Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Mater ; : e2401137, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38742799

RESUMEN

In contrast to biological cell membranes, it is still a major challenge for synthetic membranes to efficiently separate ions and small molecules, due to their similar sizes in the sub-nanometer range. Inspired by biological ion channels with their unique channel wall chemistry that facilitates ion sieving by ion-channel interactions, we report here the first free-standing, ultrathin (10-17 nm) nanomembranes composed entirely of polydopamine (PDA) as ion and molecular sieves. These nanomembranes are obtained via an easily scalable electropolymerization strategy and provide nanochannels with various amine and phenolic hydroxyl groups that offer a favorable chemical environment for ion-channel electrostatic and hydrogen bond interactions. They exhibit remarkable selectivity for monovalent ions over multivalent ions and larger species with K+/Mg2+ of ≈4.2, K+/[Fe(CN)6]3- of ≈10.3, and K+/Rhodamine B of ≈273.0 in a pressure-driven process, as well as cyclic reversible pH-responsive gating properties. Infrared spectra reveal hydrogen bond formation between hydrated multivalent ions and PDA, which prevents the transport of multivalent ions and facilitates high selectivity. We propose chemically rich, free-standing, and pH-responsive PDA nanomembranes with specific interaction sites as customizable high-performance sieves for a wide range of challenging separation requirements. This article is protected by copyright. All rights reserved.

2.
Angew Chem Int Ed Engl ; 63(18): e202314143, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38179812

RESUMEN

Drug safety and efficacy due to premature release into the bloodstream and poor biodistribution remains a problem despite seminal advances in this area. To circumvent these limitations, we report drug cyclization based on dynamic covalent linkages to devise a dual lock for the small-molecule anticancer drug, camptothecin (CPT). Drug activity is "locked" within the cyclic structure by the redox responsive disulfide and pH-responsive boronic acid-salicylhydroxamate and turns on only in the presence of acidic pH, reactive oxygen species and glutathione through traceless release. Notably, the dual-responsive CPT is more active (100-fold) than the non-cleavable (permanently closed) analogue. We further include a bioorthogonal handle in the backbone for functionalization to generate cyclic-locked, cell-targeting peptide- and protein-CPTs, for targeted delivery of the drug and traceless release in triple negative metastatic breast cancer cells to inhibit cell growth at low nanomolar concentrations.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Camptotecina/química , Distribución Tisular , Antineoplásicos/química , Micelas , Proteínas , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Liberación de Fármacos , Línea Celular Tumoral
3.
Bioconjug Chem ; 34(9): 1645-1652, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37665137

RESUMEN

Viral infections pose a significant threat to human health, and effective antiviral strategies are urgently needed. Antiviral peptides have emerged as a promising class of therapeutic agents due to their unique properties and mechanisms of action. While effective on their own, combining antiviral peptides may allow us to enhance their potency and to prevent viral resistance. Here, we developed an orthogonal chemical strategy to prepare a heterodimeric peptide conjugate assembled on a protein-based nanoplatform. Specifically, we combined the optimized version of two peptides inhibiting HIV-1 by distinct mechanisms. Virus-inhibitory peptide (VIRIP) is a 20 amino acid fragment of α1-antitrypsin that inhibits HIV-1 by targeting the gp41 fusion peptide. Endogenous peptide inhibitor of CXCR4 (EPI-X4) is a 16-residue fragment of human serum albumin that prevents HIV-1 entry by binding to the viral CXCR4 co-receptor. Optimized forms of both peptides are assembled on supramolecular nanoplatforms through the streptavidin-biotin interaction. We show that the construct consisting of the two different peptides (SAv-VIR-102C9-EPI-X4 JM#173-C) shows increased activity against CCR5- and CXCR4-tropic HIV-1 variants. Our results are a proof of concept that peptides with different modes of action can be assembled on nanoplatforms to enhance their antiviral activity.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/prevención & control , Péptidos/farmacología , Albúmina Sérica Humana , Antivirales
4.
Int J Mol Sci ; 23(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36499357

RESUMEN

Advanced derivatives of the Endogenous Peptide Inhibitor of CXCR4 (EPI-X4) have shown therapeutic efficacy upon topical administration in animal models of asthma and dermatitis. Here, we studied the plasma stability of the EPI-X4 lead compounds WSC02 and JM#21, using mass spectrometry to monitor the chemical integrity of the peptides and a functional fluorescence-based assay to determine peptide function in a CXCR4-antibody competition assay. Although mass spectrometry revealed very rapid disappearance of both peptides in human plasma within seconds, the functional assay revealed a significantly higher half-life of 9 min for EPI-X4 WSC02 and 6 min for EPI-X4 JM#21. Further analyses demonstrated that EPI-X4 WSC02 and EPI-X4 JM#21 interact with low molecular weight plasma components and serum albumin. Albumin binding is mediated by the formation of a disulfide bridge between Cys10 in the EPI-X4 peptides and Cys34 in albumin. These covalently linked albumin-peptide complexes have a higher stability in plasma as compared with the non-bound peptides and retain the ability to bind and antagonize CXCR4. Remarkably, chemically synthesized albumin-EPI-X4 conjugates coupled by non-breakable bonds have a drastically increased plasma stability of over 2 h. Thus, covalent coupling of EPI-X4 to albumin in vitro before administration or in vivo post administration may significantly increase the pharmacokinetic properties of this new class of CXCR4 antagonists.


Asunto(s)
Receptores CXCR4 , Albúmina Sérica Humana , Animales , Humanos , Receptores CXCR4/metabolismo , Péptidos/química , Semivida , Albúmina Sérica/metabolismo
5.
Macromol Biosci ; 22(2): e2100299, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34791790

RESUMEN

With the advent of chemical strategies that allow the design of smart bioconjugates, peptide- and protein-drug conjugates are emerging as highly efficient therapeutics to overcome limitations of conventional treatment, as exemplified by antibody-drug conjugates (ADCs). While targeting peptides serve similar roles as antibodies to recognize overexpressed receptors on diseased cell surfaces, peptide-drug conjugates suffer from poor stability and bioavailability due to their low molecular weights. Through a combination of a supramolecular protein-based assembly platform and a pH-responsive linker, the authors devise herein the convenient assembly of a trivalent protein-drug conjugate. The conjugate should ideally possess distinct features of ADCs such as 1) recognition sites that recognize cell receptor and are arranged on 2) distinct locations on a high molecular weight protein scaffold, 3) a stimuli-responsive linker, as well as 4) an attached payload such as a drug molecule. These AD-like conjugates target cancer cells that overexpress somatostatin receptors, can enable controlled release in the microenvironment of cancer cells through a new pH-responsive biotin linker, and exhibit stability in biological media.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Anticuerpos Monoclonales/química , Antígenos , Antineoplásicos/química , Biotina , Concentración de Iones de Hidrógeno , Inmunoconjugados/química , Inmunoconjugados/farmacología
6.
Chem Sci ; 12(40): 13321-13330, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34777751

RESUMEN

The development of bioconjugation chemistry has enabled the combination of various synthetic functionalities to proteins, giving rise to new classes of protein conjugates with functions well beyond what Nature can provide. Despite the progress in bioconjugation chemistry, there are no reagents developed to date where the reactivity can be tuned in a user-defined fashion to address different amino acid residues in proteins. Here, we report that 2-chloromethyl acryl reagents can serve as a simple yet versatile platform for selective protein modification at cysteine or disulfide sites by tuning their inherent electronic properties through the amide or ester linkage. Specifically, the 2-chloromethyl derivatives (acrylamide or acrylate) can be obtained via a simple and easily implemented one-pot reaction based on the coupling reaction between commercially available starting materials with different end-group functionalities (amino group or hydroxyl group). 2-Chloromethyl acrylamide reagents with an amide linkage favor selective modification at the cysteine site with fast reaction kinetics and near quantitative conversations. In contrast, 2-chloromethyl acrylate reagents bearing an ester linkage can undergo two successive Michael reactions, allowing the selective modification of disulfides bonds with high labeling efficiency and good conjugate stability.

7.
J Am Chem Soc ; 143(41): 17047-17058, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34632780

RESUMEN

Dynamic covalent chemistry (DCvC) has emerged as a versatile synthetic tool for devising stable, stimuli-responsive linkers or conjugates. The interplay of binding affinity, association and dissociation constants exhibits a strong influence on the selectivity of the reaction, the conversion rate, as well as the stability in aqueous solutions. Nevertheless, dynamic covalent interactions often exhibit fast binding and fast dissociation events or vice versa, affecting their conversion rates or stabilities. To overcome the limitation in linker design, we reported herein dual responsive dynamic covalent peptide tags combining a pH responsive boronate ester with fast association and dissociation rates, and a redox-active disulfide with slow formation and dissociation rate. Precoordination by boronic acid-catechol interaction improves self-sorting and selectivity in disulfide formation into heterodimers. The resulting bis-peptide conjugate exhibited improved complex stability in aqueous solution and acidic tumor-like extracellular microenvironment. Furthermore, the conjugate responds to pH changes within the physiological range as well as to redox conditions found inside cancer cells. Such tags hold great promise, through cooperative effects, for controlling the stability of bioconjugates under dilution in aqueous media, as well as designing intelligent pharmaceutics that react to distinct biological stimuli in cells.


Asunto(s)
Ácidos Borónicos
9.
ChemMedChem ; 16(1): 94-104, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-32667697

RESUMEN

Proteins have attracted increasing attention as biopharmaceutics and diagnostics due to their high specificity, biocompatibility, and biodegradability. The biopharmaceutical sector in particular is experiencing rapid growth, which has led to an increase in the production and sale of protein drugs and diagnostics over the last two decades. Since the first-generation biopharmaceutics dominated by native proteins, both recombinant and chemical technologies have evolved and transformed the outlook of this rapidly developing field. This review article presents updates on the fabrication of covalent and supramolecular fusion hybrids, as well as protein-polymer hybrids using solid-phase approaches that hold great promise for preparing protein hybrids with precise control at the macromolecular level to incorporate additional features. In addition, the applications of the resultant protein hybrids in medicine and diagnostics are highlighted where possible.


Asunto(s)
Proteínas/química , Técnicas de Síntesis en Fase Sólida/métodos , Anticuerpos/química , Anticuerpos/metabolismo , Sustancias Macromoleculares/síntesis química , Sustancias Macromoleculares/química , Preparaciones Farmacéuticas/síntesis química , Preparaciones Farmacéuticas/química , Polietilenglicoles/química , Polímeros/química , Multimerización de Proteína , Proteínas/síntesis química
10.
Angew Chem Int Ed Engl ; 60(25): 13757-13777, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33258535

RESUMEN

Site-selective protein functionalization serves as an invaluable tool for investigating protein structures and functions in complicated cellular environments and accomplishing semi-synthetic protein conjugates such as traceable therapeutics with improved features. Dual functionalization of proteins allows the incorporation of two different types of functionalities at distinct location(s), which greatly expands the features of native proteins. The attachment and crosstalk of a fluorescence donor and an acceptor dye provides fundamental insights into the folding and structural changes of proteins upon ligand binding in their native cellular environments. Moreover, the combination of drug molecules with different modes of action, imaging agents or stabilizing polymers provides new avenues to design precision protein therapeutics in a reproducible and well-characterizable fashion. This review aims to give a timely overview of the recent advancements and a future perspective of this relatively new research area. First, the chemical toolbox for dual functionalization of proteins is discussed and compared. The strengths and limitations of each strategy are summarized in order to enable readers to select the most appropriate method for their envisaged applications. Thereafter, representative applications of these dual-modified protein bioconjugates benefiting from the synergistic/additive properties of the two synthetic moieties are highlighted.


Asunto(s)
Proteínas/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Modelos Moleculares , Estructura Molecular , Proteínas/química
11.
Chem Commun (Camb) ; 56(68): 9858-9861, 2020 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-32717008

RESUMEN

The preparation of precise macromolecules with multiple functionalities remains a challenge in drug delivery. Here, a method to prepare stoichiometrically precise tetrafunctional streptavidin conjugates is presented with an exemplary structure combining exactly one fluorescent label, one cell targeting group, one nucleus penetrating peptide and one drug molecule.


Asunto(s)
Péptidos de Penetración Celular/química , Portadores de Fármacos/química , Estreptavidina/química , Biotinilación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Péptidos de Penetración Celular/metabolismo , Péptidos de Penetración Celular/farmacología , Doxorrubicina/química , Doxorrubicina/farmacología , Colorantes Fluorescentes/química , Ácido Fólico/química , Ácido Fólico/farmacología , Humanos , Microscopía Confocal
12.
Org Biomol Chem ; 18(6): 1140-1147, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31971218

RESUMEN

An inverse electron demand Diels-Alder reaction between tetrazine and trans-cyclooctene (TCO) holds great promise for protein modification and manipulation. Herein, we report the design and synthesis of a tetrazine-based disulfide rebridging reagent, which allows the site-selective installation of a tetrazine group into disulfide-containing peptides and proteins such as the hormone somatostatin (SST) and the antigen binding fragment (Fab) of human immunoglobulin G (IgG). The fast and efficient conjugation of the tetrazine modified proteins with three different TCO-containing substrates to form a set of bioconjugates in a site-selective manner was successfully demonstrated for the first time. Homogeneous, well-defined bioconjugates were obtained underlining the great potential of our method for fast bioconjugation in emerging protein therapeutics. The formed bioconjugates were stable against glutathione and in serum, and they maintained their secondary structure. With this work, we broaden the scope of tetrazine chemistry for site-selective protein modification to prepare well-defined SST and Fab conjugates with preserved structures and good stability under biologically relevant conditions.


Asunto(s)
Ciclooctanos/metabolismo , Disulfuros/metabolismo , Compuestos Heterocíclicos/química , Inmunoglobulina G/metabolismo , Ciclooctanos/química , Disulfuros/síntesis química , Disulfuros/química , Humanos , Inmunoglobulina G/química , Modelos Moleculares , Estructura Molecular , Procesamiento Proteico-Postraduccional
13.
Sci Rep ; 10(1): 371, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941913

RESUMEN

Acute myeloid leukemia (AML) is characterized by relapse and treatment resistance in a major fraction of patients, underlining the need of innovative AML targeting therapies. Here we analysed the therapeutic potential of an innovative biohybrid consisting of the tumor-associated peptide somatostatin and the photosensitizer ruthenium in AML cell lines and primary AML patient samples. Selective toxicity was analyzed by using CD34 enriched cord blood cells as control. Treatment of OCI AML3, HL60 and THP1 resulted in a 92, and 99 and 97% decrease in clonogenic growth compared to the controls. Primary AML cells demonstrated a major response with a 74 to 99% reduction in clonogenicity in 5 of 6 patient samples. In contrast, treatment of CD34+ CB cells resulted in substantially less reduction in colony numbers. Subcellular localization assays of RU-SST in OCI-AML3 cells confirmed strong co-localization of RU-SST in the lysosomes compared to the other cellular organelles. Our data demonstrate that conjugation of a Ruthenium complex with somatostatin is efficiently eradicating LSC candidates of patients with AML. This indicates that receptor mediated lysosomal accumulation of photodynamic metal complexes is a highly attractive approach for targeting AML cells.


Asunto(s)
Leucemia Mieloide Aguda/terapia , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Receptores de Somatostatina/metabolismo , Rutenio/uso terapéutico , Somatostatina/uso terapéutico , Adulto , Anciano , Apoptosis , Línea Celular Tumoral , Estabilidad de Medicamentos , Femenino , Sangre Fetal/metabolismo , Humanos , Lisosomas/metabolismo , Masculino , Persona de Mediana Edad , Fármacos Fotosensibilizantes/química , Especies Reactivas de Oxígeno/metabolismo
14.
Adv Healthc Mater ; 8(17): e1900665, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31318180

RESUMEN

The targeted pharmacological modulation of polymorphonuclear leukocytes (PMNs) is of major medical interest. These innate immune cells play a central role in the defense against pathogenic microorganisms. However, their excessive chemotactic recruitment into tissues after traumatic injury is detrimental due to local and systemic inflammation. Rho-GTPases, being the master regulators of the actin cytoskeleton, regulate migration and chemotaxis of PMNs, are attractive pharmacological targets. Herein, supramolecular protein complexes are assembled in a "mix-and-match" approach containing the specific Rho-inhibiting clostridial C3 enzyme and three PMN-binding peptides using an avidin platform. Selective delivery of the C3 Rho-inhibitor with these complexes into the cytosol of human neutrophil-like NB-4 cells and primary human PMNs ex vivo is demonstrated, where they catalyze the adenosine diphosphate (ADP) ribosylation of Rho and induce a characteristic change in cell morphology. Notably, the complexes do not deliver C3 enzyme into human lung epithelial cells, A549 lung cancer cells, and immortalized human alveolar epithelial cells (hAELVi), demonstrating their cell type-selectivity. The supramolecular complexes represent attractive molecular tools to decipher the role of PMNs in infection and inflammation or for the development of novel therapeutic approaches for diseases that are associated with hyperactivity and reactivity of PMNs such as post-traumatic injury.


Asunto(s)
Neutrófilos/metabolismo , Toxinas Biológicas/farmacología , ADP Ribosa Transferasas/metabolismo , Avidina/metabolismo , Biotinilación , Toxinas Botulínicas/metabolismo , Línea Celular , Citosol/metabolismo , Endocitosis/efectos de los fármacos , Humanos , Neutrófilos/efectos de los fármacos , Péptidos/síntesis química , Péptidos/química
15.
ACS Nano ; 13(8): 8749-8759, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31322856

RESUMEN

The surface of proteins is heterogeneous with sophisticated but precise hydrophobic and hydrophilic patches, which is essential for their diverse biological functions. To emulate such distinct surface patterns on macromolecules, we used rigid spherical synthetic dendrimers (polyphenylene dendrimers) to provide controlled amphiphilic surface patches with molecular precision. We identified an optimal spatial arrangement of these patches on certain dendrimers that enabled their interaction with human adenovirus 5 (Ad5). Patchy dendrimers bound to the surface of Ad5 formed a synthetic polymer corona that greatly altered various host interactions of Ad5 as well as in vivo distribution. The dendrimer corona (1) improved the ability of Ad5-derived gene transfer vectors to transduce cells deficient for the primary Ad5 cell membrane receptor and (2) modulated the binding of Ad5 to blood coagulation factor X, one of the most critical virus-host interactions in the bloodstream. It significantly enhanced the transduction efficiency of Ad5 while also protecting it from neutralization by natural antibodies and the complement system in human whole blood. Ad5 with a synthetic dendrimer corona revealed profoundly altered in vivo distribution, improved transduction of heart, and dampened vector sequestration by liver and spleen. We propose the design of bioactive polymers that bind protein surfaces solely based on their amphiphilic surface patches and protect against a naturally occurring protein corona, which is highly attractive to improve Ad5-based in vivo gene therapy applications.


Asunto(s)
Adenovirus Humanos/genética , Dendrímeros/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Transducción Genética , Adenovirus Humanos/efectos de los fármacos , Animales , Proteínas de la Cápside/química , Dendrímeros/química , Vectores Genéticos/química , Vectores Genéticos/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Hígado/química , Hígado/efectos de los fármacos , Polímeros/química , Polímeros/farmacología , Receptores Virales/antagonistas & inhibidores , Receptores Virales/química
16.
Chem Soc Rev ; 47(24): 9069-9105, 2018 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-30452046

RESUMEN

Nature has evolved an optimal synthetic factory in the form of translational and posttranslational processes by which millions of proteins with defined primary sequences and 3D structures can be built. Nature's toolkit gives rise to protein building blocks, which dictates their spatial arrangement to form functional protein nanostructures that serve a myriad of functions in cells, ranging from biocatalysis, formation of structural networks, and regulation of biochemical processes, to sensing. With the advent of chemical tools for site-selective protein modifications and recombinant engineering, there is a rapid development to develop and apply synthetic methods for creating structurally defined, functional protein nanostructures for a broad range of applications in the fields of catalysis, materials and biomedical sciences. In this review, design principles and structural features for achieving and characterizing functional protein nanostructures by synthetic approaches are summarized. The synthetic customization of protein building blocks, the design and introduction of recognition units and linkers and subsequent assembly into structurally defined protein architectures are discussed herein. Key examples of these supramolecular protein nanostructures, their unique functions and resultant impact for biomedical applications are highlighted.


Asunto(s)
Nanoestructuras/química , Nanotecnología/métodos , Proteínas/química , Animales , Catálisis , Técnicas de Química Sintética/métodos , Humanos , Modelos Moleculares , Nanoestructuras/ultraestructura , Conformación Proteica , Ingeniería de Proteínas/métodos , Proteínas/síntesis química , Proteínas/genética , Proteínas/ultraestructura
17.
Macromol Biosci ; 18(10): e1800196, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30066983

RESUMEN

Protein-based polymeric polyelectrolytes are emerging as alternative synthetic nanoparticles owing to their biodegradability and biocompatibility. However, potential in vivo toxicity remains a significant challenge. Herein an array of protein polyelectrolytes generated from cationic human serum albumin (cHSA) and polyethylene glycol (PEG) are synthesized via synthetic customization as antimicrobials for the treatment of systemic infections. By varying PEG molecular weight and chain length, in vitro hemolytic activity can be fine-tuned without significantly affecting antimicrobial potency. The optimal hybrid material, PEG (2000)18 -cHSA, with potent antimicrobial character, low hemolytic activity, and in vitro biofilm disruptive properties is identified. Surface plasmon resonance (SPR) evaluation demonstrates significantly higher binding activity of the protein nanoparticles to bacteria cell wall components and microfluidic live-cell imaging indicates that the nanoparticles act through a membranolytic mechanism. Given their low susceptibility to drug resistance and potent activity against resistant bacteria strains, these findings establish the PEGylated albumin nanoparticles as a potent weaponry against drug resistance and biofilm-related infection.


Asunto(s)
Antibacterianos , Bacterias/crecimiento & desarrollo , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Biopelículas/efectos de los fármacos , Candida albicans/fisiología , Eritrocitos/metabolismo , Hemólisis/efectos de los fármacos , Nanopartículas/química , Albúmina Sérica Humana , Antibacterianos/síntesis química , Antibacterianos/química , Antibacterianos/farmacología , Biopelículas/crecimiento & desarrollo , Eritrocitos/citología , Humanos , Polietilenglicoles/química , Albúmina Sérica Humana/química , Albúmina Sérica Humana/farmacología
18.
Adv Sci (Weinh) ; 5(8): 1701036, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30128225

RESUMEN

A facile chemical approach integrating supramolecular chemistry, site-selective protein chemistry, and molecular biology is described to engineer synthetic multidomain protein therapeutics that sensitize cancer cells selectively to significantly enhance antitumor efficacy of existing chemotherapeutics. The desired bioactive entities are assembled via supramolecular interactions at the nanoscale into structurally ordered multiprotein complexes comprising a) multiple copies of the chemically modified cyclic peptide hormone somatostatin for selective targeting and internalization into human A549 lung cancer cells expressing SST-2 receptors and b) a new cysteine mutant of the C3bot1 (C3) enzyme from Clostridium botulinum, a Rho protein inhibitor that affects and influences intracellular Rho-mediated processes like endothelial cell migration and blood vessel formation. The multidomain protein complex, SST3-Avi-C3, retargets C3 enzyme into non-small cell lung A549 cancer cells and exhibits exceptional tumor inhibition at a concentration ≈100-fold lower than the clinically approved antibody bevacizumab (Avastin) in vivo. Notably, SST3-Avi-C3 increases tumor sensitivity to a conventional chemotherapeutic (doxorubicin) in vivo. These findings show that the integrated approach holds vast promise to expand the current repertoire of multidomain protein complexes and can pave the way to important new developments in the area of targeted and combination cancer therapy.

19.
Chem Asian J ; 13(22): 3474-3479, 2018 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-30036452

RESUMEN

Dynamic covalent chemistry is a versatile and powerful tool that integrates both stable chemical bonds and stimulus responsiveness into the construction of smart biotherapeutics. With minimalistic molecular design, a dynamic covalent protein assembly that incorporates selective targeting and intracellular release upon pH stimulus is presented. The construct comprises an active enzymatic protein core (cytochrome c) self-assembled with cancer cell targeting motifs (somatostatin) through boronic acid/salicylhydroxamate chemistry. The bioorthogonal assembly takes place rapidly under neutral aqueous conditions while the release of the protein is initiated under acidic conditions found within cellular vesicles during uptake. By demonstrating that these modular components act in synergy, we show the broad applicability of such chemical strategies to advance the frontier of modern nanomedicine.


Asunto(s)
Ácidos Borónicos/química , Citocromos c/metabolismo , Salicilamidas/química , Somatostatina/metabolismo , Células A549 , Calcio/metabolismo , Citocromos c/química , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/química , Humanos , Concentración de Iones de Hidrógeno , Microscopía Confocal , Nanomedicina , Somatostatina/química
20.
Bioconjug Chem ; 29(8): 2665-2670, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29949347

RESUMEN

The development of small protein tags that exhibit bioorthogonality, bond stability, and reversibility, as well as biocompatibility, holds great promise for applications in cellular environments enabling controlled drug delivery or for the construction of dynamic protein complexes in biological environments. Herein, we report the first application of dynamic covalent chemistry both for purification and for reversible assembly of protein conjugates using interactions of boronic acid with diols and salicylhydroxamates. Incorporation of the boronic acid (BA) tag was performed in a site-selective fashion by applying disulfide rebridging strategy. As an example, a model protein enzyme (lysozyme) was modified with the BA tag and purified using carbohydrate-based column chromatography. Subsequent dynamic covalent "click-like" bioconjugation with a salicylhydroxamate modified fluorescent dye (BODIPY FL) was accomplished while retaining its original enzymatic activity.


Asunto(s)
Ácidos Borónicos/química , Química Clic , Proteínas/química , Compuestos de Boro/química , Cromatografía Liquida/métodos , Disulfuros/química , Colorantes Fluorescentes/química , Muramidasa/química , Muramidasa/metabolismo , Proteínas/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Espectrometría de Masas en Tándem/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...