Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 64(7): 4109-4116, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33761256

RESUMEN

Small molecule colloidal aggregates adsorb and partially denature proteins, inhibiting them artifactually. Oddly, this inhibition is typically time-dependent. Two mechanisms might explain this: low concentrations of the colloid and enzyme might mean low encounter rates, or colloid-based protein denaturation might impose a kinetic barrier. These two mechanisms should have different concentration dependencies. Perplexingly, when enzyme concentration was increased, incubation times actually lengthened, inconsistent with both models and with classical chemical kinetics of solution species. We therefore considered molecular crowding, where colloids with lower protein surface density demand a shorter incubation time than more crowded colloids. To test this, we grew and shrank colloid surface area. As the surface area shrank, the incubation time lengthened, while as it increased, the converse was true. These observations support a crowding effect on protein binding to colloidal aggregates. Implications for drug delivery and for detecting aggregation-based inhibition will be discussed.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coloides/metabolismo , Malato Deshidrogenasa/metabolismo , beta-Lactamasas/metabolismo , Adsorción , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Coloides/química , Pruebas de Enzimas , Fulvestrant/química , Cinética , Malato Deshidrogenasa/antagonistas & inhibidores , Malato Deshidrogenasa/química , Unión Proteica , Sorafenib/química , beta-Lactamasas/química
2.
Chembiochem ; 20(4): 526-531, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30388302

RESUMEN

Phosphatases play an important role in cell biology, but only a few probes are suitable for selectively imaging phosphatase activity in live cells, because the current probes require cell fixation or exhibit considerable cytotoxicity. Herein, we show that conjugating a d-peptide to a quinazolinone derivative generates cell-compatible, biostable probes for imaging the phosphatase activity inside live cells. Moreover, our results show that inhibiting ectophosphatases is a critical factor for imaging intracellular phosphatases. As the first example of using selective inhibitors to ensure intracellular function of molecular probes, this work illustrates a facile approach to design molecular probes for profiling the activities of enzymes in a spatial, selective manner in a complicated environment.


Asunto(s)
Nanopartículas/química , Péptidos/química , Monoéster Fosfórico Hidrolasas/metabolismo , Línea Celular Tumoral , Colorantes Fluorescentes/química , Humanos , Microscopía Confocal , Péptidos/metabolismo , Monoéster Fosfórico Hidrolasas/química , Quinazolinonas/química
3.
Angew Chem Int Ed Engl ; 57(36): 11716-11721, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-29971927

RESUMEN

In a model study to investigate the consequence of reactions of intrinsically disordered regions (IDRs) of proteins in the context of the formation of highly ordered structures, we found that enzymatic reactions control the secondary structures of peptides during assembly. Specifically, phosphorylation of an α-helix-dominant peptide results in mostly disordered conformations, which become ß-strand-dominant after enzymatic dephosphorylation to regenerate the peptide. In the presence of another peptide largely with a ß-strand conformation, direct coassembly of the peptides results in amorphous aggregates consisting of α-helix and ß-strand peptides, but the enzymatically generated peptide coassemblies (from the phosphopeptide) mainly adopt a ß-strand conformation and form ordered structures (e.g., nanofibers). These results indicate that enzymatic dephosphorylation instructs conformationally flexible peptides to adopt thermodynamically favorable conformations in homotypic or heterotypic supramolecular assemblies.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Péptidos/química , Biocatálisis , Modelos Moleculares , Nanofibras/química , Nanofibras/ultraestructura , Fosfopéptidos/química , Fosforilación , Agregado de Proteínas , Estructura Secundaria de Proteína , Termodinámica
4.
Biochemistry ; 57(32): 4867-4879, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30001488

RESUMEN

Despite the well-established biophysical principle of adhesion-guided in vitro morphogenesis, there are few single synthetic molecular species that can rapidly enable morphogenesis (e.g., a cell monolayer to cell spheroids) in a cell culture because adhesion inherently involves many signals. Here we show the use of adaptive multifunctional supramolecular assemblies of glycopeptides, consisting of cell adhesion sequence and saccharide, to induce cell spheroids rapidly from a monolayer of cells. Having a general architecture of N-terminal capping, glycosylation, and an integrin-binding sequence, the glycopeptides self-assemble to form a dynamic continuum of nanostructures (i.e., from nanoparticles to nanofibers) to affect the interactions of integrins, E-selectin, and cadherins with their natural ligands and to act adaptively according to the cellular environment. Such adaptive (i.e., context-dependent) interactions weaken cell-substratum adhesion and enhance intercellular interactions, which rapidly and transiently induce cell spheroids. This work illustrates the use of supramolecular assemblies of simple glycopeptides to modulate biophysical conditions for regulating cell functions, which is a new approach for developing biomaterials.


Asunto(s)
Glicopéptidos/química , Cadherinas/antagonistas & inhibidores , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Técnicas de Cultivo de Célula , Selectina E/antagonistas & inhibidores , Selectina E/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Fibronectinas/antagonistas & inhibidores , Fibronectinas/metabolismo , Glicopéptidos/farmacología , Humanos , Integrinas/antagonistas & inhibidores , Integrinas/metabolismo , Morfogénesis/efectos de los fármacos , Nanoestructuras/química
5.
Chem Asian J ; 13(22): 3464-3468, 2018 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29897657

RESUMEN

Nanofibers of short peptides are emerging as a promising type of agents for inhibiting cancer cells. But the proteolysis of peptides decreases the anticancer efficacy of the peptide nanofibers. Here we show that decreasing the activity of proteasomes enhance the activity of peptide nanofibers for inhibiting cancer cells. Based on the structure of galactin-3, we designed a heptapeptide, which self-assembles to form nanofibers. The nanofibers of the heptapeptide exhibit moderate cytotoxicity to three representative cancer cell lines (HeLa, MCF-7, and HepG2), largely due to the proteolysis of the peptides. Using a clinically approved proteasome inhibitor, bortezomib, to treat the cancer cells significantly decreases the proteolysis of the peptides and enhances the activity of the peptide nanofibers for inhibiting the cancer cells. This work illustrates a promising approach for enhancing the anticancer efficacy of peptide nanofibers by modulating intracellular protein degradation machinery, as well as provides insights for understanding the cytotoxicity of aberrant protein or peptide aggregates in complicated cellular environment.


Asunto(s)
Nanofibras/química , Péptidos/química , Secuencia de Aminoácidos , Supervivencia Celular/efectos de los fármacos , Galectina 3/química , Células HeLa , Células Hep G2 , Humanos , Hidrogeles/química , Células MCF-7 , Péptidos/metabolismo , Péptidos/farmacología , Proteolisis
6.
ACS Nano ; 12(4): 3804-3815, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29537820

RESUMEN

Recent studies have demonstrated that enzyme-instructed self-assembly (EISA) in extra- or intracellular environments can serve as a multistep process for controlling cell fate. There is little knowledge, however, about the kinetics of EISA in the complex environments in or around cells. Here, we design and synthesize three dipeptidic precursors (ld-1-SO3, dl-1-SO3, dd-1-SO3), consisting of diphenylalanine (l-Phe-d-Phe, d-Phe-l-Phe, d-Phe-d-Phe, respectively) as the backbone, which are capped by 2-(naphthalen-2-yl)acetic acid at the N-terminal and by 2-(4-(2-aminoethoxy)-4-oxobutanamido)ethane-1-sulfonic acid at the C-terminal. On hydrolysis by carboxylesterases (CES), these precursors result in hydrogelators, which self-assemble in water at different rates. Whereas all three precursors selectively kill cancer cells, especially high-grade serous ovarian carcinoma cells, by undergoing intracellular EISA, dl-1-SO3 and dd-1-SO3 exhibit the lowest and the highest activities, respectively, against the cancer cells. This trend inversely correlates with the rates of converting the precursors to the hydrogelators in phosphate-buffered saline. Because CES exists both extra- and intracellularly, we use kinetic modeling to analyze the kinetics of EISA inside cells and to calculate the cytotoxicity of each precursor for killing cancer cells. Our results indicate that (i) the stereochemistry of the precursors affects the morphology of the nanostructures formed by the hydrogelators, as well as the rate of enzymatic conversion; (ii) decreased extracellular hydrolysis of precursors favors intracellular EISA inside the cells; (iii) the inherent features ( e.g., self-assembling ability and morphology) of the EISA molecules largely dictate the cytotoxicity of intracellular EISA. As the kinetic analysis of intracellular EISA, this work elucidates how the stereochemistry modulates EISA in the complex extra- and/or intracellular environment for developing anticancer molecular processes. Moreover, it provides insights for understanding the kinetics and cytotoxicity of aggregates of aberrant proteins or peptides formed inside and outside cells.


Asunto(s)
Antineoplásicos/farmacología , Hidrolasas de Éster Carboxílico/metabolismo , Dipéptidos/farmacología , Nanoestructuras/química , Neoplasias Ováricas/tratamiento farmacológico , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dipéptidos/síntesis química , Dipéptidos/química , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Hidrólisis , Cinética , Neoplasias Ováricas/patología
7.
Mol Ther ; 26(2): 648-658, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29396265

RESUMEN

Most of the peptides used for promoting cellular uptake bear positive charges. In our previous study, we reported an example of taurine (bearing negative charges in physiological conditions) promoting cellular uptake of D-peptides. Taurine, conjugated to a small D-peptide via an ester bond, promotes the cellular uptake of this D-peptide. Particularly, intracellular carboxylesterase (CES) instructs the D-peptide to self-assemble and to form nanofibers, which largely disfavors efflux and further enhances the intracellular accumulation of the D-peptide, as supported by that the addition of CES inhibitors partially impaired cellular uptake of this molecule in mammalian cell lines. Using dynamin 1, 2, and 3 triple knockout (TKO) mouse fibroblasts, we demonstrated that cells took up this molecule via macropinocytosis and dynamin-dependent endocytosis. Imaging of Drosophila larval blood cells derived from endocytic mutants confirmed the involvement of multiple endocytosis pathways. Electron microscopy (EM) indicated that the precursors can form aggregates on the cell surface to facilitate the cellular uptake via macropinocytosis. EM also revealed significantly increased numbers of vesicles in the cytosol. This work provides new insights into the cellular uptake of taurine derivative for intracellular delivery and self-assembly of D-peptides.


Asunto(s)
Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Péptidos/farmacología , Pinocitosis/efectos de los fármacos , Taurina , Animales , Transporte Biológico , Línea Celular , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Estructura Molecular , Péptidos/química , Transducción de Señal/efectos de los fármacos , Taurina/química
8.
J Am Chem Soc ; 140(6): 2301-2308, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29377688

RESUMEN

As a promising molecular process for selectively inhibiting cancer cells without inducing acquired drug resistance, enzyme-instructed self-assembly (EISA) usually requires relatively high dosages. Despite its discovery 30 years ago, the translation of the knowledge about NF-κB signaling into clinic remains complicated due to the broad roles of NF-κB in cellular regulation. Here we show that integrating EISA and NF-κB targeting boosts the efficacy of EISA over an order of magnitude without compromising selectivity against cancer cells. That is, in situ enzymatic self-assembly of a tetrapeptide results in nanofibers, which hardly affect cell viability, but lead to inductive expression of tumor necrosis factor receptor 2 (TNFR2) and decreased expression of three key proteins at the upstream of NF-κB pathway in the cancer cells. Adding the inhibitors targeting NF-κB further decreases the expressions of those upstream proteins, which turns the otherwise innocuous nanofibers to being lethal to the cancer cells, likely causing necroptosis. As the first case of using supramolecular processes to enable synthetic lethality, this work illustrates a versatile approach to translate key regulatory circuits into promising therapeutic targets.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , FN-kappa B/metabolismo , Nanofibras/uso terapéutico , Oligopéptidos/uso terapéutico , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Antineoplásicos/química , Biocatálisis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , FN-kappa B/antagonistas & inhibidores , Nanofibras/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oligopéptidos/química , Transducción de Señal
9.
Interface Focus ; 7(6): 20160116, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29147550

RESUMEN

As a novel class of biomaterials, nucleopeptides, via the conjugation of nucleobases and peptides, usually self-assemble to form nanofibres driven mainly by hydrogen bonds. Containing nucleobase(s), nucleopeptides have a unique property-interacting with nucleic acids. Here we report the design and characterization of nucleopeptides that self-assemble in water and are able to interact with single-stranded DNAs (ssDNAs). Containing nucleobases on their side chains, these nucleopeptides bind with the ssDNAs, and the ssDNAs reciprocally affect the self-assembly of nucleopeptides. In addition, the interactions between nucleopeptides and ssDNAs also decrease their proteolytic resistance against proteinase K, which further demonstrates the binding with ssDNAs. The nucleopeptides also interact with plasmid DNA and deliver hairpin DNA into cells. This work illustrates a new and rational approach to create soft biomaterials by the integration of nucleobases and peptides to bind with DNA, which may lead to the development of nucleopeptides for controlling DNA in cells.

10.
ChemNanoMat ; 3(1): 17-21, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29104854

RESUMEN

Reaction-diffusion (RD) is the most important inherent feature of living organism, but it has yet to be used for developing biofunctional nanoparticles (NPs). Here we show the use of chirality to control the RD of NPs for selectively inhibiting cancer cells. We observe that L-phosphotyrosine (L-pY) decorated NPs (NP@L-pYs) are innocuous to cells, but D-pY decorated ones (NP@D-pYs) selectively inhibit cancer cells. Our study shows that alkaline phosphatases (ALP), presented in the culture and overexpressed on the cancer cells, dephosphorylates NP@L-pYs much faster than NP@D-pYs. Such a rate difference allows the NP@D-pYs to be mainly dephosphorylated on cell surface, thus adhering selectively on the cancer cells to result in poly(ADP-ribose)polymerase (PARP) hyperactivation mediated cell death. Without phosphate groups or being prematurely dephosphorylated before reaching cancer cells (as the case of NP@L-pYs), the NPs are innocuous to cells. Moreover, NP@D-pYs even exhibit more potent activity than cisplatin for inhibiting platinum-resistant ovarian cancer cells (e.g., A2780-cis). As the first example of chirality controlling RD process of NPs for inhibiting cancer cells, this work illustrates a fundamentally new way for developing nanomedicine based on RD processes and nanoparticles.

11.
Org Biomol Chem ; 15(27): 5689-5692, 2017 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-28675212

RESUMEN

Here we show the first example of an immunoreceptor tyrosine-based inhibitory motif (ITIM), LYYYYL, as well as its enantiomeric or retro-inverso peptide, to self-assemble in water via enzyme-instructed self-assembly. Upon enzymatic dephosphorylation, the phosphohexapeptides become hexapeptides, which self-assemble in water to result in supramolecular hydrogels. This work illustrates a new approach to design bioinspired soft materials from a less explored, but important pool of immunomodulatory peptides.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Péptidos/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Motivo de Inhibición del Inmunorreceptor Basado en Tirosina , Imagen Óptica , Péptidos/farmacología
12.
Biomaterials ; 129: 1-27, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28319779

RESUMEN

This review discusses supramolecular biofunctional materials, a novel class of biomaterials formed by small molecules that are held together via noncovalent interactions. The complexity of biology and relevant biomedical problems not only inspire, but also demand effective molecular design for functional materials. Supramolecular biofunctional materials offer (almost) unlimited possibilities and opportunities to address challenging biomedical problems. Rational molecular design of supramolecular biofunctional materials exploit powerful and versatile noncovalent interactions, which offer many advantages, such as responsiveness, reversibility, tunability, biomimicry, modularity, predictability, and, most importantly, adaptiveness. In this review, besides elaborating on the merits of supramolecular biofunctional materials (mainly in the form of hydrogels and/or nanoscale assemblies) resulting from noncovalent interactions, we also discuss the advantages of small peptides as a prevalent molecular platform to generate a wide range of supramolecular biofunctional materials for the applications in drug delivery, tissue engineering, immunology, cancer therapy, fluorescent imaging, and stem cell regulation. This review aims to provide a brief synopsis of recent achievements at the intersection of supramolecular chemistry and biomedical science in hope of contributing to the multidisciplinary research on supramolecular biofunctional materials for a wide range of applications. We envision that supramolecular biofunctional materials will contribute to the development of new therapies that will ultimately lead to a paradigm shift for developing next generation biomaterials for medicine.


Asunto(s)
Materiales Biocompatibles/farmacología , Secuencia de Aminoácidos , Animales , Sistemas de Liberación de Medicamentos , Humanos , Hidrogeles/química , Modelos Moleculares , Ingeniería de Tejidos
13.
Cell Death Dis ; 8(2): e2614, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28206986

RESUMEN

Nanofibrils of small molecules, as a new class of biofunctional entities, exhibit emergent properties for controlling cell fates, but the relevant mechanism remains to be elucidated and the in vivo effect has yet to be examined. Here, we show that D-peptide nanofibrils, generated by enzyme-instructed self-assembly (EISA), pleiotropically activate extrinsic death signaling for selectively killing cancer cells. Catalyzed by alkaline phosphatases and formed in situ on cancer cells, D-peptide nanofibrils present autocrine proapoptotic ligands to their cognate receptors in a juxtacrine manner, as well as directly cluster the death receptors. As multifaceted initiators, D-peptide nanofibrils induce apoptosis of cancer cells without harming normal cells in a co-culture, kill multidrug-resistant (MDR) cancer cells, boost the activities of anticancer drugs, and inhibit tumor growth in a murine model. Such a supramolecular cellular biochemical process (consisting of reaction, assembly, and binding) for multi-targeting or modulating protein-protein interaction networks ultimately may lead to new ways for combating cancer drug resistance.


Asunto(s)
Neoplasias/inmunología , Péptidos/inmunología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células HeLa , Humanos , Nanofibras , Péptidos/farmacología
14.
Adv Healthc Mater ; 6(15)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28233466

RESUMEN

Tight ligand-receptor binding, paradoxically, is a major root of drug resistance in cancer chemotherapy. To address this problem, instead of using conventional inhibitors or ligands, this paper focuses on the development of a novel process-enzyme-instructed self-assembly (EISA)-to kill cancer cells selectively. Here it is demonstrated that EISA as an intracellular process to generate nanofibrils of short peptides for selectively inhibiting cancer cell proliferation, including drug resistant ones. As the process that turns the non-self-assembling precursors into the self-assembling peptides upon the catalysis of carboxylesterases (CES), EISA occurs intracellularly to selectively inhibit a range of cancer cells that exhibit relatively high CES activities. More importantly, EISA inhibits drug resistant cancer cells (e.g., triple negative breast cancer cells (HCC1937) and platinum-resistant ovarian cells (SKOV3, A2780cis)). With the IC50 values of 28-80 and 25-44 µg mL-1 of l- and d-dipeptide precursors against cancer cells, respectively, EISA is innocuous to normal cells. Moreover, using coculture of cancer and normal cells, the selectivity of EISA is validated against cancer cells. Besides revealing that intracellular EISA cause apoptosis or necroptosis to kill the cancer cells, this work illustrates a new approach to amplify the enzymatic difference between cancer and normal cells and to expand the pool of drug candidates for potentially overcoming drug resistance in cancer therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Hidrolasas de Éster Carboxílico/metabolismo , Dipéptidos/administración & dosificación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Experimentales/patología , Resultado del Tratamiento
15.
Angew Chem Int Ed Engl ; 56(10): 2623-2627, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28128498

RESUMEN

Hydrogels consisting of carboxylic acid groups and N-isopropylacrylamide as pendants on their polymeric network usually exhibit volume expansion upon deprotonation or volume contraction when being heated. Demonstrated here is an anti-intuitive case of a hydrogel containing multiple carboxylic acid groups at each crosslinking point in the polymeric network, which shrinks upon increasing pH from 1 to 7 at 37 °C or expands upon heating from 25 to 37 °C at pH 1. The unexpected volume change originates from the high percentage of the crosslinker in the polymers, as detected by solid-state 13 C NMR spectroscopy. In addition, the volume changes are thermally reversible. As the first example of the use of functional hyper-crosslinkers to control the pH and thermal responses of nanogels, this work illustrates a new way to design soft materials with unusual behaviors.

16.
Front Chem Sci Eng ; 11(4): 509-515, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29403673

RESUMEN

Enzyme-instructed self-assembly (EISA) offers a facile approach to explore the supramolecular assemblies of small molecules in cellular milieu for a variety of biomedical applications. One of the commonly used enzymes is phosphatase, but the study of the substrates of phosphatases mainly focuses on the phosphotyrosine containing peptides. In this work, we examine the EISA of phosphoserine containing small peptides for the first time by designing and synthesizing a series of precursors containing only phosphoserine or both phosphoserine and phosphotyrosine. Conjugating a phosphoserine to the C-terminal of a well-established self-assembling peptide backbone, (naphthalene-2-ly)-acetyl-diphenylalanine (NapFF), affords a novel hydrogelation precursor for EISA. The incorporation of phosphotyrosine, another substrate of phosphatase, into the resulting precursor, provides one more enzymatic trigger on a single molecule, and meanwhile increases the precursors' propensity to aggregate after being fully dephosphorylated. Exchanging the positions of phosphorylated serine and tyrosine in the peptide backbone provides insights on how the specific molecular structures influence self-assembling behaviors of small peptides and the subsequent cellular responses. Moreover, the utilization of D-amino acids largely enhances the biostability of the peptides, thus providing a unique soft material for potential biomedical applications.

17.
J Am Chem Soc ; 139(1): 71-74, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-27997165

RESUMEN

Isolated short peptides usually are unable to maintain their original secondary structures due to the lack of the restriction from proteins. Here we show that two complementary pentapeptides from a ß-sheet motif of a protein, being connected to an aromatic motif (i.e., pyrene) at their C-terminal, self-assemble to form ß-sheet like structures upon mixing. Besides enabling the self-assembly to result in supramolecular hydrogels upon mixing, aromatic-aromatic interactions promote the pentapeptides transform from α-helix to ß-sheet conformation. As the first example of using aromatic-aromatic interactions to mimic the conformational restriction in a protein, this work illustrates a bioinspired way to generate peptide nanofibers with predefined secondary structures of the peptides by a rational design using protein structures as the blueprint.


Asunto(s)
Hidrogeles/química , Péptidos/química , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta
18.
J Am Chem Soc ; 138(47): 15397-15404, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27797504

RESUMEN

The concurrence of enzymatic reaction and ligand-receptor interactions is common for proteins, but rare for small molecules and has yet to be explored. Here we show that ligand-receptor interaction modulates the morphology of molecular assemblies formed by enzyme-instructed assembly of small molecules. While the absence of ligand-receptor interaction allows enzymatic dephosphorylation of a precursor to generate the hydrogelator that self-assembles to form long nanofibers, the presence of the ligand-receptor interaction biases the pathway to form precipitous aggregates containing short nanofibers. While the hydrogelators self-assemble to form nanofibers or nanoribbons that are unable to bind with the ligand (i.e., vancomycin), the addition of surfactant breaks up the assemblies to restore the ligand-receptor interaction. In addition, an excess amount of the ligands can disrupt the nanofibers and result in the precipitates. As the first example of the use of ligand-receptor interaction to modulate the kinetics of enzymatic self-assembly, this work not only provides a solution to evaluate the interaction between aggregates and target molecules but also offers new insight for understanding the emergent behavior of sophisticated molecular systems having multiple and parallel processes.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Vancomicina/metabolismo , Fosfatasa Alcalina/química , Biocatálisis , Cinética , Ligandos , Tamaño de la Partícula , Bibliotecas de Moléculas Pequeñas/química , Propiedades de Superficie , Tensoactivos/química , Tensoactivos/metabolismo , Vancomicina/química
19.
J Mater Chem B ; 4(7): 1318-1323, 2016 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-27134750

RESUMEN

Based on the self-assembly capability of the core segment (GNNQQNY) of yeast prion Sup35, we design and synthesis a series of structurally related precursors for enzymatic formation of hydrogels. We found that, with the catalysis of alkaline phosphatase, the precursor becomes a hydrogelator that self-assembles in water to form nanofibers with an average width less than ten nanometers. Interestingly, the introduction of amyloid segment into a cytotoxic precursor (N'ffyp: D-1P) is able to abrogate the cytotoxicity of the precursor, making the resulting peptide to be cell compatible. This work contributes a new insight to the use of enzyme to form cell compatible hydrogels of peptides cross-ß spine.

20.
PLoS One ; 11(4): e0154126, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27100780

RESUMEN

Identifying novel cancer biomarkers is important for early cancer detection as it can reduce mortality rates. The cancer secretome, the collection of all macromolecules secreted by a tumor cell, alters its composition compared to normal tissue, and this change plays an important role in the observation of cancer progression. The collection and accurate analysis of cancer secretomes could lead to the discovery of novel biomarkers, thus improving outcomes of cancer treatment. We unexpectedly discovered that enzyme-instructed self-assembly (EISA) of a D-peptide hydrogelator results in nanonets/hydrogel around cancer cells that overexpress ectophosphatases. Here we show that these nanonets are able to rapidly collect proteins in the pericellular space (i.e., near the surface) of cancer cells. Because the secretory substances are at their highest concentration near the cell surface, the use of pericellular nanonets to collect the cancer secretome maximizes the yield and quality of samples, reduces pre-analytical variations, and allows the dynamic profiling of secretome samples. Thus, this new approach has great potential in identifying the heterotypic signaling in tumor microenvironments thereby improving the understanding of tumor microenvironments and accelerating the discovery of potential biomarkers in cancer biology. Data are available via ProteomeXchange with identifier PXD003924.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Espacio Extracelular/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato , Neoplasias/metabolismo , Medios de Cultivo Condicionados/metabolismo , Electroforesis en Gel de Poliacrilamida , Células HeLa , Humanos , Espectrometría de Masas , Nanoestructuras/química , Neoplasias/patología , Proteoma/metabolismo , Proteómica/métodos , Reproducibilidad de los Resultados , Vías Secretoras
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...