Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Pept Sci ; 29(12): e3527, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37322567

RESUMEN

We report the first application of a novel amino-Li resin to water-based solid-phase peptide synthesis (SPPS) applying the Smoc-protecting group approach. We demonstrated that it is a suitable support for the sustainable water-based alternative to a classical SPPS approach. The resin possesses good swelling properties in aqueous milieu, provides significant coupling sites, and may be applicable to the synthesis of difficult sequences and aggregation-prone peptides.


Asunto(s)
Técnicas de Síntesis en Fase Sólida , Agua , Péptidos/química
2.
Angew Chem Int Ed Engl ; 61(45): e202210883, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36049110

RESUMEN

Sactipeptides are ribosomally synthesized peptides containing a unique sulfur to α-carbon crosslink. Catalyzed by sactisynthases, this thioether pattern endows sactipeptides with enhanced structural, thermal, and proteolytic stability, which makes them attractive scaffolds for the development of novel biotherapeutics. Herein, we report the in-depth study on the substrate tolerance of the sactisynthase AlbA to catalyze the formation of thioether bridges in sactipeptides. We identified a possible modification site within the sactipeptide subtilosin A allowing for peptide engineering without compromising formation of thioether bridges. A panel of natural and hybrid sactipeptides was produced to study the AlbA-mediated formation of thioether bridges, which were identified mass-spectrometrically. In a proof-of-principle study, we re-engineered subtilosin A to a thioether-bridged, specific streptavidin targeting peptide, opening the door for the functional engineering of sactipeptides.


Asunto(s)
Péptidos , Sulfuros , Sulfuros/química , Péptidos/química
3.
J Pept Sci ; 27(4): e3298, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33458922

RESUMEN

The development of novel biotherapeutics based on peptides and proteins is often limited to extracellular targets, because these molecules are not able to reach the cytosol. In recent years, several approaches were proposed to overcome this limitation. A plethora of cell-penetrating peptides (CPPs) was developed for cytoplasmic delivery of cell-impermeable cargo molecules. For many CPPs, multimerization or multicopy arrangement on a scaffold resulted in improved delivery but also higher cytotoxicity. Recently, we introduced dextran as multivalent, hydrophilic polysaccharide scaffold for multimerization of cell-targeting cargoes. Here, we investigated covalent conjugation of a CPP to dextran in multiple copies and assessed the ability of resulted molecular hybrid to enter the cytoplasm of mammalian cells without largely compromising cell viability. As a CPP, we used a novel, low-toxic cationic amphiphilic peptide L17E derived from M-lycotoxin. Here, we show that cell-penetrating properties of L17E are retained upon multivalent covalent linkage to dextran. Dextran-L17E efficiently mediated cytoplasmic translocation of an attached functional peptide and a peptide nucleic acid (PNA). Moreover, a synthetic route was established to mask the lysine side chains of L17E with a photolabile protecting group thus opening avenues for light-triggered activation of cellular uptake.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Citosol/metabolismo , Dextranos/metabolismo , Colorantes Fluorescentes/metabolismo , Péptidos de Penetración Celular/síntesis química , Péptidos de Penetración Celular/química , Citosol/química , Dextranos/química , Colorantes Fluorescentes/química , Células HeLa , Humanos , Estructura Molecular , Imagen Óptica , Células Tumorales Cultivadas
4.
Chembiochem ; 20(24): 3006-3012, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31206933

RESUMEN

Triggering apoptosis of tumor cells has been in focus of cancer-inspired research since decades. As clustering of death receptor 5 (DR5), which is overexpressed on various cancer cells, leads to formation of the death-inducing signaling cascade (DISC), DR5 has recently become a promising target for tumor treatment. Herein, we demonstrate that covalent multimerization of a death receptor targeting peptide (DR5TP) on a dextran scaffold generates potent apoptosis-inducing conjugates (EC50 =2-20 nm). A higher conformational flexibility compared to reported DR5TP multimerization approaches, introduced by the polysaccharide framework compensates the reported need for the defined ligand orientation that was considered as essential prerequisite for effective receptor clustering and apoptosis induction. Enzyme-catalyzed ligation of a hydrophilic dextran conjugate bearing multiple DR5-targeting sites to a human fragment crystallizable (Fc) receptor did not affect the potency (EC50 =2-7 nm), providing an option for improved in vivo half-life and prospective conjugation to an antibody of interest in view of bispecific tumor targeting.


Asunto(s)
Apoptosis/efectos de los fármacos , Dextranos/química , Peptidomiméticos/química , Peptidomiméticos/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Secuencia de Aminoácidos , Supervivencia Celular/efectos de los fármacos , Humanos , Células Jurkat
5.
Chemistry ; 24(57): 15195-15200, 2018 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-30047596

RESUMEN

Microbial transglutaminase from Streptomyces mobaraensis (mTG) has emerged as a useful biotechnological tool due to its ability to crosslink a side chain of glutamine and primary amines. To date, the substrate specificity of mTG is not fully understood, which poses an obvious challenge when mTG is used to address novel targets. To that end, a viable strategy providing an access to tailor-made transglutaminases is required. This work reports an ultrahigh-throughput screening approach based on yeast surface display and fluorescence-activated cell sorting (FACS) that enabled the evolution of microbial transglutaminase towards enhanced activity. Five rounds of FACS screening followed by recombinant expression of the most potent variants in E. coli yielded variants that possessed, compared to the wild type enzyme, improved enzymatic performance and labeling behavior upon conjugation with an engineered therapeutic anti-HER2 antibody. This robust and generally applicable platform enables tailoring of the catalytic efficiency of mTG.


Asunto(s)
Evolución Molecular Dirigida/métodos , Ingeniería de Proteínas/métodos , Streptomyces/enzimología , Streptomyces/genética , Transglutaminasas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Inmunoconjugados/genética , Inmunoconjugados/metabolismo , Modelos Moleculares , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Fluorescencia , Streptomyces/metabolismo , Transglutaminasas/metabolismo
6.
Chemistry ; 22(2): 506-10, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26612739

RESUMEN

The activation of element-hydrogen bonds by means of metal-ligand cooperation has received increasing attention as alternative to classical activation processes, which exclusively occur at the metal center. Carbene complexes derived from methandiide precursors have been applied in this chemistry enabling the activation of a series of E-H bonds by addition reactions across the M-C bond. However, no chiral carbene complexes have been applied to realize stereoselective transformations to date. Herein, we report the isolation and structure elucidation of an enantiomerically pure dilithiomethane, which could be prepared by direct double deprotonation. The obtained dilithium salt was used for the preparation of the first chiral methandiide-derived carbene complex, which was applied in stereoselective cooperative S-H bond activation.

7.
Angew Chem Int Ed Engl ; 53(47): 12941-5, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-25296746

RESUMEN

A series of novel bioactive derivatives of the sunflower trypsin inhibitor-1 (SFTI-1) suitable for hyperpolarization by parahydrogen-induced polarization (PHIP) was developed. The PHIP activity was achieved by labeling with L-propargylglycine, O-propargyl-L-tyrosine, or 4-pentynoic acid. (1) H NMR signal enhancements (SE) of up to a factor of 70 were achieved in aqueous solution. We found that an isolated spatial location of the triple bond within the respective label and its accessibility for the hydrogenation catalyst are essential factors for the degree of signal enhancement.


Asunto(s)
Alquinos/química , Ácidos Grasos Insaturados/química , Glicina/análogos & derivados , Espectroscopía de Resonancia Magnética/métodos , Péptidos Cíclicos/química , Tirosina/análogos & derivados , Catálisis , Glicina/química , Hidrogenación , Modelos Moleculares , Estructura Molecular , Protones , Tirosina/química
8.
Front Chem ; 9: 693097, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34368077

RESUMEN

Herein, we present the design, synthesis, and biological evaluation of novel integrin-targeting molecular hybrids combining RGD peptides and a potent cytotoxin presented on dextran polysaccharides. Based on an aglycosylated Fc as a centerpiece, endosomal-cleavable cytotoxic agent monomethyl auristatin E (MMAE) and dextran as multimerization site were covalently connected by two bioorthogonal enzyme-mediated reactions site-specifically. Decoration of dextran with cyclic RGD peptides, introduced by copper "click" reaction, resulted in the final constructs with the potential to kill integrin-overexpressing tumor cells. We found that these modifications had little impact on the stability of the Fc scaffold and the RGD-bearing construct showed good binding properties of αvß3-expressing U87MG cells. Furthermore, the construct showed a remarkable antiproliferative activity. These results demonstrate the general capability of our design to provoke receptor-mediated endocytosis upon binding to the cellular surface, followed by endosomal cleavage of the linkage between Fc-dextran and MMAE and its subsequent release. Our approach opens new avenues to transcribe small molecule binders into tailor-made multimeric molecular hybrids with antitumor potential.

9.
ChemistryOpen ; 8(3): 354-357, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30976476

RESUMEN

Antibody-drug conjugates (ADCs) are multicomponent biomolecules that have emerged as a powerful tool for targeted tumor therapy. Combining specific binding of an immunoglobulin with toxic properties of a payload, they however often suffer from poor hydrophilicity when loaded with a high amount of toxins. To address these issues simultaneously, we developed dextramabs, a novel class of hybrid antibody-drug conjugates. In these architectures, the therapeutic antibody trastuzumab is equipped with a multivalent dextran polysaccharide that enables efficient loading with a potent toxin in a controllable fashion. Our modular chemoenzymatic approach provides an access to synthetic dextramabs bearing monomethyl auristatin as releasable cytotoxic cargo. They possess high drug-to-antibody ratios, remarkable hydrophilicity, and high toxicity in vitro.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA