances in the field of ribosomal polypeptide synthesis containing ncAAs.


Asunto(s)
Aminoácidos/metabolismo , Péptidos/metabolismo , Aminoacil-ARNt Sintetasas/metabolismo , Ribosomas/metabolismo
13.
Bull Chem Soc Jpn ; 97(5): uoae018, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38828441

RESUMEN

Due to their constrained conformations, cyclic ß2,3-amino acids (cßAA) are key building blocks that can fold peptides into compact and rigid structures, improving peptidase resistance and binding affinity to target proteins, due to their constrained conformations. Although the translation efficiency of cßAAs is generally low, our engineered tRNA, referred to as tRNAPro1E2, enabled efficient incorporation of cßAAs into peptide libraries using the flexible in vitro translation (FIT) system. Here we report on the design and application of a macrocyclic peptide library incorporating 3 kinds of cßAAs: (1R,2S)-2-aminocyclopentane carboxylic acid (ß1), (1S,2S)-2-aminocyclohexane carboxylic acid (ß2), and (1R,2R)-2-aminocyclopentane carboxylic acid. This library was applied to an in vitro selection against the SARS-CoV-2 main protease (Mpro). The resultant peptides, BM3 and BM7, bearing one ß2 and two ß1, exhibited potent inhibitory activities with IC50 values of 40 and 20 nM, respectively. BM3 and BM7 also showed remarkable serum stability with half-lives of 48 and >168 h, respectively. Notably, BM3A and BM7A, wherein the cßAAs were substituted with alanine, lost their inhibitory activities against Mpro and displayed substantially shorter serum half-lives. This observation underscores the significant contribution of cßAA to the activity and stability of peptides. Overall, our results highlight the potential of cßAA in generating potent and highly stable macrocyclic peptides with drug-like properties.

14.
Nat Chem ; 15(7): 998-1005, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37217786

RESUMEN

γ-Amino acids can play important roles in the biological activities of natural products; however, the ribosomal incorporation of γ-amino acids into peptides is challenging. Here we report how a selection campaign employing a non-canonical peptide library containing cyclic γ2,4-amino acids resulted in the discovery of very potent inhibitors of the SARS-CoV-2 main protease (Mpro). Two kinds of cyclic γ2,4-amino acids, cis-3-aminocyclobutane carboxylic acid (γ1) and (1R,3S)-3-aminocyclopentane carboxylic acid (γ2), were ribosomally introduced into a library of thioether-macrocyclic peptides. One resultant potent Mpro inhibitor (half-maximal inhibitory concentration = 50 nM), GM4, comprising 13 residues with γ1 at the fourth position, manifests a 5.2 nM dissociation constant. An Mpro:GM4 complex crystal structure reveals the intact inhibitor spans the substrate binding cleft. The γ1 interacts with the S1' catalytic subsite and contributes to a 12-fold increase in proteolytic stability compared to its alanine-substituted variant. Knowledge of interactions between GM4 and Mpro enabled production of a variant with a 5-fold increase in potency.


Asunto(s)
Aminoácidos , COVID-19 , Aminoácidos/química , Antivirales/química , Ácidos Carboxílicos , Péptidos/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Conformación Proteica , SARS-CoV-2/metabolismo
15.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 11): 1414-6, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22102245

RESUMEN

The cytidine at the first anticodon position of archaeal tRNA(Ile2), which decodes the isoleucine AUA codon, is modified to 2-agmatinylcytidine (agm(2)C) to guarantee the fidelity of protein biosynthesis. This post-transcriptional modification is catalyzed by tRNA(Ile)-agm(2)C synthetase (TiaS) using ATP and agmatine as substrates. Archaeoglobus fulgidus TiaS was overexpressed in Escherichia coli cells and purified. tRNA(Ile2) was prepared by in vitro transcription with T7 RNA polymerase. TiaS was cocrystallized with both tRNA(Ile2) and ATP by the vapour-diffusion method. The crystals of the TiaS-tRNA(Ile2)-ATP complex diffracted to 2.9 Å resolution using synchrotron radiation at the Photon Factory. The crystals belonged to the primitive hexagonal space group P3(2)21, with unit-cell parameters a = b = 131.1, c = 86.6 Å. The asymmetric unit is expected to contain one TiaS-tRNA(Ile2)-ATP complex, with a Matthews coefficient of 2.8 Å(3) Da(-1) and a solvent content of 61%.


Asunto(s)
Adenosina Trifosfato/química , Archaeoglobales/enzimología , Isoleucina-ARNt Ligasa/química , ARN de Transferencia de Isoleucina/química , Adenosina Trifosfato/metabolismo , Cristalización , Cristalografía por Rayos X , Isoleucina-ARNt Ligasa/metabolismo , Unión Proteica , ARN de Transferencia de Isoleucina/metabolismo
16.
iScience ; 24(11): 103302, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34805784

RESUMEN

Lasso-grafting (LG) technology is a method for generating de novo biologics (neobiologics) by genetically implanting macrocyclic peptide pharmacophores, which are selected in vitro against a protein of interest, into loops of arbitrary protein scaffolds. In this study, we have generated a neo-capsid that potently binds the hepatocyte growth factor receptor MET by LG of anti-MET peptide pharmacophores into a circularly permuted variant of Aquifex aeolicus lumazine synthase (AaLS), a self-assembling protein nanocapsule. By virtue of displaying multiple-pharmacophores on its surface, the neo-capsid can induce dimerization (or multimerization) of MET, resulting in phosphorylation and endosomal internalization of the MET-capsid complex. This work demonstrates the potential of the LG technology as a synthetic biology approach for generating capsid-based neobiologics capable of activating signaling receptors.

17.
ACS Appl Mater Interfaces ; 13(46): 54817-54829, 2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34766499

RESUMEN

Fast and selective recognition of molecules at the nanometer scale without labeling is a much desired but still challenging goal to achieve. Here, we show the use of high-speed atomic force microscopy (HS-AFM) for real-time and real-space recognition of unlabeled membrane receptors using tips conjugated with small synthetic macrocyclic peptides. The single-molecule recognition method is validated by experiments on the human hepatocyte growth factor receptor (hMET), which selectively binds to the macrocyclic peptide aMD4. By testing and comparing aMD4 synthesized with linkers of different lengths and rigidities, we maximize the interaction between the functionalized tip and hMET added to both a mica surface and supported lipid bilayers. Phase contrast imaging by HS-AFM enables us to discriminate nonlabeled hMET against the murine MET homologue, which does not bind to aMD4. Moreover, using ligands and linkers of small size, we achieve minimal deterioration of the spatial resolution in simultaneous topographic imaging. The versatility of macrocyclic peptides in detecting unlimited types of membrane receptors with high selectivity and the fast imaging by HS-AFM broaden the range of future applications of this method for molecular recognition without labeling.


Asunto(s)
Compuestos Macrocíclicos/química , Péptidos/química , Proteínas Proto-Oncogénicas c-met/análisis , Silicatos de Aluminio/química , Animales , Humanos , Ligandos , Membrana Dobles de Lípidos/química , Compuestos Macrocíclicos/síntesis química , Ratones , Microscopía de Fuerza Atómica , Estructura Molecular , Nanotecnología , Imagen Óptica , Péptidos/síntesis química , Propiedades de Superficie
18.
Science ; 372(6547): 1220-1224, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34112695

RESUMEN

Viruses are ubiquitous pathogens of global impact. Prompted by the hypothesis that their earliest progenitors recruited host proteins for virion formation, we have used stringent laboratory evolution to convert a bacterial enzyme that lacks affinity for nucleic acids into an artificial nucleocapsid that efficiently packages and protects multiple copies of its own encoding messenger RNA. Revealing remarkable convergence on the molecular hallmarks of natural viruses, the accompanying changes reorganized the protein building blocks into an interlaced 240-subunit icosahedral capsid that is impermeable to nucleases, and emergence of a robust RNA stem-loop packaging cassette ensured high encapsidation yields and specificity. In addition to evincing a plausible evolutionary pathway for primordial viruses, these findings highlight practical strategies for developing nonviral carriers for diverse vaccine and delivery applications.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cápside/metabolismo , Evolución Molecular Dirigida , ARN Mensajero/metabolismo , Sustitución de Aminoácidos , Aquifex/enzimología , Proteínas Bacterianas/química , Cápside/química , Microscopía por Crioelectrón , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Nucleocápside/química , Nucleocápside/genética , Nucleocápside/metabolismo , Dominios Proteicos , Estructura Secundaria de Proteína , Subunidades de Proteína , ARN Mensajero/química , ARN Mensajero/genética , Ribonucleasas/metabolismo
19.
Cell Chem Biol ; 27(9): 1181-1191.e7, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32640189

RESUMEN

Medium-sized macrocyclic peptides are an alternative to small compounds and large biomolecules as a class of pharmaceutics. The CD47-SIRPα signaling axis functions as an innate immune checkpoint that inhibits phagocytosis in phagocytes and has been implicated as a promising target for cancer immunotherapy. The potential of macrocyclic peptides that target this signaling axis as immunotherapeutic agents has remained unknown, however. Here we have developed a macrocyclic peptide consisting of 15 amino acids that binds to the ectodomain of mouse SIRPα and efficiently blocks its interaction with CD47 in an allosteric manner. The peptide markedly promoted the phagocytosis of antibody-opsonized tumor cells by macrophages in vitro as well as enhanced the inhibitory effect of anti-CD20 or anti-gp75 antibodies on tumor formation or metastasis in vivo. Our results suggest that allosteric inhibition of the CD47-SIRPα interaction by macrocyclic peptides is a potential approach to cancer immunotherapy.


Asunto(s)
Antígeno CD47/metabolismo , Péptidos Cíclicos/metabolismo , Receptores Inmunológicos/metabolismo , Regulación Alostérica , Animales , Antígenos CD20/inmunología , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno CD47/química , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Péptidos Cíclicos/química , Péptidos Cíclicos/uso terapéutico , Fagocitosis , Unión Proteica , Receptores Inmunológicos/química , Rituximab/inmunología , Rituximab/uso terapéutico , Tasa de Supervivencia
20.
Nat Commun ; 10(1): 5403, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31776333

RESUMEN

Glycosylation of proteins profoundly impacts their physical and biological properties. Yet our ability to engineer novel glycoprotein structures remains limited. Established bacterial glycoengineering platforms require secretion of the acceptor protein to the periplasmic space and preassembly of the oligosaccharide substrate as a lipid-linked precursor, limiting access to protein and glycan substrates respectively. Here, we circumvent these bottlenecks by developing a facile glycoengineering platform that operates in the bacterial cytoplasm. The Glycoli platform leverages a recently discovered site-specific polypeptide glycosyltransferase together with variable glycosyltransferase modules to synthesize defined glycans, of bacterial or mammalian origin, directly onto recombinant proteins in the E. coli cytoplasm. We exploit the cytoplasmic localization of this glycoengineering platform to generate a variety of multivalent glycostructures, including self-assembling nanomaterials bearing hundreds of copies of the glycan epitope. This work establishes cytoplasmic glycoengineering as a powerful platform for producing glycoprotein structures with diverse future biomedical applications.


Asunto(s)
Citoplasma/metabolismo , Glicoproteínas/biosíntesis , Ingeniería Metabólica/métodos , Benzazepinas , Epítopos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Glucosa/química , Glucosa/metabolismo , Glucosiltransferasas/metabolismo , Glicoproteínas/genética , Glicoproteínas/inmunología , Glicosilación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Monosacáridos , Polisacáridos/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA